Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Mol Sci ; 24(23)2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38069161

ABSTRACT

The angiotensin II type 2 (AT2) receptor has a role in promoting insulin sensitivity. However, the mechanisms underlying the AT2 receptor-induced facilitation of insulin are still not completely understood. Therefore, we investigated whether acute in vivo administration of AT2 receptor agonist compound 21 (C21) could activate insulin signaling molecules in insulin-target tissues. We report that, in male C57BL/6 mice, an acute (5 min, 0.25 mg/kg; i.v.) injection of C21 induces the phosphorylation of Akt and ERK1/2 at activating residues (Ser473 and Thr202/Tyr204, respectively) in both epididymal white adipose tissue (WAT) and heart tissue. In WAT, the extent of phosphorylation (p) of Akt and ERK1/2 induced by C21 was approximately 65% of the level detected after a bolus injection of a dose of insulin known to induce maximal activation of the insulin receptor (IR). In the heart, C21 stimulated p-Akt to a lesser extent than in WAT and stimulated p-ERK1/2 to similar levels to those attained by insulin administration. C21 did not modify p-IR levels in either tissue. We conclude that in vivo injection of the AT2 receptor agonist C21 activates Akt and ERK1/2 through a mechanism that does not involve the IR, indicating the participation of these enzymes in AT2R-mediated signaling.


Subject(s)
Insulins , Proto-Oncogene Proteins c-akt , Mice , Animals , Male , Phosphorylation , MAP Kinase Signaling System , Mice, Inbred C57BL , Adipose Tissue/metabolism , Receptor, Angiotensin, Type 2/metabolism
2.
Clin Sci (Lond) ; 126(9): 613-30, 2014 May.
Article in English | MEDLINE | ID: mdl-24450744

ABSTRACT

The prevalence of Type 2 diabetes mellitus is predicted to increase dramatically over the coming years and the clinical implications and healthcare costs from this disease are overwhelming. In many cases, this pathological condition is linked to a cluster of metabolic disorders, such as obesity, systemic hypertension and dyslipidaemia, defined as the metabolic syndrome. Insulin resistance has been proposed as the key mediator of all of these features and contributes to the associated high cardiovascular morbidity and mortality. Although the molecular mechanisms behind insulin resistance are not completely understood, a negative cross-talk between AngII (angiotensin II) and the insulin signalling pathway has been the focus of great interest in the last decade. Indeed, substantial evidence has shown that anti-hypertensive drugs that block the RAS (renin-angiotensin system) may also act to prevent diabetes. Despite its long history, new components within the RAS continue to be discovered. Among them, Ang-(1-7) [angiotensin-(1-7)] has gained special attention as a counter-regulatory hormone opposing many of the AngII-related deleterious effects. Specifically, we and others have demonstrated that Ang-(1-7) improves the action of insulin and opposes the negative effect that AngII exerts at this level. In the present review, we provide evidence showing that insulin and Ang-(1-7) share a common intracellular signalling pathway. We also address the molecular mechanisms behind the beneficial effects of Ang-(1-7) on AngII-mediated insulin resistance. Finally, we discuss potential therapeutic approaches leading to modulation of the ACE2 (angiotensin-converting enzyme 2)/Ang-(1-7)/Mas receptor axis as a very attractive strategy in the therapy of the metabolic syndrome and diabetes-associated diseases.


Subject(s)
Angiotensin I/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Insulin/metabolism , Peptide Fragments/metabolism , Renin-Angiotensin System , Signal Transduction , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/therapeutic use , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects
3.
Am J Physiol Renal Physiol ; 302(12): F1606-15, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22492942

ABSTRACT

Angiotensin (ANG)-(1-7) is known to attenuate diabetic nephropathy; however, its role in the modulation of renal inflammation and oxidative stress in type 2 diabetes is poorly understood. Thus in the present study we evaluated the renal effects of a chronic ANG-(1-7) treatment in Zucker diabetic fatty rats (ZDF), an animal model of type 2 diabetes and nephropathy. Sixteen-week-old male ZDF and their respective controls [lean Zucker rats (LZR)] were used for this study. The protocol involved three groups: 1) LZR + saline, 2) ZDF + saline, and 3) ZDF + ANG-(1-7). For 2 wk, animals were implanted with subcutaneous osmotic pumps that delivered either saline or ANG-(1-7) (100 ng·kg(-1)·min(-1)) (n = 4). Renal fibrosis and tissue parameters of oxidative stress were determined. Also, renal levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), ED-1, hypoxia-inducible factor-1α (HIF-1α), and neutrophil gelatinase-associated lipocalin (NGAL) were determined by immunohistochemistry and immunoblotting. ANG-(1-7) induced a reduction in triglyceridemia, proteinuria, and systolic blood pressure (SBP) together with a restoration of creatinine clearance in ZDF. Additionally, ANG-(1-7) reduced renal fibrosis, decreased thiobarbituric acid-reactive substances, and restored the activity of both renal superoxide dismutase and catalase in ZDF. This attenuation of renal oxidative stress proceeded with decreased renal immunostaining of IL-6, TNF-α, ED-1, HIF-1α, and NGAL to values similar to those displayed by LZR. Angiotensin-converting enzyme type 2 (ACE2) and ANG II levels remained unchanged after treatment with ANG-(1-7). Chronic ANG-(1-7) treatment exerts a renoprotective effect in ZDF associated with a reduction of SBP, oxidative stress, and inflammatory markers. Thus ANG-(1-7) emerges as a novel target for treatment of diabetic nephropathy.


Subject(s)
Angiotensin I/therapeutic use , Diabetic Nephropathies/drug therapy , Kidney/drug effects , Peptide Fragments/therapeutic use , Proteinuria/drug therapy , Acute-Phase Proteins/metabolism , Animals , Blood Pressure/drug effects , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Fibrosis , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Kidney/pathology , Lipocalin-2 , Lipocalins/metabolism , Male , Oxidative Stress/drug effects , Proteinuria/metabolism , Proteinuria/pathology , Proto-Oncogene Proteins/metabolism , Rats , Rats, Zucker , Tumor Necrosis Factor-alpha/metabolism
4.
Hepatology ; 53(6): 2097-106, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21391228

ABSTRACT

UNLABELLED: Galectin-1 (Gal-1), a widely expressed ß-galactoside-binding protein, exerts pleiotropic biological functions. Gal-1 is up-regulated in hepatocarcinoma cells, although its role in liver pathophysiology remains uncertain. We investigated the effects of Gal-1 on HepG2 hepatocellular carcinoma (HCC) cell adhesion and polarization. Soluble and immobilized recombinant Gal-1 (rGal-1) promoted HepG2 cell adhesion to uncoated plates and also increased adhesion to laminin. Antibody-mediated blockade experiments revealed the involvement of different integrins as critical mediators of these biological effects. In addition, exposure to rGal-1 markedly accelerated the development of apical bile canaliculi as shown by TRITC-phalloidin labeling and immunostaining for multidrug resistance associated-protein 2 (MRP2). Notably, rGal-1 did not interfere with multidrug resistance protein 1/P-glycoprotein or MRP2 apical localization, neither with transfer nor secretion of 5-chloromethylfluorescein diacetate through MRP2. Stimulation of cell adhesion and polarization by rGal-1 was abrogated in the presence of thiodigalactoside, a galectin-specific sugar, suggesting the involvement of protein-carbohydrate interactions in these effects. Additionally, Gal-1 effects were abrogated in the presence of wortmmanin, PD98059 or H89, suggesting involvement of phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinase and cyclic adenosine monophosphate-dependent protein kinase signaling pathways in these functions. Finally, expression levels of this endogenous lectin correlated with HCC cell adhesion and polarization and up-regulation of Gal-1-favored growth of hepatocarcinoma in vivo. CONCLUSION: Our results provide the first evidence of a role of Gal-1 in modulating HCC cell adhesion, polarization, and in vivo tumor growth, with critical implications in liver pathophysiology.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cell Polarity/physiology , Cell Proliferation , Galectin 1/physiology , Liver Neoplasms/physiopathology , Cell Adhesion/physiology , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/physiology , Humans , Mitogen-Activated Protein Kinases/physiology , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology
5.
J Mol Endocrinol ; 69(2): 357-376, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35608964

ABSTRACT

Growth hormone (GH) exerts major actions in cardiac growth and metabolism. Considering the important role of insulin in the heart and the well-established anti-insulin effects of GH, cardiac insulin resistance may play a role in the cardiopathology observed in acromegalic patients. As conditions of prolonged exposure to GH are associated with a concomitant increase of circulating GH, IGF1 and insulin levels, to dissect the direct effects of GH, in this study, we evaluated the activation of insulin signaling in the heart using four different models: (i) transgenic mice overexpressing GH, with chronically elevated GH, IGF1 and insulin circulating levels; (ii) liver IGF1-deficient mice, with chronically elevated GH and insulin but decreased IGF1 circulating levels; (iii) mice treated with GH for a short period of time; (iv) primary culture of rat cardiomyocytes incubated with GH. Despite the differences in the development of cardiomegaly and in the metabolic alterations among the three experimental mouse models analyzed, exposure to GH was consistently associated with a decreased response to acute insulin stimulation in the heart at the receptor level and through the PI3K/AKT pathway. Moreover, a blunted response to insulin stimulation of this signaling pathway was also observed in cultured cardiomyocytes of neonatal rats incubated with GH. Therefore, the key novel finding of this work is that impairment of insulin signaling in the heart is a direct and early event observed as a consequence of exposure to GH, which may play a major role in the development of cardiac pathology.


Subject(s)
Acromegaly , Human Growth Hormone , Animals , Growth Hormone/metabolism , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Mice , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Signal Transduction
6.
Am J Physiol Renal Physiol ; 300(1): F272-82, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962118

ABSTRACT

Angiotensin (ANG)-(1-7) constitutes an important functional end-product of the renin-angiotensin-aldosterone system that acts to balance the physiological actions of ANG II. In the kidney, ANG-(1-7) exerts beneficial effects by inhibiting growth-promoting pathways and reducing proteinuria. We examined whether a 2-wk treatment with a daily dose of ANG-(1-7) (0.6 mg·kg(-1)·day(-1)) exerts renoprotective effects in salt-loaded stroke-prone spontaneously hypertensive rats (SHRSP). Body weight, glycemia, triglyceridemia, cholesterolemia, as well as plasma levels of Na+ and K+ were determined both at the beginning and at the end of the treatment. Also, the weekly evolution of arterial blood pressure, proteinuria, and creatinine clearance was evaluated. Renal fibrosis was determined by Masson's trichrome staining. Interleukin (IL)-6, tumor necrosis factor (TNF)-α, and nuclear factor-κB (NF-κB) levels were determined by immunohistochemistry and confirmed by Western blotting analysis. The levels of glomerular nephrin were assessed by immunofluorescence. Chronic administration of ANG-(1-7) normalized arterial pressure, reduced glycemia and triglyceridemia, improved proteinuria, and ameliorated structural alterations in the kidney of SHRSP as shown by a restoration of glomerular nephrin levels as detected by immunofluorescence. These results were accompanied with a decrease in both the immunostaining and abundance of IL-6, TNF-α, and NF-κB. In this context, the current study provides strong evidence for a protective role of ANG-(1-7) in the kidney.


Subject(s)
Angiotensin I/therapeutic use , Interleukin-6/metabolism , Kidney/drug effects , NF-kappa B/metabolism , Peptide Fragments/therapeutic use , Proteinuria/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood Pressure/drug effects , Kidney/pathology , Male , Membrane Proteins/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sodium Chloride/pharmacology
7.
Am J Physiol Heart Circ Physiol ; 298(3): H1003-13, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20061544

ABSTRACT

The present study examined whether chronic treatment with angiotensin (ANG)-(1-7) reduces cardiac remodeling and inhibits growth-promoting signaling pathways in the heart of fructose-fed rats (FFR), an animal model of insulin resistance. Sprague-Dawley rats were fed either normal rat chow (control) or the same diet plus 10% fructose in drinking water. For the last 2 wk of a 6-wk period of the corresponding diet, control and FFR were implanted with osmotic pumps that delivered ANG-(1-7) (100 ng.kg(-1).min(-1)). A subgroup of each group of animals (control or FFR) underwent a sham surgery. We determined heart weight, myocyte diameter, interstitial fibrosis, and perivascular collagen type III deposition as well as the phosphorylation degree of ERK1/2, JNK1/2, and p38MAPK. FFR showed a mild hypertension that was significantly reduced after ANG-(1-7) treatment. Also, FFR displayed higher ANG II circulating and local levels in the heart that remained unaltered after chronic ANG-(1-7) infusion. An increased heart-to-body weight ratio, myocyte diameter, as well as left ventricular fibrosis and perivascular collagen type III deposition were detected in the heart of FFR. Interestingly, significant improvements in these cardiac alterations were obtained after ANG-(1-7) treatment. Finally, FFR that received ANG-(1-7) chronically displayed significantly lower phosphorylation levels of ERK1/2, JNK1/2, and p38MAPK. The beneficial effects obtained by ANG-(1-7) were associated with normal values of Src-homology 2-containing protein-tyrosine phosphatase-1 (SHP-1) activity in the heart. In conclusion, chronic ANG-(1-7) treatment ameliorated cardiac hypertrophy and fibrosis and attenuated the growth-promoting pathways in the heart. These findings show an important protective role of ANG-(1-7) in the heart of insulin-resistant rats.


Subject(s)
Angiotensin I/pharmacology , Fructose/adverse effects , Hypertension/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Insulin Resistance , Peptide Fragments/pharmacology , Ventricular Remodeling/drug effects , Angiotensin II/metabolism , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/physiology , Dietary Carbohydrates/adverse effects , Disease Models, Animal , Hypertension/etiology , Hypertension/metabolism , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/physiopathology , Insulin/blood , Male , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Rats , Rats, Sprague-Dawley
8.
Mol Cell Endocrinol ; 498: 110587, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31539597

