Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Acta Physiol (Oxf) ; 210(4): 889-98, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24410878

ABSTRACT

AIM: We aimed to investigate the histological and clinical presentations of experimental autoimmune myocarditis (EAM) induced by different immunization schemes. METHODS: Male young Lewis rats were divided into five groups immunized by porcine myocardial myosin: subcutaneously (SC) 2 mg (in two 1-mg doses on day 0 and 7), 0 mg (sham group) subcutaneously into rear footpads (RF), 0.25 mg RF, 0.5 mg RF or 1 mg RF (all RF once on day 0). On day 21, left ventricular (LV) function was assessed by cardiac magnetic resonance imaging and cardiac catheterization. The type and degree of myocardial inflammatory infiltrates were determined by conventional histology and immunohistochemistry. RESULTS: In the SC immunized rats and in the RF sham group, we observed 0% mortality, while in the actively RF immunized rats, mortality was 20, 20 and 44% for the 0.25 mg, 0.5 mg and 1 mg myosin doses respectively. Morbidity as defined by inflammatory infiltrates on haematoxylin and eosin (HE) staining was 22% in the SC immunized rats, 0% in the RF sham group and 100% in all actively RF immunized groups. We observed augmented relative ventricle weight and spleen weight, increased LV end-diastolic pressure, reduced LV developed pressure and reduced LV ejection fraction in all with myosin-immunized RF groups without any systematic dose effect. CONCLUSION: Subcutaneous immunization to the neck and flanks did not induce a reproducible EAM, while RF myosin administration reliably led to EAM. Lower myosin doses seem to induce the complete histological and clinical picture of EAM while being associated with lower mortality, non-specific symptoms and animal distress.


Subject(s)
Autoimmune Diseases , Myocarditis/immunology , Myosins/immunology , Animals , Immunization , Male , Myocardium/immunology , Myocardium/pathology , Myosins/administration & dosage , Random Allocation , Rats , Rats, Inbred Lew
3.
Dtsch Med Wochenschr ; 136(47): 2418, 2011 Nov.
Article in German | MEDLINE | ID: mdl-22048949

ABSTRACT

This commentary summarizes the expert consensus and recommendations of the working group 'Herz und Niere' of the German Society of Cardiology (DGK), the German Society of Nephrology (DGfN) and the German Hypertension League (DHL) on renal denervation for antihypertensive treatment. Renal denervation is a new, interventional approach to selectively denervate renal afferent and efferent sympathetic fibers. Renal denervation has been demonstrated to reduce office systolic and diastolic blood pressure in patients with resistant hypertension, defined as systolic office blood pressure ≥ 160 mm Hg and ≥ 150 mm Hg in patients with diabetes type 2, which should currently be used as blood pressure thresholds for undergoing the procedure. Exclusion of secondary hypertension causes and optimized antihypertensive drug treatment is mandatory in every patient with resistant hypertension. In order to exclude pseudoresistance, 24-hour blood pressure measurements should be performed. Preserved renal function was an inclusion criterion in the Symplicity studies, therefore, renal denervation should be only considered in patients with a glomerular filtration rate > 45 ml/min. Adequate centre qualification in both, treatment of hypertension and interventional expertise are essential to ensure correct patient selection and procedural safety. Long-term follow-up after renal denervation and participation in the German Renal Denervation (GREAT) Registry are recommended to assess safety and efficacy after renal denervation over time.


Subject(s)
Catheter Ablation , Hypertension, Renal/surgery , Renal Artery/innervation , Sympathectomy/methods , Adolescent , Adult , Aged , Aged, 80 and over , Angiography , Blood Glucose/metabolism , Blood Pressure , Blood Pressure Monitoring, Ambulatory , Diagnosis, Differential , Follow-Up Studies , Heart Rate , Humans , Hypertension, Renal/diagnosis , Hypertension, Renal/drug therapy , Hypertension, Renal/etiology , Middle Aged , Randomized Controlled Trials as Topic , Registries , Young Adult
5.
Diabetes Obes Metab ; 11(11): 1060-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19732122

