Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Antioxid Redox Signal ; 14(4): 543-58, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20649491

ABSTRACT

Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme in the pentose phosphate pathway and a major source of nicotinamide adenine dinucleotide phosphate reduced (NADPH), which regulates numerous enzymatic (including glutathione reductase and NADPH oxidase that, respectively, generates reduced glutathione and reactive oxygen species) reactions involved in various cellular actions, yet its physiological function is seldom investigated. We, however, recently showed that inhibiting G6PD causes precontracted coronary artery (CA) to relax in an endothelium-derived relaxing factor- and second messenger-independent manner. Here we assessed the role of G6PD in regulating CA contractility. Treating bovine CAs for 20 min with potassium chloride (KCl; 30 mM), amphotericin B (50 µM), or U46619 (100 nM) significantly (p < 0.05) increased both G6PD activity and glucose flux through the pentose phosphate pathway. The effect was Ca(2+) independent, and there was a corresponding increase in protein kinase C (PKC) activity. Activation of G6PD by KCl was blocked by the PKCδ inhibitor rottlerin (10 µM) or by knocking down PKCδ expression using siRNA. Phorbol 12, 13-dibutyrate (10 µM), a PKC activator, significantly increased G6PD phosphorylation and activity, whereas single (S210A, T266A) and double (S210A/T266A) mutations at sites flanking the G6PD active site significantly inhibited phosphorylation, shifted the isoelectric point, and reduced enzyme activity. Knocking down G6PD decreased NADPH and reactive oxygen species generation, and reduced KCl-evoked increases in [Ca(2+)](i) and myosin light chain phosphorylation, thereby reducing CA contractility. Similarly, aortas from G6PD-deficient mice developed less KCl/phorbol 12, 13-dibutyrate-evoked force than those from their wild-type littermates. Conversely, overexpression of G6PD augmented KCl-evoked increases in [Ca(2+)](i), thereby augmenting CA contraction. Our findings demonstrate that G6PD activity and NADPH is increased in activated CA in a PKCδ-dependent manner and that G6PD modulates Ca(2+) entry and CA contractions evoked by membrane depolarization.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Glucosephosphate Dehydrogenase/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Smooth, Vascular/metabolism , Phosphorylation
2.
J Biol Chem ; 285(25): 19561-71, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20363753

ABSTRACT

Hypoxic pulmonary vasoconstriction (HPV) is a physiological response to a decrease in airway O(2) tension, but the underlying mechanism is incompletely understood. We studied the contribution of glucose-6-phosphate dehydrogenase (Glc-6-PD), an important regulator of NADPH redox and production of reactive oxygen species, to the development of HPV. We found that hypoxia (95% N(2), 5% CO(2)) increased contraction of bovine pulmonary artery (PA) precontracted with KCl or serotonin. Depletion of extracellular glucose reduced NADPH, NADH, and HPV, substantiating the idea that glucose metabolism and Glc-6-PD play roles in the response of PA to hypoxia. Our data also show that inhibition of glycolysis and mitochondrial respiration (indicated by an increase in NAD(+) and decrease in the ATP-to-ADP ratio) by hypoxia, or by inhibitors of pyruvate dehydrogenase or electron transport chain complexes I or III, increased generation of reactive oxygen species, which in turn activated Glc-6-PD. Inhibition of Glc-6-PD decreased Ca(2+) sensitivity to the myofilaments and diminished Ca(2+)-independent and -dependent myosin light chain phosphorylation otherwise increased by hypoxia. Silencing Glc-6-PD expression in PA using a targeted small interfering RNA abolished HPV and decreased extracellular Ca(2+)-dependent PA contraction increased by hypoxia. Similarly, Glc-6-PD expression and activity were significantly reduced in lungs from Glc-6-PD(mut(-/-)) mice, and there was a corresponding reduction in HPV. Finally, regression analysis relating Glc-6-PD activity and the NADPH-to-NADP(+) ratio to the HPV response clearly indicated a positive linear relationship between Glc-6-PD activity and HPV. Based on these findings, we propose that Glc-6-PD and NADPH redox are crucially involved in the mechanism of HPV and, in turn, may play a key role in increasing pulmonary arterial pressure, which is involved in the development of pulmonary hypertension.


Subject(s)
Enzyme Activation , Glucosephosphate Dehydrogenase/metabolism , Hypoxia , Pulmonary Artery/enzymology , Vasoconstriction , Animals , Blood Pressure , Calcium/metabolism , Cattle , Glucose/metabolism , Lung/pathology , NADP/metabolism , Oxidation-Reduction , Phosphorylation
3.
Am J Physiol Heart Circ Physiol ; 297(1): H153-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19429815

ABSTRACT

Increased oxidative stress is a known cause of cardiac dysfunction in animals and patients with diabetes, but the sources of reactive oxygen species [e.g., superoxide anion (O(2)(-))] and the mechanisms underlying O(2)(-) production in diabetic hearts are not clearly understood. Our aim was to determine whether NADPH oxidase (Nox) is a source of O(2)(-) and whether glucose-6-phosphate dehydrogenase (G6PD)-derived NADPH plays a role in augmenting O(2)(-) generation in diabetes. We assessed cardiac function, Nox and G6PD activities, NADPH levels, and the activities of antioxidant enzymes in heart homogenates from young (9-11 wk old) Zucker lean and obese (fa/fa) rats. We found that myocardial G6PD activity was significantly higher in fa/fa than in lean rats, whereas superoxide dismutase and glutathione peroxidase activities were decreased (P < 0.05). O(2)(-) levels were elevated (70-90%; P < 0.05) in the diabetic heart, and this elevation was blocked by the Nox inhibitor gp-91(ds-tat) (50 microM) or by the mitochondrial respiratory chain inhibitors antimycin (10 microM) and rotenone (50 microM). Inhibition of G6PD by 6-aminonicotinamide (5 mM) and dihydroepiandrosterone (100 microM) also reduced (P < 0.05) O(2)(-) production. Notably, the activities of Nox and G6PD in the fa/fa rat heart were inhibited by chelerythrine, a protein kinase C inhibitor. Although we detected no changes in stroke volume, cardiac output, or ejection fraction, left ventricular diameter was slightly increased during diastole and systole, and left ventricular posterior wall thickness was decreased during systole (P < 0.05) in Zucker fa/fa rats. Our findings suggest that in a model of severe hyperlipidema and hyperglycemia Nox-derived O(2)(-) generation in the myocardium is fueled by elevated levels of G6PD-derived NADPH. Similar mechanisms were found to activate O(2)(-) production and induce endothelial dysfunction in aorta. Thus G6PD may be a useful therapeutic target for treating the cardiovascular disease associated with type 2 diabetes, if second-generation drugs specifically reducing the activity of G6PD to near normal levels are developed.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Hyperglycemia/metabolism , Mitochondria, Heart/metabolism , NADPH Oxidases/metabolism , NADP/metabolism , Obesity/metabolism , Superoxides/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/physiopathology , Fatty Acids, Nonesterified/blood , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , Glucosephosphate Dehydrogenase/biosynthesis , Glucosephosphate Dehydrogenase/genetics , Glutathione/metabolism , Heart Diseases/etiology , Heart Diseases/physiopathology , Hydrogen Peroxide/metabolism , Hyperglycemia/genetics , Insulin/blood , Isometric Contraction/physiology , Mitochondria, Heart/enzymology , Myocardium/enzymology , Myocardium/metabolism , Obesity/genetics , Rats , Rats, Zucker , Triglycerides/blood , Up-Regulation
4.
Free Radic Biol Med ; 47(3): 219-28, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19230846

