Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
J Appl Physiol (1985) ; 112(5): 711-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22174407

ABSTRACT

Endurance training has been shown to increase pancreatic ß-cell function and mass. However, whether exercise modulates ß-cell growth and survival pathways signaling is not completely understood. This study investigated the effects of exercise on growth and apoptotic markers levels in rat pancreatic islets. Male Wistar rats were randomly assigned to 8-wk endurance training or to a sedentary control group. After that, pancreatic islets were isolated; gene expression and the total content and phosphorylation of several proteins related to growth and apoptotic pathways as well as the main antioxidant enzymes were determined by real-time polymerase chain reaction and Western blot analysis, respectively. Reactive oxygen species (ROS) production was measured by fluorescence. Endurance training increased the time to reach fatigue by 50%. Endurance training resulted in increased protein phosphorylation content of AKT (75%), AKT substrate (AS160; 100%), mTOR (60%), p70s6k (90%), and ERK1/2 (50%), compared with islets from control group. Catalase protein content was 50% higher, whereas ROS production was 49 and 77% lower in islets from trained rats under basal and stimulating glucose conditions, respectively. Bcl-2 mRNA and protein levels increased by 46 and 100%, respectively. Bax and cleaved caspase-3 protein contents were reduced by 25 and 50% in islets from trained rats, respectively. In conclusion, these results demonstrate that endurance training favors the ß-cell growth and survival by activating AKT and ERK1/2 pathways, enhancing antioxidant capacity, and reducing ROS production and apoptotic proteins content.


Subject(s)
Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Physical Endurance/physiology , Signal Transduction/physiology , Animals , Antioxidants/metabolism , Apoptosis/physiology , Body Weight , Fatigue/genetics , Fatigue/metabolism , Fatigue/physiopathology , Gene Expression , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Oxidation-Reduction , Phosphorylation , Physical Conditioning, Animal , Physical Endurance/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
3.
Eur J Appl Physiol ; 111(9): 2369-74, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21287194

ABSTRACT

Endurance exercise has been shown to reduce pancreatic islets glucose-stimulated insulin secretion (GSIS). Anaplerotic/cataplerotic pathways are directly related to GSIS signaling. However, the effect of endurance training upon pancreatic islets anaplerotic enzymes is still unknown. In this sense, we tested the hypothesis that endurance exercise decreases GSIS by reducing anaplerotic/cataplerotic enzymes content. Male Wistar rats were randomly assigned to one of the four experimental groups as follows: control sedentary group (CTL), trained 1 day per week (TRE1×), trained 3 days per week (TRE3×) and trained 5 days per week (TRE5x) and submitted to an 8 weeks endurance-training protocol. After the training protocol, pancreatic islets were isolated and incubated with basal (2.8 mM) and stimulating (16.7 mM) glucose concentrations for GSIS measurement by radioimmunoassay. In addition, pyruvate carboxylase (PYC), pyruvate dehydrogenase (PDH), pyruvate dehydrogenase kinase 4 (PDK4), ATP-citrate lyase (ACL) and glutamate dehydrogenase (GDH) content were quantified by western blotting. Our data showed that 8 weeks of chronic endurance exercise reduced GSIS by 50% in a dose-response manner according to weekly exercise frequency. PYC showed significant twofold increase in TRE3×. PYC enhancement was even higher in TRE5× (p < 0.0001). PDH and PDK4 reached significant 25 and 50% enhancement, respectively compared with CTL. ACL and GDH also reported significant 50 and 75% increase, respectively. The absence of exercise-induced correlations among GSIS and anaplerotic/cataplerotic enzymes suggests that exercise may control insulin release by activating other signaling pathways. The observed anaplerotic and cataplerotic enzymes enhancement might be related to ß-cell surviving rather than insulin secretion.


Subject(s)
Enzymes/metabolism , Insulin/metabolism , Islets of Langerhans/enzymology , Physical Conditioning, Animal/physiology , ATP Citrate (pro-S)-Lyase/metabolism , Animals , Enzymes/analysis , Glucose/pharmacology , Glutamate Dehydrogenase/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Physical Conditioning, Animal/adverse effects , Physical Endurance/physiology , Protein Kinases/metabolism , Pyruvate Carboxylase/metabolism , Rats , Rats, Wistar , Secretory Pathway/drug effects , Up-Regulation/drug effects
4.
J Endocrinol ; 208(3): 257-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21212094

ABSTRACT

Endurance exercise is known to enhance peripheral insulin sensitivity and reduce insulin secretion. However, it is unknown whether the latter effect is due to the reduction in plasma substrate availability or alterations in ß-cell secretory machinery. Here, we tested the hypothesis that endurance exercise reduces insulin secretion by altering the intracellular energy-sensitive AMP-activated kinase (AMPK) signaling pathway. Male Wistar rats were submitted to endurance protocol training one, three, or five times per week, over 8 weeks. After that, pancreatic islets were isolated, and glucose-induced insulin secretion (GIIS), glucose transporter 2 (GLUT2) protein content, total and phosphorylated calmodulin kinase kinase (CaMKII), and AMPK levels as well as peroxisome proliferator-activated receptor-γ coactivator-1-α (PGC-1α) and uncoupling protein 2 (UCP2) content were measured. After 8 weeks, chronic endurance exercise reduced GIIS in a dose-response manner proportionally to weekly exercise frequency. Contrariwise, increases in GLUT2 protein content, CaMKII and AMPK phosphorylation levels were observed. These alterations were accompanied by an increase in UCP2 content, probably mediated by an enhancement in PGC-1α protein expression. In conclusion, chronic endurance exercise induces adaptations in ß-cells leading to a reduction in GIIS, probably by activating the AMPK signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Insulin/metabolism , Ion Channels/metabolism , Islets of Langerhans/metabolism , Mitochondrial Proteins/metabolism , Physical Conditioning, Animal , Physical Endurance , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glucose Transporter Type 2/metabolism , Insulin Secretion , Male , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Transcription Factors/metabolism , Uncoupling Protein 2
5.
J Biol Chem ; 286(15): 12870-80, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21257748

ABSTRACT

Type 2 diabetes mellitus results from the complex association of insulin resistance and pancreatic ß-cell failure. Obesity is the main risk factor for type 2 diabetes mellitus, and recent studies have shown that, in diet-induced obesity, the hypothalamus becomes inflamed and dysfunctional, resulting in the loss of the perfect coupling between caloric intake and energy expenditure. Because pancreatic ß-cell function is, in part, under the control of the autonomic nervous system, we evaluated the role of hypothalamic inflammation in pancreatic islet function. In diet-induced obesity, the earliest markers of hypothalamic inflammation are present at 8 weeks after the beginning of the high fat diet; similarly, the loss of the first phase of insulin secretion is detected at the same time point and is restored following sympathectomy. Intracerebroventricular injection of a low dose of tumor necrosis factor α leads to a dysfunctional increase in insulin secretion and activates the expression of a number of markers of apoptosis in pancreatic islets. In addition, the injection of stearic acid intracerebroventricularly, which leads to hypothalamic inflammation through the activation of tau-like receptor-4 and endoplasmic reticulum stress, produces an impairment of insulin secretion, accompanied by increased expression of markers of apoptosis. The defective insulin secretion, in this case, is partially dependent on sympathetic signal-induced peroxisome proliferator receptor-γ coactivator Δα and uncoupling protein-2 expression and is restored after sympathectomy or following PGC1α expression inhibition by an antisense oligonucleotide. Thus, the autonomic signals generated in concert with hypothalamic inflammation can impair pancreatic islet function, a phenomenon that may explain the early link between obesity and defective insulin secretion.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Hypothalamic Diseases/complications , Hypothalamic Diseases/metabolism , Hypothalamus/metabolism , Islets of Langerhans/metabolism , Animals , Diabetes Mellitus, Type 2/pathology , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Hypothalamic Diseases/chemically induced , Hypothalamic Diseases/pathology , Hypothalamus/pathology , Inflammation/chemically induced , Inflammation/complications , Inflammation/metabolism , Inflammation/pathology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/pathology , Male , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Stearic Acids/adverse effects , Stearic Acids/pharmacology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Time Factors , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/pharmacology
6.
Am J Physiol Heart Circ Physiol ; 300(3): H902-12, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148763

ABSTRACT

We studied the implication of focal adhesion kinase (FAK) in cardiac mitochondrial biogenesis induced by mechanical stress. Prolonged stretching (2-12 h) of neonatal rat ventricular myocytes (NRVM) upregulated the main components of mitochondrial transcription cascade [peroxisome proliferator-activated receptor coactivator-1 (PGC-1α), nuclear respiratory factor (NRF-1), and mitochondrial transcription factor A]. Concomitantly, prolonged stretching enhanced mitochondrial biogenesis [copy number of mitochondrial DNA (mtDNA), content of the subunit IV of cytochrome oxidase, and mitochondrial staining-green fluorescence intensity of Mitotracker green] and induced the hypertrophic growth (cell size and atrial natriuretic peptide transcripts) of NRVM. Furthermore, the stretching of NRVM enhanced phosphorylation, nuclear localization, and association of FAK with PGC-1α. Recombinant FAK COOH-terminal, but not the NH(2)-terminal or kinase domain, precipitated PGC-1α from nuclear extracts of NRVM. Depletion of FAK by RNA interference suppressed the upregulation of PGC-1α and NRF-1 and markedly attenuated the enhanced mitochondrial biogenesis and hypertrophic growth of stretched NRVM. In the context of energy metabolism, FAK depletion became manifest by a reduction of ATP levels in stretched NRVM. Complementary studies in adult mice left ventricle demonstrated that pressure overload upregulated PGC-1α, NRF-1, and mtDNA. In vivo FAK silencing transiently attenuated the upregulation of PGC-1α, NRF-1, and mtDNA, as well as the left ventricular hypertrophy induced by pressure overload. In conclusion, activation of FAK signaling seems to be important for conferring enhanced mitochondrial biogenesis coupled to the hypertrophic growth of cardiomyocytes in response to mechanical stress, via control of mitochondrial transcription cascade.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Stress, Mechanical , Animals , Animals, Newborn , Cells, Cultured , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/physiology , Mice , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/physiology , Myocytes, Cardiac/physiology , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 1/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Rats , Rats, Wistar , Transcription Factors/metabolism , Transcription Factors/physiology , Up-Regulation
7.
J Physiol Sci ; 60(6): 425-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20848345

ABSTRACT

In rats, the acute central dipsogenic and natriuretic action of angiotensin II (AngII) seems to be independent of the hemodynamic effects of the peptide; however, in genetically hypertensive models, this relationship has not yet been investigated. It has been demonstrated that AngII induces the suppressor of cytokine signaling (SOCS-3) expression in the brain that, in turn, modulates further activation of the pathway, leading to desensitization to AngII stimuli with regard to its dipsogenic effect. This study investigates age-related Janus kinase (JAK-2) and SOCS-3 hypothalamic expression, by immunoblotting, and the involvement of SOCS-3 expression in urinary sodium handling and dipsogenic response in spontaneously hypertensive rats (SHR), compared with age-matched Wistar-Kyoto (WKy) rats. The intracerebroventricular (i.c.v.) application of AngII significantly enhanced the dipsogenic response, reduced C(Cr), and reciprocally promoted increased absolute and fractional rates of excretion of sodium in WKy rats. The central AngII-induced dipsogenic effect in WKy and SHR was significantly attenuated by prior i.c.v. administration of DUP753. In addition, the magnitude of the dipsogenic and renal response to AngII was significantly attenuated in age-matched SHR. Blocking of hypothalamic SOCS-3 expression by an antisense oligonucleotide resulted in partial reversal of the refractory nature of AngII in thirst responses in SHR. The altered centrally applied AngII response in SHR associated with increased hypothalamic JAK-2/SOCS-3 expression may suggest that abnormal regulation of the central angiotensin pathways may contribute to dysfunction of water-electrolyte homeostasis in SHR.


Subject(s)
Angiotensin II/pharmacology , Drinking/drug effects , Hypothalamus/metabolism , Sodium/urine , Suppressor of Cytokine Signaling Proteins/biosynthesis , Angiotensin II/administration & dosage , Animals , Injections, Intraventricular , Janus Kinase 2/biosynthesis , Kidney/physiology , Losartan/pharmacology , Male , Oligonucleotides, Antisense/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Suppressor of Cytokine Signaling 3 Protein
8.
Endocrinology ; 149(8): 3870-80, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18467448

ABSTRACT

Protein tyrosine phosphatase (PTP1B) has been implicated in the negative regulation of insulin and leptin signaling. PTP1B knockout mice are hypersensitive to insulin and leptin and resistant to obesity when fed a high-fat diet. We investigated the role of hypothalamic PTP1B in the regulation of food intake, insulin and leptin actions and signaling in rats through selective decreases in PTP1B expression in discrete hypothalamic nuclei. We generated a selective, transient reduction in PTP1B by infusion of an antisense oligonucleotide designed to blunt the expression of PTP1B in rat hypothalamic areas surrounding the third ventricle in control and obese rats. The selective decrease in hypothalamic PTP1B resulted in decreased food intake, reduced body weight, reduced adiposity after high-fat feeding, improved leptin and insulin action and signaling in hypothalamus, and may also have a role in the improvement in glucose metabolism in diabetes-induced obese rats.


Subject(s)
Drug Resistance/drug effects , Hypothalamus/drug effects , Insulin/pharmacology , Leptin/pharmacology , Obesity/pathology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Adiposity/drug effects , Adiposity/genetics , Animals , Body Weight/drug effects , Body Weight/genetics , Diet, Atherogenic , Drug Resistance/genetics , Eating/drug effects , Eating/genetics , Glucose/metabolism , Hypothalamus/enzymology , Hypothalamus/metabolism , Injections, Intraventricular , Male , Obesity/etiology , Obesity/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Tyrosine Phosphatases/genetics , Rats , Rats, Wistar , Satiation/drug effects , Signal Transduction/drug effects
9.
FEBS Lett ; 580(19): 4625-31, 2006 Aug 21.
Article in English | MEDLINE | ID: mdl-16876161

ABSTRACT

TNF-alpha acts on the hypothalamus modulating food intake and energy expenditure through mechanisms incompletely elucidated. Here, we explore the hypothesis that, to modulate insulin-induced anorexigenic signaling in hypothalamus, TNF-alpha requires the synthesis of NO. TNF-alpha activates signal transduction through JNK and p38 in hypothalamus, peaking at 10(-8) M. This is accompanied by the induction of expression of the inducible and neuronal forms of NOS, in both cases peaking at 10(-12) M. In addition, TNF-alpha stimulates NOS catalytic activity. Pre-treatment with TNF-alpha at a low dose (10(-12) M) inhibits insulin-dependent anorexigenic signaling, and this effect is abolished in iNOS but not in nNOS knockout mice.


Subject(s)
Feeding Behavior/physiology , Hypothalamus/drug effects , Insulin/physiology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Dose-Response Relationship, Drug , Hypothalamus/physiology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/administration & dosage
10.
Endocrinology ; 146(2): 579-88, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15514089

ABSTRACT

Angiotensin II inhibits insulin-induced activation of phosphatidylinositol 3-kinase through a mechanism, at least in part, dependent on serine phosphorylation of the insulin receptor and insulin receptor substrates (IRS)-1/2. Recent evidence shows that suppressor of cytokine signaling-3 (SOCS-3) is induced by insulin and angiotensin II and participates in the negative control of further stimulation of each of these signaling systems independently. In the present study, we evaluated the interaction of angiotensin II-induced SOCS-3 with the insulin signaling pathway in the heart of living rats. A single iv dose of angiotensin II promotes a significant increase of SOCS-3 in heart, an effect that lasts up to 180 min. Once induced, SOCS-3 interacts with the insulin receptor, JAK-2, IRS-1, and IRS-2. The inhibition of SOCS-3 expression by a phosphorthioate-modified antisense oligonucleotide partially restores angiotensin II-induced inhibition of insulin-induced insulin receptor, IRS-1 and IRS-2 tyrosine phosphorylation, and IRS-1 and IRS-2 association with p85-phosphatidylinositol 3-kinase and [Ser473] phosphorylation of Akt. Moreover, the inhibition of SOCS-3 expression partially reverses angiotensin II-induced inhibition of insulin-stimulated glucose transporter-4 translocation to the cell membrane. These results are reproduced in isolated cardiomyocytes. Thus, SOCS-3 participates, as a late event, in the negative cross-talk between angiotensin II and insulin, producing an inhibitory effect on insulin-induced glucose transporter-4 translocation.


Subject(s)
Angiotensin II/metabolism , Insulin/metabolism , Myocytes, Cardiac/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/metabolism , Gene Expression , Glucose Transporter Type 4 , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Janus Kinase 2 , Male , Milk Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Receptor Cross-Talk/physiology , Repressor Proteins/genetics , STAT5 Transcription Factor , Serine/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/metabolism , Transcription Factors/genetics
11.
J Endocrinol ; 181(1): 117-28, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15072572

ABSTRACT

Angiotensin II exerts a potent dypsogenic stimulus on the hypothalamus, which contributes to its centrally mediated participation in the control of water balance and blood pressure. Repetitive intracerebroventricular (i.c.v.) injections of angiotensin II lead to a loss of effect characterized as physiological desensitization to the peptide's action. In the present study, we demonstrate that angiotensin II induces the expression of suppressor of cytokine signaling (SOCS)-3 via angiotensin receptor 1 (AT1) and JAK-2, mostly located at the median preoptic lateral and anterodorsal preoptic nuclei. SOCS-3 produces an inhibitory effect upon the signal transduction pathways of several cytokines and hormones that employ members of the JAK/STAT families as intermediaries. The partial inhibition of SOCS-3 translation by antisense oligonucleotide was sufficient to significantly reduce the refractoriness of repetitive i.c.v. angiotensin II injections, as evaluated by water ingestion. Thus, by acting through AT1 on the hypothalamus, angiotensin II induces the expression of SOCS-3 which, in turn, blocks further activation of the pathway and consequently leads to desensitization to angiotensin II stimuli concerning its dypsogenic effect.


Subject(s)
Angiotensin II/pharmacology , Drinking Behavior/drug effects , Hypothalamus/metabolism , Proto-Oncogene Proteins , Repressor Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Depression, Chemical , Hypothalamus/chemistry , Hypothalamus/drug effects , Immunohistochemistry , Injections, Intraventricular , Janus Kinase 2 , Male , Oligonucleotides, Antisense/pharmacology , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Transcription Factors/genetics
12.
Endocrinology ; 144(12): 5604-14, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12960006

ABSTRACT

Insulin and angiotensin II (AngII) may act through overlapping intracellular pathways to promote cardiac myocyte growth. In this report insulin and AngII signaling, through the phosphatidylinositol 3-kinase (PI 3-kinase) and MAPK pathways, were compared in cardiac tissues of control and obese Zucker rats. AngII induced Janus kinase 2 tyrosine phosphorylation and coimmunoprecipitation with insulin receptor substrate 1 (IRS-1) and IRS-2 as well as an increase in tyrosine phosphorylation of IRS and its association with growth factor receptor-binding protein 2. Simultaneous treatment with both hormones led to marked increases in the associations of IRS-1 and -2 with growth factor receptor-binding protein 2 and in the dual phosphorylation of ERK1/2 compared with the administration of AngII or insulin alone. In contrast, an acute inhibition of both basal and insulin-stimulated PI 3-kinase activity was induced by both hormones. Insulin stimulated the phosphorylation of MAPK equally in lean and obese rats. Conversely, insulin-induced phosphorylation of Akt in heart was decreased in obese rats. Pretreatment with losartan did not change insulin-induced activation of ERK1/2 and attenuated the reduction of Akt phosphorylation in the heart of obese rats. Thus, the imbalance between PI 3-kinase-Akt and MAPK signaling pathways in the heart may play a role in the development of cardiovascular abnormalities observed in insulin-resistant states, such as in obese Zucker rats.


Subject(s)
Adaptor Proteins, Signal Transducing , Angiotensin II/metabolism , Hypoglycemic Agents/metabolism , Insulin Resistance/physiology , Insulin/metabolism , MAP Kinase Signaling System/physiology , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Vasoconstrictor Agents/metabolism , Angiotensin II/pharmacology , Animals , GRB2 Adaptor Protein , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Janus Kinase 2 , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Obesity/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Rats, Zucker , Receptor Cross-Talk/physiology , Vasoconstrictor Agents/pharmacology
13.
Endocrinology ; 144(10): 4586-96, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12960061

ABSTRACT

Angiotensin II (Ang II) exerts a potent growth stimulus on the heart and vascular wall. Activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) intracellular signaling pathway by Ang II mediates at least some of the mitogenic responses to this hormone. In other signaling systems that use the JAK/STAT pathway, proteins of the suppressor of cytokine signaling (SOCS) family participate in signal regulation. In the present study it is demonstrated that SOCS3 is constitutively expressed at a low level in rat heart and neonatal rat ventricular myocytes. Ang II at a physiological concentration enhances the expression of SOCS3 mRNA and protein, mainly via AT1 receptors. After induction, SOCS3 associates with JAK2 and impairs further activation of the JAK2/STAT1 pathway. Pretreatment of rats with a specific phosphorthioate antisense oligonucleotide to SOCS3, reverses the desensitization to angiotensin signaling, as detected by a fall in c-Jun expression after repetitive infusions of the hormone. Thus, SOCS3 is induced by Ang II in rat heart and neonatal rat ventricular myocytes and participates in the modulation of the signal generated by this hormone.


Subject(s)
Angiotensin II/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Proteins/metabolism , Proto-Oncogene Proteins , Repressor Proteins , Transcription Factors , Animals , Heart/drug effects , Janus Kinase 2 , Male , Oligonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Tissue Distribution , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...