Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 660
Filter
1.
Cell Stress Chaperones ; 22(3): 371-376, 2017 05.
Article in English | MEDLINE | ID: mdl-28321764

ABSTRACT

The objective of the study was to evaluate the neuroprotective effects of bacoside A and bromelain against dichlorvos-incited toxicity. Healthy 6-8-week old, male Swiss mice were administered subacute doses of dichlorvos (40 mg/kg bw), bacoside A (5 mg/kg bw) and bromelain (70 mg/kg bw). AChE, BChE, GABA, serotonin and total protein content and their expressions were used for determination of toxic action of dichlorvos. Protective effects of bacoside A and bromelain were evaluated on the same parameters. Exposure to dichlorvos leads to significant decline in activities of AChE (p < 0.01, p < 0.001), BChE (p < 0.05) and GABA (p < 0.01) and total protein levels (p < 0.01). Antioxidant treatment significantly increased the activities of AChE (p < 0.01, p < 0.001), BChE (p < 0.05), GABA (p < 0.01) and total protein level (p < 0.05) compared to those in dichlorvos-treated mice. Overexpression of Hsp 70 protein and underexpression of phosphorylase a and b, catalase SOD and GPx were observed after dichlorvos exposure which suggests the oxidative stress. The results indicate that dichlorvos-induced neuronal damage which results in the generation of molecular expression of proteins is in agreement with the biochemical data ameliorated by bacoside A and bromelain.


Subject(s)
Brain/drug effects , Cholinesterases/metabolism , Dichlorvos/toxicity , Protective Agents/pharmacology , Serotonin/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Antioxidants/pharmacology , Brain/metabolism , Bromelains/pharmacology , Catalase/metabolism , Down-Regulation/drug effects , HSP70 Heat-Shock Proteins/metabolism , Male , Mice , Oxidative Stress/drug effects , Phosphorylase a/metabolism , Phosphorylase b/metabolism , Saponins/pharmacology , Superoxide Dismutase/metabolism , Triterpenes/pharmacology , Up-Regulation/drug effects
2.
Carbohydr Polym ; 106: 209-16, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24721070

ABSTRACT

A series of linear glucan saccharides (GS) with defined quantity and degree of polymerization (DP) were synthesized from α-d-glucose 1-phosphate (α-d-Glc 1-P) by phosphorylase-a. The GS product fractions with average DP 11, 22, 38, 52, 60, 70, and 79 were measured by HPSEC-ELSD system. Then the same seven fractions were resolved into individual peaks with DP: 6-14, 10-32, 27-55, 37-67, 44-75, 49-83 and 53-89 by HPAEC-PAD system. Results showed that measurement of α-d-Glc 1-P amount consuming during GS synthesis by both systems enable calculation of reaction yield. The reaction yield for the 24h biosynthesis of the GS product was 25.3% (measured by HPSEC-ELSD) or 29.1% (measured by HPAEC-PAD). The HPSEC-ELSD and HPAEC-PAD systems were also successfully used for phosphorylase-a activity measurement in order to perform its kinetic characterization. This study established feasible systems for preparation of various sizes of the GS with defined DP and quantity as well as characterization of phosphorylase-a kinetics.


Subject(s)
Glucans/chemical synthesis , Glucosephosphates/chemistry , Phosphorylase a/chemistry , Polymers/chemical synthesis , Polysaccharides/chemistry , Animals , Calibration , Chromatography, Gel/methods , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange/methods , Glucans/isolation & purification , Nuclear Magnetic Resonance, Biomolecular , Polymerization
3.
PLoS One ; 8(2): e50479, 2013.
Article in English | MEDLINE | ID: mdl-23418414

ABSTRACT

Folliculogenesis is tightly controlled by a series of hormones, growth factors and cytokines, many of which are secreted as proproteins and require processing by proteases before becoming functional. Furin is a member of the subtilisin-like proteases that activate large numbers of proprotein substrates and is ubiquitously expressed and implicated in many physiological and pathological processes. However, the precise role of furin during folliculogenesis has not been thoroughly investigated. The goal of the present work is to identify the role of furin in the development of granulosa cells during folliculogenesis, using immunohistochemistry, RT-PCR, Western blot and functional studies in primary cultured rat granulosa cells. Our results demonstrate that furin is highly expressed in granulosa cells and oocytes of the ovary with very limited expression in other ovarian cells such as the epithelial, stromal or theca cells. Furin siRNA significantly increases apoptosis of the granulosa cells from large antral/preovulatory follicles, in part via downregulation of the anti-apoptotic proteins, XIAP and p-AKT. On the contrary, furin siRNA markedly decreases proliferation of granulosa cells based on the downregulation of proliferation cell nuclear antigen (PCNA). Taken together, these data suggest that furin may play an important role in regulating apoptosis and proliferation of granulosa cells.


