Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Eur J Heart Fail ; 11(8): 739-48, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19633101

ABSTRACT

AIMS: Activation of phosphoinositide-3 kinase (PI3K) is essential for cell growth, relating to adaptive and maladaptive cardiac hypertrophy. This longitudinal canine study was designed to investigate the role of PI3Kalpha and PI3Kgamma in cardiac remodelling during congestive heart failure (CHF) and cardiac recovery (CR). METHODS AND RESULTS: All dogs were surgically instrumented. Congestive heart failure was induced by cardiac pacing for 3-4 weeks and CR was allowed by terminating pacing for 5-6 weeks after induction of HF. Control dogs had sham surgery, but did not undergo pacing. Left ventricular (LV) contractile function was depressed in CHF and restored to 80-90% of the normal level in CR, with a 25% increase in LV weight. The expression of PI3Kgamma was increased four-fold in CHF, but returned to control levels in CR. In contrast, the expression of PI3Kalpha in CHF was not different from that in controls, but increased three-fold in CR and was accompanied by increases in phosphorylation of Akt (five-fold), GSK-3beta (five-fold), beta-catenin (three-fold), mTOR (two-fold), and P70S6K (two-fold). CONCLUSION: Our results indicate that PI3K isoforms are regulated differently during the course of CHF/CR and that the selective activation of PI3Kalpha, through Akt, GSK-3beta, and mTOR signalling pathways, may be involved in the development of cardiac compensatory hypertrophy and functional restoration.


Subject(s)
Glycogen Synthase Kinase 3 , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Analysis of Variance , Animals , Dobutamine , Dogs , Glycogen Synthase Kinase 3 beta , Hemodynamics , Longitudinal Studies , Male , Models, Animal , RNA, Messenger , Signal Transduction
2.
Biochem Biophys Res Commun ; 367(3): 642-8, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18190779

ABSTRACT

Inflammation is critically involved in atherogenesis. Signaling from innate immunity receptors TLR2 and 4, IL-1 and IL-18 is mediated by MyD88 and further by interleukin-1 receptor activated kinases (IRAK) 4 and 1. We hypothesized that IRAK4 kinase activity is critical for development of atherosclerosis. IRAK4 kinase-inactive knock-in mouse was crossed with the ApoE-/- mouse. Lesion development was stimulated by carotid ligation. IRAK4 functional deficiency was associated with down-regulation of several pro-inflammatory genes, inhibition of macrophage infiltration, smooth muscle cell and lipid accumulation in vascular lesions. Reduction of plaque size and inhibition of outward remodeling were also observed. Similar effects were observed when ApoE-/- mice subjected to carotid ligation were treated with recombinant IL-1 receptor antagonist thereby validating the model in the relevant pathway context. Thus, IRAK4 functional deficiency inhibits vascular lesion formation in ApoE-/- mice, which further unravels mechanisms of vascular inflammation and identifies IRAK4 as a potential therapeutic target.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/prevention & control , Disease Models, Animal , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1 Receptor-Associated Kinases/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/pathology , C-Reactive Protein/analysis , C-Reactive Protein/biosynthesis , C-Reactive Protein/genetics , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Crosses, Genetic , Diet, Atherogenic , Disease Progression , Gene Expression Regulation/drug effects , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/prevention & control , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/pharmacology , Interleukin-6/blood , Ligation , Mice , Mice, Knockout , Mice, Transgenic , Vascular Patency/drug effects , Vascular Patency/genetics
3.
Blood ; 109(2): 560-5, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16990598

ABSTRACT

Phosphatidylinositol-3-kinase (PI3K), and its downstream effector Akt, or protein kinase Balpha (PKBalpha), play a major regulatory role in control of apoptosis, proliferation, and angiogenesis. PI3K and Akt are amplified or overexpressed in a number of malignancies, including sarcomas, ovarian cancer, multiple myeloma, and melanoma. This pathway regulates production of the potent angiogenic factor vascular endothelial growth factor (VEGF), and protects tumor cells against both chemotherapy and reactive oxygen-induced apoptosis through phosphorylation of substrates such as apoptotic peptidase-activating factor-1 (APAF-1), forkhead proteins, and caspase 9. Given its diverse actions, compounds that suppress the PI3K/Akt pathway have potential pharmacologic utility as angiogenesis inhibitors and antineoplastic agents. Using the SVR angiogenesis assay, a screen of natural products, we isolated the alkaloid solenopsin, and found that it is a potent angiogenesis inhibitor. We also found that solenopsin inhibits the PI3K signaling pathway in cells upstream of PI3K, which may underlie its affects on angiogenesis. Consistent with inhibition of the activation of PI3K, solenopsin prevented the phosphorylation of Akt and the phosphorylation of its substrate forkhead box 01a (FOXO1a), a member of the forkhead family of transcription factors. Interestingly, solenopsin also inhibited Akt-1 activity in an ATP-competitive manner in vitro without affecting 27 of 28 other protein kinases tested.


Subject(s)
Alkaloids/pharmacology , Neovascularization, Physiologic/drug effects , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Alkaloids/chemical synthesis , Alkaloids/chemistry , Animals , Ants , Cell Line , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Mice , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Protein Kinases/drug effects , Protein Kinases/metabolism , Zebrafish/embryology
4.
Bioorg Med Chem Lett ; 16(9): 2518-21, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16464575

ABSTRACT

Fluorescent analogs were synthesized of the potent PI 3-kinase inhibitors, wortmannin and demethoxyviridin. The esterification of 11-deacetylwortmannin, 17-hydroxywortmannin, and demethoxyviridin with the fluorescent carboxylic acids NBD-sarcosine and 7-dimethylaminocoumarin-4-acetic acid generated six novel fluorescent esters. Potent inhibition of PI 3-kinase-alpha was observed for the derivatives of 11-desacetylwortmannin and demethoxyviridin.


Subject(s)
Androstadienes/chemical synthesis , Androstenes/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fluorescent Dyes/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Androstadienes/chemistry , Androstadienes/pharmacology , Androstenes/chemistry , Androstenes/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Molecular Conformation , Molecular Probe Techniques , Stereoisomerism , Structure-Activity Relationship , Wortmannin
5.
J Biol Chem ; 280(19): 19298-305, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15737997

ABSTRACT

Mixed lineage kinase 7 (MLK7) is a mitogen-activated protein kinase kinase kinase (MAPKKK) that activates the pro-apoptotic signaling pathways p38 and JNK. A library of potential kinase inhibitors was screened, and a series of dihydropyrrolopyrazole quinolines was identified as highly potent inhibitors of MLK7 in vitro catalytic activity. Of this series, an aryl-substituted dihydropyrrolopyrazole quinoline (DHP-2) demonstrated an IC50 of 70 nM for inhibition of pJNK formation in COS-7 cell MLK7/JNK co-transfection assays. In stimulated cells, DHP-2 at 200 nM or MLK7 small interfering RNA completely blocked anisomycin and UV induced but had no effect on interleukin-1beta or tumor necrosis factor-alpha-induced p38 and JNK activation. Additionally, the compound blocked anisomycin and UV-induced apoptosis in COS-7 cells. Heart tissue homogenates from MLK7 transgenic mice treated with DHP-2 at 30 mg/kg had reduced JNK and p38 activation with no apparent effect on ERK activation, demonstrating that this compound can be used to block MLK7-driven MAPK pathway activation in vivo. Taken together, these data demonstrate that MLK7 is the MAPKKK required for modulation of the stress-activated MAPKs downstream of anisomycin and UV stimulation and that DHP-2 can be used to block MLK7 pathway activation in cells as well as in vivo.


Subject(s)
Anisomycin/antagonists & inhibitors , Anisomycin/chemistry , Cytokines/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Pyrazoles/pharmacology , Quinolines/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anisomycin/pharmacology , Apoptosis , Blotting, Western , COS Cells , Catalysis , DNA Fragmentation , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glutathione Transferase/metabolism , Humans , Inhibitory Concentration 50 , Interleukin-1/metabolism , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinases/metabolism , Mice , Models, Chemical , Muscle Proteins/metabolism , Myocardium/metabolism , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Plasmids/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemistry , Quinolines/chemistry , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors , Transfection , Transgenes , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays
6.
Int Immunopharmacol ; 5(3): 495-502, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15683846

ABSTRACT

Phosphoinositide 3-kinase (PI3K) exhibits broad functional effects in immune cells. We investigated the role of PI3K in allergic airway inflammation using LY294002, a specific PI3K inhibitor, in a mouse asthma model. BALB/c mice were sensitized and challenged with ovalbumin (OVA), and developed airway eosinophilia, mucus hypersecretion, elevation in cytokine levels, and airway hyperresponsiveness. Intratracheal administration of LY294002 significantly inhibited OVA-induced increases in total cell counts, eosinophil counts, and IL-5, IL-13, and eotaxin levels in bronchoalveolar lavage fluid. Histological studies show that LY294002 dramatically inhibited OVA-induced lung tissue eosinophilia and airway mucus production. In addition, LY294002 significantly suppressed OVA-induced airway hyperresponsiveness to inhaled methacholine. Western blot analysis of whole lung lysates shows that LY294002 markedly attenuated OVA-induced serine phosphorylation of Akt, a direct downstream substrate of PI3K. Taken together, our findings suggest that inhibition of PI3K signaling pathway can suppress T-helper type 2 (Th2) cytokine production, eosinophil infiltration, mucus production, and airway hyperresponsiveness in a mouse asthma model and may have therapeutic potential for the treatment of allergic airway inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Asthma/drug therapy , Chromones/pharmacology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Animals , Asthma/chemically induced , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Movement/drug effects , Cytokines/metabolism , Disease Models, Animal , Eosinophils/cytology , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Mucus/metabolism , Ovalbumin/immunology , Ovalbumin/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pulmonary Eosinophilia/drug therapy , Pulmonary Eosinophilia/pathology , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/drug therapy
7.
Anal Biochem ; 338(1): 136-42, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15707944

ABSTRACT

Protein kinases play important roles in many disease processes and are primary targets for drug development. Because cellular phosphorylation cascades are complex multidirectional pathways, the behavior of a drug in a biochemical enzyme assay may not accurately reflect its performance in the context of a whole cell. We have developed a near-infrared cytoblot assay that can be used to investigate both kinase signaling and effects of kinase inhibitors. Adherent cells were grown in either 96- or 384-well plates. Following stimulation, protein phosphorylation was detected immunohistochemically by simultaneous staining with two primary antibodies: a phospho-specific primary and normalization antibody that recognized either the target protein regardless of phosphorylation status (pan protein) or a housekeeping protein. Secondary antibodies labeled with two spectrally distinct near-infrared dyes were used for visualization. Nuclear staining with TO-PRO-3 was also used in place of the normalization antibody. Normalization for well-to-well variability was accomplished by ratiometric analysis of the two wavelengths. The near-infrared cytoblot was used to analyze phosphorylation of EGFR, Akt, Stat3, MEK 1, and ERK1/2. This assay format was also able to simultaneously assess the phosphorylation of multiple signaling proteins in response to known kinase inhibitors. We observed that the IC50 for the EGFR inhibitor PD168393 was similar for EGFR and Stat3 but was significantly higher for ERK1/2, a downstream modulator of EGFR function. The observation that the receptor and its effectors show different IC50 values for the same inhibitory drug could be important for target selection in drug development.


Subject(s)
Protein Kinases/analysis , Signal Transduction/physiology , Animals , Butadienes/pharmacology , Cells, Cultured , Chromones/pharmacology , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique, Direct/methods , Immunohistochemistry/methods , Mice , Morpholines/pharmacology , NIH 3T3 Cells , Nitriles/pharmacology , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor , Signal Transduction/drug effects , Spectrophotometry, Infrared , Trans-Activators/metabolism
9.
Crit Care Med ; 32(5 Suppl): S302-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15118535

ABSTRACT

OBJECTIVE: To explore whether the improvement in organ function and the vasoactive effect observed in the clinical studies of drotrecogin alfa (activated) (recombinant human activated protein C, rhAPC) in sepsis are a result of rhAPC's effect on endothelial cell (EC) permeability and modulation of the intracellular cytoskeleton via the Rho kinase signaling pathway. DESIGN: Findings regarding dose and duration of exposure to the drug with sequential addition of rhAPC and mediators (thrombin, histamine, interleukin-1 beta). SETTING: Research laboratory in a pharmaceutical company. SUBJECTS: Cultured primary human EC from different tissues and vascular beds. INTERVENTIONS: A monolayer of EC was incubated with either rhAPC, thrombin, histamine, or interleukin-1 beta alone or with rhAPC in combination with thrombin or interleukin-beta. The effect of rhAPC and mediators on EC permeability was monitored with measurement of electrical resistance. The effect on Rho kinase pathway signaling was monitored by the levels of phosphorylated myosin light chain and blockage with the Rho kinase specific inhibitor, Y27632. MEASUREMENTS AND MAIN RESULTS: Thrombin alone induced an early, concentration-dependent, and transient leakiness of EC. Interleukin-1 beta (0.5 ng/mL) induced an early, irreversible leakiness of EC. rhAPC (0.05-0.2 microg/mL, approximate median therapeutic blood levels) alone had no effect on EC permeability. rhAPC at > or=1 microg/mL induced an early EC leakage. rhAPC (0.19 microg/mL) attenuated the leakage induced by 0.5 ng/mL interleukin-1beta on microvascular EC derived from lung and skin and partially attenuated the leakage induced by 0.25 nM thrombin on human coronary arterial ECs. Levels of phosphorylated myosin light chain increased rapidly in human coronary arterial ECs when stimulated with thrombin or rhAPC (about 100-fold less potent) in a concentration-dependent manner via the Rho kinase signaling pathway. Short (5 mins) preconditioning of human coronary arterial ECs with 0.19 microg/mL rhAPC partially blocked the increase in phosphorylated myosin light chain levels induced by thrombin (0.06-0.2 nM). CONCLUSIONS: At concentrations exceeding physiologic and therapeutic levels, rhAPC increases EC permeability, an effect not seen at lower concentrations. The data suggest that interpretation of published in vitro and in vivo data of rhAPC and EC permeability should take into consideration the concentrations of rhAPC used or achieved. Other preliminary novel observations suggest that studying the effects of rhAPC on EC permeability and intracellular cytoskeletal organization may provide understanding of the effect of rhAPC on EC function.


Subject(s)
Anti-Infective Agents/pharmacology , Cell Membrane Permeability/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Protein C/pharmacology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/physiology , Recombinant Proteins/pharmacology , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins , Signal Transduction/drug effects , rho-Associated Kinases
10.
Am J Physiol Heart Circ Physiol ; 286(2): H796-805, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14563664

ABSTRACT

The phosphoinositide 3-kinase (PI3K) inhibitor LY-294002 decreased steady-state contraction in neonatal rat ventricular myocytes (NRVM). To determine whether the effect on steady-state contraction could be due to decreased intracellular Ca(2+) content, Ca(2+) content was assessed with fluorescent plate reader analysis by using the caffeine-releasable Ca(2+) stores as an index of sarcoplasmic reticulum (SR) Ca(2+) content. Caffeine-releasable Ca(2+) content was diminished in a dose-dependent manner with LY-294002, suggesting that the decrease in steady-state contraction was due to diminished intracellular Ca(2+) content. Activation of the L-type Ca(2+) channel by BAY K 8644 was attenuated by LY-294002, suggesting the effect of LY-294002 is to reduce Ca(2+) influx at this channel. To investigate whether additional proteins involved in excitation-contraction (EC) coupling are likewise regulated by PI3K activity, the effects of compounds acting at sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA2a), the ryanodine receptor, and the Na/Ca exchanger (NCX) were compared with LY-294002. Inhibition of SERCA2a by thapsigargin increased basal Ca(2+) levels in contrast to LY-294002, indicating that SERCA2a activity is sustained in the presence of LY-294002. Ryanodine decreased SR Ca(2+) content. The additive effect with coadministration of LY-294002 could be attributed to a decrease in Ca(2+) influx at the L-type Ca(2+) channel. The NCX inhibitor Ni(2+) was used to investigate whether the decrease in intracellular Ca(2+) content with LY-294002 could be due to inhibition of the NCX reverse-mode activity. The minimal effect of LY-294002 with Ni(2+) suggests that the primary effect of LY-294002 on EC coupling occurs through inhibition of PI3K-mediated L-type Ca(2+) channel activity.


Subject(s)
Calcium/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Phosphatidylinositol 3-Kinases/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Animals, Newborn , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/physiology , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Chromones/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Enzyme Inhibitors/pharmacology , Heart Ventricles/drug effects , Kinetics , Morpholines/pharmacology , Myocytes, Cardiac/enzymology , Phosphoinositide-3 Kinase Inhibitors , Piperazines/pharmacology , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/metabolism , Ventricular Function
11.
Am J Physiol Cell Physiol ; 284(6): C1645-53, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12606308

ABSTRACT

AlphaENaC-EGFP (enhanced green fluorescent protein-tagged alpha-subunit of the epithelial Na(+) channel) stably transfected clonal lines derived from the A6 parental cell line were used to study the physical mechanisms of insulin-stimulated Na(+) transport. Within 1 min of insulin stimulation, ENaC migrates from a diffuse cytoplasmic localization to the apical and lateral membranes. Concurrently, after insulin stimulation, phosphatidylinositol 3-kinase (PI 3-kinase) is colocalized with ENaC on the lateral but not apical membrane. An inhibitor of PI 3-kinase, LY-294002, does not inhibit ENaC/PI 3-kinase colocalization but does alter the intracellular site of the colocalization, preventing the translocation of ENaC to the lateral and apical membranes. These data show that insulin stimulation causes the migration of ENaC to the lateral and apical cell membranes and that this trafficking is dependent on PI 3-kinase activity.


Subject(s)
Biological Transport/physiology , Insulin/metabolism , Kidney/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sodium Channels/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Polarity , Chromones/metabolism , Electrophysiology , Enzyme Inhibitors/metabolism , Epithelial Sodium Channels , Kidney/cytology , Microscopy, Fluorescence , Morpholines/metabolism , Phosphoinositide-3 Kinase Inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium/metabolism , Sodium Channels/genetics , Xenopus laevis
13.
Cardiovasc Res ; 57(1): 139-46, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12504823

ABSTRACT

OBJECTIVE: Members of the protein kinase C (PKC) family are important mediators of cell signaling underlying multiple aspects of myocardial function. Activation of the betaII isoform of PKC is thought to be involved in the development of congestive heart failure. To investigate the biological effect of PKC-betaII, we measured gene expression of angiotensin converting enzyme (ACE) and angiotensin II (AngII) receptors AT(1A) and AT(1B) in cardiomyocytes overexpressing PKC-betaII. METHODS: An adenovirus construct expressing PKC-betaII was introduced into cultured neonatal rat ventricular myocytes (NRVMs). Western blot and in situ kinase assay was used to measure PKC-betaII level and activity in NRVMs. Real time quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis was used to measure the mRNA levels of several genes following PMA stimulation of either un-infected or ad-PKC-betaII infected cells. RESULTS: Our data show that activation of PKC-betaII in cardiomyocytes leads to elevated expression of angiotensin-converting enzyme (ACE) gene. Treatment of adeno-PKC-betaII infected cardiomyocytes with phorbol 12-myristate 13-acetate (PMA) resulted in an 8-fold increase of ACE mRNA expression, whereas ACE mRNA levels only increased around 2-fold in uninfected or adeno-GFP (green fluorescent protein) infected cardiomyocytes with similar PMA treatment. The induction of ACE mRNA was blocked by the PKC-beta-specific antagonist LY379196. No significant change of angiotensin II receptors AT1a and AT1b could be detected in the cardiomyocytes expressing PKC-betaII. CONCLUSION: These data indicate that ACE is a transcription target of PKC-betaII activation in cardiomyocytes, and also suggest a mechanism for the involvement of PKC in cardiac hypertrophy and fibrosis through increased activity of angiotensin converting enzyme in the myocardium.


Subject(s)
Isoenzymes/metabolism , Myocytes, Cardiac/enzymology , Peptidyl-Dipeptidase A/metabolism , Protein Kinase C/metabolism , Angiotensin I , Animals , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Expression , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Mesylates/pharmacology , Peptidyl-Dipeptidase A/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C beta , Pyrroles/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transduction, Genetic/methods
14.
Diabetes ; 51(9): 2709-18, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12196463

ABSTRACT

Protein kinase C (PKC) beta isoform activity is increased in myocardium of diabetic rodents and heart failure patients. Transgenic mice overexpressing PKCbeta2 (PKCbeta2Tg) in the myocardium exhibit cardiomyopathy and cardiac fibrosis. In this study, we characterized the expression of connective tissue growth factor (CTGF) and transforming growth factor beta (TGFbeta) with the development of fibrosis in heart from PKCbeta2Tg mice at 4-16 weeks of age. Heart-to-body weight ratios of transgenic mice increased at 8 and 12 weeks, indicating hypertrophy, and ratios did not differ at 16 weeks. Collagen VI and fibronectin mRNA expression increased in PKCbeta2Tg hearts at 4-12 weeks. Histological examination revealed myocyte hypertrophy and fibrosis in 4- to 16-week PKCbeta2Tg hearts. CTGF expression increased in PKCbeta2Tg hearts at all ages, whereas TGFbeta increased only at 8 and 12 weeks. In 8-week diabetic mouse heart, CTGF and TGFbeta expression increased two- and fourfold, respectively. Similarly, CTGF expression increased in rat hearts at 2-8 weeks of diabetes. This is the first report of increased CTGF expression in myocardium of diabetic rodents suggesting that cardiac injury associated with PKCbeta2 activation, diabetes, or heart failure is marked by increased CTGF expression. CTGF could act independently or together with other cytokines to induce cardiac fibrosis and dysfunction.


Subject(s)
Cardiomyopathies/metabolism , Diabetes Mellitus, Experimental/enzymology , Growth Substances/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Isoenzymes/metabolism , Myocardium/metabolism , Protein Kinase C/metabolism , Animals , Cardiomyopathies/pathology , Connective Tissue Growth Factor , Enzyme Activation , Extracellular Matrix/metabolism , Fibrosis , Heart Ventricles , Immunologic Techniques , Male , Mice , Mice, Transgenic , Myocardium/pathology , Protein Kinase C beta , Rats , Staining and Labeling , Transforming Growth Factor beta/metabolism
15.
Circulation ; 105(3): 373-9, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11804995

ABSTRACT

BACKGROUND: Inadequate angiogenic response to ischemia in the myocardium of diabetic patients could result in poor collateral formation. Yet, excessive neovascularization in the retina causes proliferative diabetic retinopathy. Since vascular endothelial growth factor (VEGF) is the major angiogenic factor expressed in response to hypoxia, we have characterized expression of VEGF and its receptors in retina, renal glomeruli, aorta, and myocardium in insulin-resistant and diabetic states. Methods and Results- The expression of mRNA and protein for VEGF and its receptors, VEGF-R1 and VEGF-R2, in the myocardium was decreased significantly by 40% to 70% in both diabetic and insulin-resistant nondiabetic rats. Twofold reductions in VEGF and VEGF-R2 were observed in ventricles from diabetic patients compared with nondiabetic donors. In contrast, expression of VEGF and its receptors were increased 2-fold in retina and glomeruli from diabetic or insulin-resistant rats. Insulin treatment of diabetic rats normalized changes in both cardiac and microvascular tissues. Insulin increased VEGF mRNA expression in cultured rat neonatal cardiac myocytes. CONCLUSIONS: The results documented for the first time that differential regulation of VEGF and its receptors exist between microvascular and cardiac tissues, which can be regulated by insulin. These results provide a potential explanation for concomitant capillary leakage and neovascularization in the retina and inadequate collateral formation in the myocardium of insulin-resistant and diabetic patients.


Subject(s)
Diabetes Mellitus/metabolism , Endothelial Growth Factors/metabolism , Insulin Resistance , Lymphokines/metabolism , Myocardium/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Animals , Cells, Cultured , Collateral Circulation , Coronary Circulation , Diabetes Mellitus/blood , Endothelial Growth Factors/genetics , Female , Gene Expression Regulation/drug effects , Humans , Insulin/pharmacology , Kidney Glomerulus/metabolism , Lymphokines/genetics , Male , Middle Aged , Neovascularization, Pathologic , Obesity , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Retina/metabolism , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL