Elsevier

Neuropeptides

Volume 76, August 2019, 101937
Neuropeptides

The additive effect of allopregnanolone on ghrelin's orexigenic effect in rats

https://doi.org/10.1016/j.npep.2019.101937Get rights and content

Highlights

  • Allopregnanolone (AlloP) further increases food-intake in the presence of ghrelin in rats.

  • At physiological concentrations, AlloP potentiates hypothalamic inhibitory transmission in the lateral ARC and medial PVN.

  • AlloP's effect on food-intake appears to depend on the level of activity in GABAergic circuits involved in eating behavior.

Abstract

The progesterone metabolite, allopregnanolone (AlloP), is a GABAA receptor modulating steroid and is known to have orexigenic and pro-obesity effects. The neurobiological mechanisms underpinning these effects are most likely due to enhanced GABAergic signaling in the lateral arcuate nucleus (ARC) and medial paraventricular nucleus (PVN) of the hypothalamus. Inspired by the finding that GABAergic signaling is also important for the orexigenic effects of the circulating hormone, ghrelin, we sought to determine the extent to which AlloP (one of the most potent endogenous GABAA-receptor modulators) operates alongside ghrelin to enhance food intake. Male rats with ad libitum access to standard chow were injected intravenously with AlloP and/or ghrelin, alone or in combination. The intake of the standard chow was greater after AlloP 1 mg/kg together with ghrelin 30 μg/kg than with 30 μg/kg ghrelin alone. Food intake was also increased for the combined treatment of AlloP 0.5 mg/kg + ghrelin 10 μg/kg, AlloP 1 mg/kg + ghrelin 10 μg/kg, and AlloP 0.5 mg/kg + ghrelin 30 μg/kg. There was no significant difference in food intake between the two ghrelin doses or between the two doses of AlloP and the vehicle. In electrophysiological studies, physiologically relevant concentrations of AlloP prolonged the current decay time of spontaneous inhibitory post-synaptic current of dissociated cells of the ARC and PVN. We conclude that AlloP enhances the hyperphagic effect of ghrelin, findings of potential relevance for the hyperphagia associated with the luteal phase of the reproductive cycle.

Introduction

It is well established that GABAA receptor modulating steroids (GAMS) have orexigenic and pro-obesity effects, especially progestogens, and the metabolite allopregnanolone (AlloP). During the luteal phase of the menstrual cycle, that can be considered a preparatory phase for a forthcoming pregnancy, energy intake is higher and food cravings are more common (Barr et al., 1995; Cross et al., 2001; Dalvit, 1981; Hormes and Rozin, 2009; Johnson et al., 1994; Reed et al., 2008). This fact underscores the important role of progestogens in ensuring adequate nutrition during pregnancy and lactation (Beksinska et al., 2010; Butte and King, 2005). It appears that some factor formed by the corpus luteum is implicated in these effects since the fluctuation in energy intake is abolished in anovulatory cycles (Ottander et al., 2005). Indeed, some hormonal contraceptives have the side effect of increasing body weight (Bahamondes et al., 2001; Beksinska et al., 2010; Berenson and Rahman, 2009; Bonny et al., 2006; Risser et al., 1999), and the progestogen, medroxyprogesterone-acetate has even been used to improve appetite and hence, the body weight in malnourished patients (Simons et al., 1996). Notably, circulating AlloP levels have been associated with binge eating in women and obesity in both men and women (Menozzi et al., 2002; Monteleone et al., 2003; Predieri et al., 2007). Stress is a known risk factor for obesity (Vieweg et al., 2007), and stress also stimulates the production of AlloP and other GABAA-receptor active steroids in male rats (Purdy et al., 1991) and humans of both sexes (Droogleever Fortuyn et al., 2004).

In rodents, hyperphagia can be induced by the administration of AlloP, and the effect is dose-dependent (Chen et al., 1996; Reddy and Kulkarni, 1998, Reddy and Kulkarni, 1999). Their meal size is increased, which correlates with obesity in rats (Farley et al., 2003; Furnes et al., 2009), and weight gain is also achieved after long term treatment with AlloP (Holmberg et al., 2015). In a choice situation, AlloP does also promote intake of more calorie-dense foods (Holmberg et al., 2014).

Another potent orexigenic substance is ghrelin, which is a stomach-derived peptide hormone (Kojima et al., 1999; Wren et al., 2000) with pro-obesity effects (Tschop et al., 2000). Circulating levels of ghrelin increase pre-prandial, and it has been suggested that ghrelin plays an important role in hunger and meal initiation (Cummings et al., 2001). Ghrelin activates circuits involved in energy homeostasis and appetitive behavior that includes the hypothalamus (Hewson and Dickson, 2000), brainstem (Bailey et al., 2000; Faulconbridge et al., 2008) and mesolimbic reward circuits (Abizaid et al., 2006; Dickson et al., 2010; Egecioglu et al., 2010; Jerlhag et al., 2007; Skibicka et al., 2011; Skibicka et al., 2012). In the arcuate nucleus (ARC), a key target for ghrelin is the orexigenic neuropeptide Y/agouti-related peptide (NPY/AgRP)-expressing cells (Dickson and Luckman, 1997; Qi et al., 2015), which inhibit the anorexigenic pro-opiomelanocortin (POMC) population in the arcuate nucleus (ARC) by a mechanism involving increased GABAergic transmission (Cowley et al., 2003; Gao and Horvath, 2008; Jobst et al., 2004). Indeed, important roles for GABAergic transmission in feeding and ghrelin's orexigenic effects are highlighted by the finding that specific deletion of the vesicular GABA transporter in AgRP-expressing neurons leads to a strongly reduced orexigenic effect of ghrelin (Tong et al., 2008). Both NPY/AgRP- and POMC-expressing neurons project to “second-order neurons” located in, for example, the paraventricular nucleus (PVN), where anorexigenic peptides are released thereby activating catabolic circuits (Gil-Campos et al., 2006; Valassi et al., 2008). Even at this site, inhibitory GABAergic transmission appears to be of importance for food intake (Dos-Santos et al., 2018; Pu et al., 1999). AlloP potentiates the effect of GABA at the GABAA-receptor (Majewska et al., 1986) in a manner similar to that of benzodiazepine, which has a well-documented hyperphagic effect (Cooper, 2005).

Inspired by the important role of GABAergic transmission for the orexigenic effect of ghrelin, we sought to determine what effect AlloP, perhaps the most potent positive endogenous modulator of the GABAA-receptor, would have on ghrelin's orexigenic effects. Additionally, given the large fluctuations in AlloP in normal physiology (Hill et al., 2007; Holzbauer, 1975; Nyberg et al., 2007), together with the large differences in sensitivity thresholds for the GABAA-receptor between different brain areas (Herd et al., 2007), we also investigated the sensitivity of GABAA-receptors to AlloP on cells from the lateral ARC and the medial PVN, two important hypothalamic areas for food intake regulation.

Section snippets

Animals

Food intake studies were performed on 24 male Wistar rats (Taconic™, Lille Skensved, Denmark) weighing 150 ± 3.4 g (mean ± st.dev.) upon arrival to the facility. The animals were housed in triads in cages measuring 55 × 35 × 20 cm with ad libitum access to food and water. For the first seven days, the animals were acclimatized to the facility and, thereafter, handled on four separate occasions. On two occasions, the rats were habituated to the intravenous (i.v.) injection procedure first by

Impact of AlloP on ghrelin-induced food intake in rats

Food intake during the first 30 min was significantly increased by AlloP and ghrelin (F(8,111) = 4,71; p = .001, Fig. 1). The food intake was larger in the presence of 1 mg/kg AlloP together with 30 μg/kg ghrelin than with 30 μg/kg ghrelin alone (p = .033) (Fig. 1). A significant increase in food intake relative to vehicle treated animals was also detected for all of the following groups: ghrelin 10 μg/kg (p = .033), ghrelin 30 μg/kg (p = .010), AlloP 0.5 mg/kg + ghrelin 10 μg/kg (p = .003),

Discussion

Here we demonstrate that AlloP enhances the orexigenic effects of ghrelin. This finding implies that ghrelin and AlloP exert their orexigenic effects, at least in part, by different mechanisms. This effect remained and was even more pronounced when the animal's individual satiety and hunger drive was considered. Ghrelin increased food intake which is in compliance with its established effect on meal initiation (Cummings, 2006; Cummings et al., 2001) and motivated behavior for food (Egecioglu et

Acknowledgment

Dr. David Haage for excellent assistance with conducting the experiments, and for great overall support.

The research leading to these results has received funding from the European Community's Seventh Framework Programme (FP7/2007-2013 under grant agreement no 245009 and FP7-HEALTH-2009-241592), the Swedish Medical Research Council (2006-5663, 2009-5266, 4x-11198), Novo Nordisk Fonden (GeA/AIR), ALF Göteborg (SU7601) and the Swedish Foundation for Strategic Research of the Sahlgrenska Center

Declaration of interest

Professor Torbjörn Bäckström is a shareholder and board member of Umecrine AB. None of the other authors have any competing interest.

References (80)

  • Y. Date et al.

    The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats

    Gastroenterology

    (2002)
  • V.H. Demaria-Pesce et al.

    Mathematical determination of feeding patterns and its consequence on correlational studies

    Physiol. Behav.

    (1998)
  • S.L. Dickson et al.

    Blockade of central nicotine acetylcholine receptor signaling attenuate ghrelin-induced food intake in rodents

    Neuroscience

    (2010)
  • H.A. Droogleever Fortuyn et al.

    Effects of PhD examination stress on allopregnanolone and cortisol plasma levels and peripheral benzodiazepine receptor density

    Psychoneuroendocrinology

    (2004)
  • L.F. Faulconbridge et al.

    Caudal brainstem delivery of ghrelin induces fos expression in the nucleus of the solitary tract, but not in the arcuate or paraventricular nuclei of the hypothalamus

    Brain Res.

    (2008)
  • M.A. Fudge et al.

    Allopregnanolone produces hyperphagia by reducing neophobia without altering food palatability

    Eur. Neuropsychopharmacol.

    (2006)
  • Q. Gao et al.

    Neuronal control of energy homeostasis

    FEBS Lett.

    (2008)
  • D. Haage et al.

    Allopregnanolone modulates spontaneous GABA release via presynaptic Cl- permeability in rat preoptic nerve terminals

    Brain Res.

    (2002)
  • M.B. Herd et al.

    Neurosteroid modulation of synaptic and extrasynaptic GABA(A) receptors

    Pharmacol. Ther.

    (2007)
  • S. Higgs et al.

    Antineophobic effect of the neuroactive steroid 3alpha-hydroxy-5beta-pregnan-20-one in male rats

    Pharmacol. Biochem. Behav.

    (1998)
  • M. Hill et al.

    Circulating levels of pregnanolone isomers during the third trimester of human pregnancy

    J. Steroid Biochem. Mol. Biol.

    (2007)
  • E. Holmberg et al.

    Repeated allopregnanolone exposure induces weight gain in schedule fed rats on high fat diet

    Physiol. Behav.

    (2015)
  • E. Holmberg et al.

    Allopregnanolone involvement in feeding regulation, overeating and obesity

    Front. Neuroendocrinol.

    (2018)
  • M. Holzbauer

    Physiological variations in the ovarian production of 5alpha-pregnane derivatives with sedative properties in the rat

    J Steroid Biochem

    (1975)
  • J.M. Hormes et al.

    Perimenstrual chocolate craving. What happens after menopause?

    Appetite

    (2009)
  • E.E. Jobst et al.

    The electrophysiology of feeding circuits

    Trends Endocrinol. Metab.

    (2004)
  • S. Johansson et al.

    Graded action potentials generated by neurons in rat hypothalamic slices

    Brain Res.

    (1995)
  • W.G. Johnson et al.

    Energy regulation over the menstrual cycle

    Physiol. Behav.

    (1994)
  • U. Karlsson et al.

    Glutamate-evoked currents in acutely dissociated neurons from the rat medial preoptic nucleus

    Brain Res.

    (1997)
  • U. Ottander et al.

    Allopregnanolone and pregnanolone are produced by the human corpus luteum

    Mol. Cell. Endocrinol.

    (2005)
  • M. Perello et al.

    Ghrelin increases the rewarding value of high-fat diet in an orexin-dependent manner

    Biol. Psychiatry

    (2010)
  • B. Przewlocka et al.

    Evidence that GABA in the nucleus dorsalis raphe induces stimulation of locomotor activity and eating behavior

    Life Sci.

    (1979)
  • Y. Qi et al.

    Chronic overproduction of ghrelin in the hypothalamus leads to temporal increase in food intake and body weight

    Neuropeptides

    (2015)
  • D.S. Reddy et al.

    Sex and estrous cycle-dependent changes in neurosteroid and benzodiazepine effects on food consumption and plus-maze learning behaviors in rats

    Pharmacol. Biochem. Behav.

    (1999)
  • S.C. Reed et al.

    Changes in mood, cognitive performance and appetite in the late luteal and follicular phases of the menstrual cycle in women with and without PMDD (premenstrual dysphoric disorder)

    Horm. Behav.

    (2008)
  • W.L. Risser et al.

    Weight change in adolescents who used hormonal contraception

    J. Adolesc. Health

    (1999)
  • K.P. Skibicka et al.

    Ghrelin directly targets the ventral tegmental area to increase food motivation

    Neuroscience

    (2011)
  • S. Tsujii et al.

    GABA-related feeding control in genetically obese rats

    Brain Res.

    (1991)
  • E. Valassi et al.

    Neuroendocrine control of food intake

    Nutr. Metab. Cardiovasc. Dis.

    (2008)
  • V.S. Vorobjev

    Vibrodissociation of sliced mammalian nervous tissue

    J. Neurosci. Methods

    (1991)
  • Cited by (7)

    • Medroxyprogesterone acetate positively modulates specific GABA<inf>A</inf>-receptor subtypes - affecting memory and cognition

      2022, Psychoneuroendocrinology
      Citation Excerpt :

      MPA has even been used as treatment for cachexia (Simons et al., 1998). We have earlier shown that the neuroactive steroid allopregnanolone has a pronounced effect in hypothalamic cells and it is therefore of interested to use the same brain region for confirming MPA experiment as the region is very sensitive to allopregnanolone with significant effects already at 2 nM (Löfgren et al., 2019). This effect is at least partly mediated by the GABAA receptor subtype α2 in the hypothalamus and is involved in stimulating hyperphagia. (

    • The effects of neurosteroid allopregnanolone on synaptic dysfunction in the hippocampus in experimental parkinsonism rats: An electrophysiological and molecular study

      2022, Neuropeptides
      Citation Excerpt :

      Investigations have shown the neuroprotective effects of neurosteroids. Progesterone is a neuroactive steroid that displays important modulatory effects on normal functions such as responses to stress, memory, emotion and cognition (Frye and Lacey, 2001; Melcangi and Mensah-Nyagan, 2008) and exhibits significant roles in the pathology and treatment of psychological disorders such as anxiety, depression, schizophrenia, epilepsy and other neurodegenerative diseases (Landgren et al., 1987; Löfgren et al., 2019). Allopregnanolone (3-hydroxy-5-pregnan-20-one; Allo) is a reduced metabolite of progesterone and is synthesized de novo in the central nervous system (CNS) throughout one's lifetime (Löfgren et al., 2019; Pomata et al., 2000).

    View all citing articles on Scopus
    View full text