scholarly journals Acylation Type Determines Ghrelin's Effects on Energy Homeostasis in Rodents

Endocrinology ◽  
2012 ◽  
Vol 153 (10) ◽  
pp. 4687-4695 ◽  
Author(s):  
Kristy M. Heppner ◽  
Nilika Chaudhary ◽  
Timo D. Müller ◽  
Henriette Kirchner ◽  
Kirk M. Habegger ◽  
...  

Abstract Ghrelin is a gastrointestinal polypeptide that acts through the ghrelin receptor (GHSR) to promote food intake and increase adiposity. Activation of GHSR requires the presence of a fatty-acid (FA) side chain on amino acid residue serine 3 of the ghrelin molecule. However, little is known about the role that the type of FA used for acylation plays in the biological action of ghrelin. We therefore evaluated a series of differentially acylated peptides to determine whether alterations in length or stability of the FA side chain have an impact on the ability of ghrelin to activate GHSR in vitro or to differentially alter food intake, body weight, and body composition in vivo. Fatty acids principally available in the diet (such as palmitate C16) and therefore representing potential substrates for the ghrelin-activating enzyme ghrelin O-acyltransferase (GOAT) were used for dose-, time-, and administration/route-dependent effects of ghrelin on food intake, body weight, and body composition in rats and mice. Our data demonstrate that altering the length of the FA side chain of ghrelin results in the differential activation of GHSR. Additionally, we found that acylation of ghrelin with a long-chain FA (C16) delays the acute central stimulation of food intake. Lastly, we found that, depending on acylation length, systemic and central chronic actions of ghrelin on adiposity can be enhanced or reduced. Together our data suggest that modification of the FA side-chain length can be a novel approach to modulate the efficacy of pharmacologically administered ghrelin.

2006 ◽  
Vol 190 (2) ◽  
pp. 515-525 ◽  
Author(s):  
Anthony P Coll ◽  
Martin Fassnacht ◽  
Steffen Klammer ◽  
Stephanie Hahner ◽  
Dominik M Schulte ◽  
...  

Pro-opiomelanocortin (POMC) is a polypeptide precursor that undergoes extensive processing to yield a range of peptides with biologically diverse functions. POMC-derived ACTH is vital for normal adrenal function and the melanocortin α-MSH plays a key role in appetite control and energy homeostasis. However, the roles of peptide fragments derived from the highly conserved N-terminal region of POMC are less well characterized. We have used mice with a null mutation in the Pomc gene (Pomc−/−) to determine the in vivo effects of synthetic N-terminal 1–28 POMC, which has been shown previously to possess adrenal mitogenic activity. 1–28 POMC (20 μg) given s.c. for 10 days had no effect on the adrenal cortex of Pomc−/− mice, with resultant cortical morphology and plasma corticosterone levels being indistinguishable from sham treatment. Concurrent administration of 1–28 POMC and 1–24 ACTH (30 μg/day) resulted in changes identical to 1–24 ACTH treatment alone, which consisted of upregulation of steroidogenic enzymes, elevation of corticosterone levels, hypertrophy of the zona fasciculate, and regression of the X-zone. However, treatment of corticosterone-depleted Pomc−/− mice with 1–28 POMC reduced cumulative food intake and total body weight. These anorexigenic effects were ameliorated when the peptide was administered to Pomc−/− mice with circulating corticosterone restored either to a low physiological level by corticosterone-supplemented drinking water (CORT) or to a supraphysiological level by concurrent 1–24 ACTH administration. Further, i.c.v. administration of 1–28 POMC to CORT-treated Pomc−/− mice had no effect on food intake or body weight. In wild-type mice, the effects of 1–28 POMC upon food intake and body weight were identical to sham treatment, but 1–28 POMC was able to ameliorate the hyperphagia induced by concurrent 1–24 ACTH treatment. In a mouse model which lacks all endogenous POMC peptides, s.c. treatment with synthetic 1–28 POMC alone can reduce food intake and body weight, but has no impact upon adrenal growth or steroidogenesis.


Endocrinology ◽  
2009 ◽  
Vol 150 (11) ◽  
pp. 4920-4930 ◽  
Author(s):  
Pia Steen Petersen ◽  
David P. D. Woldbye ◽  
Andreas Nygaard Madsen ◽  
Kristoffer L. Egerod ◽  
Chunyu Jin ◽  
...  

The receptor for the orexigenic peptide, ghrelin, is one of the most constitutively active 7TM receptors known, as demonstrated under in vitro conditions. Change in expression of a constitutively active receptor is associated with change in signaling independent of the endogenous ligand. In the following study, we found that the expression of the ghrelin receptor in the hypothalamus was up-regulated approximately 2-fold in rats both during 48-h fasting and by streptozotocin-induced hyperphagia. In a separate experiment, to probe for the effect of the high basal signaling of the ghrelin receptor in vivo, we used intracerebroventricular administration by osmotic pumps of a peptide [D-Arg1, D-Phe5, D-Trp7,9, Leu11]-substance P. This peptide selectively displays inverse agonism at the ghrelin receptor as compared with an inactive control peptide with just a single amino acid substitution. Food intake and body weight were significantly decreased in the group of rats treated with the inverse agonist, as compared with the groups treated with the control peptide or the vehicle. In the hypothalamus, the expression of neuropeptide Y and uncoupling protein 2 was decreased by the inverse agonist. In a hypothalamic cell line that endogenously expresses the ghrelin receptor, we observed high basal activity of the cAMP response element binding protein, an important signaling transduction pathway for appetite regulation. The activation was further increased by ghrelin administration and decreased by administration of the inverse agonist. It is suggested that the high constitutive signaling activity is important for the in vivo function of the ghrelin receptor in the control of food intake and body weight.


2021 ◽  
Author(s):  
◽  
Roshan Kumari ◽  

Introduction: Obesity and its associated metabolic syndrome are major medical problems worldwide including United States. Adipose tissue is the primary site of energy storage, playing important roles in health. Adipose tissue also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety, inflammation, and total energy homeostasis. Activin A and activin B play important roles in maintaining body composition and energy homeostasis. This dissertation highlights the role of activin/SMADs signaling in adipose tissue development, function, and maintenance. SMAD2/3 proteins are downstream mediators of transforming growth factor-β (TGFβ) family signaling, including activins, which regulate critical preadipocyte and mature adipocyte functions. Previous studies have demonstrated that Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity and the direct contributions of Smad2 and Smad3 in adipose tissues individually or in combination and the responses of these tissues to activin signaling are unknown. Additionally, our lab demonstrated that the combined loss of activin A and activin B have reduced adiposity in mice and appearance of brown-like cells in visceral white adipose tissue. However, the cell-autonomous role of activins on cell proliferation and differentiation remained unknown in vitro. My hypothesis was that activin signaling regulate adipocyte differentiation and functions via SMAD2/3-mediated mechanism(s) and that the individual or combined adipose-specific deletion of SMAD2/SMAD3 would result in reduced adiposity similar to activin deficient mice. Objective: Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 individually and in combination, on diet-induced adiposity and to establish whether preadipocytes isolated from subcutaneous and visceral white adipose tissues differ in their differentiation capacity. We also assessed the role of activins on cell proliferation and differentiation using an in vitro model. Research Design: To assess the adipose-selective requirements of Smad2, Smad3 and Smad2/3, we generated three lines of adipose-selective conditional knockout (cKO) mice including Smad2cKO, Smad3cKO, and Smad2/3 double cKO mice using Smad2 and/or Smad3 “floxed” mice intercrossed with Adiponectin-Cre mice. Additionally, we isolated preadipocytes and examined adipogenic activity of visceral and subcutaneous preadipocyte and the effects of activin on preadipocyte proliferation and differentiation in vitro. Furthermore, we used mouse embryonic fibroblasts (MEFs) from wild type mice and activin double knockout mice to study the cell autonomous role of activin on differentiation and cell fate. Results: Our results demonstrated that subcutaneous preadipocytes differentiate uniformly and almost all wildtype subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A promoted proliferation and suppressed differentiation of subcutaneous preadipocytes more robustly given that visceral adipocytes differentiate poorly at baseline. Additionally, global knockout of activin A and B promoted differentiation and browning in differentiated MEFs in vitro consistent with in vivo studies. Furthermore, we showed that Smad2cKO mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3cKO male mice displayed a trend of reduced body weight on high fat diet. On both (LFD and HFD) diets, Smad3cKO male mice displayed an adipose depot-selective phenotype, with significant reduction in subcutaneous fat mass but not visceral fat mass. Smad2/3cKO male mice did not show any difference in body weight or fat mass compared to control mice. Female mice with adipose-selective combined deletion of Smad2/3, displayed reduced body weight and reduction of fat mass in both visceral and subcutaneous depot with higher metabolic rate on HFD compared to control littermates. Conclusions: Our study demonstrated that Smad3 is an important contributor to the development and/or maintenance of subcutaneous white adipose tissue in a sex-selective fashion. Combined reduction of Smad2/3 protects female mice from diet induced obesity and is important for visceral and subcutaneous depots in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation. Activin treatment promoted proliferation of preadipocytes, while activin deficiency promoted differentiation and altered the phenotypic characteristics of White adipocytes to brown-like cells in vitro consistent with in vivo.


2007 ◽  
Vol 292 (3) ◽  
pp. E891-E899 ◽  
Author(s):  
Raul M. Luque ◽  
Zhi H. Huang ◽  
Bhumik Shah ◽  
Theodore Mazzone ◽  
Rhonda D. Kineman

Leptin-deficient obese mice ( ob/ob) have decreased circulating growth hormone (GH) and pituitary GH and ghrelin receptor (GHS-R) mRNA levels, whereas hypothalamic GH-releasing hormone (GHRH) and somatostatin (SST) expression do not differ from lean controls. Given the fact that GH is suppressed in diet-induced obesity (a state of hyperleptinemia), it remains to be determined whether the absence of leptin contributes to changes in the GH axis of ob/ob mice. Therefore, to study the impact of leptin replacement on the hypothalamic-pituitary GH axis of ob/ob mice, leptin was infused for 7 days (sc), resulting in circulating leptin levels that were similar to wild-type controls (∼1 ng/ml). Leptin treatment reduced food intake, body weight, and circulating insulin while elevating circulating n-octanoyl ghrelin concentrations. Leptin treatment did not alter hypothalamic GHRH, SST, or GHS-R mRNA levels compared with vehicle-treated controls. However, leptin significantly increased pituitary GH and GHRH-R expression and tended to enhance circulating GH levels, but this latter effect did not reach statistical significance. In vitro, leptin (1 ng/ml, 24 h) did not affect pituitary GH, GHRH-R, or GHS-R mRNA but did enhance GH release. The in vivo effects of leptin on circulating hormone and pituitary mRNA levels were not replicated by pair feeding ob/ob mice to match the food intake of leptin-treated mice. However, leptin did prevent the fall in hypothalamic GHRH mRNA and circulating IGF-I levels observed in pair-fed mice. These results demonstrate that leptin replacement has positive effects on multiple levels of GH axis function in ob/ob mice.


2019 ◽  
Author(s):  
Jason A. West ◽  
Soumitra S. Ghosh ◽  
David G. Parkes ◽  
Anastasia Tsakmaki ◽  
Rikke V. Grønlund ◽  
...  

ABSTRACTObjectiveCombinatorial gut hormone therapy is one of the more promising strategies for identifying improved treatments for metabolic disease. Many approaches combine the established benefits of glucagon-like peptide-1 (GLP-1) agonism with one or more additional molecules with the aim of improving metabolic outcomes. Recent attention has been drawn to the glucose-dependent insulinotropic polypeptide (GIP) system due to compelling pre-clinical evidence describing the metabolic benefits of antagonising the GIP receptor (GIPR). We rationalised that benefit might be accrued from combining GIPR antagonism with GLP-1 agonism. To this end we investigated the metabolic effects of co-administration of previously reported peptide-based GIPR antagonists with the GLP-1 agonist liraglutide.MethodsTwo GIPR peptide antagonists, GIPA-1 (mouse GIP(3-30)NH2) and GIPA-2 (NαAc-K10[γEγE-C16]-Arg18-hGIP(5–42)), were pharmacologically characterised in vitro in an assay measuring cAMP production in CHO-K1 cells overexpressing the mouse GIPR. These peptides were then characterised in vivo in lean mice for their effect on oral glucose tolerance, as well as their ability to antagonize exogenous GIP action. Finally, a mouse model of diet-induced obesity (DIO) was used to investigate the potential metabolic benefits of chronic dosing of peptide-based GIPR antagonists, alone or in combination with liraglutide.ResultsIn vitro, both GIPR peptides exhibited potent antagonistic properties, with GIPA-2 being the more potent of the two. Acute in vivo administration of GIPA-1 during an oral glucose tolerance test (OGTT) had negligible effects on glucose tolerance and circulated insulin in lean mice. In contrast, GIPA-2 impaired glucose tolerance and attenuated circulating insulin levels, with offsetting effects on glycemia noted with co-administration with exogenous mouse GIP, suggesting true antagonism via GIPA-2 at the GIP receptor. Chronic administration studies in a DIO mouse model showed expected effects of GLP-1 agonism (via liraglutide), lowering food intake, body weight, fasting blood glucose and plasma insulin concentrations while improving glucose sensitivity, whereas delivery of either GIPR antagonist alone had negligible effects on these parameters. Interestingly, chronic dual therapy with the GIPR antagonists and GLP-1 showed separation from single intervention arms though augmented insulin sensitizing effects (modestly lowering insulin and HOMA-IR) and lowering plasmas triglycerides and free-fatty acids, with more notable effects observed with GIPA-1 compared to GIPA-2.ConclusionWe conclude that, in contrast to the well-documented effects of GLP-1R agonism, systemic administration of peptide-based GIPR antagonists demonstrate minimal benefit on metabolic parameters in DIO mice, exhibiting no major effects on body weight, food intake and glycaemic parameters. However, the co-administration of both a GIPR antagonist together with a GLP1 agonist uncovers interesting synergistic and beneficial effects on measures of insulin sensitivity, circulating lipids and certain adipose stores that seem influenced by the degree or nature of GIP receptor antagonism.


2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Adelaide A Bernard ◽  
Irene Ojeda Naharros ◽  
Florence Bourgain Guglielmetti ◽  
Xinyu Yue ◽  
Christian Vaisse

Abstract Genetic studies in humans and mice have demonstrated that the Melanocortin 4 Receptor (MC4R) is essential for adequate regulation of food intake and body weight. MC4R is expressed in a small population of hypothalamic neurons and very little is known about its molecular and cellular dynamics in vivo. We have recently demonstrated that MC4R localizes to and functions at the primary cilia of select hypothalamic neurons to control energy homeostasis. The primary cilium is a solitary hair-like organelle that serves as an antenna sensing extracellular environment. Defective primary cilia lead to a series of conditions known as ciliopathies, that can manifest through a variety of clinical features, including hyperphagia and obesity. Here we establish that the ciliary localization and the body weight regulating activity of MC4R is dependent on a single-pass transmembrane accessory protein: the Melanocortin Receptor Associated Protein 2 (MRAP2). Specifically, we show that deleting MRAP2 specifically from MC4R neurons (MC4RMRAP2-/-) leads to early onset obesity and hyperphagia. In vitro, co-expression of MRAP2 in ciliated IMCD3 cells increases MC4R localization to the primary cilium. We further demonstrate that MRAP2 and MC4R colocalize specifically at the primary cilium in vivo, and that MC4R fails to localize to the primary cilium when MRAP2 is deleted. These findings highlight the role of the primary cilium in the control of energy homeostasis, and the importance of accessory proteins for the localization of GPCRs to the primary cilium where they exert their function, in this case being critical for the regulation of energy homeostasis.


2007 ◽  
Vol 292 (1) ◽  
pp. R235-R241 ◽  
Author(s):  
Yingkui Yang ◽  
Min Chen ◽  
Keith E. Georgeson ◽  
Carroll M. Harmon

The increase in the prevalence of human obesity highlights the need to identify molecular and cellular mechanisms involved in control of feeding and energy balance. Oleoylethanolamide (OEA), an endogenous lipid produced primarily in the small intestine, has been identified to play an important role in the regulation of animal food intake and body weight. Previous studies indicated that OEA activates peroxisome proliferator-activated receptor-α, which is required to mediate the effects of appetite suppression, reduces blood lipid levels, and enhances peripheral fatty acid catabolism. However, the effect of OEA on enterocyte function is unclear. In this study, we have examined the effect of OEA on intestinal fatty acid uptake and FAT/CD36 expression in vivo and in vitro. We intraperitoneally administered OEA to rats and examined FAT/CD36 mRNA level and fatty acid uptake in enterocytes isolated from the proximal small intestine, as well as in adipocytes. Our results indicate that OEA treatment significantly increased FAT/CD36 mRNA expression in intestinal mucosa and isolated jejunal enterocytes. In addition, we also found that OEA treatment significantly increases fatty acid uptake in isolated enterocytes in vitro. These results suggest that in addition to appetite regulation, OEA may regulate body weight by altered peripheral lipid metabolism, including increased lipolysis in adipocytes and enhanced fatty acid uptake in enterocytes, both in conjunction with increased expression of FAT/CD36. This study may have important implications in understanding the mechanism of OEA in the regulation of fatty acid absorption in human physiological and pathophysiological conditions.


2018 ◽  
Vol 9 (12) ◽  
pp. 6257-6267 ◽  
Author(s):  
Ting Luo ◽  
Omar Miranda-Garcia ◽  
Geoff Sasaki ◽  
Jinling Wang ◽  
Neil F. Shay

Genistein and daidzein decrease mice food intake, ameliorate symptoms of metabolic syndrome, including decreasing body weight gain, and improving glucose metabolism, and appear to produce differential effects, possibly via the regulation of LXR-mediated pathways.


2019 ◽  
Vol 18 (7) ◽  
pp. 516-522
Author(s):  
Néstor F. Díaz ◽  
Héctor Flores-Herrera ◽  
Guadalupe García-López ◽  
Anayansi Molina-Hernández

The brain histaminergic system plays a pivotal role in energy homeostasis, through H1- receptor activation, it increases the hypothalamic release of histamine that decreases food intake and reduces body weight. One way to increase the release of hypothalamic histamine is through the use of antagonist/inverse agonist for the H3-receptor. Histamine H3-receptors are auto-receptors and heteroreceptors located on the presynaptic membranes and cell soma of neurons, where they negatively regulate the synthesis and release of histamine and other neurotransmitters in the central nervous system. Although several compounds acting as H3-receptor antagonist/inverse agonists have been developed, conflicting results have been reported and only one has been tested as anti-obesity in humans. Animal studies revealed the opposite effect in food intake, energy expeditor, and body weight, depending on the drug, spice, and route of administration, among others. The present review will explore the state of art on the effects of H3-receptor ligands on appetite and body-weight, going through the following: a brief overview of the circuit involved in the control of food intake and energy homeostasis, the participation of the histaminergic system in food intake and body weight, and the H3-receptor as a potential therapeutic target for obesity.


Nutrients ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 2223
Author(s):  
Manon Dominique ◽  
Nicolas Lucas ◽  
Romain Legrand ◽  
Illona-Marie Bouleté ◽  
Christine Bôle-Feysot ◽  
...  

CLPB (Caseinolytic peptidase B) protein is a conformational mimetic of α-MSH, an anorectic hormone. Previous in vivo studies have already shown the potential effect of CLPB protein on food intake and on the production of peptide YY (PYY) by injection of E. coli wild type (WT) or E. coli ΔClpB. However, until now, no study has shown its direct effect on food intake. Furthermore, this protein can fragment naturally. Therefore, the aim of this study was (i) to evaluate the in vitro effects of CLPB fragments on PYY production; and (ii) to test the in vivo effects of a CLPB fragment sharing molecular mimicry with α-MSH (CLPB25) compared to natural fragments of the CLPB protein (CLPB96). To do that, a primary culture of intestinal mucosal cells from male Sprague–Dawley rats was incubated with proteins extracted from E. coli WT and ΔCLPB after fragmentation with trypsin or after a heat treatment of the CLPB protein. PYY secretion was measured by ELISA. CLPB fragments were analyzed by Western Blot using anti-α-MSH antibodies. In vivo effects of the CLPB protein on food intake were evaluated by intraperitoneal injections in male C57Bl/6 and ob/ob mice using the BioDAQ® system. The natural CLPB96 fragmentation increased PYY production in vitro and significantly decreased cumulative food intake from 2 h in C57Bl/6 and ob/ob mice on the contrary to CLPB25. Therefore, the anorexigenic effect of CLPB is likely the consequence of enhanced PYY secretion.


Sign in / Sign up

Export Citation Format

Share Document