ABSTRACT

The renin-angiotensin system modulates insulin action. Pharmacological stimulation of angiotensin type 2 receptor (AT2R) was shown to have beneficial metabolic effects in various animal models of insulin resistance and type 2 diabetes and also to increase insulin sensitivity in wild type mice. In this study we further explored the role of the AT2R on insulin action and glucose homeostasis by investigating the glycemic profile and in vivo insulin signaling status in insulin-target tissues from both male and female AT2R knockout (KO) mice. When compared to the respective wild-type (WT) group, glycemia and insulinemia was unaltered in AT2RKO mice regardless of sex. However, female AT2RKO mice displayed decreased insulin sensitivity compared to their WT littermates. This was accompanied by a compensatory increase in adiponectinemia and with a specific attenuation of the activity of main insulin signaling components (insulin receptor, Akt and ERK1/2) in adipose tissue with no apparent alterations in insulin signaling in either liver or skeletal muscle. These parameters remained unaltered in male AT2RKO mice as compared to male WT mice. Present data show that the AT2R has a physiological role in the conservation of insulin action in female but not in male mice. Our results suggest a sexual dimorphism in the control of insulin action and glucose homeostasis by the AT2R and reinforce the notion that pharmacological modulation of the balance between the AT1R and AT2R receptor could be important for treatment of metabolic syndrome and type 2 diabetes.


Subject(s)
Adiponectin/blood , Biomarkers/blood , Blood Glucose/metabolism , Insulin Resistance , Insulin/blood , Receptor, Angiotensin, Type 2/physiology , Sex Characteristics , Adipose Tissue/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors , Signal Transduction
9.
Front Pharmacol ; 10: 146, 2019.
Article in English | MEDLINE | ID: mdl-30853914

ABSTRACT

The MasR receptor (MasR) is an orphan G protein-coupled receptor proposed as a candidate for mediating the angiotensin (Ang)-converting enzyme 2-Ang-(1-7) protective axis of renin-angiotensin system. This receptor has been suggested to participate in several physiological processes including cardio- and reno-protection and regulation of the central nervous system function. Although the knowledge of the signaling mechanisms associated with MasR is essential for therapeutic purposes, these are still poorly understood. Accordingly, in the current study we aimed to characterize the signaling pathways triggered by the MasR. To do that, we measured cAMP and Ca2+ levels in both naïve and MasR transfected cells in basal conditions and upon incubation with putative MasR ligands. Besides, we evaluated activation of ERK1/2 by Ang-(1-7) in MasR transfected cells. Results indicated the existence of a high degree of MasR constitutive activity toward cAMP modulation. This effect was not mediated by the PDZ-binding motif of the MasR but by receptor coupling to Gαi-adenylyl cyclase signaling pathway. Incubation of MasR transfected cells with Ang-(1-7) or the synthetic ligand AVE 0991 amplified MasR negative modulation of cAMP levels. On the other hand, we provided evidence for lack of MasR-associated modulation of Ca2+ levels by Ang-(1-7). Finally, it was determined that the MasR attenuated Ang-(1-7)-induced ERK1/2 phosphorylation mediated by AT1R. We provided further characterization of MasR signaling mechanisms regarding its constitutive activity and response to putative ligands. This information could prove useful to better describe MasR physiological role and development of therapeutic agents that could modulate its action.

10.
J Gerontol A Biol Sci Med Sci ; 63(8): 788-97, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18772466

ABSTRACT

Calorie restriction (CR) improves insulin sensitivity and increases life span in normal but not in long-lived growth hormone-resistant knockout (GHRKO) mice. In this study, we examined interactive effects of GH resistance and long-term CR on cardiac insulin action. GHRKO mice exhibited marked increases in the insulin-induced phosphorylation of the insulin receptor (IR), insulin receptor substrate-1 (IRS-1), Akt, and ERK1/2 along with elevated insulin-stimulated IRS-1-associated regulatory subunit of phosphatidylinositol 3-kinase in the heart. These changes were associated with elevated protein levels of IR, IRS-1, and Akt and with a down-regulation of cardiac glucose transporter 4 (GLUT4). In normal mice, CR induced an important increase in the phosphorylation of cardiac Akt without elevation of Akt protein, reaching activation levels similar to those seen in GHRKO mice. This change may be cardioprotective and thus contribute to increased longevity in response to CR. Interestingly, the insulin signaling cascade in the heart of GHRKO mice was unaffected by CR.


Subject(s)
Caloric Restriction , Heart/physiology , Insulin-Like Growth Factor I/metabolism , Receptors, Somatotropin/physiology , Adaptor Proteins, Signal Transducing/physiology , Adenylate Kinase/metabolism , Animals , Down-Regulation/physiology , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins , Longevity , Mice , Mice, Knockout , Phosphorylation , Receptor, Insulin/physiology , Receptors, Somatotropin/metabolism , Signal Transduction/physiology
11.
Physiol Rep ; 6(16): e13824, 2018 08.
Article in English | MEDLINE | ID: mdl-30156060

