Central administration of obestatin fails to show inhibitory effects on food and water intake in mice

https://doi.org/10.1016/j.regpep.2009.04.014Get rights and content

Abstract

Obestatin is a ghrelin-associated peptide hormone with presumed anorexigenic and inhibitory effect on gastric propulsive motility activity. Recent literature, however, discloses much contestation over satiety and gastrointestinal motility-related functionalities of obestatin. In addition, antidipsinogenic effects in rodents by obestatin were recently reported. The present study was set up to bring more clarity into the contested effects of obestatin on food and water intake. Additionally, the stability of obestatin in brain tissue homogenate was investigated. The in vitro incubation of obestatin in brain homogenates revealed disappearance half-life times of 19 min for crude brain homogenate to 27 min for brain membrane homogenate. For the behavioural studies, male C57Bl/6 mice were intracerebroventricularly treated with 0.2 nmol murine amidated obestatin or vehicle at the age of 3 months. An additional group of mice was treated with 0.3 nmol of corticotropin releasing factor (CRF) as a positive control of suppression of food intake. Food and water intake were studied over a period of 5 h in metabolic cages. Under our experimental conditions, no suppressive effects of obestatin on food or water intake were observed, whereas CRF evoked a significant suppression of food intake, which proves the internal validity of the study design.

Introduction

A complex network of peptide hormones secreted from various tissues and organs like gut, brain, adipose tissue, pancreas and thyroid gland regulates food intake and energy metabolism [1]. Obestatin is a recently identified 23-amino acid gut-derived peptide encoded by the preproghrelin gene. Although originating from the same precursor as the widely studied appetite-stimulating peptide ghrelin, obestatin was reported to bring on antithetical physiological effects with regards to food intake and body weight regulation. The initial publication by Zhang et al. [2] reported that human obestatin suppressed food intake in fasted mice and reduced spontaneous body weight gain in lean mice after intraperitoneal (i.p.) or intracerebroventricular (i.c.v.) administration. Furthermore, peripheral administration of obestatin was shown to suppress gastric emptying. In vitro, obestatin suppressed jejunal activity and counteracted the stimulatory effect of ghrelin. Obestatin was hypothesized to exert its physiological effects through activation of the orphan G-protein coupled receptor GPR39.

Several subsequent studies, with few exceptions [3], [4], [5], [6], failed to corroborate the initially reported anorexigenic properties of obestatin [[7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18], [19], [20]; for a summary; see Table 1]. In addition, the putative effect of exogenous obestatin on gastrointestinal motility has been questioned [7], [13], [21], [22], [23], [24], [25], as well as the presumed interaction between ghrelin and obestatin in the regulation of food intake and energy metabolism [8], [10]. Furthermore, the interrelationship between obestatin, obesity and diabetes has been reviewed [26].

The GPR39 receptor may not be deorphanised yet based on negative studies employing GPR39 transfected cell lines [27], [28], GPR39 knockout mice [11], and lack of GPR39 receptor expression in the murine hypothalamus [29].

In addition, other physiological actions of obestatin were reported. Samson et al. [20] reported that i.c.v. administration of obestatin inhibits water consumption related to food intake or stimulated by the dipsogenic peptide hormone angiotensin II. The same study failed to show effects of obestatin administration on food intake, locomotor activity and stereotypic behaviours. Other studies showed that obestatin regulated sleep [30], affected cell proliferation [31], [32], [33], increased the secretion of pancreatic juice enzymes [34], promoted survival of pancreatic β-cells [35], and inhibited glucose-induced insulin secretion [36].

As stated by Gourcerol et al. [37], above-mentioned observations on food and water intake await further confirmation. Therefore, the aim of the present study was to analyse the effect of i.c.v. administration of murine amidated obestatin in C57BL/6 mice on feeding and drinking behaviour during 5 h, employing metabolic cages. Corticotropin releasing factor (CRF) was used as a positive control for suppression of food intake [38], [39], in order to verify the internal validity of our study design. In addition, the in vitro brain metabolic stability of obestatin was determined.

Section snippets

Animals

For the ex vivo in vitro brain metabolisation experiments, female CD-1 mice (Harlan, Horst, The Netherlands) weighing 26–30 g, were used. The in vivo experiments were conducted on adult male C57Bl/6J mice (Charles River Laboratories, Brussels, Belgium) aged 12 weeks at the start of the experiment. Animals were housed under standard laboratory conditions: constant room temperature and humidity, food and water ad libitum, 12-h light: 12-h dark cycle (lights on at 08:00 h, lights off at 20:00 h).

Metabolic stability of mouse obestatin in brain homogenates

The in vitro incubation of obestatin in both brain homogenates revealed disappearance half-life times of 19 min for crude brain homogenate to 27 min for brain membrane homogenate, based upon UV (195 nm) and fluorescence detection (λEX = 230 nm with λEM = 296 nm). The addition of some protease inhibitors could increase the ex vivo in vitro stability by a factor of 3 in the presence of a commercially available inhibitor cocktail (P8340) and with a factor of 2 with EDTA. All other individual protease

Discussion

As the P8340 protease-inhibitor cocktail carries the risk of showing a physiological activity itself, and EDTA will complex divalent ions, like Ca2+ and Mg2+, from the aCSF, both active protease inhibitors are expected to have confounding activities in the in vivo test. Therefore, they were not used for these experiments. Even in the absence of protease inhibitor, the in vitro half-life time was found to be around 19–27 min, contrasting with a half-life time for some peptides of only a few

Acknowledgements

This work was supported by the Fund for Scientific Research-Flanders (FWO grant G.0038.05), Interuniversity Poles of Attraction (IUAP Network P6/43), the Flemish Institute supporting Scientific-Technological Research in industry (IWT; SBO grant IWT-50164), agreement between Institute Born-Bunge and University of Antwerp, the Medical Research Foundation Antwerp, the Thomas Riellaerts research fund, and Neurosearch Antwerp. DVD is a postdoctoral fellow of the FWO.

References (51)

  • A.J. Ren et al.

    Inhibitory effect of obestatin on glucose-induced insulin secretion in rats

    Biochem Biophys Res Commun

    (2008)
  • G. Gourcerol et al.

    Lack of obestatin effects on food intake: Should obestatin be renamed ghrelin-associated peptide (GAP)? Regul

    Pept

    (2007)
  • M.J. Rosenthal et al.

    Corticotropin releasing factor (CRF) and age-related differences in behaviour of mice

    Neurobiol aging

    (1989)
  • B. De Spiegeleer et al.

    Impurity profiling QC testing of synthetic peptides using LC-PDA-fluorescence and LC-ESI-MS: the obestatin case

    Anal Biochem

    (2008)
  • V. Vergote et al.

    In vitro metabolic stability of obestatin: kinetics and identification of cleavage products

    Peptides

    (2008)
  • L. Prokai

    Electrospray ionization mass spectrometric and liquid chromatographic-mass spectrometric studies on the metabolism of synthetic dynorphin A peptides in brain tissue in vitro and in vivo

    J Chromatogr A

    (1998)
  • Y. Gao et al.

    Opioid receptor binding and antinociceptive activity of the analogues of endomorphin-2 and morphiceptin with phenylalanine mimics in the position 3 or 4

    Bioorg Med Chem Lett

    (2006)
  • L. Murillo et al.

    Brain processing of hemorphin-7 peptides in various subcellular fractions from rats

    Peptides

    (2006)
  • N.J. Rothwell

    Central effects of CRF on metabolism and energy balance

    Neurosci Biobehav Rev

    (1990)
  • S.C. Heinrichs et al.

    The role of corticotrophin-releasing factor and urocortin in the modulation of ingestive behaviour

    Neuropeptides

    (1999)
  • W. Pan et al.

    Differential BBB interactions of three ingestive peptides: obestatin, ghrelin, and adiponectin

    Peptides

    (2006)
  • F.Y. Che et al.

    Peptidomics of Cpefat/fat mouse hypothalamus. Effect of food deprivation and exercise on peptide levels

    J Biol Chem

    (2005)
  • W.S. Dhillo

    Appetite regulation: an overview

    Thyroid

    (2007)
  • J.V. Zhang et al.

    Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin's effects on food intake

    Science

    (2005)
  • E. Bresciani et al.

    Obestatin inhibits feeding but does not modulate GH and corticosterone secretion in the rat

    J Endocrinol Invest

    (2006)
  • Cited by (33)

    • Obestatin may affect the GnRH/KNDy gene network in sheep hypothalamus

      2019, Research in Veterinary Science
      Citation Excerpt :

      It is involved in the complex gut-brain neurohormonal networks, whereby hormones from the gastrointestinal tract signalize the feeling of satiety and/or hunger to the hypothalamus (Lacquaniti et al., 2011). First studies conducted on rodents shown that obestatin could act via the GPR39 receptor, but at the same time there were findings which did not confirm the obestatin affinity to this receptor (Chartrel et al., 2007; Granata et al., 2008; Gurriarán-Rodríguez et al., 2015; Lauwers et al., 2006; Tremblay et al., 2007; Van Dijck et al., 2009; Zhang, 2005). However, recent data from nuclear magnetic resonance (NMR) analysis of human obestatin structure showed that this hormone have the necessary domain required for full activation of the GPR39 (Alén et al., 2012).

    • Obestatin stimulates the somatotrophic axis activity in sheep

      2018, Brain Research
      Citation Excerpt :

      Obestatin is generally considered as a ligand for the orphan receptor GPR39 and/or glucagon-like peptide 1 receptor (GLP-1R), the expression of which was found in hypothalamic structures. Nevertheless, the studies evaluating this issue performed on rodents are controversial (Chartrel et al., 2007; Granata et al., 2008; Tremblay et al., 2007; Van Dijck et al., 2009). However, recent studies using nuclear magnetic resonance analysis clearly showed that obestatin contains a domain necessary for the GPR39 activation (Alén et al., 2012).

    View all citing articles on Scopus
    View full text