ABSTRACT

BACKGROUND AND AIM: Results on the cardiovascular effects of PPAR-gamma agonists are conflicting. On one hand, it was suggested that the PPAR-gamma agonist rosiglitazone may increase the risk of cardiovascular events. On the other hand, PPAR-gamma agonists reduce myocardial infarct size and improve myocardial function during ischemia/reperfusion in animal studies in vivo. However, the mechanism of this effect is unclear, and it is open if PPAR-gamma agonists have a direct effect on cardiac myocyte survival in ischemia/reperfusion. The aim of this study was to determine the effect of the PPAR-gamma agonist rosiglitazone on hypoxia/reoxygenation-induced apoptosis of isolated cardiomyocytes. METHODS: Isolated rat cardiac myocytes were pretreated with rosiglitazone or vehicle for 30 min before they were subjected to hypoxia for 4 h followed by different times of reoxygenation (5 min to 12 h). Apoptosis was determined by in situ hybridization for DNA fragmentation (TUNEL) as well as detection of cytoplasmic accumulation of histone-associated DNA fragments by enzyme-linked immunosorbent assay (ELISA). Activation of apoptosis regulating intracellular signalling pathways was studied by immunoblotting using phosphospecific antibodies. RESULTS: Rosiglitazone significantly reduced apoptosis of isolated cardiomyocytes subjected to hypoxia/reoxygenation, independently determined with two methods. After 4 h of hypoxia and 12 h of reoxygenation, 34 +/- 3.6% of the vehicle treated cardiac myocytes stained positive for DNA fragmentation in the TUNEL staining. Rosiglitazone treatment reduced this effect by 23% (p < 0.01). Even more pronounced, cytoplasmic accumulation of histone-associated DNA fragments detected by ELISA was reduced by 35% (p < 0.05) in the presence of rosiglitazone. This inhibition of hypoxia/reoxygenation-induced apoptosis was associated with an increased reoxygenation-induced rephosphorylation of the protein kinase Akt, a crucial mediator of cardiomyocyte survival in ischemia/reperfusion of the heart. This effect was reversed by GW-9662, an irreversible PPAR-gamma antagonist. However, rosiglitazone did not alter phosphorylation of the MAP kinases ERK1/2 and c-Jun N-terminal kinase (JNK). CONCLUSION: It can be concluded that cardiac myocytes are direct targets of PPAR-gamma agonists promoting its survival in ischemia/reperfusion, at least in part by facilitating Akt rephosphorylation. This effect may be of clinical relevance inhibiting the reperfusion-induced injury in patients suffering from myocardial infarction or undergoing cardiac surgery.


Subject(s)
Apoptosis/drug effects , Cell Hypoxia/drug effects , Hypoglycemic Agents/pharmacology , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Thiazolidinediones/pharmacology , Analysis of Variance , Animals , Myocytes, Cardiac/physiology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Rosiglitazone
6.
Dtsch Med Wochenschr ; 133(24): 1308-12, 2008 Jun.
Article in German | MEDLINE | ID: mdl-18465684

ABSTRACT

Aliskiren, a renin inhibitor, is the first in a new class of drugs interfering with the renin angiotensin system. Aliskiren was approved by the US Food and Drug Administration (FDA) in March 2007, and in Europe in August 2007 for the treatment of hypertension (marketed as Tekturna and Rasilez, respectively). Several clinical trials demonstrated effective blood pressure reduction due to aliskiren treatment. Whether aliskiren exhibits morbidity and mortality benefits for patients beyond its blood pressure reduction capability, can only be judged after realization of comparative long-term clinical trials. Furthermore, it remains to be seen, whether the use of aliskiren will be indicated for treatment of additional diseases, as it was the case for other inhibitors of the renin angiotensin system. In fact, recent and ongoing clinical trials regarding heart failure and diabetic nephropathy demonstrated first beneficial effects of aliskiren in these conditions (reduction of urinary albumin/creatinine-ratio and NTproBNP, respectively).


Subject(s)
Amides/therapeutic use , Antihypertensive Agents/therapeutic use , Fumarates/therapeutic use , Hypertension/drug therapy , Renin/antagonists & inhibitors , Amides/adverse effects , Animals , Antihypertensive Agents/adverse effects , Cardiovascular Diseases/drug therapy , Contraindications , Fumarates/adverse effects , Gastrointestinal Diseases/chemically induced , Humans , Kidney Diseases/drug therapy
7.
Acta Diabetol ; 42 Suppl 1: S26-32, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15868116

ABSTRACT

Vascular protection is key to reducing the morbidity associated with diabetes. Angiotensin II is known to exert a variety of deleterious effects on the vasculature, and this is likely to be a major explanation of the protective benefits observed with blockade of the renin-angiotensin-aldosterone system (RAAS). Intriguingly, RAAS blockade also appears to reduce the onset of new diabetes, which points to a fundamental effect on metabolism. Recent developments have thrown new light onto the mechanism of these effects. The importance of unopposed stimulation of the angiotensin II type 2 (AT(2)) receptor in vascular protection is recognised, and recent studies have revealed that some angiotensin II type 1 (AT(1)) receptor blockers (ARBs) show partial peroxisome proliferator-activated receptor-gamma (PPARgamma) agonistic activity in vitro, an effect that is at least partly due to direct interaction with PPARgamma itself. There is a clear order of potency among the ARBs, with telmisartan the most potent and the only ARB to show an effect at physiologically achievable plasma concentrations. Adiponectin, an adipokine closely involved with glucose sensitisation, is also modulated by the relative activation of AT(1) and AT(2) receptors. Such effects would suggest that important benefits may result from the use of ARBs in clinical practice, although confirmation of the clinical relevance will depend upon the results of numerous ongoing studies.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Antihypertensive Agents/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/prevention & control , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antihypertensive Agents/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Benzoates/pharmacology , Benzoates/therapeutic use , Humans , Insulin Resistance/physiology , PPAR gamma/physiology , Renin-Angiotensin System/drug effects , Telmisartan
8.
J Cardiovasc Pharmacol ; 38(6): 909-21, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11707695

ABSTRACT

Vascular smooth muscle cell (VSMC) migration involves adhesion, locomotion, and invasion regulated by various signaling molecules, among which the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinases (MAPK) play a critical role. We have shown that the peroxisome proliferator-activated receptor-gamma (PPAR-gamma) ligands troglitazone and rosiglitazone inhibit VSMC migration downstream of ERK MAPK. The purpose of the current study was to more specifically determine which step(s) in VSMC migration are targeted by inhibition of the ERK MAPK pathway or activation of PPAR-gamma. VSMC adhesion was not affected by the ERK MAPK pathway inhibitor PD98059 or PPAR-gamma ligands. Phosphorylation and activation of myosin light chain kinase (MLCK) play important roles in cell locomotion. Platelet-derived growth factor (PDGF)-induced MLCK phosphorylation (1.7-fold) was completely blocked by PD98059 at 30 microM (p < 0.05), but not by troglitazone or rosiglitazone. PDGF-directed migration (5.8-fold) was inhibited by PD98059 (-88% at 30 microM) and the MLCK inhibitor ML9 (0.1-1 microM, -84% at 1 microM) (all p < 0.05). The transcription factor Ets-1 mediates matrix metalloproteinase induction required for tissue invasion by VSMC. PDGF (20 ng/ml) stimulated an Ets-1 protein expression (14-fold at 60 min) in VSMC, which was inhibited by PD98059 (-72% at 30 microM), troglitazone (-69% at 20 microM), and rosiglitazone (-54% at 10 microM) (all p < 0.05). Immunohistochemistry of rat aortae 2 h after balloon injury showed a dramatic upregulation of Ets-1, which was markedly inhibited in animals that had received troglitazone treatment. In contrast, phosphorylated ERK MAPK was not affected by troglitazone. These data are consistent with PPAR-gamma ligands exerting their anti-migratory effects downstream of ERK MAPK activation by blocking nuclear events, such as Ets-1 expression, required for cell invasion in response to arterial injury.


Subject(s)
Cell Movement , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Receptors, Cytoplasmic and Nuclear/agonists , Thiazolidinediones , Transcription Factors/agonists , Animals , Aortic Diseases/etiology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Nucleus/enzymology , Cells, Cultured , Chromans/pharmacology , Cytosol/enzymology , Enzyme Inhibitors/pharmacology , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/metabolism , Graft Occlusion, Vascular/pathology , Ligands , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Rosiglitazone , Thiazoles/pharmacology , Transcription Factors/metabolism , Transcriptional Activation , Troglitazone
9.
Atherosclerosis ; 159(1): 93-101, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11689211

ABSTRACT

Migration, proliferation and differentiation of vascular smooth muscle cells (VSMC) and macrophages are important pathological responses that contribute to the development and progression of vascular lesions. Cytokines such as TNFalpha are present at sites of vascular injury and regulate a variety of cellular functions of inflammatory cells and VSMC. Cell migration, proliferation and differentiation require de novo gene transcription resulting from extracellular signals being transduced to the nucleus, where multiple genes are regulated to participate in lesion formation. In VSMC and macrophages, TNFalpha induces activation of the extracellular signal-regulated kinases 1/2 (ERK 1/2), which transmit signals from the cytosol to the nucleus. Potential nuclear targets of TNFalpha-activated ERK 1/2 include the transcription factors Ets-1, Egr-1, and c-fos, which are known to regulate cellular growth, differentiation, and migration. The aim of this study was to investigate the expression of the transcription factors Ets-1, Egr-1 and c-fos in different types of vascular lesions, their regulation by TNFalpha and the role of ERK 1/2 in these signaling events. Atherosclerotic lesions from fructose-fed LDL-receptor deficient mice and neointimal lesions from rat aortae 2 weeks post balloon injury demonstrated the presence and colocalization of TNFalpha, phosphorylated and activated ERK 1/2, and transcription factors Ets-1, Egr-1 and c-fos. Neointimal lesions consisted primarily of VSMC, whereas atherosclerotic lesions predominantly contained macrophages. In cultured rat aortic VSMC, TNFalpha (100 U/ml) stimulated a rapid and transient expression of Ets-1, Egr-1 and c-fos with a maximal induction 1 h after stimulation. In cultured RAW 264.7 mouse macrophages, TNFalpha similarly induced the expression of Ets-1, Egr-1, and c-fos. Induction of these transcription factors was mediated via ERK 1/2 activation, since the ERK 1/2-pathway inhibitor PD98059 (10-30 microM) significantly inhibited their TNFalpha-induced expression. TNFalpha induced ERK 1/2 activation in both cell types. These findings underscore the importance of the ERK 1/2 pathway in the expression of TNFalpha-regulated transcription factors, which may participate in different forms of vascular lesion formation.


