Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
2.
Acta Biomater ; 10(7): 2877-93, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24675108

ABSTRACT

Tissue engineered heart valves offer a promising alternative for the replacement of diseased heart valves avoiding the limitations faced with currently available bioprosthetic and mechanical heart valves. In the paradigm of tissue engineering, a three-dimensional platform - the so-called scaffold - is essential for cell proliferation, growth and differentiation, as well as the ultimate generation of a functional tissue. A foundation for success in heart valve tissue engineering is a recapitulation of the complex design and diverse mechanical properties of a native valve. This article reviews technological details of the scaffolds that have been applied to date in heart valve tissue engineering research.


Subject(s)
Heart Valves , Tissue Engineering , Humans
3.
Int J Cardiol ; 153(2): 202-6, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-20843569

ABSTRACT

BACKGROUND: Enhanced external counterpulsation (EECP) is associated with improvement in endothelial function, angina and quality of life in patients with symptomatic coronary artery disease, although the mechanisms underlying the observed clinical benefits are not completely clear. The purpose of this study was to examine the effects of EECP on circulating haematopoietic progenitor cells (HPCs) and endothelial progenitor cells (EPCs) in patients with refractory angina. We compared HPC and EPC counts between patients scheduled for EECP and patients with normal angiographic coronary arteries, with and without coronary endothelial dysfunction. We hypothesized that an increase in circulating bone marrow derived progenitor cells in response to EECP may be part of the mechanism of action of EECP. METHODS: Thirteen consecutive patients scheduled to receive EECP treatment were prospectively enrolled. Clinical characteristics were recorded and venous blood (5 ml) was drawn on day 1, day 17, day 35 (final session) and one month post completion of EECP therapy. Buffy coat was extracted and HPCs and EPCs were counted by flow cytometry. RESULTS: Median Canadian Cardiovascular Society (CCS) angina class decreased and Duke Activity Status Index (DASI) functional score increased significantly (both, p < 0.05) in response to EECP, an effect that was maintained at one month after termination of treatment. Flow cytometric analysis revealed an accompanying significant increase in CD34+, CD133+ and CD34+, CD133+ CPC counts over the course of treatment (p < 0.05). DASI scores correlated significantly with CD34+ (R = 0.38 p = 0.02), CD133+ (R = 0.5, p = 0.006) and CD34+, CD133+ (R = 0.47, p = 0.01) CPC counts. CONCLUSION: This study shows that HPCs, but not EPCs are significantly increased in response to EECP treatment and correlate with reproducible measures of clinical improvement. These findings are the first to link the functional improvement observed with EECP treatment with increased circulating progenitor cells.


Subject(s)
Antigens, CD34/biosynthesis , Counterpulsation/methods , Stem Cells/metabolism , Aged , Aged, 80 and over , Antigens, CD34/blood , Cohort Studies , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Flow Cytometry/methods , Hematopoietic Stem Cells/metabolism , Humans , Male , Middle Aged , Prospective Studies , Treatment Outcome
4.
Heart ; 95(16): 1315-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19447837

ABSTRACT

BACKGROUND: B-type natriuretic peptide (BNP, nesiritide) has anti-fibrotic, anti-hypertrophic, anti-inflammatory, vasodilating, lusitropic and aldosterone-inhibiting properties but conventional doses of BNP cause hypotension, limiting its use in heart failure. OBJECTIVE: To determine whether infusion of low-dose BNP within 24 h of successful reperfusion for anterior acute myocardial infarction (AMI) would prevent adverse left ventricular (LV) remodelling and suppress aldosterone. METHODS: A translational proof-of-concept study was carried out to determine tolerability and biological activity of intravenous BNP at 0.003 and 0.006 microg/kg/min, without bolus started within 24 h of successful reperfusion for anterior AMI. 24 patients with first anterior wall ST elevation AMI and successful revascularisation were randomly assigned to receive 0.003 (n = 12) or 0.006 (n = 12) microg/kg/min of IV BNP for 72 h in addition to standard care during hospitalisation for anterior AMI. RESULTS: Baseline characteristics, drugs and peak cardiac biomarkers for myocardial damage were similar between both groups. Infusion of BNP at 0.006 microg/kg/min resulted in greater biological activity than infusion at 0.003 microg/kg/min as measured by higher mean (SEM) plasma cGMP levels (8.6 (1) vs 5.5 (1) pmol/ml, p<0.05) and suppression of plasma aldosterone (8.0 (2) to 4.6 (1) ng/dl, p<0.05), which was not seen in the 0.003 microg/kg/min group. LV ejection fraction (LVEF) improved significantly from baseline to 1 month (40 (4)% to 54 (5)%, p<0.05) in the 0.006 group but not in the 0.003 group. Infusion of BNP at 0.006 microg/kg/min was associated with a decrease of LV end-systolic volume index (61 (9) to 43 (8) ml/m(2), p<0.05) at 1 month, which was not seen in the 0.003 group. No drug-related serious adverse events occurred in either group. CONCLUSIONS: 72 h infusion of low BNP at the time of anterior AMI is well tolerated and biologically active. Patients treated with low-dose BNP had improved LVEF and smaller LV end-systolic volume at 1 month.


Subject(s)
Mineralocorticoid Receptor Antagonists/administration & dosage , Myocardial Infarction/drug therapy , Natriuretic Agents/administration & dosage , Natriuretic Peptide, Brain/administration & dosage , Vasodilator Agents/administration & dosage , Female , Humans , Male , Middle Aged , Myocardial Infarction/physiopathology , Recombinant Proteins/administration & dosage , Stroke Volume/drug effects , Ventricular Remodeling/drug effects
5.
J Thromb Haemost ; 7(7): 1106-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19422457

ABSTRACT

BACKGROUND: Mouse tissue factor pathway inhibitor (TFPI) is produced in three alternatively spliced isoforms that differ in domain structure and mechanism for cell surface binding. Tissue expression of TFPI isoforms in mice was characterized as an initial step for identification of their physiological functions. METHODS AND RESULTS: Sequence homology demonstrates that TFPIalpha existed over 430 Ma while TFPIbeta and TFPIgamma evolved more recently. In situ hybridization studies of heart and lung did not reveal any cells exclusively expressing a single isoform. Although our previous studies have demonstrated that TFPIalpha mRNA is more prevalent than TFPIbeta or TFPIgamma mRNA in mouse tissues, western blot studies demonstrated that TFPIbeta is the primary protein isoform produced in adult tissues, while TFPIalpha is expressed during embryonic development and in placenta. Consistent with TFPIbeta as the primary isoform produced within adult vascular beds, the TFPI isoform in mouse plasma migrates like TFPIbeta in SDS-PAGE and mice have a much smaller heparin-releasable pool of plasma TFPIalpha than humans. CONCLUSIONS: The data demonstrate that alternatively spliced isoforms of TFPI are temporally expressed in mouse tissues at the level of protein production. TFPIalpha and TFPIbeta are produced in embryonic tissues and in placenta while adult tissues produce almost exclusively TFPIbeta.


Subject(s)
Alternative Splicing , Lipoproteins/genetics , Amino Acid Sequence , Animals , Blotting, Western , Conserved Sequence , Enzyme-Linked Immunosorbent Assay , Female , In Situ Hybridization , Lipoproteins/chemistry , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Myocardium/metabolism , Placenta/metabolism , RNA, Messenger/genetics , Sequence Homology, Amino Acid
7.
Gene Ther ; 10(5): 443-52, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12601399

ABSTRACT

We describe a mechanical method for delivery of adenoviral vector to the adventitial surface of arteries and to other tissues. Our goal was to characterize, principally in intact carotid artery, the morphological, biochemical, and functional effects of mechanical delivery of a recombinant beta-galactosidase-expressing adenoviral vector following its direct application using a small paintbrush. Our ex vivo and in vivo data demonstrate efficient, accurate, and rapid transduction of arteries without compromise of their morphological, biochemical, and functional integrity. We also demonstrate the general applicability of this technique in vivo via transduction of skeletal muscle, fibrotendinous tissue, peritoneum, serosal surface of bowel, and wounded skin. We conclude that direct mechanical delivery of an adenoviral vector to tissues using a suitable paintbrush represents an intuitive, accurate, and effective means of augmenting gene transfer efficiency, and may be a useful adjunct to other delivery methods.


Subject(s)
Adenoviridae/genetics , Carotid Arteries , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Administration, Topical , Animals , Carotid Arteries/enzymology , Dogs , Models, Animal , beta-Galactosidase/analysis , beta-Galactosidase/genetics
8.
Blood ; 98(10): 2980-7, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698280

ABSTRACT

Lipoprotein (a) [Lp(a)] has been associated with both anti-fibrinolytic and atherogenic effects. However, no direct link currently exists between this atherogenic lipoprotein and intravascular coagulation. The current study examined the binding and functional effects of Lp(a), its lipoprotein constituents, apoliprotein (a) [apo(a)] and low-density lipoprotein (LDL), and lysine-plasminogen (L-PLG), which shares significant homology with apo(a), on tissue factor pathway inhibitor (TFPI), a major regulator of tissue factor-mediated coagulation. Results indicate that Lp(a), apo(a), and PLG but not LDL bound recombinant TFPI (rTFPI) in vitro and that apo(a) bound to a region spanning the last 37 amino acid residues of the c-terminus of TFPI. The apparent binding affinity for TFPI was much higher for Lp(a) (KD approximately 150 nM) compared to PLG (KD approximately 800 nM) and nanomolar concentrations of apo(a) (500 nM) inhibited PLG binding to TFPI. Lp(a) also inhibited in a concentration-dependent manner rTFPI activity and endothelial cell surface TFPI activity in vitro, whereas PLG had no such effect. Moreover physiologic concentrations of PLG (2 microM) had no effect on the concentration-dependent inhibition of TFPI activity induced by Lp(a). In human atherosclerotic plaque, apo(a) and TFPI immunostaining were shown to coexist in smooth muscle cell-rich areas of the intima. These data suggest a novel mechanism whereby Lp(a) through its apo(a) moiety may promote thrombosis by binding and inactivating TFPI.


Subject(s)
Lipoprotein(a)/metabolism , Lipoproteins/antagonists & inhibitors , Models, Biological , Thrombosis/metabolism , Animals , Apolipoproteins A/metabolism , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Binding Sites , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Endothelium, Vascular/cytology , Fibrinolysis , Humans , Lipoprotein(a)/chemistry , Lipoprotein(a)/pharmacology , Lipoproteins/genetics , Lipoproteins/metabolism , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/ultrastructure , Peptide Fragments/metabolism , Plasminogen/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Thrombosis/etiology
9.
Thromb Haemost ; 86(3): 923-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11583328

ABSTRACT

Plasmin is an important protease that mediates clot fibrinolysis and vessel wall extracellular matrix proteolysis. Recently, in vitro studies have suggested that plasmin can cleave and inactivate recombinant TFPI, a major inhibitor of TF-mediated coagulation. We hypothesized that such an interaction may occur in vascular cells expressing TFPI, or in the vessel wall, with implications for thrombolysis. In a series of experiments, we examined the effects of plasmin on cell surface and extracellular matrix (ECM) associated TFPI in endothelial cells (EC) in culture and on EC and smooth muscle cells (SMC) in the vessel wall. Plasmin (0.2 microM) decreased cell surface and matrix associated TFPI activity in cultured endothelial cells by 77 +/- 5% and 69 +/- 6% respectively (p < 0.01). Plasminogen, the proenzyme form of plasmin had no such effect on cell surface TFPI or matrix TFPI. Cell surface TFPI antigen measured by fluorescence activated cell sorter (FACS) was also significantly reduced by plasmin. Proteolysis of conditioned medium TFPI was suggested by loss of a approximately 45kD TFPI on Western Blot analysis following plasmin treatment. Plasmin also proteolysed a approximately 45kD TFPI protein in the intact ECM of EC, an effect which was inhibited by preincubation with aprotinin, a plasmin inhibitor. Incubation of similar concentrations of plasmin, with homogenates of normal vessel decreased a approximately 45kD TFPI immunoreactive band on Western blot analysis. Plasmin also decreased surface TFPI activity on frozen sections of normal vessel as measured by an amidolytic assay. Finally, plasmin treatment of atherosclerotic plaque sections caused complete removal of TFPI immunoreactivity associated with luminal EC and intimal SMC, when compared to control treated plaque (n = 3). Together these data suggest that plasmin proteolyses the majority of EC-associated (surface and matrix) TFPI and may remove TFPI from the luminal surface and intima of the vessel wall. TFPI proteolysis in cultured EC was associated with significant reduction in TFPI anticoagulant activity. These data provide evidence that plasmin degradation of TFPI occurs in vascular cells and in the vessel wall and may have implications for re-thrombosis following thrombolysis in vivo.


Subject(s)
Coronary Artery Disease/metabolism , Endothelium, Vascular/drug effects , Extracellular Matrix Proteins/metabolism , Fibrinolysin/pharmacology , Lipoproteins/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Animals , Blotting, Western , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media, Conditioned , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , In Vitro Techniques , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Plasminogen/pharmacology , Recurrence , Thrombolytic Therapy , Thrombosis/drug therapy , Tunica Intima/cytology , Tunica Intima/drug effects , Tunica Intima/metabolism
10.
Biochem Biophys Res Commun ; 286(5): 968-72, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11527395

ABSTRACT

Nuclear factor kappaB (NF-kappaB) is a transcriptional factor which may be pivotal in the pathogenesis of atherosclerosis. Endothelin-1 (ET-1) is a peptide with proatherogenic properties. We hypothesized that ET-1 may act through activation of NF-kappaB and degradation of IkappaB-alpha, the cytosolic inhibitor of NF-kappaB activation, to create an atherogenic environment. The human monocytic cell line THP-1 was stimulated with ET-1 +/- the ET antagonist, BQ788 and the proteosome inhibitor, PSI. LPS was used as a positive control. Gel shift assays for NF-kappaB activity and Western blot analysis for IkappaB-alpha were performed. Both LPS and ET-1 led to activation of NF-kappaB in nuclear extracts [3.4 +/- 0.45 (LPS) and 2.9 +/- 0.26 (ET-1) fold increase in Arbitrary Densitometric Units (ADU) compared with negative control (P < 0.005 in both cases)]. In the presence of the ETB antagonist, BQ788, NF-kappaB activation was attenuated and not different from control (1.7 +/- 0.24 fold DU compared with negative control; P = NS). In addition, both LPS and ET-1 mediated NF-kappaB activation were attenuated by preincubation with the proteosome inhibitor, PSI (1.3 +/- 0.58 and 1.1 +/- 0.3 fold increase in ADU compared with negative control respectively). Both LPS and ET-1 led to a decrease in the amount of IkappaB-alpha present in the THP-1 cytoplasmic extracts (2.1 +/- 1.5% and 54 +/- 15.7% of ADU vs negative control (P < 0.05). NF-kappaB is activated by ET-1 in human THP-1 monocytes. This data supports a role for the ETs in the development of inflammation in the vessel wall in atherosclerosis.


Subject(s)
Endothelin-1/pharmacology , I-kappa B Proteins , Macrophages/metabolism , NF-kappa B/metabolism , Antihypertensive Agents/pharmacology , Arteriosclerosis/metabolism , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cysteine Endopeptidases , Cytoplasm/metabolism , Cytosol/metabolism , DNA-Binding Proteins/metabolism , Humans , Inflammation/metabolism , Monocytes/metabolism , Multienzyme Complexes/antagonists & inhibitors , NF-KappaB Inhibitor alpha , Oligopeptides/pharmacology , Peptides/chemistry , Piperidines/pharmacology , Proteasome Endopeptidase Complex , Protein Binding
11.
Circ Res ; 89(1): 71-6, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11440980

ABSTRACT

Tissue factor (TF) is a low-molecular-weight glycoprotein that initiates the extrinsic clotting cascade and is considered a major regulator of arterial thrombogenicity. TF pathway inhibitor (TFPI) is a major physiological inhibitor of TF-initiated coagulation. The aim of this study was to define the complex interplay between TF and TFPI and the regulation of vascular thrombogenicity in a model of vascular remodeling. To determine the levels and pattern of vascular expression of TF and TFPI associated with vascular remodeling, a murine model of flow cessation was studied. TF activity of the arteries increased after ligation (P<0.05). Quantitative analysis of homogenates of remodeled carotid arteries revealed increased TF expression but unchanged TFPI expression compared with normal carotid arteries, resulting in enhanced TF activity. To determine the potential therapeutic role of TFPI in this thrombogenic state, mice were treated with intravascular adenoviral delivery of either murine TFPI (Ad-mTFPImyc) or a control adenovirus (Ad-DeltaE1). Overexpression of TFPI decreased vascular TF activity compared with viral control (P<0.01). Overexpression of TFPI inhibited neointimal formation (P=0.038), resulting in enhanced luminal area (P=0.001) 4 weeks after flow cessation. In this murine model of vascular remodeling, an imbalance between TF and TFPI expression is generated, resulting in increased TF activity. Overexpression of TFPI in this model inhibits vascular TF activity and results in attenuation of vascular remodeling associated with flow interruption.


Subject(s)
Arteriosclerosis/etiology , Carotid Artery Thrombosis/etiology , Lipoproteins/physiology , Thromboplastin/physiology , Animals , Arteriosclerosis/metabolism , Arteriosclerosis/therapy , Carotid Artery Thrombosis/metabolism , Carotid Artery Thrombosis/therapy , Genetic Therapy , Lipoproteins/genetics , Mice , Mice, Inbred C57BL
12.
Arterioscler Thromb Vasc Biol ; 21(1): 122-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145943

ABSTRACT

Recent evidence suggests that some benefit from the 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors may occur independent of lipid lowering. We aimed to determine the effect of simvastatin on coronary endothelial function, endothelial NO synthase (eNOS) expression, and oxidative stress in experimental hypercholesterolemia (HC) in the absence of cholesterol lowering. Pigs were randomized to 3 experimental groups: normal diet (N group), high cholesterol diet (HC group), and HC diet with simvastatin (HC+S group) for 12 weeks. Low density lipoprotein cholesterol was similarly increased in the HC and HC+S groups compared with the N group. In vitro analysis of coronary large- and small-vessel endothelium-dependent vasorelaxation was performed. The mean vasorelaxation of epicardial vessels to bradykinin was significantly attenuated in the HC group compared with the N group (32.3+/-1.2% versus 42.9+/-1.6%, respectively; P<0.0001). This attenuation was significantly reversed in the HC+S group (38.7+/-1.5%, P<0.005 versus HC group). The maximal vasorelaxation to substance P was significantly attenuated in the HC group compared with the N group (50.5+/-11.9% versus 79.3+/-5.3%, respectively; P<0.05). This attenuated response was normalized in the HC+S group (74.9+/-4.1%, P<0.05 versus HC group). The maximal arteriolar vasorelaxation to bradykinin was also significantly attenuated in the HC group compared with the N group (71.9+/-4.9% versus 96.8+/-1.34%, respectively; P<0.005). This was reversed in the HC+S group (98.4+/-0.6%, P<0.0001 versus HC group). Western blotting of coronary tissue homogenates for eNOS demonstrated a decrease in protein levels in the HC group compared with the N group, with normalization in the HC+S group. Elevation of plasma F(2)-isoprostanes and thiobarbituric acid-reactive substances, markers of oxidative stress, occurred in the HC compared with the N group. These changes were reversed in the HC+S group. In summary, simvastatin preserves endothelial function in coronary epicardial vessels and arterioles in experimental HC (in the absence of cholesterol lowering) in association with an increase in coronary eNOS levels and a decrease in oxidative stress. These alterations may play a role in the reduction in cardiac events after treatment with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors.


Subject(s)
Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Hypercholesterolemia/prevention & control , Hypercholesterolemia/physiopathology , Lipid Metabolism , Simvastatin/pharmacology , Animals , Arterioles/metabolism , Arterioles/physiopathology , Blotting, Western , Bradykinin/metabolism , Catalase/analysis , Coronary Vessels/enzymology , Dinoprost/analogs & derivatives , Dinoprost/blood , Endothelium, Vascular/enzymology , F2-Isoprostanes , Female , Glutathione Peroxidase/analysis , Hydroxymethylglutaryl CoA Reductases/blood , Hypercholesterolemia/metabolism , Immunohistochemistry , In Vitro Techniques , Lipid Peroxidation/drug effects , Lipids/blood , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type III , Swine , Thiobarbituric Acid Reactive Substances/analysis , Vasodilation/drug effects
13.
Mayo Clin Proc ; 75(8): 831-4, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10943238

ABSTRACT

Cardiovascular disease is the leading cause of death in the Western world, and gene therapy approaches to several cardiovascular disorders have been proposed. One of the major stumbling blocks to be overcome before widespread clinical use of this technology is how to deliver DNA efficiently and safely to cells in vivo. While delivery of DNA alone is inefficient, use of viral vectors may overcome this problem. Adenoviral vectors are most commonly used in cardiovascular gene delivery, but toxicity related to these vectors remains a concern. In addition, duration of gene expression with use of these vectors is limited, which may be advantageous in settings in which transient expression is satisfactory to obtain a therapeutic effect. Gene therapy has been suggested as an approach to multiple conditions, including restenosis after angioplasty, therapeutic neovascularization, and bypass graft restenosis. Phase 1 clinical trials were recently reported. While proof of principle has been established in preclinical animal models, convincing efficacy data in humans do not yet exist. Improvements in vector technology and methods of catheter-mediated vascular gene delivery are needed before widespread clinical application of this therapy.


Subject(s)
Arteriosclerosis/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Adenoviridae , Animals , Arteriosclerosis/genetics , Clinical Trials, Phase I as Topic , Coronary Artery Disease/therapy , DNA/therapeutic use , Endothelial Growth Factors/genetics , Gene Expression Regulation , Humans , Liposomes , Lymphokines/genetics , Plasmids , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Biol Chem ; 275(26): 20210-6, 2000 Jun 30.
Article in English | MEDLINE | ID: mdl-10770953

ABSTRACT

Liver injury during cholestasis reflects a balance between the effects of toxic and nontoxic bile acids. However, the critical distinction between a toxic and nontoxic bile acid remains subtle and unclear. For example, the glycine conjugate of chenodeoxycholate (GCDC) induces hepatocyte apoptosis, whereas the taurine conjugate (TCDC) does not. We hypothesized that the dissimilar cellular responses may reflect differential activation of a phosphatidylinositol 3-kinase (PI3K)-dependent signaling pathway. In the bile acid-transporting McNtcp.24 rat hepatoma cell line, TCDC, but not GCDC, stimulated PI3K activity. Consistent with this observation, inhibition of PI3K rendered TCDC cytotoxic, and constitutive activation of PI3K rendered GCDC nontoxic. Both Akt and the atypical protein kinase C isoform zeta (PKCzeta) have been implicated in PI3K-dependent survival signaling. However, TCDC activated PKCzeta, but not Akt. Moreover, inhibition of PKCzeta converted TCDC into a cytotoxic agent, whereas overexpression of wild-type PKCzeta blocked GCDC-induced apoptosis. We also demonstrate that TCDC activated nuclear factor kappaB (NF-kappaB) in a PI3K- and PKCzeta-dependent manner. Moreover, inhibition of NF-kappaB by an IkappaB super-repressor rendered TCDC cytotoxic, suggesting that NF-kappaB is also necessary to prevent the cytotoxic effects of TCDC. Collectively, these data suggest that some hydrophobic bile acids such as TCDC activate PI3K-dependent survival pathways, which prevent their otherwise inherent toxicity.


Subject(s)
Bile Acids and Salts/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Taurochenodeoxycholic Acid/physiology , Adenoviridae/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Glycochenodeoxycholic Acid/metabolism , Immunoblotting , Luciferases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Oncogene Protein v-akt , Plasmids , Protein Kinase C/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Retroviridae Proteins, Oncogenic/metabolism , Retroviridae Proteins, Oncogenic/physiology , Taurochenodeoxycholic Acid/metabolism , Transfection , Tumor Cells, Cultured
15.
J Am Coll Cardiol ; 35(3): 796-801, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10716485

ABSTRACT

OBJECTIVES: We sought to define the vascular actions of the cardiac hormone brain natriuretic peptide (BNP) on cellular proliferation and cyclic guanosine monophosphate (cGMP) in human aortic vascular smooth muscle cells (HAVSMCs). Secondly, we investigated BNP and acetylcholine (ACh) vasorelaxations in aortic rings from normal and atherosclerotic rabbits in the presence and absence of long-term oral inhibition of neutral endopeptidase (NEP). BACKGROUND: The vascular actions of BNP are not well defined, despite the presence of its receptor in vascular smooth muscle and the upregulation of NEP, the ectoenzyme that degrades BNP, in the vascular wall in atherosclerosis. METHODS: HAVSMCs stimulated with fetal calf serum (FCS) were pulsed with bromodeoxyuridine (BrdU) with and without BNP. The HAVSMCs were incubated in the presence and absence of BNP to assess cGMP. Vasorelaxations to BNP and ACh were assessed in rings in normal and atherosclerotic rabbits in the presence and absence of long-term oral inhibition of NEP, together with assessment of atheroma formation. RESULTS: FCS-stimulated BrdU uptake in HAVSMCs was suppressed with BNP. BNP potentiated cGMP in HAVSMCs. BNP resulted in potent vasorelaxation in normal isolated aortic rings, which were impaired in atherosclerotic versus normal rabbits and preserved with NEP inhibition, which also decreased atheroma formation. Relaxations to ACh, which were also impaired in atherosclerosis, were preserved with inhibition of NEP. CONCLUSIONS: We conclude that BNP potently inhibits vascular smooth muscle cell proliferation and potentiates the generation of cGMP. BNP potently relaxes the normal rabbit aorta, and this response is impaired in atherosclerosis but preserved with inhibition of NEP, together with a reduction in atheroma formation and preservation of relaxations to ACh.


Subject(s)
Arteriosclerosis/physiopathology , Muscle, Smooth, Vascular/drug effects , Natriuretic Peptide, Brain/pharmacology , Neprilysin/metabolism , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Cell Division/drug effects , Cells, Cultured , Humans , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Phenylephrine/pharmacology , Rabbits , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
16.
Atherosclerosis ; 148(1): 23-30, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10580167

ABSTRACT

BACKGROUND: Experimental hypercholesterolemia (HC) is characterized by a decrease in nitric oxide (NO) bioavailability and cellular proliferation. Nuclear factor-kappaB (NF-kappaB) is a transcriptional factor which plays a coordinating role in inflammation and cellular proliferation and may be involved in early atherosclerosis. We examined whether activated NF-kappaB was present in experimental hypercholesterolemia in the coronary vasculature in association with a decrease in NO bioavailability. METHODS: A total of 14 juvenile domestic crossbred pigs were placed on a HC diet and six pigs on a normal diet for 10-12 weeks. A monoclonal antibody to the activated form of the p65 subunit of NF-kappaB was used to detect immunoreactivity in coronary artery sections. Coronary tissue homogenates were analyzed for activated NF-kappaB and endothelial nitric oxide synthase (eNOS) using Western blotting. In vitro coronary endothelium-dependent relaxation was performed in response to bradykinin, as a measure of NO bioavailability. RESULTS: Intimal staining for activated NF-kappaB was present in 12/14 HC pigs as compared with 0/6 controls (P<0.001). Confocal microscopy confirmed the presence of NF-kappaB in the nucleus of intimal cells although the majority of the staining was cytoplasmic. In the HC group, Western blotting revealed an increase in activated NF-kappaB in the vessel wall compared to the normal group, in association with a decrease in the presence of eNOS protein and an attenuated vasorelaxation response to bradykinin. CONCLUSION: This study suggests a potential role for activation of NF-kappaB, in association with a decrease in NO bioavailability, in the initial stages of atherosclerosis in the coronary vasculature.


Subject(s)
Coronary Vessels/metabolism , Hypercholesterolemia/metabolism , NF-kappa B/metabolism , Animals , Blotting, Western , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Female , Hypercholesterolemia/blood , Hypercholesterolemia/pathology , Hypercholesterolemia/physiopathology , Immunohistochemistry , Lipids/blood , Protein Isoforms/metabolism , Swine
17.
Biochem Biophys Res Commun ; 265(3): 722-7, 1999 Nov 30.
Article in English | MEDLINE | ID: mdl-10600487

ABSTRACT

Caveolae have been implicated in growth factor receptor and G-protein coupled receptor signaling in vascular cells. It has been postulated that caveolin, the structural protein of caveolae, may act as a general tyrosine kinase inhibitor by binding and inhibiting signaling molecules involved in the activation of the MAP kinase proliferation cascade. Using an in vitro model of VSMC proliferation, we found that serum stimulation caused a dose dependent decrease in both caveolin-1 and caveolin-2 protein levels in human coronary artery smooth muscle cells. Heparin, an inhibitor of VSMC proliferation, inhibited the serum-induced loss of caveolin-1 and caveolin-2. In addition, heparin caused an increase in both caveolin-1 and caveolin-2 localization to caveolae-enriched sucrose gradient membrane fractions when compared to serum alone. Taken together, caveolin may play an important role in the regulation of VSMC proliferation and heparin and serum have opposing effects on caveolin expression and localization in VSMC.


Subject(s)
Caveolins , Heparin/pharmacology , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Caveolin 1 , Caveolin 2 , Cell Division/drug effects , Cells, Cultured , Culture Media , Humans , Microscopy, Fluorescence , Muscle, Smooth, Vascular/cytology
18.
Circulation ; 100(19 Suppl): II392-6, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10567335

ABSTRACT

BACKGROUND: Proliferation of the intima is an early lesion of saphenous vein graft disease. Early patency rates of radial artery grafts are acceptable, but little is known about their risk of intimal hyperplasia. METHODS AND RESULTS: To develop a model of intimal hyperplasia, we incubated human saphenous veins, internal mammary arteries, and radial arteries (n=6, 8, and 10, respectively) in an organ culture with Roswell Park Memorial Institute 1640 (30% serum) for 0, 4, 7, 10, and 14 days. Quantitative histological studies were performed, and the average intimal-to-medial (I/M) ratio was calculated for each incubation interval. After 10 and 14 days of culture, the I/M ratio increased in the saphenous veins (P=0. 03, P=0.04 versus 0 day, respectively). No significant increase occurred in the I/M ratio in either the internal mammary or radial arteries. Next, the ability of adenoviral gene transfers to inhibit intimal hyperplasia in the saphenous veins was evaluated. Adenoviral-mediated gene transfer of nitric oxide synthase significantly reduced the I/M ratio at 14 days compared with vehicle (P=0.001) and virus (P=0.004) controls. CONCLUSIONS: The human saphenous vein has a greater propensity for intimal hyperplasia than arterial grafts; the human radial artery behaves similarly to the internal mammary artery. In the future, gene therapy may augment nitric oxide synthase, limiting vein graft disease.


Subject(s)
Cardiopulmonary Bypass , Genetic Therapy , Nitric Oxide/physiology , Tunica Media/pathology , Adenoviridae , Gene Transfer Techniques , Genetic Vectors , Humans , Hyperplasia/genetics , Hyperplasia/therapy , Mammary Arteries/pathology , Organ Culture Techniques , Radial Artery/pathology , Saphenous Vein/pathology
20.
Atherosclerosis ; 142(2): 317-25, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10030383

ABSTRACT

The altered coronary vasoactivity detected in experimental hypercholesterolemia before lesion formation is presumably due to an imbalance between vasodilating and vasoconstricting factors. Apoptosis, which has been previously described in advanced atherosclerosis, is modulated by vascular derived peptides with vasoactive properties. We hypothesized that coronary apoptosis occurs in experimental hypercholesterolemia prior to lesion formation. Pigs were sacrificed after being on either a high-cholesterol diet for 10-16 weeks (n = 17) or a normal diet (n = 9). Identification of apoptosis in each layer of coronary arteries and arterioles was performed by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end-labeling (TUNEL). In additional animals, ligation-mediated polymerase chain reaction (PCR) and transmission electron and confocal microscopy were done. Plasma cholesterol levels were higher in the cholesterol-fed animals (86+/-9 mg/dl versus 342+/-20 mg/dl, P < 0.001). Atheromatous plaques were not evident in the high-cholesterol group. TUNEL was positive in 11 of 17 hypercholesterolemic animals, primarily in the intima (1-2% of cells) and adventitia (3% of cells), but not in control vessels. A similar distribution was detected in arterioles. DNA bands were detected only in experimental animals, as were morphological features of apoptosis by transmission electron and confocal microscopy. In experimental hypercholesterolemia, apoptosis occurred in coronary arteries and arterioles before lesion formation. Apoptosis may be an integral process of early coronary atherosclerosis.


Subject(s)
Apoptosis , Coronary Vessels/ultrastructure , Hypercholesterolemia/pathology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Division , Coloring Agents , Coronary Artery Disease/etiology , Coronary Artery Disease/pathology , DNA/analysis , DNA Primers/chemistry , Disease Models, Animal , Hypercholesterolemia/complications , In Situ Nick-End Labeling , Microscopy, Confocal , Polymerase Chain Reaction , Proliferating Cell Nuclear Antigen/genetics , Propidium , Swine , Tunica Intima/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL