Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 156(3): 858-68, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25535832

ABSTRACT

Recent evidence suggests that peripheral ghrelin regulates glucose metabolism. Here, we designed experiments to examine how central acyl ghrelin infusion affects peripheral glucose metabolism under pair-fed or ad libitum feeding conditions. Mice received intracerebroventricular (icv) infusion of artificial cerebrospinal fluid (aCSF), ghrelin, and allowed to eat ad libitum (icv ghrelin ad lib) or ghrelin and pair-fed to the aCSF group (icv ghrelin pf). Minipumps delivered acyl ghrelin at a dose of 0.25 µg/h at 0.5 µL/h for 7 days. There was no difference in daily blood glucose, insulin, glucagon, triglycerides, or nonesterified fatty acids. Body weight gain and food intake was significantly higher in icv ghrelin ad lib mice. However, both icv ghrelin ad lib and icv ghrelin pf groups exhibited heavier white adipose mass. Icv ghrelin pf mice exhibited better glucose tolerance than aCSF or icv ghrelin ad lib mice during a glucose tolerance test, although both icv ghrelin ad lib and icv ghrelin pf increased insulin release during the glucose tolerance test. Central acyl ghrelin infusion and pair feeding also increased breakdown of liver glycogen and triglyceride, and regulated genes involved in hepatic lipid and glucose metabolism. Icv ghrelin pf mice had an increase in plasma blood glucose during a pyruvate tolerance test relative to icv ghrelin ad lib or aCSF mice. Our results suggest that under conditions of negative energy (icv ghrelin pf), central acyl ghrelin engages a neural circuit that influences hepatic glucose function. Metabolic status affects the ability of central acyl ghrelin to regulate peripheral glucose homeostasis.


Subject(s)
Ghrelin/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Liver/drug effects , Liver/metabolism , Acylation , Animals , Blood Glucose/metabolism , Enzyme-Linked Immunosorbent Assay , Ghrelin/blood , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Leptin/blood , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Pyruvic Acid/metabolism
2.
Endocrinology ; 154(12): 4737-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24105484

ABSTRACT

The WNT pathway has been well characterized in embryogenesis and tumorigenesis. In humans, specific polymorphisms in the T cell-specific transcription factor 7 and the WNT coreceptor, low-density lipoprotein receptor-related protein-6 (LRP-6), both prominent components of this pathway, correlate with a higher incidence of type 2 diabetes, suggesting that the WNT pathway might be involved in the control of adult glucose homeostasis. We previously demonstrated that glycogen-synthase-kinase-3ß (GSK-3ß), the key enzyme of the WNT pathway, is increased in the hypothalamus during obesity and exacerbates high-fat diet-induced weight gain as well as glucose intolerance. These data suggest that WNT action in the hypothalamus might be required for normal glucose homeostasis. Here we characterized whether WNT signaling in general is altered in the hypothalamus of adult obese mice relative to controls. First we identified expression of multiple components of this pathway in the murine arcuate nucleus by in situ hybridization. In this region mRNA of ligands and target genes of the WNT pathway were down-regulated in obese and glucose-intolerant leptin-deficient mice. Similarly, the number of cells immunoreactive for the phosphorylated (active) form of the WNT-coreceptor LRP-6 was also decreased in leptin-deficient mice. Leptin treatment normalized expression of the WNT-target genes Axin-2 and Cylin-D1 and increased the number of phospho-LRP-6-immunoreactive cells reaching levels of lean controls. Leptin also increased the levels of phosphorylated (inactive) GSK-3ß in the arcuate nucleus, and this effect was colocalized to neuropeptide Y neurons, suggesting that inactivation of GSK-3ß may contribute to the neuroendocrine control of energy homeostasis. Taken together our findings identify hypothalamic WNT signaling as an important novel pathway that integrates peripheral information of the body's energy status encoded by leptin.


Subject(s)
Hypothalamus/physiology , Leptin/pharmacology , Obesity/drug therapy , Obesity/metabolism , Wnt Signaling Pathway/physiology , Aging/physiology , Animals , Animals, Newborn , Chromatin , Female , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Inbred Strains , Myocardium/metabolism , Obesity/genetics , Pregnancy , Prenatal Exposure Delayed Effects , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Testis/metabolism , Testosterone/pharmacology
3.
J Neurosci ; 30(48): 16180-7, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123564

ABSTRACT

Obesity is associated with resistance to the actions of both leptin and insulin via mechanisms that remain incompletely understood. To investigate whether leptin resistance per se contributes to insulin resistance and impaired glucose homeostasis, we investigated the effect of acute leptin administration on glucose homeostasis in normal as well as leptin- or leptin receptor-deficient mice. In hyperglycemic, leptin-deficient Lep(ob/ob) mice, leptin acutely and potently improved glucose metabolism, before any change of body fat mass, via a mechanism involving the p110α and ß isoforms of phosphatidylinositol-3-kinase (PI3K). Unlike insulin, however, the anti-diabetic effect of leptin occurred independently of phospho-AKT, a major downstream target of PI3K, and instead involved enhanced sensitivity of the hypothalamus to insulin action upstream of PI3K, through modulation of IRS1 (insulin receptor substrate 1) phosphorylation. These data suggest that leptin resistance, as occurs in obesity, reduces the hypothalamic response to insulin and thereby impairs peripheral glucose homeostasis, contributing to the development of type 2 diabetes.


Subject(s)
Glucose/metabolism , Homeostasis/physiology , Hypothalamus/metabolism , Insulin Resistance/physiology , Leptin/deficiency , Obesity/metabolism , Adipose Tissue/enzymology , Adipose Tissue/physiopathology , Animals , Blood Glucose/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/physiology , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/physiology , Homeostasis/genetics , Hypothalamus/enzymology , Insulin Resistance/genetics , Isoenzymes/genetics , Isoenzymes/physiology , Leptin/genetics , Male , Mice , Mice, Knockout , Mice, Obese , Obesity/enzymology , Obesity/genetics , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...