ABSTRACT

The renin-angiotensin system modulates insulin action. Angiotensin type 1 receptor exerts a deleterious effect, whereas the angiotensin type 2 receptor (AT2R) appears to have beneficial effects providing protection against insulin resistance and type 2 diabetes. To further explore the role of the AT2R on insulin action and glucose homeostasis, in this study we administered C57Bl/6 mice with the synthetic agonist of the AT2R C21 for 12 weeks (1 mg/kg per day; ip). Vehicle-treated animals were used as control. Metabolic parameters, glucose, and insulin tolerance, in vivo insulin signaling in main insulin-target tissues as well as adipose tissue levels of adiponectin, and TNF-α were assessed. C21-treated animals displayed decreased glycemia together with unaltered insulinemia, increased insulin sensitivity, and increased glucose tolerance compared to nontreated controls. This was accompanied by a significant decrease in adipocytes size in epididymal adipose tissue and significant increases in both adiponectin and UCP-1 expression in this tissue. C21-treated mice showed an increase in both basal Akt and ERK1/2 phosphorylation levels in the liver, and increased insulin-stimulated Akt activation in adipose tissue. This positive modulation of insulin action induced by C21 appeared not to involve the insulin receptor. In C21-treated mice, adipose tissue and skeletal muscle became unresponsive to insulin in terms of ERK1/2 phosphorylation levels. Present data show that chronic pharmacological activation of AT2R with C21 increases insulin sensitivity in mice and indicate that the AT2R has a physiological role in the conservation of insulin action.


Subject(s)
Insulin Resistance/physiology , Receptor, Angiotensin, Type 2/agonists , Sulfonamides/pharmacology , Thiophenes/pharmacology , Adipocytes/drug effects , Adiponectin/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Cell Size/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Drug Administration Schedule , Drug Evaluation, Preclinical/methods , Glucose Tolerance Test , MAP Kinase Signaling System/physiology , Male , Mice, Inbred C57BL , Receptor, Angiotensin, Type 2/physiology , Signal Transduction , Sulfonamides/administration & dosage , Thiophenes/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
12.
J Hypertens ; 24(8): 1607-17, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16877964

ABSTRACT

BACKGROUND: Angiotensin II (AII) has been shown to contribute to the pathogenesis of hypertension and insulin resistance. In addition, the administration of selective AII type 1 receptor blockers has been shown to improve insulin sensitivity. However, only a few studies have addressed the molecular mechanisms involved in this association. Furthermore, in a previous study we illustrated that obese Zucker rats (OZR) present increased serine 994 (Ser994) phosphorylation of hepatic insulin receptor, and this event seems to be implicated in the regulation of the intrinsic IRK in this model of insulin resistance. OBJECTIVE AND DESIGN: We examined the effects of chronic treatment with irbesartan (50 mg/kg a day for 6 months) on the hepatic insulin signaling system of OZR. METHODS: The extent of phosphorylation of several components of the insulin signaling system was assessed by immunoprecipitation, followed by immunoblotting with phosphospecific antibodies. In addition, liver AII levels and fat deposits were determined by immunohistochemistry and Oil red O, respectively. RESULTS: OZR displayed a marked attenuation in the in-vivo phosphorylation of several components of the insulin signaling pathways in the liver, together with significantly higher hepatic AII levels and hepatic steatosis when compared with lean Zucker rats. We found that in the livers of OZR long-term administration of irbesartan is associated with: (i) increased insulin-stimulated insulin receptor tyrosine phosphorylation; (ii) decreased insulin receptor Ser994 phosphorylation; (iii) augmented insulin receptor substrate (IRS) 1 and 2 abundance and tyrosine phosphorylation; (iv) augmented association between IRS and the p85 regulatory subunit of phosphatidylinositol 3-kinase; (v) increased insulin-induced Akt phosphorylation; and (vi) decreased hepatic steatosis. CONCLUSION: The present study provides substantial information that demonstrates that long-term selective AII blockade by irbesartan improves insulin signaling and is associated with decreased insulin receptor Ser994 phosphorylation in the liver of a representative animal model of the human metabolic syndrome.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Antihypertensive Agents/pharmacology , Biphenyl Compounds/pharmacology , Insulin Resistance , Obesity/metabolism , Obesity/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tetrazoles/pharmacology , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Analysis of Variance , Angiotensin II/drug effects , Angiotensin II/metabolism , Animals , Biomarkers/blood , Biomarkers/urine , Blood Pressure , Disease Models, Animal , Enzyme Activation/drug effects , Fatty Liver/metabolism , Fatty Liver/physiopathology , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Irbesartan , Male , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Rats , Rats, Zucker , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
13.
Oncotarget ; 7(11): 11889-98, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26933812

ABSTRACT

Thioredoxin-1 (Trx1) protects the heart from ischemia/reperfusion (I/R) injury. Given that the age at which the first episode of coronary disease takes place has considerably decreased, life at middle-aged (MA) emerges as a new field of study. The aim was determine whether infarct size, Trx1 expression and activity, Akt and GSK-3ß were altered in young (Y) and MA mice overexpressing cardiac Trx1, and in a dominant negative (DN-Trx1) mutant of Trx1. Langendorff-perfused hearts were subjected to 30 minutes of ischemia and 120 minutes of reperfusion (R). We used 3 and 12 month-old male of wild type (WT), Trx1, and DN-Trx1. Trx1 overexpression reduced infarct size in young mice (WT-Y: 46.8±4.1% vs. Trx1-Y: 27.6±3.5%, p < 0.05). Trx1 activity was reduced by 52.3±3.2% (p < 0.05) in Trx1-MA, accompanied by an increase in nitration by 17.5±0.9%, although Trx1 expression in transgenic mice was similar between young and middle-aged. The expression of p-Akt and p-GSK-3ß increased during reperfusion in Trx1-Y. DN-Trx1 mice showed neither reduction in infarct size nor Akt and GSK-3ß phosphorylation. Our data suggest that the lack of protection in Trx1 middle-aged mice even with normal Trx1 expression may be associated to decreased Trx1 activity, increased nitration and inhibition of p-Akt and p-GSK-3ß.


Subject(s)
Gene Expression Regulation, Developmental , Heart/physiology , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress , Thioredoxins/metabolism , Animals , Humans , Mice , Mice, Transgenic , Myocardial Reperfusion Injury/pathology , Phosphorylation , Thioredoxins/genetics
14.
Growth Horm IGF Res ; 15(5): 324-36, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16112592

ABSTRACT

Growth hormone (GH) is an important modulator of insulin sensitivity. Multiple mechanisms appear to be involved in this modulatory effect. GH does not interact directly with the insulin receptor (IR), but conditions of GH excess are associated in general with hyperinsulinemia that induces a reduction of IR levels and impairment of its kinase activity. Several post-receptor events are shared between GH and insulin. This signaling crosstalk could be involved in the diabetogenic effects of GH. The utilization of animal models of GH excess, deficiency or resistance provided evidence that the signaling pathway leading to stimulation of the phosphatidylinositol 3-kinase (PI3K)/Akt cascade is an important site of regulation, and pointed to the liver as the major site of GH-induced insulin resistance. In skeletal muscle, GH-induced insulin resistance might involve an increase in the amount of the p85 subunit of PI3K that plays a negative role in insulin signalling. GH also reduces insulin sensitivity by enhancing events that negatively modulate insulin signaling such as stimulation of serine phosphorylation of IRS-1, which prevents its recruitment to the IR and induction of the suppressor of cytokine signalling (SOCS)-1 and SOCS-3 which modulate the signalling potential of the IRS proteins. In addition, GH has been shown to decrease the expression of the insulin-sensitizing adipo-cytokines adiponectin and visfatin. Finally, genetic manipulation of mice indicated that whereas GH plays a major role in reducing insulin sensitivity, circulating IGF-I also participates in the control of insulin sensitivity and plays an important role in the hormonal balance between GH and insulin.


Subject(s)
Growth Hormone/physiology , Insulin Resistance , Insulin/physiology , Signal Transduction/physiology , Animals , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptor, Insulin/metabolism
15.
J Endocrinol ; 182(3): 433-44, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15350185

ABSTRACT

Serine phosphorylation of the insulin receptor (IR) has been proposed to exert an inhibitory influence on its tyrosine kinase activity. Previous works using site-directed mutagenesis suggested that serine 994 of the IR (IR Ser 994) might be part of an inhibitory domain of the receptor. In this study we examined whether this residue is subjected to phosphorylation in vivo. We used a site-phosphospecific antibody to determine the extent of phosphorylation of IR Ser 994 in insulin target tissues from two animal models of insulin resistance with different IR kinase (IRK) activity: obese (fa/fa) Zucker rats and transgenic mice overexpressing bovine growth hormone (PEPCK-bGH mice). Phosphorylation at IR Ser 994 was markedly increased in liver of obese rats. This alteration appeared to be tissue-selective since no phosphorylation on Ser 994 was detected in IRs isolated from skeletal muscle of these animals. On the other hand, the phosphorylation level of IR Ser 994 was very low in liver of PEPCK-bGH mice and did not differ from that of the control group. We have also demonstrated that protein kinase (PK) C isoforms alpha, betaI and zeta are able to promote the in vitro phosphorylation of the IR on Ser 994. Differential findings in these two models of insulin resistance might thus reflect increased PKC activity resulting from increased lipid availability in obese Zucker rats. Our results suggest that Ser 994 is a novel in vivo IR phosphorylation site that might be involved in the regulation of the IRK in some states of insulin resistance.


Subject(s)
Insulin Resistance , Liver/metabolism , Obesity/metabolism , Receptor, Insulin/metabolism , Serine/metabolism , Animals , Cattle , Growth Hormone/genetics , Immunoblotting , Male , Mice , Mice, Transgenic , Models, Animal , Phosphorylation , Precipitin Tests , Rats , Rats, Zucker
16.
J Endocrinol ; 221(2): 215-27, 2014 May.
Article in English | MEDLINE | ID: mdl-24756097

ABSTRACT

The renin-angiotensin system (RAS) plays a crucial role in the regulation of physiological homeostasis and diseases such as hypertension, coronary artery disease, and chronic renal failure. In this cascade, the angiotensin-converting enzyme (ACE)/angiotensin II (Ang II)/AT1 receptor axis induces pathological effects, such as vasoconstriction, cell proliferation, and fibrosis, while the ACE2/Ang-(1-7)/Mas receptor axis is protective for end-organ damage. The altered function of the RAS could be a contributing factor to the cardiac and renal alterations induced by GH excess. To further explore this issue, we evaluated the consequences of chronic GH exposure on the in vivo levels of Ang II, Ang-(1-7), ACE, ACE2, and Mas receptor in the heart and the kidney of GH-transgenic mice (bovine GH (bGH) mice). At the age of 7-8 months, female bGH mice displayed increased systolic blood pressure (SBP), a high degree of both cardiac and renal fibrosis, as well as increased levels of markers of tubular and glomerular damage. Angiotensinogen abundance was increased in the liver and the heart of bGH mice, along with a concomitant increase in cardiac Ang II levels. Importantly, the levels of ACE2, Ang-(1-7), and Mas receptor were markedly decreased in both tissues. In addition, Ang-(1-7) administration reduced SBP to control values in GH-transgenic mice, indicating that the ACE2/Ang-(1-7)/Mas axis is involved in GH-mediated hypertension. The data indicate that the altered expression profile of the ACE2/Ang-(1-7)/Mas axis in the heart and the kidney of bGH mice could contribute to the increased incidence of hypertension, cardiovascular, and renal alterations observed in these animals.


Subject(s)
Angiotensin I/metabolism , Growth Hormone/metabolism , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin I/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Down-Regulation/genetics , Female , Growth Hormone/genetics , Hypertension/genetics , Hypertension/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
17.
Regul Pept ; 177(1-3): 1-11, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22561450

ABSTRACT

Angiotensin (Ang)-(1-7) stimulates proteins belonging to the insulin signaling pathway and ameliorates the Ang II negative effects at this level. However, up to date, receptors involved and mechanisms behind these observations remain unknown. Accordingly, in the present study, we explored the in vivo effects of antagonism of the Ang-(1-7) specific Mas receptor on insulin signal transduction in rat insulin-target tissues. We evaluated the acute modulation of insulin-stimulated phosphorylation of Akt, GSK-3ß (Glycogen synthase kinase-3ß) and AS160 (Akt substrate of 160kDa) by Ang-(1-7) and/or Ang II in the presence and absence of the selective Mas receptor antagonist A-779 in insulin-target tissues of normal rats. Also using A-779, we determined whether the Mas receptor mediates the improvement of insulin sensitivity exerted by chronic Ang-(1-7) treatment in fructose-fed rats (FFR), a model of insulin resistance, dyslipidemia and mild hypertension. The two major findings of the present work are as follows; 1) Ang-(1-7) attenuates acute Ang II-mediated inhibition of insulin signaling components in normal rats via a Mas receptor-dependent mechanism; and 2). The Mas receptor appears to be involved in beneficial effects of Ang-(1-7) on the phosphorylation of crucial insulin signaling mediators (Akt, GSK-3ß and AS160), in liver, skeletal muscle and adipose tissue of FFR. These results shed light into the mechanism by which Ang-(1-7) exerts its positive physiological modulation of insulin actions in classical metabolic tissues and reinforces the central role of Akt in these effects.


Subject(s)
Angiotensin I/pharmacology , Insulin/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Angiotensin I/administration & dosage , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Blood Pressure , Dyslipidemias/metabolism , Dyslipidemias/pathology , Fructose/administration & dosage , GTPase-Activating Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hypertension/metabolism , Hypertension/pathology , Insulin Resistance , Liver/drug effects , Liver/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Peptide Fragments/administration & dosage , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors
18.
Growth Horm IGF Res ; 22(6): 224-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22947377

ABSTRACT

OBJECTIVE: Growth hormone (GH) resistance leads to enhanced insulin sensitivity, decreased systolic blood pressure and increased lifespan. The aim of this study was to determine if there is a shift in the balance of the renin-angiotensin system (RAS) towards the ACE2/Ang-(1-7)/Mas receptor axis in the heart and the kidney of a model of GH resistance and retarded aging, the GH receptor knockout (GHR-/-) mouse. DESIGN: RAS components were evaluated in the heart and the kidney of GHR-/- and control mice by immunohistochemistry and Western blotting (n=12 for both groups). RESULTS: The immunostaining of Ang-(1-7) was increased in both the heart and the kidney of GHR-/- mice. These changes were concomitant with an increased immunostaining of the Mas receptor and ACE2 in both tissues. The immunostaining of AT1 receptor was reduced in heart and kidney of GHR-/- mice while that of AT2 receptor was increased in the heart and unaltered in the kidney. Ang II, ACE and angiotensinogen levels remained unaltered in the heart and the kidney of GH resistant mice. These results were confirmed by Western blotting and correlated with a significant increase in the abundance of the endothelial nitric oxide synthase in both tissues. CONCLUSIONS: The shift within the RAS towards an exacerbation of the ACE2/Ang-(1-7)/Mas receptor axis observed in GHR-/- mice could be related to a protective role in cardiac and renal function; and thus, possibly contribute to the decreased incidence of cardiovascular diseases displayed by this animal model of longevity.


Subject(s)
Angiotensin I/genetics , Kidney/metabolism , Myocardium/metabolism , Peptide Fragments/genetics , Peptidyl-Dipeptidase A/genetics , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Somatotropin/genetics , Up-Regulation , Angiotensin I/biosynthesis , Angiotensin-Converting Enzyme 2 , Animals , Mice , Mice, Knockout , Peptide Fragments/biosynthesis , Peptidyl-Dipeptidase A/biosynthesis , Proto-Oncogene Mas , Proto-Oncogene Proteins/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis
19.
J Hypertens ; 29(8): 1613-23, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21720265

ABSTRACT

OBJECTIVE: Atenolol, a first-generation ß-blocker, effectively reduces blood pressure, although its use in metabolic syndrome remains controversial. Accordingly, this study evaluated the renal effects of nebivolol, a third-generation ß-blocker with additional vasodilating activity, versus those of atenolol in an animal model of diabetic nephropathy. METHODS: Zucker diabetic fatty (ZDF) rats and control lean Zucker rats (LZRs) were treated for 6 months with either nebivolol or atenolol. Blood pressure, circulating insulin, triglycerides, cholesterol and glucose, as well as proteinuria and creatinine clearance were evaluated. Thiobarbituric acid-reactive species, reduced glutathione (GSH)/oxidized glutathione (GSSG) ratio, CuZn superoxide dismutase, catalase and glutathione peroxidase were determined as biomarkers of oxidative stress in kidney homogenates. Expression of transforming growth factor-ß1 (TGF-ß1), α-smooth muscle actin (α-SMA), collagen type I and III, plasminogen activator inhibitor-1 (PAI-1), vascular and platelet endothelial cell adhesion molecule-1 (VCAM-1 and PECAM-1, respectively) were determined by immunohistochemistry. Fibrosis was evaluated by light microscopy. RESULTS: Both drugs induced a similar control of blood pressure throughout the study. Contrary to atenolol, nebivolol showed a beneficial impact on lipid profile, preserved glomerular filtration rate, reduced proteinuria and induced a positive regulation of structural podocyte proteins (nephrin and podocin) expression. Additionally nebivolol decreased oxidative stress biomarkers, induced a substantial reduction in the accumulation of extracellular matrix proteins, down-regulated the renal expression of VCAM-1, monocyte chemotactic protein-1 (MCP-1), ED1, α-SMA, TGF-ß1 and PAI-1 and up-regulated the expression of PECAM-1. CONCLUSION: Our current finding underscores the importance of this therapy in hypertensive states concomitant with altered lipid and glucose metabolism.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/therapeutic use , Benzopyrans/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/prevention & control , Ethanolamines/therapeutic use , Metabolic Syndrome/complications , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Atenolol/pharmacology , Atenolol/therapeutic use , Benzopyrans/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Ethanolamines/pharmacology , Fibrosis , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Glucose/metabolism , Kidney/pathology , Kidney/physiopathology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Male , Nebivolol , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Zucker , Time Factors
20.
Regul Pept ; 161(1-3): 1-7, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20188769

ABSTRACT

The in vivo effect of angiotensin (ANG)-(1-7) on the activation of insulin signaling transduction in rat extracardiac tissues is unknown. Thus, in the present study, we evaluated the ability of ANG-(1-7) to stimulate the phosphorylation of Akt, a main mediator of insulin action in rat extracardiac tissues (adipose tissue, liver and skeletal muscle). We proved that ANG-(1-7) induces the phosphorylation of Akt at both threonine 308 and serine 473 in all tissues analyzed. Selective antagonism of the Mas receptor with A779 blocked the ANG-(1-7)-induced Akt phosphorylation in extracardiac tissues. Reinforcing this evidence, we determined that ANG-(1-7) induces the in vivo activation of the downstream target of Akt, glycogen synthase kinase-3beta in liver and skeletal muscle. Moreover, in every tissue analyzed, the presence of the Mas receptor was detected by immunohistochemical analysis. Based on the current results, we postulate that ANG-(1-7) could be a positive physiological contributor to the actions of insulin in extracardiac tissues. Therefore, our findings extend the possibilities for new approaches in the study of ANG-(1-7)/Mas receptor axis and show the therapeutic potential of ANG-(1-7) in the treatment of metabolic disorders such as insulin resistance as well as other disorders associated with diminished Akt activity.


Subject(s)
Angiotensin I/pharmacology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Adipose Tissue/metabolism , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunoblotting , Immunohistochemistry , In Vitro Techniques , Liver/drug effects , Liver/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL