Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
QJM ; 115(12): 830-836, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-35866641

ABSTRACT

BACKGROUND: While progress is evident in gender and ethnic representation in the workplace, this disparity remains prevalent in academic positions. OBJECTIVES: We examined gender and Asian ethnic representation in editorial boards of cardiology journals. METHODS: A cross-sectional analysis was conducted using publicly available data on Cardiology and Cardiovascular medicine journals in the first quartile of the 2020 Scimago Journal & Country Rank indicator. The proportions of female and Asian editorial board members, associate editors and editors-in-chief were assessed. Subgroup analyses were conducted based on the journal's geographical origin, subspecialty and demographic of the editor-in-chief. RESULTS: Seventy-six cardiology journals, involving 8915 editorial board members, were included. Only 19.6% of editorial board members were female, 20.8% Asians and 4.0% Asian females. There were less female representation amongst editors-in-chief (9.9%) compared to associate editors (22.3%). European (18.1%) and North American-based journals (21.1%) had higher female representation compared to Asian-based journals (8.7%). There was lower Asian representation in European (18.1%) and North American-based journals (19.9%) compared to Asian-based journals (72.3%). Females were underrepresented in interventional (14.5%) journals, while Asians were underrepresented in general cardiology (18.3%) and heart failure (18.3%) journals. Journals led by female editors-in-chief had significantly higher female representation compared to male-led ones, while journals with Asian editors-in-chief had greater Asian representation compared to non-Asian led ones. CONCLUSION: This study highlights the female and Asian ethnic underrepresentation in academic roles in cardiology journal editorial boards. Further analysis is needed for other ethnicities, while the community pushes towards gender-balanced and ethnic diversity across editorial boards.


Subject(s)
Asian , Gender Equity , Female , Humans , Male , Cross-Sectional Studies
2.
Diabetologia ; 56(8): 1856-66, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23616239

ABSTRACT

AIM/HYPOTHESIS: Monocytes/macrophages play important roles in adipose and vascular tissues and can be polarised as inflammatory M1 or anti-inflammatory M2. We sought to analyse monocyte polarisation status in type 2 diabetes, which is characterised by chronic inflammation. METHODS: We enrolled 60 individuals without diabetes and 53 patients with type 2 diabetes. We quantified standard monocyte subsets defined by cluster of differentiation (CD)14 and CD16. In addition, based on the phenotype of polarised macrophages in vitro, we characterised and quantified more definite M1 (CD68(+)CCR2(+)) and M2 (CX3CR1(+)CD206(+)/CD163(+)) monocytes. We also analysed bone marrow (BM) samples and the effects of granulocyte-colony stimulating factor (G-CSF) stimulation in diabetic and control individuals. RESULTS: We found no alterations in standard monocyte subsets (classical, intermediate and non-classical) when comparing groups. For validation of M1 and M2 phenotypes, we observed that M2 were enriched in non-classical monocytes and had lower TNF-α content, higher LDL scavenging and lower transendothelial migratory capacity than M1. Diabetic patients displayed an imbalanced M1/M2 ratio compared with the control group, attributable to a reduction in M2. The M1/M2 ratio was directly correlated with waist circumference and HbA1c and, among diabetic patients, M2 reduction and M1/M2 increase were associated with microangiopathy. A decrease in M2 was also found in the BM from diabetic patients, with a relative M2 excess compared with the bloodstream. BM stimulation with G-CSF mobilised M2 macrophages in diabetic but not in healthy individuals. CONCLUSIONS/INTERPRETATION: We show that type 2 diabetes markedly reduces anti-inflammatory M2 monocytes through a dysregulation in bone-marrow function. This defect may have a negative impact on microangiopathy.


Subject(s)
Bone Marrow/immunology , Diabetes Mellitus, Type 2/immunology , Diabetic Angiopathies/immunology , Monocytes/cytology , Adult , Aged , Aged, 80 and over , Bone Marrow/metabolism , Cells, Cultured , Female , Humans , Male , Middle Aged , Monocytes/immunology
5.
Clin Res Cardiol ; 95(1): 13-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16598441

ABSTRACT

BACKGROUND: Coronary microvascular dysfunction contributes to infarct extension and poor prognosis after an acute myocardial infarction (AMI). Recently, progenitor cell application has been demonstrated to improve neovascularization and myocardial function after experimental myocardial infarction. Therefore, we investigate coronary blood flow regulation in patients after AMI treated with intracoronary progenitor cell therapy. METHODS AND RESULTS: In the TOPCARE-AMI trial, patients received either bone marrow-derived or circulating progenitor cells into the infarct-related artery 3-7 days after AMI. The present substudy investigates in 40 patients coronary blood flow regulation at the time of progenitor cell therapy and at 4-month follow-up by i.c. Doppler in the infarct artery as well as a reference vessel. At the initial measurement, coronary flow reserve (CFR) was reduced in the infarct artery compared to the reference vessel (median 2.5 vs. 3.4, p<0.001). At 4-month follow-up, intracoronary progenitor cell therapy was associated with a normalization of CFR in the infarct artery (median 3.9 vs. reference vessel 3.8, p=0.15). CFR also improved in the reference vessel, but mechanisms were different: reference vessel increase in CFR was secondary to an increased basal vascular resistance, probably due to reduced need for hypercontractility. In contrast, in the infarct artery, adenosine-induced minimal vascular resistance profoundly decreased, indicating an increased maximal coronary vascular conductance capacity. In addition, in a non-randomized matched control group (n=8), minimal vascular resistance in the infarct artery was significantly elevated compared to progenitor cell treated patients 4 months after AMI (p=0.012). CONCLUSIONS: Intracoronary progenitor cell therapy after AMI is associated with complete restoration of coronary flow reserve due to a substantial improvement of maximal coronary vascular conductance capacity. The clinical importance of improved microcirculation by progenitor cell therapy in patients after AMI has to be established in further randomized trials.


Subject(s)
Blood Flow Velocity , Coronary Circulation , Coronary Vessels/diagnostic imaging , Coronary Vessels/surgery , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/surgery , Stem Cell Transplantation/methods , Coronary Vessels/physiopathology , Echocardiography, Doppler , Female , Humans , Male , Middle Aged , Myocardial Infarction/physiopathology , Prognosis , Recovery of Function , Treatment Outcome
6.
Handb Exp Pharmacol ; (174): 283-98, 2006.
Article in English | MEDLINE | ID: mdl-16370332

ABSTRACT

Postinfarction congestive heart failure with impaired systolic left ventricular function is a loss of cardiomyocyte disease. Adult stem or progenitor cells from the bone marrow and the peripheral blood have been experimentally shown to differentiate towards endothelial cells and cardiomyocytes under the appropriate conditions. The use of autologous adult stem cells for neovascularization and cardiac regeneration is a promising concept and has shown benefit in pilot clinical trails enrolling postinfarction patients with coronary artery disease. Cell therapy may act through differentiation into and thus replacement of cardiomyocytes and/or neovascularization, the formation of new vessels in the adult organism. Moreover, the release of factors acting in a paracrine manner may contribute to neovascularization and scar remodelling. In this review, the experimental data regarding neovascularization and cardiomyocyte formation from adult stem/progenitor cells are discussed.


Subject(s)
Bone Marrow Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic/physiology , Stem Cells/cytology , Stem Cells/physiology , Wound Healing/physiology , Animals , Cell Differentiation , Humans , Myocytes, Cardiac/physiology
7.
J Intern Med ; 257(5): 415-22, 2005 May.
Article in English | MEDLINE | ID: mdl-15836657

ABSTRACT

OBJECTIVE: Enhanced expression of CXCL16 has been demonstrated in atherosclerotic plaques and several properties have been attributed to CXCL16 that could influence the atherosclerotic process. CXCL16 exists in transmembrane and soluble forms. The transmembrane form acts as a scavenger receptor for oxidised LDL whereas the soluble form acts a chemoattractant for mainly CD8+ T cells. In addition, the soluble form of CXCL16 influences human aortic smooth muscle cell proliferation in vitro. In the present work, a human molecular genetic approach employing a common polymorphism within exon 4 of CXCL16 (181 Ala>Val) was used to investigate whether CXCL16 may be involved in the development of coronary artery disease. The polymorphism is located within the spacer region between the chemokine and transmembrane region and potentially influences an Ala/Val cleavage site, a site commonly used for the release of chemokines by tumour necrosis factor-alpha converting enzyme. DESIGN AND SUBJECTS: We first genotyped 387 unselected survivors of a first myocardial infarction aged <60 years and 387 sex- and age-matched controls. A subset of patients (n = 236) was evaluated by quantitative coronary angiography. Secondly, a cohort of 468 patients undergoing percutaneous transluminal coronary angioplasty (PTCA) with stent implantation was genotyped. RESULTS: No significant difference in allele frequency between patient and controls of the 181 A>V polymorphism was detected. However, the V-allele was associated with increased severity of coronary stenoses. Secondly, the V-allele was associated with smaller minimal luminal diameter in the coronary segment subjected to intervention in a second cohort of patients undergoing PTCA with stent implantation. CONCLUSIONS: The present work provides evidence that CXCL16 is involved in processes leading to enhanced stenosis in atherosclerotic coronary arteries.


Subject(s)
Chemokines, CXC/genetics , Coronary Stenosis/genetics , Membrane Proteins/genetics , Myocardial Infarction/genetics , Receptors, Immunologic/genetics , Amino Acid Sequence , Analysis of Variance , Angioplasty, Balloon, Coronary , Case-Control Studies , Chemokine CXCL16 , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Chi-Square Distribution , Coronary Angiography , Coronary Stenosis/pathology , Female , Follow-Up Studies , Gene Frequency , Humans , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Middle Aged , Molecular Sequence Data , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Polymorphism, Genetic , Polymorphism, Restriction Fragment Length , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Scavenger , Sequence Analysis, DNA , Stents
9.
Circulation ; 108(18): 2212-8, 2003 Nov 04.
Article in English | MEDLINE | ID: mdl-14557356

ABSTRACT

BACKGROUND: Experimental and initial clinical studies suggest that transplantation of circulating blood- (CPC) or bone marrow-derived (BMC) progenitor cells may beneficially affect postinfarction remodeling processes after acute myocardial infarction (AMI). To relate functional characteristics of the infused cells to quantitative measures of outcome at 4-month follow-up, we performed serial contrast-enhanced MRI and assessed the migratory capacity of the transplanted progenitor cells immediately before intracoronary infusion. METHODS AND RESULTS: In 28 patients with reperfused AMI receiving either BMCs or CPCs into the infarct artery 4.7+/-1.7 days after AMI, serial contrast-enhanced MRI performed initially and after 4 months revealed a significant increase in global ejection fraction (from 44+/-10% to 49+/-10%; P=0.003), a decrease in end-systolic volume (from 69+/-26 to 60+/-28 mL; P=0.003), and unchanged end-diastolic volumes (122+/-34 versus 117+/-37 mL; P=NS). Infarct size, measured as late enhancement (LE) volume, decreased significantly, from 46+/-32 to 37+/-28 mL (P<0.05). There was a significant correlation between the reduction in LE volume and global ejection fraction improvement. The migratory capacity of transplanted cells as assessed ex vivo toward a gradient of vascular endothelial growth factor for CPCs and stromal cell derived factor-1 for BMCs was closely correlated with the reduction of LE volume. By multivariate analysis, migratory capacity remained the most important independent predictor of infarct remodeling. CONCLUSIONS: Analysis of serial contrast-enhanced MRI suggests that intracoronary infusion of adult progenitor cells in patients with AMI beneficially affects postinfarction remodeling processes. The migratory capacity of the infused cells is a major determinant of infarct remodeling, disclosing a causal effect of progenitor cell therapy on regeneration enhancement.


Subject(s)
Coronary Vessels , Magnetic Resonance Imaging , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Ventricular Remodeling , Cell Movement/drug effects , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Coronary Angiography , Female , Follow-Up Studies , Humans , Image Enhancement , Infusions, Intra-Arterial , Magnetic Resonance Imaging/methods , Male , Middle Aged , Myocardial Reperfusion , Recovery of Function , Stents , Stroke Volume , Treatment Outcome , Vascular Endothelial Growth Factor A/pharmacology , Ventricular Function, Left
10.
J Am Coll Cardiol ; 38(7): 2063-70, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11738316

ABSTRACT

OBJECTIVES: The aim of this study was to investigate whether the caspase-3 inhibitor Ac-DEVD-CHO functionally improves stunned myocardium. BACKGROUND: Degradation of troponin I contributes to the pathogenesis of myocardial stunning, whereas the role of apoptosis is unknown. Caspase-3 is an essential apoptotic protease that is specifically inhibited by Ac-DEVD-CHO. METHODS: Isolated working hearts of rats were exposed to 30 min of low-flow ischemia, followed by 30 min of reperfusion. Ac-DEVD-CHO (0.1 to 1 micromol/l) was added 15 min before ischemia/reperfusion or 5 min before reperfusion. Cardiac output, external heart power, left ventricular (LV) developing pressure and contractility (dp/dt(max)) were measured. Apoptosis was assessed by TUNEL staining and internucleosomal deoxyribonucleic acid fragmentation. Caspase-3 processing and troponin I cleavage were determined by immunoblotting. Caspase-3 activity was measured using a fluorogenic substrate. RESULTS: The addition of Ac-DEVD-CHO before ischemia/reperfusion or before reperfusion dose-dependently and significantly (p < 0.05) improved post-ischemic recovery of cardiac output, external heart power, LV developing pressure and dp/dt(max), compared with the vehicle (0.01% dimethyl sulfoxide). Ac-DEVD-CHO was similarly effective when given before reperfusion. Ac-DEVD-CHO blocked ischemia/reperfusion-induced caspase-3 activation, but cardiomyocyte apoptosis was unaffected. Troponin I cleavage was not inhibited by Ac-DEVD-CHO. CONCLUSIONS: Caspase-3 is activated in stunned myocardium. Inhibition of caspase-3 by Ac-DEVD-CHO significantly improves post-ischemic contractile recovery of stunned myocardium, even when given after the onset of ischemia. The mechanism(s) of protection by Ac-DEVD-CHO appear to be independent of apoptosis. Inhibition of caspase-3 is a novel therapeutic strategy to improve functional recovery of stunned myocardium.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Myocardial Contraction/drug effects , Myocardial Stunning/physiopathology , Oligopeptides/pharmacology , Animals , Apoptosis/physiology , Cardiac Output/drug effects , Cardiac Output/physiology , Caspase 3 , Caspases/physiology , Dose-Response Relationship, Drug , In Situ Nick-End Labeling , Male , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/physiopathology , Perfusion , Rats , Rats, Sprague-Dawley , Troponin I/metabolism , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
11.
Circulation ; 104(25): 3023-5, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11748093

ABSTRACT

BACKGROUND: Anti-tumor necrosis factor (TNF)-alpha therapy with etanercept, a recombinant TNF receptor that binds to and functionally inactivates TNF-alpha, was shown to improve the functional status of patients with congestive heart failure (CHF). Because administration of TNF-alpha has been shown experimentally to depress endothelium-dependent relaxation, we hypothesized that TNF-alpha antagonism with etanercept might improve the depressed systemic endothelial vasodilator function, which importantly contributes to increased peripheral vascular resistance in patients with advanced CHF. METHODS AND RESULTS: Endothelium-dependent (acetylcholine, ACH; 10 to 50 microgram/min) and endothelium-independent (sodium nitroprusside, SNP; 2 to 8 microgram/min) forearm blood flow (FBF) responses were measured by venous occlusion plethysmography in 13 patients with documented CHF (New York Heart Association class III) before, 6 hours after, and 7 days after subcutaneous injection of a single dose of 25 mg etanercept. Maximum ACH-induced FBF increased significantly from 6.9+/-1.0 to 13.0+/-1.6 mL/min per 100 mL of forearm tissue (P<0.05) 6 hours after administration of etanercept and returned to 7.0+/-1.1 mL/min per 100 mL of forearm tissue after 7 days (P=NS), whereas SNP-induced FBF responses were not significantly affected. In contrast, FBF responses were not altered in control CHF patients, who did not receive etanercept (n=5). Etanercept-induced increases in ACH-mediated FBF were closely correlated with baseline TNF-alpha serum levels (r=0.66; P<0.02). CONCLUSIONS: The administration of etanercept profoundly improves systemic endothelial vasodilator capacity in patients with advanced heart failure, suggesting an important role of inflammatory mediators for impaired endothelial vasoreactivity in CHF. Improvement of systemic endothelial function might importantly contribute to the beneficial effects of etanercept on the functional status of patients with CHF.


Subject(s)
Endothelium, Vascular/drug effects , Heart Failure/drug therapy , Immunoglobulin G/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Acetylcholine/pharmacology , Adult , Dose-Response Relationship, Drug , Endothelium, Vascular/physiopathology , Etanercept , Female , Forearm/blood supply , Heart Failure/blood , Heart Failure/physiopathology , Humans , Immunoglobulin G/therapeutic use , Male , Middle Aged , Nitroprusside/pharmacology , Plethysmography , Receptors, Tumor Necrosis Factor/therapeutic use , Regional Blood Flow/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vasodilator Agents/pharmacology
12.
J Clin Invest ; 108(9): 1341-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696579

ABSTRACT

Endothelial nitric oxide synthase (eNOS) is activated by phosphorylation of serine 1177 by the protein kinase Akt/PKB. Since hyperglycemia-induced mitochondrial superoxide overproduction increases O-linked N-acetylglucosamine modification and decreases O-linked phosphorylation of the transcription factor Sp1, the effect of hyperglycemia and the hexosamine pathway on eNOS was evaluated. In bovine aortic endothelial cells, hyperglycemia inhibited eNOS activity 67%, and treatment with glucosamine had a similar effect. Hyperglycemia-associated inhibition of eNOS was accompanied by a twofold increase in O-linked N-acetylglucosamine modification of eNOS and a reciprocal decrease in O-linked serine phosphorylation at residue 1177. Both the inhibition of eNOS and the changes in its post-translational modifications were reversed by antisense inhibition of glutamine:fructose-6-phosphate amidotransferase, the rate-limiting enzyme of the hexosamine pathway, or by blocking mitochondrial superoxide overproduction with uncoupling protein-1 (UCP-1) or manganese superoxide dismutase (MnSOD). Immunoblot analysis of cells expressing myc-tagged wild-type human eNOS confirmed the reciprocal increase in O-linked N-acetylglucosamine and decrease in O-linked serine 1177 phosphorylation in response to hyperglycemia. In contrast, when myc-tagged human eNOS carried a mutation at the Akt phosphorylation site (Ser1177), O-linked N-acetylglucosamine modification was unchanged by hyperglycemia and phospho-eNOS was undetectable. Similar changes in eNOS activity and covalent modification were found in aortae from diabetic animals. Chronic impairment of eNOS activity by this mechanism may partly explain the accelerated atherosclerosis of diabetes.


Subject(s)
Hyperglycemia/enzymology , Nitric Oxide Synthase/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Acetylglucosamine/metabolism , Animals , Cattle , Cells, Cultured , Diabetes Mellitus, Experimental/enzymology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Humans , Immunoblotting , Membrane Potentials , Mutation , Nitric Oxide Synthase Type III , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , Superoxide Dismutase/metabolism
13.
J Clin Invest ; 108(10): 1451-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714736

ABSTRACT

Atherosclerosis is an inflammatory disease of large arteries that is initiated through the activation of endothelium by proinflammatory mediators. CD40 receptor stimulation has been implicated in the pathogenesis of atherosclerosis. One of the most important atheroprotective stimuli is the viscous drag (shear stress) generated by the streaming blood acting on the endothelial monolayer. Here, we demonstrate that shear stress prevents CD40 ligand-induced endothelial cell activation, and we identify upregulation of TNF receptor-associated factor-3 (TRAF-3) as a potent CD40-inhibitory mechanism. Shear stress specifically upregulates TRAF-3 in cultured endothelial cells. Moreover, in the endothelial cells overlying human atherosclerotic plaques, TRAF-3 expression is upregulated in areas with high shear stress. Overexpression of TRAF-3 inhibits endothelial expression of proinflammatory cytokines and tissue factor and blocks DNA-binding activity of the transcription factor AP-1; it thereby prevents CD40-induced endothelial activation. Thus, upregulation of TRAF-3 represents a novel mechanism for preserving the functional integrity of the endothelial monolayer.


Subject(s)
CD40 Antigens/metabolism , Proteins/metabolism , Up-Regulation , Base Sequence , Blotting, Northern , Blotting, Western , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Signal Transduction , TNF Receptor-Associated Factor 3
14.
Circulation ; 104(18): 2182-7, 2001 Oct 30.
Article in English | MEDLINE | ID: mdl-11684628

ABSTRACT

BACKGROUND: Proinflammatory cytokines like tumor necrosis factor-alpha and oxidative stress induce apoptotic cell death in endothelial cells (ECs). Systemic inflammation and increased oxidative stress in congestive heart failure (CHF) coincide with enhanced EC apoptosis and the development of endothelial dysfunction. Therefore, we investigated the effects of antioxidative vitamin C therapy on EC apoptosis in CHF patients. METHODS AND RESULTS: Vitamin C dose dependently suppressed the induction of EC apoptosis by tumor necrosis factor-alpha and angiotensin II in vitro as assessed by DNA fragmentation, DAPI nuclear staining, and MTT viability assay. The antiapoptotic effect of vitamin C was associated with reduced cytochrome C release from mitochondria and the inhibition of caspase-9 activity. To assess EC protection by vitamin C in CHF patients, we prospectively randomized CHF patients in a double-blind trial to vitamin C treatment versus placebo. Vitamin C administration to CHF patients markedly reduced plasma levels of circulating apoptotic microparticles to 32+/-8% of baseline levels, whereas placebo had no effect (87+/-14%, P<0.005). In addition, vitamin C administration suppressed the proapoptotic activity on EC of the serum of CHF patients (P<0.001). CONCLUSIONS: Administration of vitamin C to CHF patients suppresses EC apoptosis in vivo, which might contribute to the established functional benefit of vitamin C supplementation on endothelial function.


Subject(s)
Apoptosis/drug effects , Ascorbic Acid/administration & dosage , Endothelium, Vascular/drug effects , Heart Failure/drug therapy , Administration, Oral , Adult , Aged , Angiotensin II/pharmacology , Biomarkers/blood , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Administration Schedule , Endothelium, Vascular/cytology , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Female , Heart Failure/physiopathology , Humans , Injections, Intravenous , Male , Middle Aged , Oxidative Stress/drug effects , Prospective Studies , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
15.
Circ Res ; 89(8): 709-15, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11597994

ABSTRACT

Advanced aging leads to impaired endothelial NO synthesis and enhanced endothelial cell apoptosis; therefore, we investigated the sensitivity of aged endothelial cells toward apoptotic stimuli and determined the role of NO. Human umbilical vein endothelial cells (HUVECs) were cultured until 14th passage. In aged cells, oxLDL and tumor necrosis factor-alpha-induced apoptosis and caspase-3-like activity were significantly enhanced more than 3-fold compared with young cells (passage 3). Because NO contributes to protection against endothelial cell death via S-nitrosylation of caspases, we determined endothelial NO synthase (eNOS) protein expression and the content of S-nitrosylated proteins. Aged HUVECs showed significantly reduced eNOS expression (35+/-10%) and a decrease in the overall S-NO content (33+/-3%), suggesting that eNOS downregulation may be involved in age-dependent increase of apoptosis sensitivity. Indeed, eNOS knockout endothelial cells showed a significantly enhanced apoptosis induction. Exogenous NO donors abolished increased apoptosis and caspase-3-like activity. In contrast, the application of shear stress, which exerts a profound apoptosis inhibitory effect via upregulation of NO synthesis in young cells, failed to inhibit apoptosis in aged cells. Moreover, no upregulation of eNOS protein expression and S-NO content in response to shear stress was detected in aged cells. Overexpression of wild-type eNOS completely restored the antiapoptotic effect of shear stress, whereas only a partial inhibitory effect was detected under steady conditions. Strikingly, transfection of constitutively active phosphomimetic eNOS (S1177D) further abrogated apoptosis in aged HUVECs. Thus, aging of endothelial cells is associated with decreased NO synthesis and concomitantly increased sensitivity of apoptosis, which may contribute to functional impairment of the endothelial monolayer.


Subject(s)
Aging/metabolism , Apoptosis/physiology , Endothelium, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases , Aging/drug effects , Amino Acid Substitution , Animals , Aorta , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Activation/genetics , Humans , Lipoproteins, LDL/pharmacology , Male , Mice , Mice, Knockout , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroso Compounds/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Stress, Mechanical , Sulfhydryl Compounds/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology
16.
J Clin Invest ; 108(3): 391-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489932

ABSTRACT

HMG-CoA reductase inhibitors (statins) have been developed as lipid-lowering drugs and are well established to reduce morbidity and mortality from coronary artery disease. Here we demonstrate that statins potently augment endothelial progenitor cell differentiation in mononuclear cells and CD34-positive hematopoietic stem cells isolated from peripheral blood. Moreover, treatment of mice with statins increased c-kit(+)/Sca-1(+)--positive hematopoietic stem cells in the bone marrow and further elevated the number of differentiated endothelial progenitor cells (EPCs). Statins induce EPC differentiation via the PI 3-kinase/Akt (PI3K/Akt) pathway as demonstrated by the inhibitory effect of pharmacological PI3K blockers or overexpression of a dominant negative Akt construct. Similarly, the potent angiogenic growth factor VEGF requires Akt to augment EPC numbers, suggesting an essential role for Akt in regulating hematopoietic progenitor cell differentiation. Given that statins are at least as potent as VEGF in increasing EPC differentiation, augmentation of circulating EPC might importantly contribute to the well-established beneficial effects of statins in patients with coronary artery disease.


Subject(s)
Endothelium, Vascular/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Coronary Disease/drug therapy , Coronary Disease/pathology , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , In Vitro Techniques , Lymphokines/pharmacology , Mice , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-akt , Stem Cells/cytology , Stem Cells/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Biochem Biophys Res Commun ; 286(2): 419-25, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11500055

ABSTRACT

The effect of prolonged hypoxia as well as the molecular mechanisms on cardiac cell death is not well established. A possible role of Bcl-2 and Bax in hypoxia-induced apoptosis in different cell types has been proposed. Here we demonstrate the effect of hypoxia on the induction of apoptosis and the expression of Bcl-2-like proteins in vivo and in vitro. Hearts from rats exposed to chronic hypoxia (n = 4) showed an increased rate of apoptosis compared to normoxic hearts (n = 4). The induction of apoptosis in hypoxic hearts correlated with a significant decrease of Bcl-2 protein level, whereas Bax protein expression was increased. Exposure of isolated neonatal rat cardiac myocytes to hypoxia also resulted in a significant increase in apoptosis. However, Bcl-2 and Bax protein levels essentially remained unchanged. Our results may suggest a different molecular mechanism of hypoxia-induced apoptosis in vivo and in vitro.


Subject(s)
Apoptosis , Cardiomyopathies/etiology , Hypoxia/pathology , Myocardium/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Animals, Newborn , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Hypoxia , Cells, Cultured , Hypoxia/metabolism , Myocardium/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein
18.
J Biol Chem ; 276(44): 41383-7, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11524431

ABSTRACT

Nitric oxide (NO) plays an important role in the regulation of the functional integrity of the endothelium. The intracellular reaction of NO with reactive cysteine groups leads to the formation of S-nitrosothiols. To investigate the regulation of S-nitrosothiols in endothelial cells, we first analyzed the composition of the S-nitrosylated molecules in endothelial cells. Gel filtration revealed that more than 95% of the detected S-nitrosothiols had a molecular mass of more than 5000 Da. Moreover, inhibition of de novo synthesis of glutathione using N-butyl-sulfoximine did not diminish the overall cellular S-NO content suggesting that S-nitrosylated glutathione quantitatively plays only a minor role in endothelial cells. Having demonstrated that most of the S-nitrosothiols are proteins, we determined the regulation of the S-nitrosylation by pro-inflammatory and pro-atherogenic factors, such as TNFalpha and mildly oxidized low density lipoprotein (oxLDL). TNFalpha and oxLDL induced denitrosylation of various proteins as assessed by Saville-Griess assay, by immunostaining with an anti-S-nitrosocysteine antibody, and by a Western blot approach. Furthermore, the caspase-3 p17 subunit, which has previously been shown to be S-nitrosylated and thereby inhibited, was denitrosylated by TNFalpha treatment suggesting that S-nitrosylation and denitrosylation are important regulatory mechanisms in endothelial cells contributing to the integrity of the endothelial cell monolayer.


Subject(s)
Endothelium, Vascular/metabolism , Lipoproteins, LDL/metabolism , S-Nitrosothiols/metabolism , Tumor Necrosis Factor-alpha/metabolism , Blotting, Western , Chromatography, Gel , Endothelium, Vascular/cytology , Humans , Molecular Weight , S-Nitrosothiols/chemistry
19.
Mol Cell Biol ; 21(16): 5644-57, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463845

ABSTRACT

The protein kinase Akt is activated by growth factors and promotes cell survival and cell cycle progression. Here, we demonstrate that Akt phosphorylates the cell cycle inhibitory protein p21(Cip1) at Thr 145 in vitro and in intact cells as shown by in vitro kinase assays, site-directed mutagenesis, and phospho-peptide analysis. Akt-dependent phosphorylation of p21(Cip1) at Thr 145 prevents the complex formation of p21(Cip1) with PCNA, which inhibits DNA replication. In addition, phosphorylation of p21(Cip1) at Thr 145 decreases the binding of the cyclin-dependent kinases Cdk2 and Cdk4 to p21(Cip1) and attenuates the Cdk2 inhibitory activity of p21(Cip1). Immunohistochemistry and biochemical fractionation reveal that the decrease of PCNA binding and regulation of Cdk activity by p21(Cip1) phosphorylation is not caused by altered intracellular localization of p21(Cip1). As a functional consequence, phospho-mimetic mutagenesis of Thr 145 reverses the cell cycle-inhibitory properties of p21(Cip1), whereas the nonphosphorylatable p21(Cip1) T145A construct arrests cells in G(0) phase. These data suggest that the modulation of p21(Cip1) cell cycle functions by Akt-mediated phosphorylation regulates endothelial cell proliferation in response to stimuli that activate Akt.


Subject(s)
Cyclins/physiology , Endothelium, Vascular/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Cell Division/physiology , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Endothelium, Vascular/cytology , Humans , Phosphorylation , Proliferating Cell Nuclear Antigen/physiology , Protein Binding , Proto-Oncogene Proteins c-akt , Signal Transduction
20.
Circ Res ; 89(1): E1-7, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11440984

ABSTRACT

Recent studies provide increasing evidence that postnatal neovascularization involves bone marrow-derived circulating endothelial progenitor cells (EPCs). The regulation of EPCs in patients with coronary artery disease (CAD) is unclear at present. Therefore, we determined the number and functional activity of EPCs in 45 patients with CAD and 15 healthy volunteers. The numbers of isolated EPCs and circulating CD34/kinase insert domain receptor (KDR)-positive precursor cells were significantly reduced in patients with CAD by approximately 40% and 48%, respectively. To determine the influence of atherosclerotic risk factors, a risk factor score including age, sex, hypertension, diabetes, smoking, positive family history of CAD, and LDL cholesterol levels was used. The number of risk factors was significantly correlated with a reduction of EPC levels (R=-0.394, P=0.002) and CD34-/KDR-positive cells (R=-0.537, P<0.001). Analysis of the individual risk factors demonstrated that smokers had significantly reduced levels of EPCs (P<0.001) and CD34-/KDR-positive cells (P=0.003). Moreover, a positive family history of CAD was associated with reduced CD34-/KDR-positive cells (P=0.011). Most importantly, EPCs isolated from patients with CAD also revealed an impaired migratory response, which was inversely correlated with the number of risk factors (R=-0.484, P=0.002). By multivariate analysis, hypertension was identified as a major independent predictor for impaired EPC migration (P=0.043). The present study demonstrates that patients with CAD revealed reduced levels and functional impairment of EPCs, which correlated with risk factors for CAD. Given the important role of EPCs for neovascularization of ischemic tissue, the decrease of EPC numbers and activity may contribute to impaired vascularization in patients with CAD. The full text of this article is available at http://www.circresaha.org.


Subject(s)
Coronary Disease/etiology , Coronary Disease/pathology , Endothelium, Vascular/physiology , AC133 Antigen , Antigens, CD , Antigens, CD34/analysis , Cell Count , Cell Movement , Cells, Cultured , Coronary Artery Disease/etiology , Coronary Artery Disease/pathology , Endothelium, Vascular/cytology , Female , Glycoproteins/analysis , Hematopoietic Stem Cells/cytology , Humans , Hypertension/complications , Male , Middle Aged , Neovascularization, Pathologic , Peptides/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Growth Factor/analysis , Receptors, Vascular Endothelial Growth Factor , Risk Factors , Smoking
SELECTION OF CITATIONS
SEARCH DETAIL
...