Obestatin, a newly discovered peptide encoded by the ghrelin gene, was originally identified as the ligand for the orphan G protein-coupled receptor GPR39. In vivo, obestatin was reported to have opposite actions to ghrelin in the regulation of food intake, gastric emptying and body weight; however, these effects, as well as obestatin binding to GPR39, have been questioned by several groups. At the cellular level, we recently showed that in β-cells and human pancreatic islets, obestatin enhances proliferation, reduces apoptosis and promotes insulin secretion, suggesting a role in β-cell function and glucose metabolism. Moreover, other authors have described obestatin-induced c-fos expression in cultured pre-adipocytes, suggesting a role in adipocyte function. Aim of this study was to determine the role of obestatin on glucose uptake and isoproterenol-induced lipolysis in adipocytes. Obestatin protein was expressed in and secreted by human adipocytes. Cytofluorimetric and confocal microscopy analysis evidenced obestatin binding on 3T3-L1 cells and human adipocytes. Moreover, both GPR39 mRNA and protein were found expressed in 3T3-L1 adipocytes and human abdominal subcutaneous and visceral adipose tissue. Obestatin displayed survival action in 3T3-L1 preadipocytes, by reducing serum starvation-induced apoptosis through increased AC/cAMP/PKA signaling and activation of ERK1/2 and PI3K/Akt pathways. No effect of obestatin was observed with regard to adipocyte differentiation. On the other hand, in differentiated 3T3-L1 adipocytes, obestatin increased GLUT4 mRNA expression, induced glucose transporter-4 (GLUT4) translocation to the plasma membrane and stimulated insulin-induced glucose uptake either alone or in the presence of insulin. Accordingly, in these cells obestatin also activated AMP-activated protein kinase (AMPK), which is known to play a key role in the regulation of glucose uptake. Finally, obestatin strongly reduced isoproterenol-induced lypolisis in either 3T3-L1 adipocytes or mature human subcutaneous and visceral adipocytes, isolated from both obese and nonobese subjects. These results firstly indicate that obestatin plays a role in adipocyte metabolism, possessing insulin-mimetic properties, such as stimulation of GLUT-4 translocation, glucose uptake and inhibition of lypolisis. Together with the data previously shown in pancreatic β-cells, these findings reinforce the concept that obestatin exerts specific and relevant metabolic actions.

Obestatin promotes GLUT-4 translocation and glucose uptake in 3T3-L1 adipocytes and inhibits lipolysis in 3T3-L1 and human visceral and subcutaneous adipocytes

SCARLATTI, FRANCESCA;GALLO, Davide;GARETTO, STEFANO;SETTANNI, Fabio;BERGANDI, Loredana;VOLANTE, Marco;ANNUNZIATA, Marta;TALIANO, Marina;ROCCHIETTO, STEFANO;MATERA, Lina;MORINO, Mario;BOSIA, Amalia;PAPOTTI, Mauro Giulio;GHIGO, Ezio;GRANATA, Riccarda
2009-01-01

Abstract

Obestatin, a newly discovered peptide encoded by the ghrelin gene, was originally identified as the ligand for the orphan G protein-coupled receptor GPR39. In vivo, obestatin was reported to have opposite actions to ghrelin in the regulation of food intake, gastric emptying and body weight; however, these effects, as well as obestatin binding to GPR39, have been questioned by several groups. At the cellular level, we recently showed that in β-cells and human pancreatic islets, obestatin enhances proliferation, reduces apoptosis and promotes insulin secretion, suggesting a role in β-cell function and glucose metabolism. Moreover, other authors have described obestatin-induced c-fos expression in cultured pre-adipocytes, suggesting a role in adipocyte function. Aim of this study was to determine the role of obestatin on glucose uptake and isoproterenol-induced lipolysis in adipocytes. Obestatin protein was expressed in and secreted by human adipocytes. Cytofluorimetric and confocal microscopy analysis evidenced obestatin binding on 3T3-L1 cells and human adipocytes. Moreover, both GPR39 mRNA and protein were found expressed in 3T3-L1 adipocytes and human abdominal subcutaneous and visceral adipose tissue. Obestatin displayed survival action in 3T3-L1 preadipocytes, by reducing serum starvation-induced apoptosis through increased AC/cAMP/PKA signaling and activation of ERK1/2 and PI3K/Akt pathways. No effect of obestatin was observed with regard to adipocyte differentiation. On the other hand, in differentiated 3T3-L1 adipocytes, obestatin increased GLUT4 mRNA expression, induced glucose transporter-4 (GLUT4) translocation to the plasma membrane and stimulated insulin-induced glucose uptake either alone or in the presence of insulin. Accordingly, in these cells obestatin also activated AMP-activated protein kinase (AMPK), which is known to play a key role in the regulation of glucose uptake. Finally, obestatin strongly reduced isoproterenol-induced lypolisis in either 3T3-L1 adipocytes or mature human subcutaneous and visceral adipocytes, isolated from both obese and nonobese subjects. These results firstly indicate that obestatin plays a role in adipocyte metabolism, possessing insulin-mimetic properties, such as stimulation of GLUT-4 translocation, glucose uptake and inhibition of lypolisis. Together with the data previously shown in pancreatic β-cells, these findings reinforce the concept that obestatin exerts specific and relevant metabolic actions.
2009
The Endocrine Society's 91st Annual Meeting
Washington DC,
June 10-13, 2009
Endocrine Society Annual Meeting Abstract Book
Endocrine Society
-
-
-
-
http://www.endo-society.org/endo09/
Francesca Scarlatti; Davide Gallo; Stefano Garetto; Fabio Settanni; Loredana Bergandi; Marco Volante; Marta Annunziata; Marina Taliano; Stefano Rocchietto; Lina Matera; Giacomo Datta; Mario Morino; Amalia Bosia; Mauro Papotti; Ezio Ghigo; Riccarda Granata
File in questo prodotto:
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/2318/76655
Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
social impact