Subject(s)
DNA-Binding Proteins/biosynthesis , Immediate-Early Proteins , Mitogen-Activated Protein Kinases/physiology , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Transcription Factors/biosynthesis , Tumor Necrosis Factor-alpha/physiology , Animals , Arteriosclerosis/metabolism , Blotting, Western , Cells, Cultured , Early Growth Response Protein 1 , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Immunohistochemistry , Macrophages/metabolism , Male , Mice , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Rats , Rats, Sprague-Dawley
10.
J Biol Chem ; 276(50): 47650-7, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11598137

ABSTRACT

The cyclin-dependent kinase inhibitor p21(Cip1) is up-regulated in response to mitogenic stimulation in various cells. PPARgamma ligands troglitazone (TRO, 10 microm) and rosiglitazone (RSG, 10 microm) attenuated the induction of p21(Cip1) protein by platelet-derived growth factor (PDGF) and insulin without affecting cognate mRNA levels in rat aortic smooth muscle cells (RASMC). The protein kinase Cdelta (PKCdelta) inhibitor rottlerin also blocked the induction of p21(Cip1) protein, whereas the conventional PKC isotype inhibitor Gö 6976 had no effect. Kinetic studies using the protein synthesis inhibitor cycloheximide showed that TRO, RSG, and rottlerin shortened the half-life of p21(Cip1) protein. TRO, RSG, and rottlerin inhibited PDGF-induced expression of p21(Cip1), but they did not affect insulin-induced expression of p21(Cip1). Both ligands inhibited PKCdelta enzymatic activity in PDGF-stimulated RASMC but not in insulin-stimulated cells. Adenovirus-mediated overexpression of PKCdelta rescued the down-regulation of p21(Cip1) expression both by TRO and RSG in PDGF-treated RASMC. These data suggested that the PKCdelta pathway plays a critical role in PDGF-induced expression of p21(Cip1) in RASMC and may be the potential target for PPARgamma ligand effects. Src kinase-dependent tyrosine phosphorylation of PKCdelta was decreased substantially by TRO and RSG. Tyrosine phosphorylation and activation of c-Src in response to PDGF were unaffected by either PPARgamma ligand. Protein-tyrosine-phosphatase inhibitors sodium orthovanadate and dephostatin prevented PPARgamma ligand effects on PKCdelta tyrosine phosphorylation and enzymatic activity. Both inhibitors also reversed PPARgamma ligand effects on p21(Cip1) expression in PDGF-treated RASMC. PPARgamma ligands enhanced protein-tyrosine-phosphatase activity in RASMC, which may be the mechanism for decreased PKCdelta tyrosine phosphorylation and activity. PPARgamma ligands regulate p21(Cip1) at a post-translational level by blocking PKCdelta signaling and accelerating p21(Cip1) turnover.


Subject(s)
Cyclins/metabolism , Isoenzymes/metabolism , Ligands , Mitogens/pharmacology , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazolidinediones , Transcription Factors/metabolism , Acetophenones/pharmacology , Adenoviridae/genetics , Animals , Aorta, Thoracic/cytology , Apoptosis , Benzopyrans/pharmacology , Blotting, Western , Carbazoles/pharmacology , Cell Division , Cells, Cultured , Chromans/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Inhibitors/pharmacology , Hydroquinones/pharmacology , Indoles/pharmacology , Insulin/metabolism , Kinetics , Mice , Models, Biological , Muscle, Smooth, Vascular/cytology , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Precipitin Tests , Protein Kinase C-delta , Protein Processing, Post-Translational , Protein Synthesis Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , RNA/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Rosiglitazone , Signal Transduction , Thiazoles/pharmacology , Time Factors , Troglitazone , Tyrosine/metabolism , Up-Regulation , Vanadates/pharmacology
11.
Pest Manag Sci ; 57(7): 577-86, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11464788

ABSTRACT

Acute oral and contact toxicity tests of imidacloprid, an insecticide acting agonistically on nicotinic acetylcholine receptors (nAChR), to adult honeybees, Apis mellifera L var carnica, were carried out by seven different European research facilities. Results indicated that the 48-h oral LD50 of imidacloprid is between 41 and > 81 ng per bee, and the contact LD50 between 49 and 102 ng per bee. The ingested amount of imidacloprid-containing sucrose solution decreased with increasing imidacloprid concentrations and may be attributed to dose-related sub-lethal intoxication symptoms or to antifeedant responses. Some previously reported imidacloprid metabolites occurring at low levels in planta after seed dressing, i.e. olefine-, 5-OH- and 4,5-OH-imidacloprid, showed lower oral LD50 values (> 36, > 49 and 159 ng per bee, respectively) compared with the concurrently tested parent molecule (41 ng per bee). The urea metabolite and 6-chloronicotinic acid (6-CNA) exhibited LD50 values of > 99,500 and > 121,500 ng per bee, respectively. The pharmacological profile of the [3H]imidacloprid binding site in honeybee head membrane preparations is consistent with that anticipated for a nAChR. IC50 values for the displacement of [3H]imidacloprid by several metabolites such as olefine, 5-OH-, 4,5-OH-imidacloprid, urea and 6-CNA were 0.45, 24, 6600, > 100,000, and > 100,000 nM, respectively. Displacement of [3H]imidacloprid by imidacloprid revealed an IC50 value of 2.9 nM, thus correlating well with the observed acute oral toxicity of the compounds in honeybees. Neurons isolated from the antennal lobe of A mellifera and subjected to whole-cell voltage clamp electrophysiology responded to the application of 100 microM acetylcholine with a fast inward current of between 30 and 1600 pA at -70 mV clamp potential. Imidacloprid and two of the metabolites (olefine- and 5-OH-imidacloprid) acted agonistically on these neurons, whereas the others did not induce currents at test concentrations up to 3 mM. The electrophysiological data revealed Hill coefficients of approximately 1, indicating a single binding site responsible for an activation of the receptor and no direct cooperativity or allosteric interaction with a second binding site.


Subject(s)
Bees , Imidazoles , Insecticides , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Bees/metabolism , Biological Assay , Electrophysiology , Imidazoles/chemistry , Imidazoles/metabolism , Insecticides/chemistry , Insecticides/metabolism , Lethal Dose 50 , Neonicotinoids , Neurons/drug effects , Nitro Compounds
12.
Arterioscler Thromb Vasc Biol ; 21(5): 746-51, 2001 May.
Article in English | MEDLINE | ID: mdl-11348869

ABSTRACT

Retinoids inhibit rat vascular smooth muscle cell (VSMC) proliferation in vitro and intimal hyperplasia in vivo. We examined the mechanism of the antiproliferative effect of retinoids on human coronary artery smooth muscle cells (human CASMCs). The RAR ligands all-trans-retinoic acid (atRA) and ethyl-p-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthyl)-l-propenyl]-benzoic acid (TTNPB); a pan-RXR/RAR agonist, 9-cis-retinoic acid (9cRA); and the RXR-selective ligand AGN4204 all inhibited DNA synthesis stimulated with platelet-derived growth factor and insulin (IC(50): TTNPB 63 nmol/L, atRA 120 nmol/L, AGN4204 460 nmol/L, 9cRA 1.5 micromol/L). All retinoids blocked cell cycle progression as determined by flow cytometry and inhibited retinoblastoma protein (Rb) phosphorylation. TTNPB, atRA, and AGN4204 inhibited the mitogenic induction of cyclin D1, whereas 9cRA had no effect. None of the retinoids affected the expression of CDK 2, 4, or 6 or cyclin E. All retinoids attenuated mitogen-induced downregulation of CDKI p27(Kip1), a major negative regulator of Rb phosphorylation, partly through stabilizing p27(Kip1) turnover. These data demonstrate that retinoids have antiproliferative activity by modulating G(1) --> S cell cycle regulators in human CASMCs through inhibition of Rb phosphorylation and elevation of p27(Kip1) levels.


Subject(s)
Cell Cycle Proteins/metabolism , Coronary Vessels/cytology , DNA Replication/drug effects , Muscle, Smooth, Vascular/drug effects , Retinoids/pharmacology , Tumor Suppressor Proteins , Cells, Cultured , Coronary Vessels/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , DNA/biosynthesis , Down-Regulation , Humans , Microtubule-Associated Proteins/metabolism , Mitogens/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phenotype , Phosphorylation , Retinoblastoma Protein/metabolism , S Phase
13.
J Cardiovasc Pharmacol ; 37(5): 532-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11336104

ABSTRACT

Monocyte chemotactic protein 1 (MCP-1)-directed transendothelial migration of monocytes plays a key role in the early development of atherosclerosis. Migration of monocytes requires degradation of extracellular matrices, a process that involves matrix metalloproteinases (MMP) and tissue inhibitors of MMPs (TIMP). Recent studies suggest that the alpha1-adrenergic receptor antagonist doxazosin (Dox) might have antiatherosclerotic effects, although the underlying mechanisms are poorly understood. The purpose of the present study was to determine the effects of Dox on MCP-1-directed monocyte migration, MMP-9 activity, and TIMP-1 expression. MCP-1 (50 ng/ml) stimulated migration of human peripheral blood monocytes (HPBM) 2.7+/-0.42-fold and THP-1 human monocytes 5.9+/-0.83-fold compared with unstimulated control. Dox inhibited MCP-1-induced migration in a dose-dependent manner, with a maximal reduction at 10 microM of 69.5+/-5.9% in HPBM and 72.2+/-3.2% in THP-1 cells. Dox blocked migration even after pretreatment with phenoxybenzamine, an irreversible alpha1-adrenergic receptor antagonist (HPBM: phenoxybenzamine 1 microM + Dox 10 microM, 71.9+/-2.2% inhibition; THP-1 cells: phenoxybenzamine 1 microM + Dox 10 microM: 78+/-7.7% inhibition), suggesting that the antimigratory activity of Dox is mediated through a novel mechanism unrelated to its blocking of the alpha1-adrenergic receptor. Dox (10 microM) inhibited MMP-9 activity by 67.6+/-10.5%, whereas MMP-9 protein levels were not affected. Also, Dox increased PMA-induced-tissue inhibitor of MMPs-1 (TIMP-1) expression by 134.4+/-6.6%. Dox 10 microM. The present study demonstrates a potential novel antiatherosclerotic action of Dox by blocking MCP-1-directed monocyte migration, which might be partly mediated by inhibition of MMP-9 activity.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Cell Movement/drug effects , Chemokine CCL2/antagonists & inhibitors , Doxazosin/pharmacology , Monocytes/drug effects , Cell Movement/physiology , Chemokine CCL2/pharmacology , Humans , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Monocytes/metabolism , Tissue Inhibitor of Metalloproteinase-1/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Cells, Cultured
14.
Arterioscler Thromb Vasc Biol ; 21(3): 365-71, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231915

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a ligand-activated nuclear receptor expressed in all of the major cell types found in atherosclerotic lesions: monocytes/macrophages, endothelial cells, and smooth muscle cells. In vitro, PPARgamma ligands inhibit cell proliferation and migration, 2 processes critical for vascular lesion formation. In contrast to these putative antiatherogenic activities, PPARgamma has been shown in vitro to upregulate the CD36 scavenger receptor, which could promote foam cell formation. Thus, it is unclear what impact PPARgamma activation will have on the development and progression of atherosclerosis. This issue is important because thiazolidinediones, which are ligands for PPARgamma, have recently been approved for the treatment of type 2 diabetes, a state of accelerated atherosclerosis. We report herein that the PPARgamma ligand, troglitazone, inhibited lesion formation in male low density lipoprotein receptor-deficient mice fed either a high-fat diet, which also induces type 2 diabetes, or a high-fructose diet. Troglitazone decreased the accumulation of macrophages in intimal xanthomas, consistent with our in vitro observation that troglitazone and another thiazolidinedione, rosiglitazone, inhibited monocyte chemoattractant protein-1-directed transendothelial migration of monocytes. Although troglitazone had some beneficial effects on metabolic risk factors (in particular, a reduction of insulin levels in the diabetic model), none of the systemic cardiovascular risk factors was consistently improved in either model. These observations suggest that the inhibition of early atherosclerotic lesion formation by troglitazone may result, at least in part, from direct effects of PPARgamma activation in the artery wall.


Subject(s)
Arteriosclerosis/prevention & control , Chromans/pharmacology , Diabetes Mellitus, Type 2/complications , Receptors, LDL/deficiency , Thiazoles/pharmacology , Thiazolidinediones , Vasodilator Agents/pharmacology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/etiology , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemokine CCL2/pharmacology , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Endothelium, Vascular/cytology , Flavonoids/pharmacology , Fructose/administration & dosage , Humans , Insulin/blood , Lipids/blood , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Receptors, LDL/genetics , Rosiglitazone , Troglitazone , Tumor Cells, Cultured
15.
Hypertension ; 37(2 Pt 2): 587-93, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230339

ABSTRACT

Angiotensin (Ang) II has been shown to enhance the development of atherosclerotic lesions. Migration of monocytes is an early critical step in the atherosclerotic process. To elucidate mechanisms by which Ang II promotes atherogenesis, we investigated its effects on human monocyte migration. Ang II induced migration of human peripheral blood monocytes (HPBM) and human THP-1 monocytes at concentrations between 0.01 and 1 micromol/L, with a 3.6+/-0.6-fold induction in HPBM and a 4.8+/-0.9-fold induction in THP-1 cells at 1 micromol/L Ang II (both P<0.01 versus unstimulated cells). Addition of the Ang II receptor type 1 (AT1-R) antagonist losartan (1 to 100 micromol/L) suppressed Ang II-induced migration of HPBM and THP-1 monocytes in a dose-dependent manner, demonstrating an AT1-R-mediated mechanism. Ang II-directed migration was also blocked by the Src kinase inhibitor PP2 (10 micromol/L), by the extracellular-regulated protein kinase (ERK 1/2) inhibitor PD98059 (30 micromol/L), and by the p38-MAPK inhibitor SB203580 (10 micromol/L), indicating that Src, ERK 1/2, and p38 are all involved in Ang II-induced migration of HPBM and human THP-1 monocytes. The proline-rich tyrosine kinase 2 (Pyk2) and paxillin are 2 cytoskeleton-associated proteins involved in cell movement, phosphorylated by Ang II in other cell types, and abundantly expressed in monocytes. Ang II (1 micromol/L) induced Pyk2 and paxillin phosphorylation in human THP-1 monocytes, peaking after 10 minutes for Pyk2 with a 6.7+/-0.9-fold induction and after 2 minutes for paxillin with a 3.2+/-0.4-fold induction. Ang II-induced phosphorylation of both proteins was suppressed by losartan and the Src inhibitor PP2, whereas no effect was observed with PD98059 and SB203580. This study demonstrates a novel proatherogenic action of Ang II on human monocytes by stimulating their migration, through an AT1-R-dependent process, involving signaling through Src, ERK 1/2, and p38. Furthermore, the promigratory actions of Ang II in human monocytes are associated with the phosphorylation of 2 cytoskeleton-associated proteins, Pyk2 and paxillin.


Subject(s)
Angiotensin II/pharmacology , Monocytes/drug effects , Angiotensin Receptor Antagonists , Arteriosclerosis/metabolism , CSK Tyrosine-Protein Kinase , Cell Line , Cell Movement/drug effects , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/metabolism , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/biosynthesis , Isoenzymes/metabolism , Losartan/pharmacology , Monocytes/physiology , Paxillin , Phosphopeptides/pharmacology , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/metabolism , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Signal Transduction , src-Family Kinases
16.
Eur J Pharmacol ; 401(3): 259-70, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10936484

ABSTRACT

Monocyte chemotactic protein-1 (MCP-1)-directed transendothelial migration of monocytes plays a key role in the development of inflammatory diseases. Infiltration of tissues by monocytes requires degradation of extracellular matrices, a process that involves matrix metalloproteinases. We studied the effects of peroxisome proliferator-activated receptor (PPAR) gamma, alpha, and retinoid X receptor alpha (RXRalpha) ligands on MCP-1-directed migration and matrix metalloproteinase expression of a human acute monocytic leukemia cell line (THP-1). PPARgamma ligands attenuated MCP-1-induced migration, with 50% inhibition (IC(50)) at 2.8 microM for troglitazone and 4.8 microM for rosiglitazone. PPARalpha ligands WY-14643 (IC(50): 0.9 microM) and 5,8,11,14-eicosatetranoic acid (IC(50): 9.9 microM), and the potent RXRalpha ligand AGN 4204 (IC(50): 3.6 nM) also blocked monocyte migration. Troglitazone, rosiglitazone, or AGN 4204 inhibited phorbol 12-myristate 13-acetate (PMA)-induced matrix metalloproteinase-9 expression. PPARalpha activators WY-14643 and 5,8,11,14-eicosatetraynoic acid, however, had no inhibitory effect. AGN 4204 increased PMA-induced tissue inhibitor of matrix metalloproteinases-1 (TIMP-1) expression, whereas all PPAR ligands showed no effect. All PPAR and RXRalpha ligands blocked chemotaxis of THP-1 monocytes in the absence of a matrix barrier. This study demonstrates that activated PPARs and RXRalpha, block MCP-1-directed monocyte migration, mediated, at least in part, through their effects on matrix metalloproteinase-9 or TIMP-1 production, or chemotaxis.


Subject(s)
Cell Movement/drug effects , Chemokine CCL2/pharmacology , Monocytes/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Retinoic Acid/agonists , Thiazolidinediones , Transcription Factors/agonists , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Chemotaxis/drug effects , Chromans/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Monocytes/cytology , Pyrimidines/pharmacology , Retinoic Acid Receptor alpha , Rosiglitazone , Tetradecanoylphorbol Acetate/pharmacology , Thiazoles/pharmacology , Tissue Inhibitor of Metalloproteinase-1/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Troglitazone , Tumor Cells, Cultured
17.
Arterioscler Thromb Vasc Biol ; 20(5): 1216-24, 2000 May.
Article in English | MEDLINE | ID: mdl-10807736

ABSTRACT

Previous studies have demonstrated that the alpha(1)-adrenergic receptor antagonist doxazosin (Dox) inhibits multiple mitogenic signaling pathways in human vascular smooth muscle cells. This broad antiproliferative activity of Dox occurs through a novel mechanism unrelated to its blocking the alpha(1)-adrenergic receptor. Flow cytometry demonstrated that Dox prevents mitogen-induced G(1)-->S progression of human coronary artery smooth muscle cells (CASMCs) in a dose-dependent manner, with a maximal reduction of S-phase transition by 88+/-10.5% in 20 ng/mL platelet-derived growth factor and 1 micromol/L insulin (P+I)-stimulated cells (P<0.01 for 10 micromol/L Dox versus P+I alone) and 52+/-18.7% for 10% FBS-induced mitogenesis (P<0.05 for 10 micromol/L Dox versus 10% FBS alone). Inhibition of G(1) exit by Dox was accompanied by a significant blockade of retinoblastoma protein (Rb) phosphorylation. Hypophosphorylated Rb sequesters the E2F transcription factor, leading to G(1) arrest. Adenoviral overexpression of E2F-1 stimulated quiescent CASMCs to progress through G(1) and enter the S phase. E2F-mediated G(1) exit was not affected by Dox, suggesting that it targets events upstream from Rb hyperphosphorylation. Downregulation of the cyclin-dependent kinase inhibitory protein p27 is important for maximal activation of G(1) cyclin/cyclin-dependent kinase holoenzymes to overcome the cell cycle inhibitory activity of Rb. In Western blot analysis, p27 levels decreased after mitogenic stimulation (after P+I, 43+/-1.8% of quiescent cells [P<0.01 versus quiescent cells]; after 10% FBS, 55+/-7.7% of quiescent cells [P<0. 05 versus quiescent cells]), whereas the addition of Dox (10 micromol/L) markedly attenuated its downregulation (after P+I, 90+/-8.3% of quiescent cells [P<0.05 versus P+I alone]; after 10% FBS, 78+/-8.3% of quiescent cells [P<0.05 versus 10% FBS alone]). Furthermore, Dox inhibited cyclin A expression, an E2F regulated gene that is essential for cell cycle progression into the S phase. The present study demonstrates that Dox inhibits CASMC proliferation by blocking cell cycle progression from the G(0)/G(1) phase to the S phase. This G(1)-->S blockade likely results from an inhibition of mitogen-induced Rb hyperphosphorylation through prevention of p27 downregulation.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Cell Cycle/drug effects , Coronary Vessels/cytology , Doxazosin/pharmacology , Muscle, Smooth, Vascular/cytology , Retinoblastoma Protein/metabolism , Coronary Vessels/metabolism , Cyclin A/metabolism , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , G1 Phase/drug effects , Humans , Muscle, Smooth, Vascular/metabolism , Phosphorylation , S Phase/drug effects
18.
J Biol Chem ; 275(29): 22435-41, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10801895

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a member of the nuclear receptor superfamily that is activated by binding certain fatty acids, eicosanoids, and insulin-sensitizing thiazolidinediones (TZD). The TZD troglitazone (TRO) inhibits vascular smooth muscle cell proliferation and migration both in vitro and in vivo. The precise mechanism of its antiproliferative activity, however, has not been elucidated. We report here that PPARgamma ligands inhibit rat aortic vascular smooth muscle cell proliferation by blocking the events critical for G(1) --> S progression. Flow cytometry demonstrated that both TRO and another TZD, rosiglitazone, prevented G(1) --> S progression induced by platelet-derived growth factor and insulin. Movement of cells from G(1) --> S was also inhibited by the non-TZD, natural PPARgamma ligand 15-deoxy-(12,14)Delta prostaglandin J(2) (15d-PGJ(2)), and the mitogen-activated protein kinase pathway inhibitor PD98059. Inhibition of G(1) --> S exit by these compounds was accompanied by a substantial blockade of retinoblastoma protein phosphorylation. TRO and rosiglitazone attenuated both the mitogen-induced degradation of p27(kip1) and the mitogenic induction of p21(cip1). 15d-PGJ(2) and PD98059 inhibited both the degradation of p27(kip1) and the induction of cyclin D1 in response to mitogens. These effects resulted in the inhibition of mitogenic stimulation of cyclin-dependent kinases activated by cyclins D1 and E. These data demonstrate that PPARgamma ligands are antiproliferative drugs that act by modulating cyclin-dependent kinase inhibitors; they may provide a new therapeutic approach for proliferative vascular diseases.


Subject(s)
Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Cells, Cultured , G1 Phase , Ligands , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , S Phase , Transcription Factors/agonists
19.
J Biol Chem ; 275(24): 18279-83, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10764814

ABSTRACT

Tumor necrosis factor alpha (TNFalpha) interferes with insulin signaling in adipose tissue and may promote insulin resistance. Insulin binding to the insulin receptor (IR) triggers its autophosphorylation, resulting in phosphorylation of Shc and the downstream activation of p42/p44 extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (ERK1/2), which mediates insulin-induced proliferation in vascular smooth muscle cells (VSMC). Since insulin resistance is a risk factor for vascular disease, we examined the effects of TNFalpha on mitogenic signaling by insulin. In rat aortic VSMC, insulin induced rapid phosphorylation of the IR and Shc and caused a 5.3-fold increase in activated, phosphorylated ERK1/2 at 10 min. Insulin induced a biphasic ERK1/2 activation with a transient peak at 10 min and a sustained late phase after 2 h. Preincubation (30-120 min) with TNFalpha had no effect on insulin-induced IR phosphorylation. In contrast, TNFalpha transiently suppressed insulin-induced ERK1/2 activation. Insulin-induced phosphorylation of Shc was inhibited by TNFalpha in a similar pattern. Since mitogenic signaling by insulin in VSMC requires ERK1/2 activation, we examined the effect of TNFalpha on insulin-induced proliferation. Insulin alone induced a 3.4-fold increase in DNA synthesis, which TNFalpha inhibited by 48%. TNFalpha alone was not mitogenic. Inhibition of ERK1/2 activation with PD98059 also inhibited insulin-stimulated DNA synthesis by 57%. TNFalpha did not inhibit platelet-derived growth factor-induced ERK1/2 activation or DNA synthesis in VSMC. Thus, TNFalpha selectively interferes with insulin-induced mitogenic signaling by inhibiting the phosphorylation of Shc and the downstream activation of ERK1/2.


Subject(s)
Insulin/pharmacology , Mitosis , Muscle, Smooth, Vascular/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Androstadienes/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Tyrosine/metabolism , Wortmannin
20.
Eur J Pharmacol ; 390(1-2): 75-87, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10708709

ABSTRACT

human umbilical venous endothelial cells. 7E3 binding correlated with alphavbeta3-expression in all cell types. Integrin-mediated cell functions were analysed with adhesion and spreading assays on vitronectin. In human umbilical venous endothelial cells, these functions were mediated by alphavbeta3 and in human iliac arterial smooth muscle cells by alphavbeta5. In human umbilical venous smooth muscle cells, both vitronectin receptors were involved. Abciximab potently inhibited alphavbeta3-mediated cell adhesion and spreading. With tirofiban, no significant inhibition of vascular cell functions was observed. The present data demonstrate that vitronectin-cell interactions in vascular cells are mediated via two distinct integrin-receptors, alphavbeta3 and alphavbeta5. Abciximab, which solely inhibits alphavbeta3-mediated cell functions, may be particularly effective in human endothelium and in beta3-integrin expressing vascular smooth muscle cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Endothelium, Vascular/metabolism , Fibrinolytic Agents/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Muscle, Smooth, Vascular/metabolism , Receptors, Vitronectin/drug effects , Tyrosine/analogs & derivatives , Abciximab , Cell Adhesion/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Flow Cytometry , Humans , Iliac Artery/cytology , Iliac Artery/drug effects , Iliac Artery/metabolism , Immunohistochemistry , Integrins/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oligopeptides/isolation & purification , Oligopeptides/pharmacology , Receptors, Vitronectin/biosynthesis , Receptors, Vitronectin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tirofiban , Tyrosine/pharmacology , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vitronectin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...