Subject(s)
Apoptosis/physiology , Cell Proliferation , Furin/metabolism , Granulosa Cells/physiology , Ovary/metabolism , Animals , Cells, Cultured , Down-Regulation , Female , Furin/genetics , Granulosa Cells/cytology , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/cytology , Phosphorylase a , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , X-Linked Inhibitor of Apoptosis Protein/metabolism
4.
Blood ; 118(1): 156-66, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21527517

ABSTRACT

JAK-STAT signaling is involved in the regulation of cell survival, proliferation, and differentiation. JAK tyrosine kinases can be transiently activated by cytokines or growth factors in normal cells, whereas they become constitutively activated as a result of mutations that affect their function in tumors. Specifically, the JAK2V617F mutation is present in the majority of patients with myeloproliferative disorders (MPDs) and is implicated in the pathogenesis of these diseases. In the present study, we report that the kinase CK2 is a novel interaction partner of JAKs and is essential for JAK-STAT activation. We demonstrate that cytokine-induced activation of JAKs and STATs and the expression of suppressor of cytokine signaling 3 (SOCS-3), a downstream target, are inhibited by CK2 small interfering RNAs or pharmacologic inhibitors. Endogenous CK2 is associated with JAK2 and JAK1 and phosphorylates JAK2 in vitro. To extend these findings, we demonstrate that CK2 interacts with JAK2V617F and that CK2 inhibitors suppress JAK2V617F autophosphorylation and downstream signaling in HEL92.1.7 cells (HEL) and primary cells from polycythemia vera (PV) patients. Furthermore, CK2 inhibitors potently induce apoptosis of HEL cells and PV cells. Our data provide evidence for novel cross-talk between CK2 and JAK-STAT signaling, with implications for therapeutic intervention in JAK2V617F-positive MPDs.


Subject(s)
Casein Kinase II/metabolism , Hematologic Neoplasms/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Polycythemia Vera/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Mice , Phosphorylase a/physiology , Polycythemia Vera/drug therapy , Polycythemia Vera/pathology
5.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R661-71, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042684

ABSTRACT

The transcription factor hypoxia-inducible factor-1 (HIF-1) plays an essential role in regulating gene expression in response to hypoxia-ischemia. Ischemia causes the tissue not only to be hypoxic but also to be hypothermic because of the hypoperfusion under certain circumstances. On the other hand, the induced hypothermia is one of the most common therapeutic modalities to extend tolerance to hypoxia. Although hypoxia elicits a variety of cellular and systemic responses at different organizational levels in the body, little is known about how hypoxia-induced responses are affected by low temperature. We examined the influence of mild hypothermic conditions (28-32 degrees C) on HIF-1 in both in vitro and in vivo settings. In vitro experiments adopting cultured cells elucidated that hypoxia-induced HIF-1 activation was resistant to 4-h exposure to the low temperature. In contrast, exposure to the low temperature as long as 24 h suppressed HIF-1 activation and the subsequent upregulation of HIF-1 target genes such as VEGF or GLUT-1. HIF-1alpha protein stability in the cell was not affected by hypothermic treatment. Furthermore, intracellular ATP content was reduced under 1% O(2) conditions but was not largely affected by hypothermic treatment. The evidence indicates that reduction of oxygen consumption is not largely involved in suppression of HIF-1. In addition, we demonstrated that HIF-1 DNA-binding activity and HIF-1-dependent gene expressions induced under 10% O(2) atmosphere in mouse brain were not influenced by treatment under 3-h hypothermic temperature but were inhibited under 5-h treatment. On the other hand, we indicated that warming ischemic legs of mice for 24 h preserved HIF-1 activity. In this report we describe for the first time that persisting low temperature significantly reduced HIF-1alpha neosynthesis under hypoxic conditions, leading to a decrease in gene expression for adaptation to hypoxia in both in vitro and in vivo settings.


Subject(s)
Hypothermia/genetics , Hypothermia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Brain/physiology , Brain Neoplasms , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation/physiology , Glioblastoma , Hindlimb/blood supply , Hindlimb/physiology , Humans , Hypoxia/genetics , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/genetics , Ischemia/physiopathology , Male , Mice , Mice, Inbred BALB C , Phosphorylase a/physiology , Ribosomal Protein S6/metabolism , Severity of Illness Index , Temperature
6.
J Biochem ; 146(1): 71-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19279194

ABSTRACT

The action of glycogen phosphorylase (GP) is essentially reversible, although GP is generally classified as a glycogen-degrading enzyme. In this study, we developed a highly sensitive and convenient assay for GP activity by analysing its chain-lengthening action on a fluorogenic maltooligosaccharide derivative in a glucose-1-phosphate-rich medium. Characterization of the substrate specificity of GP using pyridylaminated (PA-) maltooligosaccharides of various sizes revealed that a maltotetraosyl (Glc(4)) residue comprising the non-reducing-end of a PA-maltooligosaccharide is indispensable for the chain-lengthening action of GP, and PA-maltohexaose is the most suitable substrate for the purpose of this study. By using a high-performance liquid chromatograph equipped with a fluorescence spectrophotometer, PA-maltoheptaose produced by the chain elongation of PA-maltohexaose could be isolated and quantified at 10 fmol. This method was used to measure the GP activities of crude and purified GP preparations, and was demonstrated to have about 1,000 times greater sensitivity than the spectrophotometric orthophosphate assay.


Subject(s)
Chromatography, High Pressure Liquid/methods , Fluorescent Dyes/metabolism , Glycogen Phosphorylase/metabolism , Maltose/analogs & derivatives , Oligosaccharides/metabolism , Aminopyridines , Animals , Fluorescent Dyes/chemical synthesis , Glucosephosphates/metabolism , Glycogen Phosphorylase/blood , Glycogen Phosphorylase, Muscle Form/metabolism , Humans , Liver/enzymology , Liver Extracts/metabolism , Muscle, Skeletal/enzymology , Oligosaccharides/isolation & purification , Phosphorylase a/metabolism , Phosphorylase b/metabolism , Phosphorylation , Rabbits , Sensitivity and Specificity , Substrate Specificity , Swine , Tissue Extracts/metabolism
7.
Am J Vet Res ; 69(6): 818-23, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18518664

ABSTRACT

OBJECTIVE: To determine insulin sensitivity, proportions of muscle fiber types, and activities of glycogenolytic and glycolytic enzymes in Belgians with and without polysaccharide storage myopathy (PSSM). ANIMALS: 10 Quarter Horses (QHs) and 103 Belgians in which PSSM status had been determined. PROCEDURES: To determine insulin sensitivity, a hyperinsulinemic euglycemic clamp (HEC) technique was used in 5 Belgians with PSSM and 5 Belgians without PSSM. Insulin was infused i.v. at 3 mU/min/kg for 3 hours, and concentrations of blood glucose and plasma insulin were determined throughout. An i.v. infusion of glucose was administered to maintain blood glucose concentration at 100 mg/dL. Activities of glycogenolytic and glycolytic enzymes were assessed in snap-frozen biopsy specimens of gluteus medius muscle obtained from 4 Belgians with PSSM and 5 Belgians without PSSM. Percentages of type 1, 2a, and 2b muscle fibers were determined via evaluation of >or= 250 muscle fibers in biopsy specimens obtained from each Belgian used in the aforementioned studies and from 10 QHs (5 with PSSM and 5 without PSSM). RESULTS: Belgians with and without PSSM were not significantly different with respect to whole-body insulin sensitivity, muscle activities of glycogenolytic and glycolytic enzymes, or proportions of muscle fiber types. However, Belgians had an increased proportion of type 2a and decreased proportion of type 2b muscle fibers, compared with proportions in QHs, regardless of PSSM status. CONCLUSIONS AND CLINICAL RELEVANCE: PSSM in Belgians may be attributable to excessive glycogen synthesis rather than decreased glycogen utilization or enhanced glucose uptake into muscle cells.


Subject(s)
Glycogen Storage Disease/veterinary , Horse Diseases/blood , Insulin Resistance/physiology , Musculoskeletal Diseases/veterinary , Animals , Biopsy/veterinary , Blood Glucose/metabolism , Female , Glucose Clamp Technique/veterinary , Glycogen Storage Disease/blood , Glycogen Storage Disease/enzymology , Histocytochemistry/veterinary , Horses , Insulin/blood , L-Lactate Dehydrogenase/metabolism , Male , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Musculoskeletal Diseases/blood , Musculoskeletal Diseases/enzymology , Phosphofructokinase-1, Muscle Type/metabolism , Phosphoglucomutase/metabolism , Phosphoglycerate Mutase/metabolism , Phosphorylase a/metabolism
8.
Naunyn Schmiedebergs Arch Pharmacol ; 375(1): 11-28, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17295024

ABSTRACT

Activation of either coexisting beta1- or beta2 -adrenoceptors with noradrenaline or adrenaline, respectively, causes maximum increases of contractility of human atrial myocardium. Previous biochemical work with the beta2 -selective agonist zinterol is consistent with activation of the cascade beta2 -adrenoceptors-->Gsalpha-protein-->adenylyl cyclase-->cAMP-->protein kinase (PKA)-->phosphorylation of phospholamban, troponin I, and C-protein-->hastened relaxation of human atria from nonfailing hearts. However, in feline and rodent myocardium, catecholamines and zinterol usually do not hasten relaxation through activation of beta2 -adrenoceptors, presumably because of coupling of the receptors to Gi protein. It is unknown whether the endogenously occurring beta2 -adrenoceptor agonist adrenaline acts through the above cascade in human atrium and whether its mode of action could be changed in heart failure. We assessed the effects of (-)-adrenaline, mediated through beta2 -adrenoceptors (in the presence of CGP 20712A 300 nM to block beta1 -adrenoceptors), on contractility and relaxation of right atrial trabecula obtained from nonfailing and failing human hearts. Cyclic AMP levels were measured as well as phosphorylation of phospholamban, troponin I, and protein C with Western blots and the back-phosphorylation procedure. For comparison, beta1 -adrenoceptor-mediated effects of (-)-noradrenaline were investigated in the presence of ICI 118,551 (50 nM to block beta2 -adrenoceptors). The positive inotropic effects of both (-)-noradrenaline and (-)-adrenaline were accompanied by reductions in time to peak force and time to reach 50% relaxation. (-)-Adrenaline caused similar positive inotropic and lusitropic effects in atrial trabeculae from failing hearts. However, the inotropic potency, but not the lusitropic potency, of (-)-noradrenaline was reduced fourfold in atrial trabeculae from heart failure patients. Both (-)-adrenaline and (-)-noradrenaline enhanced cyclic AMP levels and produced phosphorylation of phospholamban, troponin I, and C-protein to a similar extent in atrial trabeculae from nonfailing hearts. The hastening of relaxation caused by (-)-adrenaline together with the PKA-catalyzed phosphorylation of the three proteins involved in relaxation, indicate coupling of beta2 -adrenoceptors to Gs protein. The phosphorylation of phospholamban at serine16 and threonine17 evoked by (-)-adrenaline through beta2 -adrenoceptors and by (-)-noradrenaline through beta1 -adrenoceptors was not different in atria from nonfailing and failing hearts. Activation of beta2 -adrenoceptors caused an increase in phosphorylase a activity in atrium from failing hearts further emphasizing the presence of the beta2 -adrenoceptor-Gsalpha-protein pathway in human heart. The positive inotropic and lusitropic potencies of (-)-adrenaline were conserved across Arg16Gly- and Gln27Glu-beta2 -adrenoceptor polymorphisms in the right atrium from patients undergoing coronary artery bypass surgery, chronically treated with beta1 -selective blockers. The persistent relaxant and biochemical effects of (-)-adrenaline through beta2 -adrenoceptors and of (-)-noradrenaline through beta1 -adrenoceptors in heart failure are inconsistent with an important role of coupling of beta2 -adrenoceptors with Gialpha-protein in human atrial myocardium.


Subject(s)
Epinephrine/pharmacology , GTP-Binding Protein alpha Subunits, Gs/physiology , Heart Failure/physiopathology , Myocardial Contraction/physiology , Receptors, Adrenergic, beta-2/physiology , Adrenergic Agonists/pharmacology , Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Antagonists/pharmacology , Adult , Aged , Atrial Appendage/drug effects , Atrial Appendage/physiology , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Diastole/drug effects , Diastole/physiology , Epinephrine/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Genotype , Heart Failure/metabolism , Humans , In Vitro Techniques , Isoproterenol/pharmacology , Male , Middle Aged , Myocardial Contraction/drug effects , Norepinephrine/pharmacology , Phosphorylase a/metabolism , Phosphorylation/drug effects , Receptors, Adrenergic, beta-2/genetics , Troponin I/metabolism
9.
FEBS J ; 273(9): 1989-99, 2006 May.
Article in English | MEDLINE | ID: mdl-16640562

ABSTRACT

Hepatic insulin resistance in the leptin-receptor defective Zucker fa/fa rat is associated with impaired glycogen synthesis and increased activity of phosphorylase-a. We investigated the coupling between phosphorylase-a and glycogen synthesis in hepatocytes from fa/fa rats by modulating the concentration of phosphorylase-a. Treatment of hepatocytes from fa/fa rats and Fa/? controls with a selective phosphorylase inhibitor caused depletion of phosphorylase-a, activation of glycogen synthase and stimulation of glycogen synthesis. The flux-control coefficient of phosphorylase on glycogen synthesis was glucose dependent and at 10 mm glucose was higher in fa/fa than Fa/? hepatocytes. There was an inverse correlation between the activities of glycogen synthase and phosphorylase-a in both fa/fa and Fa/? hepatocytes. However, fa/fa hepatocytes had a higher activity of phosphorylase-a, for a corresponding activity of glycogen synthase. This defect was, in part, normalized by expression of the glycogen-targeting protein, PTG. Hepatocytes from fa/fa rats had normal expression of the glycogen-targeting proteins G(L) and PTG but markedly reduced expression of R6. Expression of R6 protein was increased in hepatocytes from Wistar rats after incubation with leptin and insulin. Diminished hepatic R6 expression in the leptin-receptor defective fa/fa rat may be a contributing factor to the elevated phosphorylase activity and/or its high control strength on glycogen synthesis.


Subject(s)
Glycogen/biosynthesis , Hepatocytes/enzymology , Insulin Resistance/genetics , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylase a/chemistry , Protein Subunits/antagonists & inhibitors , Protein Subunits/biosynthesis , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Female , Glycogen/metabolism , Glycogen/physiology , Insulin/chemistry , Intracellular Signaling Peptides and Proteins , Leptin/chemistry , Male , Obesity/enzymology , Obesity/genetics , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/metabolism , Phosphorylase a/physiology , Protein Subunits/metabolism , Rats , Rats, Wistar , Rats, Zucker , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Leptin
10.
J Biol Chem ; 280(43): 36364-71, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16131488

ABSTRACT

Transcription of human immunodeficiency virus (HIV)-1 genes is activated by HIV-1 Tat protein, which induces phosphorylation of the C-terminal domain of RNA polymerase-II by CDK9/cyclin T1. We previously showed that Tat-induced HIV-1 transcription is regulated by protein phosphatase-1 (PP1). In the present study we demonstrate that Tat interacts with PP1 and that disruption of this interaction prevents induction of HIV-1 transcription. We show that PP1 interacts with Tat in part through the binding of Val36 and Phe38 of Tat to PP1 and that Tat is involved in the nuclear and subnuclear targeting of PP1. The PP1 binding mutant Tat-V36A/F38A displayed a decreased affinity for PP1 and was a poor activator of HIV-1 transcription. Surprisingly, Tat-Q35R mutant that had a higher affinity for PP1 was also a poor activator of HIV-1 transcription, because strong PP1 binding competed out binding of Tat to CDK9/cyclin T1. Our results suggest that Tat might function as a nuclear regulator of PP1 and that interaction of Tat with PP1 is critical for activation of HIV-1 transcription by Tat.


Subject(s)
Gene Products, tat/physiology , Phosphoprotein Phosphatases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Biotinylation , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 9/metabolism , Dose-Response Relationship, Drug , Gene Products, tat/metabolism , Genes, tat/genetics , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunohistochemistry , Immunoprecipitation , Inhibitory Concentration 50 , Kinetics , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Phenylalanine/chemistry , Phosphorylase a/metabolism , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Phosphatase 1 , RNA Polymerase II/metabolism , Transcription, Genetic , Transfection , Valine/chemistry , beta-Galactosidase/metabolism
11.
Am J Med Genet A ; 137(2): 125-9, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16059941

ABSTRACT

We report on three male newborn infants of a highly inbred Lebanese family presenting with a characteristic phenotype: arthrogryposis multiplex, deafness, large inguinal hernia, hiccup-like diaphragmatic contractions, and inability to suck, requiring nasogastric gavage feeding. All three boys died from respiratory failure during the first 3 months of life. Intra vitam or post mortem examinations revealed myopathic changes and elevated glycogen content of muscle tissue. This new syndrome is probably transmitted in an autosomal recessive mode, although X-linked inheritance cannot be excluded.


Subject(s)
Abnormalities, Multiple/pathology , Arthrogryposis/pathology , Deafness/pathology , Hernia, Inguinal/pathology , 1,4-alpha-Glucan Branching Enzyme/genetics , 1,4-alpha-Glucan Branching Enzyme/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Consanguinity , Family Health , Fatal Outcome , Female , Genes, Recessive/genetics , Glycogen/metabolism , Humans , Infant , Infant, Newborn , Male , Pedigree , Phosphorylase a/metabolism , Polymorphism, Single Nucleotide
12.
Diabetes ; 54(3): 617-23, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15734835

ABSTRACT

Glucokinase and phosphorylase both have a high control strength over hepatocyte glycogen metabolism and are potential therapeutic targets for type 2 diabetes. We tested whether combined phosphorylase inactivation and glucokinase activation is a more effective strategy for controlling hepatic glycogen metabolism than single-site targeting. Activation of glucokinase by enzyme overexpression combined with selective dephosphorylation of phosphorylase-a by an indole carboxamide that favors the T conformation of phosphorylase caused a greater stimulation of glycogen synthesis than the sum of either treatment alone. This result is explained by the complementary roles of elevated glucose-6-phosphate (G6P; a positive modulator) and depleted phosphorylase-a (a negative modulator) in activating glycogen synthase and also by synergistic inactivation of phosphorylase-a by glucokinase activation and the indole carboxamide. Inactivation of phosphorylase-a by the indole carboxamide was counteracted by 5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside, which is metabolized to an AMP analog; this effect was reversed by G6P. Our findings provide further evidence for the inverse roles of G6P and AMP in regulating the activation state of hepatic phosphorylase. It is proposed that dual targeting of glucokinase and phosphorylase-a enables improved potency and efficacy in controlling hepatic glucose metabolism.


Subject(s)
Amides/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Glucokinase/metabolism , Hepatocytes/drug effects , Indoles/pharmacology , Liver Glycogen/metabolism , Phosphorylase a/antagonists & inhibitors , Adenosine Monophosphate , Aminoimidazole Carboxamide/pharmacology , Animals , Drug Interactions , Enzyme Activation/drug effects , Gene Expression , Glycogen Synthase/metabolism , Hepatocytes/enzymology , In Vitro Techniques , Male , Phosphorylase a/metabolism , Rats , Rats, Wistar , Ribonucleotides/pharmacology
13.
Am J Physiol Endocrinol Metab ; 289(3): E366-72, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15797986

ABSTRACT

Phosphorylase is regulated by a number of small-molecular-weight effectors that bind to three sites on the enzyme. Recently, a fourth site referred to as the indole-inhibitor site has been identified. Synthetic compounds bind to the site and inhibit activity. However, the effects of these compounds in the presence of other endogenous effectors are unknown. We have determined the effects of four indole derivative glycogen phosphorylase inhibitors (GPI) on recombinant human liver glycogen phosphorylase a activity. The GPIs tested were all potent inhibitors. However, the endogenous inhibitors (glucose, ADP, ATP, fructose 1-phosphate, glucose 6-phosphate, UDP-glucose) and the activator (AMP) markedly reduced the inhibitory effect of GPIs. Consistent with these in vitro findings, the IC50 for the inhibition of glycogenolysis in cells and the liver drug concentration associated with glucose-lowering activity in diabetic ob/ob mice in vivo were also significantly higher than those determined in in vitro enzyme assays. The inhibitory effect of indole-site effectors is modulated by endogenous small-molecular-weight effectors of phosphorylase a activity. However, at higher concentrations (10-30 microM), the GPI effect was dominant and resulted in inhibition of phosphorylase a activity irrespective of the presence or absence of the other modulators of the enzyme.


Subject(s)
Blood Glucose/metabolism , Hyperglycemia/drug therapy , Indoles/pharmacology , Liver/enzymology , Phosphorylase a/antagonists & inhibitors , Phosphorylase a/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amides/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fructosephosphates/metabolism , Glucosephosphates/metabolism , Glycogen/metabolism , Humans , Hyperglycemia/metabolism , Hypoglycemia/chemically induced , Hypoglycemia/metabolism , Male , Phosphorylase b/antagonists & inhibitors , Phosphorylase b/metabolism , Rats , Rats, Sprague-Dawley , Uridine Diphosphate Glucose/metabolism
14.
J Chem Inf Model ; 45(1): 136-45, 2005.
Article in English | MEDLINE | ID: mdl-15667139

ABSTRACT

Glycogen phosphorylase (GP(a)) is a specific target for the design of inhibitors and may prevent glycogenolysis under high glucose conditions in type II diabetes. The carboxamides first reported by Hoover D. J. et al. (J. Med. Chem. 1998, 41, 2934-2938) are one of the major classes of GP(a) inhibitors other than glucose derivatives. The recent, X-ray crystallographic analyses (Oikonomakos et al. Biochim. Biophys. Acta 2003, 1647, 325-332) have revealed a distinct mechanism of action for these inhibitors, which bind at a new allosteric site away from the inhibitory and catalytic sites. To elucidate the essential structural and physicochemical requirements responsible for binding to the GP(a) enzyme and to develop predictive models, CoMFA and docking studies have been carried out on a series of indole-2-carboxamide derivates. The CoMFA model developed using pharmacophoric alignments and hydrogen-bonding fields demonstrated high predictive ability against the training (r2 = 0.98, q2 = 0.68) and the test set (r2pred = 0.85). Further the superimposition of PLS coefficient contour maps from CoMFA with the GP(a) active site (PDB: 1lwo) has shown a high level of compatibility.


Subject(s)
Hypoglycemic Agents/chemistry , Phosphorylase a/antagonists & inhibitors , Binding Sites , Humans , Hypoglycemic Agents/pharmacology , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Structure , Phosphorylase a/chemistry , Protein Binding , Quantitative Structure-Activity Relationship
16.
Protein Expr Purif ; 31(1): 19-33, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12963337

ABSTRACT

The protein serine/threonine phosphatase (PP) type 2A family consists of three members: PP2A, PP4, and PP6. Specific rabbit and sheep antibodies corresponding to each catalytic subunit, as well as a rabbit antibody recognizing all three subunits, were utilized to examine the expression of these enzymes in select rat tissue extracts. PP2A, PP4, and PP6 catalytic subunits (PP2A(C), PP4(C), and PP6(C), respectively) were detected in all rat tissue extracts examined and exhibited some differences in their levels of expression. The expression of alpha4, an interacting protein for PP2A family members that may function downstream of the target of rapamycin (Tor), was also examined using specific alpha4 sheep antibodies. Like the phosphatase catalytic subunits, alpha4 was ubiquitously expressed with particularly high levels in the brain and thymus. All three PP2A family members, but not alpha4, bound to the phosphatase affinity resin microcystin-Sepharose. The phosphatase catalytic subunits were purified to apparent homogeneity (PP2A(C) and PP4(C)) or near homogeneity (PP6(C)) from bovine testes soluble extracts following ethanol precipitation and protein extraction. In contrast to PP2A(C), PP4(C) and PP6(C) exhibited relatively low phosphatase activity towards several substrates. Purified PP2A(C) and native PP2A in cellular extracts bound to GST-alpha4, and co-immunoprecipitated with endogenous alpha4 and ectopically expressed myc-tagged alpha4. The interaction of PP2A(C) with alpha4 was unaffected by rapamycin treatment of mammalian cells; however, protein serine/threonine phosphatase inhibitors such as okadaic acid and microcystin-LR disrupted the alpha4/PP2A complex. Together, these findings increase our understanding of the biochemistry of alpha4/phosphatase complexes and suggest that the alpha4 binding site within PP2A may include the phosphatase catalytic domain.


Subject(s)
Catalytic Domain , Phosphoprotein Phosphatases/isolation & purification , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Anion Exchange Resins/chemistry , Antibodies/immunology , Antibodies/isolation & purification , Antibody Specificity , Blotting, Western , Brain/enzymology , Brain Chemistry , COS Cells , Caseins/metabolism , Cattle , Chlorocebus aethiops , Chromatography, Affinity , Cloning, Molecular , Ethers, Cyclic/pharmacology , Gene Expression , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Histones/metabolism , Intercellular Signaling Peptides and Proteins , Liver/chemistry , Liver/enzymology , Male , Mice , Microcystins , Molecular Chaperones , Molecular Sequence Data , Myocardium/chemistry , Myocardium/enzymology , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/genetics , Phosphorylase a/metabolism , Phosphorylation , Precipitin Tests , Protein Binding/physiology , Protein Kinases/metabolism , Protein Phosphatase 2 , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Resins, Synthetic , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
17.
J Physiol ; 553(Pt 2): 523-31, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12963789

ABSTRACT

Skeletal muscle that is deficient in creatine kinase (CK-/-) exhibits accelerated glycogenolysis during contraction. Understanding this phenomenon could provide insight into the control of glycogenolysis during contraction. Therefore, glycogen breakdown was investigated in isolated extensor digitorum longus CK-/- muscle. Muscles were stimulated to produce repeated tetani for 20 s in the presence of sodium cyanide to block mitochondrial respiration. Accumulation of lactate after stimulation was similar in wild-type (WT) and CK-/- muscles, whereas accumulation of glucose-6-phosphate was twofold higher in CK-/- muscles, indicating greater glycogenolysis in CK-/- muscles. Total phosphorylase activity was decreased by almost 30 % in CK-/- muscle (P < 0.001). Phosphorylase fractional activity (-/+ 3.3 mM AMP) was similar in both groups in the basal state (about 10 %), but increased to a smaller extent in CK-/- muscles after stimulation (39 +/- 4 % vs. 52 +/- 4 % in WT, P < 0.05). Inorganic phosphate, the substrate for phosphorylase, increased marginally in CK-/- muscles after stimulation (basal = 25.3 +/- 2.2 micromol (g dry muscle)-1; stimulated = 33.9 +/- 2.3 micromol (g dry muscle)-1), but substantially in WT muscles (basal = 11.4 +/- 0.7 micromol (g dry muscle)-1; stimulated = 54.2 +/- 4.5 micromol (g dry muscle)-1). Kinetic studies of phosphorylase b (dephosphorylated enzyme) from muscle extracts in vitro demonstrated higher relative activities in CK-/- muscles (60-135 %) in response to low AMP concentrations (up to 50 microM) in both the basal state and after stimulation (P < 0.05), whereas no differences in activity between CK-/- and WT muscles were observed at high AMP concentrations (> 100 microM). These data indicate that allosteric activation of phosphorylase b accounts for the accelerated glycogenolysis in CK-/- muscle during contraction.


Subject(s)
Creatine Kinase/genetics , Glycogen/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Phosphorylase b/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/metabolism , Animals , Creatine/metabolism , Electric Stimulation , Enzyme Activation/drug effects , Glucose-6-Phosphate/metabolism , Glycogen Phosphorylase/metabolism , Glycogen Synthase/metabolism , In Vitro Techniques , Inosine/metabolism , Inosine Monophosphate/metabolism , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fatigue/physiology , Phosphocreatine/metabolism , Phosphorylase a/metabolism , Sodium Cyanide/pharmacology
18.
Acta Biochim Pol ; 50(2): 555-66, 2003.
Article in English | MEDLINE | ID: mdl-12833181

ABSTRACT

The cellular localization of the 35 kDa, low molecular mass acid metallophosphatase (LMW AcPase) from the frog (Rana esculenta) liver and its activity towards P-Ser and P-Tyr phosphorylated peptides were studied. This enzyme was localized to the cytoplasm of hepatocytes but did not appear in other cells of liver tissue (endothelium, macrophages, blood cells). This LMW AcPase does not display activity towards (32)P-phosphorylase a under conditions standard for the enzymes of PPP family. Proteins containing P-Ser: rabbit (32)P-phosphorylasea and phosvitin are hydrolysed only at acidic pH and are poor substrates for this enzyme. The frog AcPase is not inhibited by okadaic acid and F(-) ions, the Ser/Thr protein phosphatase inhibitors. Moreover, the frog enzyme does not cross-react with specific antisera directed against N-terminal fragment of human PP2A and C-terminal conserved fragment of the eukaryotic PP2A catalytic subunits. These results exclude LMW AcPase from belonging to Ser/Thr protein phosphatases: PP1c or PP2Ac. In addition to P-Tyr, this enzyme hydrolyses efficiently at acidic pH P-Tyr phosphorylated peptides (hirudin and gastrin fragments). K(m) value for the hirudin fragment (7.55 +/- 1.59 x 10(-6) M) is 2-3 orders of magnitude lower in comparison with other substrates tested. The enzyme is inhibited competitively by typical inhibitors of protein tyrosine phosphatases (PTPases): sodium orthovanadate, molybdate and tungstate. These results may suggest that the LMW AcPase of frog liver can act as PTPase in vivo. A different cellular localization and different response to inhibition by tetrahedral oxyanions (molybdate, vanadate and tungstate) provide further evidence that LMW AcPase of frog liver is distinct from the mammalian tartrate-resistant acid phosphatases.


Subject(s)
Acid Phosphatase/metabolism , Liver/enzymology , Metalloproteases/metabolism , Phosphoprotein Phosphatases/metabolism , Acid Phosphatase/antagonists & inhibitors , Amino Acid Sequence , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hepatocytes/enzymology , Hirudins/chemistry , Hirudins/metabolism , Humans , Immunochemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kinetics , Liver/cytology , Metalloproteases/antagonists & inhibitors , Molecular Sequence Data , Molecular Weight , Oligopeptides/chemistry , Oligopeptides/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylase a/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Phosphotyrosine/pharmacology , Phosvitin/metabolism , Protein Phosphatase 1 , Rabbits , Rana esculenta , Substrate Specificity
19.
Alcohol ; 29(2): 83-90, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12782249

ABSTRACT

We have demonstrated previously that 24 h of ethanol treatment potentiates angiotensin II (ANG II)-stimulated p42/p44 mitogen-activated protein kinase (MAPK) activity in hepatocytes. This potentiation of p42/p44 MAPK by ethanol exhibited agonist selectivity. To compare the effects of acute (24 h) versus chronic (6 weeks) ethanol treatment, ANG II-induced intracellular signaling was examined in (1) rat hepatocytes treated with ethanol for 24 h and (2) hepatocytes obtained from rats fed ethanol for 6 weeks. In hepatocytes obtained from rats fed ethanol for 6 weeks, ANG II-stimulated phosphorylase a was reduced, and this activity was calcium dependent and p42/p44 MAPK independent. Surprisingly, ANG II-stimulated p42/p44 MAPK activation was not affected in hepatocytes obtained from rats fed ethanol chronically (6 weeks). However, chronic (6 weeks) ethanol treatment decreased ethanol potentiation of p42/p44 MAPK by about 56.3% +/- 3.6% for p42 MAPK and 61.3% +/- 11.7% for p44 MAPK. Furthermore, ethanol had no effect on the expression of angiotensinogen and c-myc mRNA in hepatocytes. A decrease in ANG II-activated phosphorylase a, but not in p42/p44 MAPK activation, after chronic (6 weeks) ethanol treatment leads to the conclusion that they may not be dependent on each other.


Subject(s)
Angiotensin II/pharmacology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hepatocytes/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylase a/metabolism , Animals , Calcium/metabolism , Cell Separation , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hepatocytes/drug effects , Immunoblotting , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Phosphorylase a/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
20.
Eur J Biochem ; 270(13): 2773-81, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12823547

ABSTRACT

Multiple signalling pathways are involved in the mechanism by which insulin stimulates hepatic glycogen synthesis. In this study we used selective inhibitors of glycogen synthase kinase-3 (GSK-3) and an allosteric inhibitor of phosphorylase (CP-91149) that causes dephosphorylation of phosphorylase a, to determine the relative contributions of inactivation of GSK-3 and dephosphorylation of phosphorylase a as alternative pathways in the stimulation of glycogen synthesis by insulin in hepatocytes. GSK-3 inhibitors (SB-216763 and Li+) caused a greater activation of glycogen synthase than insulin (90% vs. 40%) but a smaller stimulation of glycogen synthesis (30% vs. 150%). The contribution of GSK-3 inactivation to insulin stimulation of glycogen synthesis was estimated to be less than 20%. Dephosphorylation of phosphorylase a with CP-91149 caused activation of glycogen synthase and translocation of the protein from a soluble to a particulate fraction and mimicked the stimulation of glycogen synthesis by insulin. The stimulation of glycogen synthesis by phosphorylase inactivation cannot be explained by either inhibition of glycogen degradation or activation of glycogen synthase alone and suggests an additional role for translocation of synthase. Titrations with the phosphorylase inactivator showed that stimulation of glycogen synthesis by insulin can be largely accounted for by inactivation of phosphorylase over a wide range of activities of phosphorylase a. We conclude that a signalling pathway involving dephosphorylation of phosphorylase a leading to both activation and translocation of glycogen synthase is a critical component of the mechanism by which insulin stimulates hepatic glycogen synthesis. Selective inactivation of phosphorylase can mimic insulin stimulation of hepatic glycogen synthesis.


Subject(s)
Glycogen/biosynthesis , Hepatocytes/metabolism , Insulin/metabolism , Phosphorylase a/metabolism , Signal Transduction/physiology , Amides/metabolism , Animals , Biological Transport/physiology , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/metabolism , Glucose/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Hepatocytes/cytology , Indoles/metabolism , Male , Phosphorylase a/antagonists & inhibitors , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL