Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 294(3): G610-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18048479

ABSTRACT

Administration of chemically synthesized ghrelin (Ghr) peptide has been shown to increase food intake and body adiposity in most species. However, the biological role of endogenous Ghr in the molecular control of energy metabolism is far less understood. Mice deficient for either Ghr or its receptor (the growth hormone secretagogue receptor, GHS-R1a) seem to exhibit enhanced protection against high-fat diet-induced obesity but do not show a substantial metabolic phenotype on a standard diet. Here we present the first mouse mutant lacking both Ghr and the Ghr receptor. We demonstrate that simultaneous genetic disruption of both genes of the Ghr system leads to an enhanced energy metabolism phenotype. Ghr/Ghr receptor double knockout (dKO) mice exhibit decreased body weight, increased energy expenditure, and increased motor activity on a standard diet without exposure to a high caloric environment. Mice on the same genetic background lacking either the Ghr or the Ghr receptor gene did not exhibit such a phenotype on standard chow, thereby confirming earlier reports. No differences in food intake, meal pattern, or lean mass were observed between dKO, Ghr-deficient, Ghr receptor-deficient, and wild-type (WT) control mice. Only dKO showed a slight decrease in body length. In summary, simultaneous deletion of Ghr and its receptor enhances the metabolic phenotype of single gene-deficient mice compared with WT mice, possibly suggesting the existence of additional, as of yet unknown, molecular components of the endogenous Ghr system.


Subject(s)
Energy Metabolism/physiology , Ghrelin/deficiency , Ghrelin/genetics , Motor Activity/physiology , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics , Alleles , Animals , Anthropometry , Blood Glucose/metabolism , Body Composition/genetics , Body Composition/physiology , Body Temperature/physiology , Body Weight/genetics , Body Weight/physiology , Eating/genetics , Eating/physiology , Gene Deletion , Genotype , Glucose Tolerance Test , Insulin Resistance/genetics , Ligands , Lipids/blood , Mice , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
Gastroenterology ; 133(5): 1534-43, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17920065

ABSTRACT

BACKGROUND & AIMS: Peptide YY (PYY) and pancreatic polypeptide (PPY) are members of the neuropeptide Y peptide family. The neuropeptide Y receptor signaling pathway has been implicated in a number of physiologic processes, including the regulation of energy balance and bone mass. To investigate the contribution of endogenous PYY and PPY to these processes, we generated both Pyy- and Ppy-deficient mice. METHODS: Pyy(-/-) and Ppy(-/-) mice and their respective wild-type littermates were studied from 8 weeks to 9 months of age. Food intake, metabolic parameters, and locomotor activity were monitored using indirect calorimetry. Body composition and bone parameters were analyzed using dual energy x-ray absorptiometry, histomorphometry, and vertebral compression testing. RESULTS: Studies in these mice showed an osteopenic phenotype specific to the Pyy-deficient line, which included a reduction in trabecular bone mass and a functional deficit in bone strength. Furthermore, female Pyy(-/-) mice showed a greater sensitivity to ovariectomy-induced bone loss compared with wild-type littermates. No food intake or metabolic phenotype was apparent in male or female Pyy(-/-) mice on standard chow. However, female Pyy(-/-) mice on a high-fat diet showed a greater propensity to gain body weight and adiposity. No metabolic or osteopenic phenotype was observed in Ppy-deficient mice. CONCLUSIONS: These results indicate that endogenous PYY plays a critical role in regulating bone mass. In comparison, its role in regulating body weight is minor and is confined to situations of high-fat feeding.


Subject(s)
Bone Density/physiology , Bone and Bones/metabolism , Energy Metabolism/physiology , Peptide YY/physiology , Absorptiometry, Photon , Adiposity/drug effects , Animals , Body Composition/genetics , Body Composition/physiology , Body Weight/drug effects , Bone Density/genetics , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/physiopathology , Dietary Fats/pharmacology , Disease Models, Animal , Energy Metabolism/genetics , Female , Gene Deletion , Male , Mice , Mice, Transgenic , Ovariectomy , Peptide YY/genetics , Phenotype , Spine/pathology , Spine/physiopathology
3.
Cell Metab ; 2(6): 421-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16330327

ABSTRACT

Endogenous modulators of the central melanocortin system, such as the agouti-related protein (AgRP), should hold a pivotal position in the regulation of energy intake and expenditure. Despite this, AgRP-deficient mice were recently reported to exhibit normal food intake, body weight gain, and energy expenditure. Here we demonstrate that 2- to 3-month-old Agrp null mice do in fact exhibit subtle changes in response to feeding challenges (fasting and MCR agonists) but, of more significance and magnitude, exhibit reduced body weight and adiposity after 6 months of age. This age-dependent lean phenotype is correlated with increased metabolic rate, body temperature, and locomotor activity and increased circulating thyroid hormone (T4 and T3) and BAT UCP-1 expression. These results provide further proof of the importance of the AgRP neuronal system in the regulation of energy homeostasis.


Subject(s)
Proteins/genetics , Proteins/physiology , Adipose Tissue/metabolism , Adrenal Glands/metabolism , Aging , Agouti-Related Protein , Animals , Body Composition , Body Temperature , Body Weight , Brain/metabolism , Calorimetry , Feeding Behavior , Gene Expression Regulation , Genetic Vectors , Intercellular Signaling Peptides and Proteins , Lac Operon , Mice , Mice, Transgenic , Models, Genetic , Neurons/metabolism , Phenotype , Thyroid Hormones/metabolism , Time Factors , beta-Galactosidase/metabolism
4.
J Clin Invest ; 115(12): 3573-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322795

ABSTRACT

The gut peptide ghrelin, the endogenous ligand for the growth hormone secretagogue receptor, has been implicated not only in the regulation of pituitary growth hormone (GH) secretion but in a number of endocrine and nonendocrine functions, including appetitive behavior and carbohydrate substrate utilization. Nevertheless, recent genetic studies have failed to show any significant defects in GH levels, food intake, or body weight in adult ghrelin-deficient (Ghrl-/-) mice. Here we demonstrate that male Ghrl-/- mice are protected from the rapid weight gain induced by early exposure to a high-fat diet 3 weeks after weaning (6 weeks of age). This reduced weight gain was associated with decreased adiposity and increased energy expenditure and locomotor activity as the animals aged. Despite the absence of ghrelin, these Ghrl-/- mice showed a paradoxical preservation of the GH/IGF-1 axis, similar to that reported in lean compared with obese humans. These findings suggest an important role for endogenous ghrelin in the metabolic adaptation to nutrient availability.


Subject(s)
Obesity/genetics , Peptide Hormones/physiology , Animal Feed , Animals , Body Composition , Body Weight , Calorimetry , Carbohydrates/chemistry , Diet , Genotype , Ghrelin , Growth Hormone/metabolism , Heterozygote , Insulin-Like Growth Factor I/metabolism , Ligands , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neuropeptides/chemistry , Oxygen/metabolism , Peptide Hormones/metabolism , Phenotype , Pituitary Gland/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
5.
Proc Natl Acad Sci U S A ; 102(7): 2496-501, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699348

ABSTRACT

We identified a glycoprotein hormone beta-subunit (OGH, also called GPB5) that, as a heterodimer with the alpha-subunit GPA2, serves as a second ligand for the thyroid-stimulating hormone receptor. Mice in which the OGH gene is deleted (OGH-/-) are indistinguishable from WT littermates in body weight, response to high-fat diet, metabolic parameters, body composition, and insulin tolerance. Mice engineered to transgenically globally overexpress OGH (OGH-TG) develop approximately 2-fold elevations in their basal thyroid levels and weigh slightly less than WT littermates despite increased food intake because of an increase in their metabolic rates. Moreover, when OGH-TG mice are challenged with a high-fat diet, they gain significantly less weight and body fat than their WT littermates. The OGH-TG mice also have reduced blood glucose, insulin, cholesterol, and triglycerides. In contrast to other approaches in which the thyroid axis is activated, OGH-TG mice exhibit only minor changes in heart rate and blood pressure. Our findings suggest that constitutive low-level activation of the thyroid axis (via OGH or other means) may provide a beneficial therapeutic approach for combating diet-induced obesity.


Subject(s)
Glycoproteins/genetics , Obesity/genetics , Peptide Hormones/genetics , Animals , Body Weight , Dietary Fats/administration & dosage , Gene Expression , Genes, Reporter , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Molecular Sequence Data , Obesity/metabolism , Obesity/pathology , Phenotype
6.
Nat Med ; 11(2): 199-205, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15654325

ABSTRACT

Genetic ablation of Inppl1, which encodes SHIP2 (SH2-domain containing inositol 5-phosphatase 2), was previously reported to induce severe insulin sensitivity, leading to early postnatal death. In the previous study, the targeting construct left the first eighteen exons encoding Inppl1 intact, generating a Inppl1(EX19-28-/-) mouse, and apparently also deleted a second gene, Phox2a. We report a new SHIP2 knockout (Inppl1(-/-)) targeted to the translation-initiating ATG, which is null for Inppl1 mRNA and protein. Inppl1(-/-) mice are viable, have normal glucose and insulin levels, and normal insulin and glucose tolerances. The Inppl1(-/-) mice are, however, highly resistant to weight gain when placed on a high-fat diet. These results suggest that inhibition of SHIP2 would be useful in the effort to ameliorate diet-induced obesity, but call into question a dominant role of SHIP2 in modulating glucose homeostasis.


Subject(s)
Dietary Fats/metabolism , Obesity/metabolism , Phosphoric Monoester Hydrolases/metabolism , Animals , Blood Chemical Analysis , Body Weight , Exons , Female , Gene Deletion , Genes, Reporter , Glucose/metabolism , Homeostasis , Inositol Polyphosphate 5-Phosphatases , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Signal Transduction , Tissue Distribution
7.
Peptides ; 25(3): 473-504, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15134868

ABSTRACT

Energy balance is maintained via a homeostatic system involving both the brain and the periphery. A key component of this system is the hypothalamus. Over the past two decades, major advances have been made in identifying an increasing number of peptides within the hypothalamus that contribute to the process of energy homeostasis. Under stable conditions, equilibrium exists between anabolic peptides that stimulate feeding behavior, as well as decrease energy expenditure and lipid utilization in favor of fat storage, and catabolic peptides that attenuate food intake, while stimulating sympathetic nervous system (SNS) activity and restricting fat deposition by increasing lipid metabolism. The equilibrium between these neuropeptides is dynamic in nature. It shifts across the day-night cycle and from day to day and also in response to dietary challenges as well as peripheral energy stores. These shifts occur in close relation to circulating levels of the hormones, leptin, insulin, ghrelin and corticosterone, and also the nutrients, glucose and lipids. These circulating factors together with neural processes are primary signals relaying information regarding the availability of fuels needed for current cellular demand, in addition to the level of stored fuels needed for long-term use. Together, these signals have profound impact on the expression and production of neuropeptides that, in turn, initiate the appropriate anabolic or catabolic responses for restoring equilibrium. In this review, we summarize the evidence obtained on nine peptides in the hypothalamus that have emerged as key players in this process. Data from behavioral, physiological, pharmacological and genetic studies are described and consolidated in an attempt to formulate a clear statement on the underlying function of each of these peptides and also on how they work together to create and maintain energy homeostasis.


Subject(s)
Body Weight/physiology , Feeding Behavior/physiology , Hypothalamus/physiology , Peptides/physiology , Animals , Homeostasis/physiology , Humans
8.
Proc Natl Acad Sci U S A ; 101(21): 8227-32, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15148384

ABSTRACT

Ghrelin is a recently identified growth hormone (GH) secretogogue whose administration not only induces GH release but also stimulates food intake, increases adiposity, and reduces fat utilization in mice. The effect on food intake appears to be independent of GH release and instead due to direct activation of orexigenic neurons in the arcuate nucleus of the hypothalamus. The effects of ghrelin administration on food intake have led to the suggestion that inhibitors of endogenous ghrelin could be useful in curbing appetite and combating obesity. To further study the role of endogenous ghrelin in appetite and body weight regulation, we generated ghrelin-deficient (ghrl(-/-)) mice, in which the ghrelin gene was precisely replaced with a lacZ reporter gene. ghrl(-/-) mice were viable and exhibited normal growth rates as well as normal spontaneous food intake patterns, normal basal levels of hypothalamic orexigenic and anorexigenic neuropeptides, and no impairment of reflexive hyperphagia after fasting. These results indicate that endogenous ghrelin is not an essential regulator of food intake and has, at most, a redundant role in the regulation of appetite. However, analyses of ghrl(-/-) mice demonstrate that endogenous ghrelin plays a prominent role in determining the type of metabolic substrate (i.e., fat vs. carbohydrate) that is used for maintenance of energy balance, particularly under conditions of high fat intake.


Subject(s)
Energy Metabolism , Feeding Behavior/physiology , Gene Deletion , Peptide Hormones/deficiency , Animals , Appetite/physiology , Body Weight , Carbohydrate Metabolism , Eating , Fasting , Fats/metabolism , Ghrelin , Mice , Peptide Hormones/genetics
9.
Regul Pept ; 117(2): 89-99, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14700744

ABSTRACT

Previous studies have indicated a relationship between cocaine- and amphetamine-related transcript (CART) and leptin. The present study used quantitative PCR and in situ hybridization to examine this CART-leptin relationship in different animal models. With CART injection, the function of this pathway was also investigated. The results demonstrate that CART mRNA in the arcuate nucleus (ARC) was significantly increased in subjects fed a high-fat diet (HFD) compared to low-fat diet (LFD). It was also elevated in obese vs. lean rats and in normal-weight obesity-prone vs. obesity-resistant rats. In each group tested, CART mRNA in the ARC was positively correlated specifically with circulating levels of leptin. Its close association specifically with leptin was further supported by a stimulatory effect of this hormone on CART expression. This leptin-CART relationship in the ARC, in contrast, was less consistent or undetectable in the paraventricular nucleus and lateral hypothalamus. Central injection of CART peptide (55-102) increased circulating non-esterified fatty acid levels and decreased lipoprotein lipase activity in adipose tissue. These results suggest that, on a fat-rich diet, this leptin-CART pathway originating in the ARC inhibits excessive body fat accrual by causing a shift from lipid storage toward lipid mobilization.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Dietary Fats/administration & dosage , Lipid Metabolism , Lipoprotein Lipase/metabolism , Nerve Tissue Proteins/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Body Composition , Body Weight/drug effects , Gene Expression , Leptin/metabolism , Lipoprotein Lipase/drug effects , Male , Nerve Tissue Proteins/genetics , Obesity/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
10.
Am J Physiol Regul Integr Comp Physiol ; 284(6): R1454-65, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12560202

ABSTRACT

Previous reports implicate the orexins in eating and body weight regulation. This study investigated possible functional relationships between hypothalamic orexins and circulating hormones or metabolites. In situ hybridization and quantitative PCR were used to examine orexin expression in the perifornical hypothalamus (PF) of rats and mice on diets varying in fat content and with differential propensity toward obesity. The results showed that orexin gene expression was stimulated by a high-fat diet in close association with elevated triglyceride levels, suggesting a functional relationship between these measures. Results obtained in obesity-prone rats and mice revealed a similar increase in orexin in close relation to triglycerides. A direct test of this orexin-triglyceride link was performed with Intralipid, which increased PF orexin expression along with circulating triglycerides. Whereas PF galanin is similarly stimulated by dietary fat, double-labeling immunofluorescence studies showed that orexin and galanin neurons are anatomically distinct. This evidence suggests that the orexins, like galanin, are "fat-responsive" peptides that respond to circulating lipids.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Hypertriglyceridemia/blood , Hypertriglyceridemia/genetics , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins , Neuropeptides/genetics , Animals , Blood Glucose/analysis , Body Weight , Colchicine/pharmacology , Dietary Fats/pharmacology , Galanin/genetics , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Insulin/blood , Leptin/blood , Male , Mice , Mice, Inbred Strains , Obesity/blood , Obesity/genetics , Orexins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...