ABSTRACT

Glucose metabolism through the glycolysis and hexosamine pathway has been shown to be altered in type 2 diabetes. However, the fate of glucose through the pentose phosphate pathway (PPP) is currently unclear. In this study, we determined whether the activity of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme in the PPP, is modulated in the liver of Zucker obese fa/fa rats (9-11 weeks of age). We found that G6PD expression and activity, NADPH levels, and 6-phosphogluconate generation were significantly increased in the liver of fa/fa rats. Inhibition of PI3 kinase and Src kinases decreased (p < 0.05) G6PD activity in the fa/fa but not in the lean rat liver, suggesting that G6PD activity is regulated by PI3/Src kinase signaling pathways. G6PD-derived NADPH increased (p < 0.05) superoxide anion levels by 70-90% in fa/fa vs lean rat liver, which was inhibited by the NADPH oxidase inhibitor gp91(ds-tat) (50 microM) and G6PD inhibitors 6-aminonicotinamide (1 mM) and dehydroepiandrosterone (100 microM), therefore indicating that elevated G6PD activity may be responsible for mediating superoxide generation. Interestingly, we also found a positive correlation between liver hypertrophy/increased G6PD activity (r2 = 0.77; p = 0.0009) and liver hypertrophy/superoxide production (r2 = 0.51; p = 0.0091) in fa/fa rats. Increased G6PD and NADPH oxidase expression and activity, in young hyperglycemic and hyperinsulinemic rats before the development of diabetes, seems to be a contributing factor in the induction of oxidative stress. Because inhibition of G6PD activity decreases oxidative stress, we conclude that G6PD behaves as a pro-oxidant in the fa/fa rat liver in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Glucosephosphate Dehydrogenase/metabolism , Liver/enzymology , NADPH Oxidases/metabolism , src-Family Kinases/metabolism , 6-Aminonicotinamide/pharmacology , Animals , Cell Extracts , Cells, Cultured , Dehydroepiandrosterone/pharmacology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gluconates/pharmacology , Glucosephosphate Dehydrogenase/genetics , Glycoproteins/pharmacology , Liver/drug effects , Liver/pathology , Male , NADPH Oxidases/genetics , Obesity , Pentose Phosphate Pathway/drug effects , Perfusion , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Zucker , Signal Transduction , Superoxides/metabolism , Up-Regulation/drug effects
5.
J Card Fail ; 13(6): 497-506, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17675065

ABSTRACT

BACKGROUND: We previously found that higher NADPH levels produced by glucose-6-phosphate dehydrogenase (G6PD) can enhance myocardial superoxide generation by NAD(P)H oxidase in a dog model of dilated cardiomyopathy. Therefore, we tested whether G6PD activity is elevated and enhances NADPH level and increases NAD(P)H oxidase-derived superoxide production in the myocardium from patients with heart failure from ischemic cardiomyopathy. METHODS AND RESULTS: Surgical discards of left ventricle were collected from 8 congestive heart failure patients undergoing surgical ventricular restoration procedures, whereas control left ventricle tissue was obtained from 5 normal donor hearts deemed not suitable for transplantation. Biochemical assays were performed in tissue homogenates. We found that superoxide and hydrogen peroxide were elevated, respectively, by 9- and 3-fold in failing versus normal hearts (P < .05). The NAD(P)H oxidase inhibitors gp91(ds-tat), apocynin, and diphenyleneiodonium, significantly inhibited superoxide generation by approximately 75%, 89%, and 91%, respectively. Superoxide production by NAD(P)H oxidase increased 10- and 3-fold by adding NADPH (100 micromol/L) and NADH (100 micromol/L), respectively, in a DPI- and gp91(ds-tat)-inhibitable manner. Interestingly, chelerythrine, a PKC inhibitor, and PP2, a Src kinase family inhibitor, reduced G6PD activity (0.29 +/- 0.04 nM x min x mg protein) by 50% and 51% and these inhibitors also decreased myocardial superoxide by 99% and 79%, respectively. Furthermore, 6-aminonicotinamide, a G6PD inhibitor, decreased myocardial superoxide production by 71%. CONCLUSIONS: These data suggest that high NAD(P)H oxidase, fueled by G6PD-derived NADPH, generates most of the superoxide in failing hearts of patients with ischemic cardiomyopathy. In addition, PKC-Src kinase signaling pathways seem to coordinate the activation of both G6PD and NAD(P)H oxidase in human cardiac muscle.


Subject(s)
Glucosephosphate Dehydrogenase/biosynthesis , Heart Failure/enzymology , Myocardium/enzymology , NADPH Oxidases/biosynthesis , Oxidative Stress/physiology , Up-Regulation/physiology , Biomarkers/metabolism , Blotting, Western , Disease Progression , Female , Heart Ventricles/enzymology , Humans , Hydrogen Peroxide/metabolism , Luminescent Measurements , Male , Middle Aged , Prognosis , Severity of Illness Index , Superoxides/metabolism
6.
J Mol Cell Cardiol ; 41(2): 340-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16828794

ABSTRACT

In the failing heart, NADPH oxidase and uncoupled NO synthase utilize cytosolic NADPH to form superoxide. NADPH is supplied principally by the pentose phosphate pathway, whose rate-limiting enzyme is glucose 6-phosphate dehydrogenase (G6PD). Therefore, we hypothesized that cardiac G6PD activation drives part of the excessive superoxide production implicated in the pathogenesis of heart failure. Pacing-induced heart failure was performed in eight chronically instrumented dogs. Seven normal dogs served as control. End-stage failure occurred after 28 +/- 1 days of pacing, when left ventricular end-diastolic pressure reached 25 mm Hg. In left ventricular tissue homogenates, spontaneous superoxide generation measured by lucigenin (5 microM) chemiluminescence was markedly increased in heart failure (1338 +/- 419 vs. 419 +/- 102 AU/mg protein, P < 0.05), as were NADPH levels (15.4 +/- 1.5 vs. 7.5 +/- 1.5 micromol/gww, P < 0.05). Superoxide production was further stimulated by the addition of NADPH. The NADPH oxidase inhibitor gp91(ds-tat) (50 microM) and the NO synthase inhibitor L-NAME (1 mM) both significantly lowered superoxide generation in failing heart homogenates by 80% and 76%, respectively. G6PD was upregulated and its activity higher in heart failure compared to control (0.61 +/- 0.10 vs. 0.24 +/- 0.03 nmol/min/mg protein, P < 0.05), while superoxide production decreased to normal levels in the presence of the G6PD inhibitor 6-aminonicotinamide. We conclude that the activation of myocardial G6PD is a novel mechanism that enhances NADPH availability and fuels superoxide-generating enzymes in heart failure.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Heart Failure/enzymology , NADP/biosynthesis , Superoxides/metabolism , 6-Aminonicotinamide/pharmacology , Animals , Blood Pressure , Cardiac Pacing, Artificial/adverse effects , Disease Models, Animal , Dogs , Enzyme Inhibitors/pharmacology , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Heart Failure/etiology , Heart Failure/physiopathology , Humans , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Teratogens/pharmacology , Time Factors
7.
Cell Cycle ; 5(6): 653-60, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16582606

ABSTRACT

We have previously demonstrated that the nuclear transport of the second subunit of the Replication Factor C complex, RFC40, by the regulatory subunit, RIalpha, of PKA is cell cycle specific and impairment in this transport results in G(1) arrest. In this study, we have investigated whether the cyclin-dependent kinases play a role in regulating the RIalpha-RFC40 complex formation. In this context, we have identified RIalpha as a novel substrate for the G(1)/S-Cyclin-dependent kinase, CDK2/Cyclin E, and found that RIalpha is specifically phosphorylated at the serine residue. Treatment of MCF7 cells with a CDK inhibitor, olomoucine, resulted in a significant accumulation in the RIalpha-RFC40 complex by 3.10 +/- 0.08 fold and a parallel decrease in the RFC40-37 complex formation by 73.73 +/- 11.81%. Furthermore, in vitro phosphorylation experiments suggest that, phosphorylation of RIalpha by CDK2/CyclinE kinase promotes the dissociation of the RIalpha-RFC40 complex and that once RIalpha is phosphorylated it cannot complex with RFC40. Inhibition of the serine-threonine phosphatase, PP1, by Calyculin A, significantly reduced the RIalpha-RFC40 complex formation, substantiating the in vitro phosphorylation data. Taken together, these findings suggest that CDK2/Cyclin E may function as downstream modulator that regulates the dissociation of the RIalpha-RFC40 complex and subsequently the association of the RFC40-RFC37 complex.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Replication Protein C/metabolism , Active Transport, Cell Nucleus/genetics , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Humans , Phosphorylation/drug effects
8.
Exp Cell Res ; 312(6): 796-806, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16413017

ABSTRACT

We have previously shown that the regulatory subunit of PKA, RIalpha, functions as a nuclear transport protein for the second subunit of the replication factor C complex, RFC40, and that this transport appears to be crucial for cell cycle progression from G1 to S phase. In this study, we found that N(6)-monobutyryl cAMP significantly up-regulates the expression of RFC40 mRNA by 1.8-fold and its endogenous protein by 2.3-fold with a subsequent increase in the RIalpha-RFC40 complex formation by 3.2-fold. Additionally, the nuclear to cytoplasmic ratio of RFC40 increased by 26% followed by a parallel increase in the percentage of S phase cells by 33%. However, there was reduction in the percentage of G1 cells by 16% and G2/M cells by 43% with a concurrent accumulation of cells in S phase. Interestingly, the higher percentage of S phase cells did not correlate with a parallel increase in DNA replication. Moreover, although cAMP did not affect the expression of the other RFC subunits, there was a significant decrease in the RFC40-37 complex formation by 81.3%, substantiating the decrease in DNA replication rate. Taken together, these findings suggest that cAMP functions as an upstream modulator that regulates the expression and nuclear translocation of RFC40.


Subject(s)
Cell Nucleus/metabolism , Cyclic AMP/physiology , Replication Protein C/genetics , Replication Protein C/metabolism , Active Transport, Cell Nucleus/physiology , Cell Line, Tumor , Cell Nucleus/drug effects , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Replication Protein C/drug effects , S Phase/drug effects , Transcription, Genetic/genetics , Tumor Cells, Cultured , Up-Regulation
9.
Cancer Biol Ther ; 4(4): 429-37, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15846072

ABSTRACT

The regulatory subunit (RIalpha) of cAMP-dependent Protein Kinase A (PKA) is overexpressed in a variety of tumors and carcinomas such as renal cell carcinomas, pituitary tumors of the rat, malignant osteoblasts, colon carcinomas, serous ovarian tumors and primary human breast carcinomas. However, the direct relation between overexpression of RIalpha and malignancy is still unclear. We have recently identified a novel interaction between RIalpha and RFC40, the second subunit of Replication Factor C (RFC), and have demonstrated that this interaction may be associated with cell survival. Coincidentally, RFC40 is overexpressed in gestational trophoblastic diseases such as choriocarcinomas. This study was undertaken to investigate a possible functional role for both these proteins together, in DNA replication and cellular proliferation. In the course of this study, a nonconventional nuclear localization signal was identified for RIalpha. Nuclear transport of RFC40 was found to be dependent on RIalpha, and this transport appeared to be a crucial step for cell cycle progression from G1 to S phase. Impairment in the nuclear transport of RFC40 by RIalpha arrested cells in G1 phase. These findings provide evidence for a previously unknown mechanism for the nuclear transport of RFC40 and also for a novel mechanism for cellular proliferation.


Subject(s)
Active Transport, Cell Nucleus/physiology , Biological Transport/physiology , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/metabolism , Replication Protein C/metabolism , Amino Acid Sequence , Breast Neoplasms/pathology , Cell Cycle , Cell Line , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Down-Regulation , Female , G1 Phase , Humans , Molecular Sequence Data , Nuclear Localization Signals/chemistry , S Phase , Sequence Homology, Amino Acid , Tumor Cells, Cultured
10.
Cell Cycle ; 4(2): 323-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15655353

ABSTRACT

Replication Factor C (RFC) is required for the loading of Proliferating Cell Nuclear Antigen (PCNA) onto DNA during DNA replication, repair and recombination. RFC40, the second subunit of the RFC complex, and PCNA have been shown to be overexpressed in gestational trophoblastic diseases. Using RFC40 as the bait in a yeast two-hybrid screening, we have identified a novel interaction between RFC40 and the regulatory subunit (RIalpha) of cAMP-dependent Protein kinase A (PKA). The interaction sites between these two proteins were investigated and mapped to the N-terminus of RIalpha and the C-terminus of RFC40. Moreover, it was demonstrated that the C-subunit of PKA was not associated with the RFC40-RIalpha complex. Furthermore, RFC37, the third subunit of the RFC complex, competes with RIalpha and displaces it from the RFC40-RIalpha complex. Interestingly, downregulation of endogenous RIalpha by 8-chloro cAMP, in MCF7 breast cancer cells led to reduction in the amount of RFC40-RIalpha complex, together with decrease in cell survival.


Subject(s)
Cell Survival/physiology , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/physiology , Multiprotein Complexes/physiology , Replication Protein C/chemistry , Replication Protein C/physiology , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/genetics , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , Immunoprecipitation , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Interaction Mapping , Replication Protein C/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...