Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Diabetol ; 11: 129, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23066908

ABSTRACT

BACKGROUND: The aim of this study was to investigate whether obestatin (OB), a peptide mediator encoded by the ghrelin gene exerting a protective effect in ischemic reperfused heart, is able to reduce cardiac dysfunctions in adult diabetic rats. METHODS: Diabetes was induced by STZ injection (50 mg/kg) in Wistar rats (DM). OB was administered (25 µg/kg) twice a day for 6 weeks. Non-diabetic (ND) rats and DM rats were distributed into four groups: untreated ND, OB-treated ND, untreated DM, OB-treated DM. Cardiac contractility and ß-adrenergic response were studied on isolated papillary muscles. Phosphorylation of AMPK, Akt, ERK1/2 and GSK3ß as well ß-1 adrenoreceptors levels were detected by western blot, while α-MHC was measured by RT-PCR. RESULTS: OB preserved papillary muscle contractility (85 vs 27% of ND), ß-adrenergic response (103 vs 65% of ND), as well ß1-adrenoreceptors and α-MHC levels in diabetic myocardial tissue. Moreover, OB up-regulated the survival kinases Akt and ERK1/2, and enhanced AMPK and GSK3ß phosphorylation. OB corrected oxidative unbalance, reduced pro-inflammatory cytokine TNF-α plasma levels, NFkB translocation and pro-fibrogenic factors expression in diabetic myocardium. CONCLUSIONS: OB displays a significant beneficial effect against the alterations of contractility and ß-adrenergic response in the heart of STZ-treated diabetic rats, which was mainly associated with the ability of OB to up-regulate the transcription of ß1-adrenergic receptors and α-MHC; this protective effect was accompanied by the ability to restore oxidative balance and to promote phosphorylation/modulation of AMPK and pro-survival kinases such as Akt, ERK1/2 and GSK3ß.


Subject(s)
Cardiotonic Agents/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Heart Diseases/drug therapy , Myocardial Contraction/drug effects , Papillary Muscles/drug effects , Peptide Hormones/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heart Diseases/etiology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Hypoglycemic Agents/pharmacology , Inflammation Mediators/blood , Male , Metformin/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Papillary Muscles/metabolism , Papillary Muscles/physiopathology , Phosphorylation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/metabolism , Recovery of Function , Time Factors , Tumor Necrosis Factor-alpha/blood , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
2.
Peptides ; 32(12): 2514-21, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22074955

ABSTRACT

Ghrelin is a gastric peptide, discovered by Kojima et al. (1999) [55] as a result of the search for an endogenous ligand interacting with the "orphan receptor" GHS-R1a (growth hormone secretagogue receptor type 1a). Ghrelin is composed of 28 aminoacids and is produced mostly by specific cells of the stomach, by the hypothalamus and hypophysis, even if its presence, as well as that of its receptors, has been demonstrated in many other tissues, not least in gonads. Ghrelin potently stimulates GH release and participates in the regulation of energy homeostasis, increasing food intake, decreasing energy output and exerting a lipogenetic effect. Furthermore, ghrelin influences the secretion and motility of the gastrointestinal tract, especially of the stomach, and, above all, profoundly affects pancreatic functions. Despite of these previously envisaged activities, it has recently been hypothesized that ghrelin regulates several aspects of reproductive physiology and pathology. In conclusion, ghrelin not only cooperates with other neuroendocrine factors, such as leptin, in the modulation of energy homeostasis, but also has a crucial role in the regulation of the hypothalamic-pituitary gonadal axis. In the current review we summarize the main targets of this gastric peptide, especially focusing on the reproductive system.


Subject(s)
Energy Metabolism , Ghrelin/physiology , Reproduction , Animals , Female , Ghrelin/blood , Ghrelin/pharmacology , Gonadotropins/metabolism , Gonads/physiology , Homeostasis , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Male , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Pregnancy , Puberty/drug effects , Puberty/physiology , Rats , Receptors, Ghrelin/genetics , Receptors, Ghrelin/physiology
3.
Biochim Biophys Acta ; 1811(6): 386-96, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21435395

ABSTRACT

The acylated peptide ghrelin (AG) and its endogenous non-acylated isoform (UAG) protect cardiomyocytes, pancreatic ß-cells, and preadipocytes from apoptosis, and induce preadipocytes differentiation into adipocytes. These events are mediated by AG and UAG binding to a still unidentified receptor, which determines the activation of phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) ERK1/2. AG and UAG also possess antilipolytic activity in vitro, but the underlying mechanism remains unknown. Thus, the objective of the current study was to characterize the molecular events involved in AG/UAG receptor signaling cascade. We treated rat primary visceral adipocytes with isoproterenol (ISO) and forskolin (FSK) to stimulate lipolysis, simultaneously incubating them with or without AG or UAG. Both peptides blocked ISO- and FSK-induced lipolysis. By direct measurement of cAMP intracellular content, we demonstrated that AG/UAG effect was associated to a reduction of ISO-induced cAMP accumulation. Moreover, the cAMP analog 8Br-cAMP abolished AG/UAG effect. As AG and UAG were ineffective against lipolysis induced by db-cAMP, another poorly hydrolyzable cAMP analog, phosphodiesterase (PDE) involvement was hypothesized. Indeed, cilostamide, a specific PDE3B inhibitor, blocked AG/UAG effect on ISO-induced lipolysis. Furthermore, the PI3K inhibitor wortmannin and AKT inhibitor 1,3-dihydro-1-(1-((4-(6-phenyl-1H-imidazo(4,5-g)quinoxalin-7-yl)phenyl)methyl)-4piperidinyl)-2H-benzimidazol-2-one trifluoroacetate also blocked AG/UAG action, suggesting a role in PDE3B activation. In particular, PI3K isoenzyme gamma (PI3Kγ) selective inhibition through the compound AS605240 prevented AG/UAG effect on ISO-stimulated lipolysis, hampering AKT phosphorylation on Ser(473). Taken together, these data demonstrate for the first time that AG/UAG attenuation of ISO-induced lipolysis involves PI3Kγ/AKT and PDE3B.


Subject(s)
Adipocytes/drug effects , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Ghrelin/pharmacology , Isoproterenol/pharmacology , Lipolysis/drug effects , Acylation , Adipocytes/cytology , Adipocytes/metabolism , Adrenergic beta-Agonists/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Benzimidazoles/pharmacology , Blotting, Western , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Ghrelin/metabolism , Glycerol/metabolism , Intra-Abdominal Fat/cytology , Isoproterenol/metabolism , Male , Phosphodiesterase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinolones/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Wistar , Thiazolidinediones/pharmacology
4.
Am J Physiol Heart Circ Physiol ; 299(2): H470-81, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20525876

ABSTRACT

Obestatin, a newly discovered peptide encoded by the ghrelin gene, induces the expression of genes regulating pancreatic beta-cell differentiation, insulin biosynthesis, and glucose metabolism. It also activates antiapoptotic signaling pathways such as phosphoinositide 3-kinase (PI3K) and ERK1/2 in pancreatic beta-cells and human islets. Since these kinases have been shown to protect against myocardial injury, we sought to investigate whether obestatin would exert cardioprotective effects. Both isolated perfused rat heart and cultured cardiomyocyte models of ischemia-reperfusion (I/R) were used to measure infarct size and cell apoptosis as end points of injury. The presence of specific obestatin receptors on cardiac cells as well as the signaling pathways underlying the obestatin effect were also studied. In the isolated heart, the addition of rat obestatin-(1-23) before ischemia reduced infarct size and contractile dysfunction in a concentration-dependent manner, whereas obestatin-(23-1), a synthetic analog with an inverse aminoacid sequence, was ineffective. The cardioprotective effect of obestatin-(1-23) was observed at concentrations of 10-50 nmol/l and was abolished by inhibiting PI3K or PKC by the addition of wortmannin (100 nmol/l) or chelerythrine, (5 micromol/l), respectively. In rat H9c2 cardiac cells or isolated ventricular myocytes subjected to I/R, 50 nmol/l obestatin-(1-23) reduced cardiomyocyte apoptosis and reduced caspase-3 activation; the antiapoptotic effect was blocked by the inhibition of PKC, PI3K, or ERK1/2 pathways. In keeping with these functional findings, radioreceptor binding results revealed the presence of specific high-affinity obestatin-binding sites, mainly localized on membranes of the ventricular myocardium and cardiomyocytes. Our data suggest that, by acting on specific receptors, obestatin-(1-23) activates PI3K, PKC-epsilon, PKC-delta, and ERK1/2 signaling and protects cardiac cells against myocardial injury and apoptosis induced by I/R.


Subject(s)
Apoptosis , Myocardial Contraction , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Peptide Hormones/metabolism , Ventricular Function, Left , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptide Hormones/administration & dosage , Peptides/pharmacology , Perfusion , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Receptors, Ghrelin/metabolism , Signal Transduction , Time Factors , Ventricular Function, Left/drug effects
5.
Endocrinology ; 151(7): 3286-98, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410201

ABSTRACT

The current study aimed to compare the effects of the peptide hormone ghrelin and des-G, its unacylated isoform, on glucose and fatty acid uptake and to identify des-G-specific binding sites in cardiomyocytes. In the murine HL-1 adult cardiomyocyte line, ghrelin and des-G had opposing metabolic effects: des-G increased medium-chain fatty acid uptake (BODIPY fluorescence intensity), whereas neither ghrelin alone nor in combination with des-G did so. Ghrelin inhibited the increase in glucose uptake normally induced by insulin (rate of 2-[(3)H]deoxy-d-glucose incorporation), but des-G did not; des-G was also able to partially reverse the inhibitory effect of ghrelin. In HL-1 cells and primary cultures of neonatal rat cardiomyocytes, des-G but not ghrelin increased insulin-induced translocation of glucose transporter-4 from nuclear to cytoplasmic compartments (immunohistochemistry and quantitative confocal analysis). AKT was phosphorylated by insulin but not affected by ghrelin or des-G, whereas neither AMP-activated protein kinase nor phosphatase and tensin homolog deleted from chromosome 10 was phosphorylated by any treatments. HL-1 and primary-cultured mouse and rat cardiomyocytes each possessed two independent specific binding sites for des-G not recognized by ghrelin (radioreceptor assays). Neither ghrelin nor des-G affected viability (dimethylthiazol diphenyltetrazolium bromide assays), whereas both isoforms were equally protective against apoptosis. Therefore, in cardiomyocytes, des-G binds to specific receptors and has effects on glucose and medium-chain fatty acid uptake that are distinct from those of ghrelin. Real-time PCR indicated that expression levels of ghrelin O-acyltransferase RNA were comparable between HL-1 cells, human myocardial tissue, and human and murine stomach tissue, indicating the possibility of des-G conversion to ghrelin within our model.


Subject(s)
Ghrelin/metabolism , Ghrelin/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Binding Sites , Biological Transport/drug effects , Cell Line , Gastric Mucosa/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Immunoblotting , Immunohistochemistry , Insulin/pharmacology , Lauric Acids/metabolism , Mice , Microscopy, Confocal , Myocytes, Cardiac/cytology , PTEN Phosphohydrolase/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Rats
6.
Diabetes ; 57(4): 967-79, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18162507

ABSTRACT

OBJECTIVE: Obestatin is a newly discovered peptide encoded by the ghrelin gene whose biological functions are poorly understood. We investigated obestatin effect on survival of beta-cells and human pancreatic islets and the underlying signaling pathways. RESEARCH DESIGN AND METHODS: beta-Cells and human islets were used to assess obestatin effect on cell proliferation, survival, apoptosis, intracellular signaling, and gene expression. RESULTS: Obestatin showed specific binding on HIT-T15 and INS-1E beta-cells, bound to glucagon-like peptide-1 receptor (GLP-1R), and recognized ghrelin binding sites. Obestatin exerted proliferative, survival, and antiapoptotic effects under serum-deprived conditions and interferon-gamma/tumor necrosis factor-alpha/interleukin-1 beta treatment, particularly at pharmacological concentrations. Ghrelin receptor antagonist [D-Lys(3)]-growth hormone releasing peptide-6 and anti-ghrelin antibody prevented obestatin-induced survival in beta-cells and human islets. beta-Cells and islet cells released obestatin, and addition of anti-obestatin antibody reduced their viability. Obestatin increased beta-cell cAMP and activated extracellular signal-related kinase 1/2 (ERK1/2) and phosphatidylinositol 3-kinase (PI 3-kinase)/Akt; its antiapoptotic effect was blocked by inhibition of adenylyl cyclase/cAMP/protein kinase A (PKA), PI 3-kinase/Akt, and ERK1/2 signaling. Moreover, obestatin upregulated GLP-1R mRNA and insulin receptor substrate-2 (IRS-2) expression and phosphorylation. The GLP-1R antagonist exendin-(9-39) reduced obestatin effect on beta-cell survival. In human islets, obestatin, whose immunoreactivity colocalized with that of ghrelin, promoted cell survival and blocked cytokine-induced apoptosis through cAMP increase and involvement of adenylyl cyclase/cAMP/PKA signaling. Moreover, obestatin 1) induced PI 3-kinase/Akt, ERK1/2, and also cAMP response element-binding protein phosphorylation; 2) stimulated insulin secretion and gene expression; and 3) upregulated GLP-1R, IRS-2, pancreatic and duodenal homeobox-1, and glucokinase mRNA. CONCLUSIONS: These results indicate that obestatin promotes beta-cell and human islet cell survival and stimulates the expression of main regulatory beta-cell genes, identifying a new role for this peptide within the endocrine pancreas.


Subject(s)
Cell Survival/drug effects , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Peptide Hormones/pharmacology , Caspase 3/metabolism , Cell Culture Techniques , Cell Division/drug effects , Cell Membrane/drug effects , Cell Membrane/physiology , Cyclic AMP/metabolism , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Insulin Receptor Substrate Proteins , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/drug effects , Peptide Hormones/metabolism , Phosphoproteins/drug effects , Phosphoproteins/metabolism
7.
Mol Biol Cell ; 18(3): 986-94, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17202410

ABSTRACT

Ghrelin is an acylated peptidyl gastric hormone acting on the pituitary and hypothalamus to stimulate appetite, adiposity, and growth hormone release, through activation of growth hormone secretagogue receptor (GHSR)-1a receptor. Moreover, ghrelin features several activities such as inhibition of apoptosis, regulation of differentiation, and stimulation or inhibition of proliferation of several cell types. Ghrelin acylation is absolutely required for both GHSR-1a binding and its central endocrine activities. However, the unacylated ghrelin form, des-acyl ghrelin, which does not bind GHSR-1a and is devoid of any endocrine activity, is far more abundant than ghrelin in plasma, and it shares with ghrelin some of its cellular activities. In here we show that both ghrelin and des-acyl ghrelin stimulate proliferating C2C12 skeletal myoblasts to differentiate and to fuse into multinucleated myotubes in vitro through activation of p38. Consistently, both ghrelin and des-acyl ghrelin inhibit C2C12 proliferation in growth medium. Moreover, the ectopic expression of ghrelin in C2C12 enhances differentiation and fusion of these myoblasts in differentiation medium. Finally, we show that C2C12 cells do not express GHSR-1a, but they do contain a common high-affinity binding site recognized by both acylated and des-acylated ghrelin, suggesting that the described activities on C2C12 are likely mediated by this novel, yet unidentified receptor for both ghrelin forms.


Subject(s)
Cell Differentiation/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Peptide Hormones/pharmacology , Animals , Binding Sites/drug effects , Biomarkers , Cell Fusion , Cell Proliferation/drug effects , Culture Media , DNA/biosynthesis , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Ghrelin , Mice , Muscle Fibers, Skeletal/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...