Elsevier

Physiology & Behavior

Volume 136, September 2014, Pages 39-46
Physiology & Behavior

Review
Amylin activates distributed CNS nuclei to control energy balance

https://doi.org/10.1016/j.physbeh.2014.01.013Get rights and content

Highlights

  • Amylin receptors are expressed in many energy balance-relevant nuclei.

  • Amylin activates hindbrain, hypothalamic, and mesolimbic sites to control feeding.

  • Amylin may be produced by structures other than the pancreas, including in the CNS.

  • Amylin-based combination pharmacotherapies may be useful for treating obesity.

Abstract

Amylin is a pancreas-derived neuropeptide that acts in the central nervous system (CNS) to reduce food intake. Much of the literature describing the anorectic effects of amylin are focused on amylin's actions in the area postrema, a hindbrain circumventricular structure. Although the area postrema is certainly an important site that mediates the intake-suppressive effects of amylin, several pieces of evidence indicate that amylin may also promote negative energy balance through action in additional CNS nuclei, including hypothalamic and mesolimbic structures. Therefore, this review highlights the distributed neural network mediating the feeding effects of amylin signaling with special attention being devoted to the recent discovery that the ventral tegmental area is physiologically relevant for amylin-mediated control of feeding. The production of amylin by alternative, extra-pancreatic sources and its potential relevance to food intake regulation is also considered. Finally, the utility of amylin and amylin-like compounds as a component of combination pharmacotherapies for the treatment of obesity is discussed.

Introduction

Amylin, also known as islet amyloid polypeptide (IAPP), is a 37-amino acid peptide hormone that is produced by pancreatic β-cells and co-secreted with insulin in response to the presence of nutrients in the gastrointestinal tract [1], [2]. Amylin has several physiological functions, but perhaps the most well-studied amylin-mediated effects are those of reduced food intake and improved glycemic control [3], [4]. In fact, the amylin analog pramlintide (Symlin) is FDA-approved for the treatment of diabetes mellitus [5], but has the additional effect of producing body weight loss in humans [6]. The actions of amylin to suppress food intake and body weight will be the focus of this review.

Amylin acts centrally to control energy balance, and this effect has been attributed primarily to activation of amylin receptors on neurons of the area postrema (AP), a hindbrain circumventricular structure. The evidence supporting the AP as an important amylin-responsive site has been extensively reviewed [2], [7], [8] and is discussed briefly below. However, the neural control of energy balance is not restricted to a select nucleus or subset of nuclei, but rather is distributed across the entire CNS [9], [10]. Given that amylin binds to sites throughout the CNS [11], [12], [13], [14], [15], the idea that amylin may act at nuclei other than the AP to control feeding has received surprisingly little attention. Recent data from our laboratory [16] and others [17], [18], [19], [20] have begun to highlight the importance of additional CNS structures in contributing to the intake- and body weight-suppressive effects of amylin. Specifically, we have recently shown that the ventral tegmental area (VTA), a mesolimbic nucleus important in food intake, food reward, and motivated behavior [21], [22], [23], is a physiologically relevant site for the actions of amylin to reduce food intake [16]. Thus, the ability of amylin to promote negative energy balance through activation of extra-AP sites is the main focus of this review.

Section snippets

The amylin receptor complex

The amylin receptor (AMY) comprises a calcitonin receptor (CTR) heterodimerized with a receptor activity modifying protein (RAMP). There are two splice variants of CTR, named CTRA and CTRB. CTRA is the more prevalent isoform and is widely expressed in the periphery and the brain [24]. CTRB expression is restricted to the CNS, but at lower levels than CTRA [12], [24]. The CTR can be activated by several closely related peptides including calcitonin, amylin, adrenomedullin, and CGRP. The CTR

Amylin activates the area postrema to reduce food intake

Although amylin is produced predominantly by the pancreas, a central site of action for the hormone is suggested by the finding that neither surgical vagotomy [43] nor chemical ablation of peripheral sensory afferent fibers via capsaicin treatment [44] attenuates the ability of peripherally administered amylin to reduce food intake. As mentioned briefly above, the most well-studied CNS site mediating the intake-suppressive effects of amylin is the area postrema (AP), a hindbrain

Alternative CNS sites of action for amylinergic control of energy balance

Although a multitude of studies have demonstrated convincingly that the AP is an important site mediating the effects of amylin on energy balance [17], [45], [55], the collective body of literature should not be interpreted to mean that the AP is the only site at which amylin acts to exert these effects. Importantly, even in rats with AP lesions, amylin- and amylin agonist-mediated behavioral and physiological effects usually attributed to AP amylin receptor activation are still observed. For

An alternative endogenous source of amylin?

If the amylin produced and secreted by the pancreatic β-cells represents the sole source of endogenous amylin in the body, it seems logical to assume that pancreas-derived amylin must travel humorally to gain access to the brain. The fact that the AP is not protected by the blood–brain barrier provides a parsimonious explanation for how amylin reaches this circumventricular structure. However, the finding that VTA amylin receptors are also physiologically relevant for the control of food intake

Amylin receptor-mediated intracellular signaling pathways

The activation of a G-protein coupled receptor, such as the amylin receptor, by binding of its ligand activates or inhibits numerous intracellular signaling molecules [36]. Broadly speaking, stimulation or suppression of these intracellular pathways can impact a variety of physiological processes that alter behavior. Activation of specific intracellular signaling molecules can induce changes both at the cellular level (e.g., alterations to gene expression; changes in intracellular calcium

Amylin-based combination pharmacotherapies for obesity treatment

The incidence of obesity has risen dramatically in many countries around the world [95], [96]. Effective anti-obesity drugs are urgently needed as behavioral therapy offers limited success, and although gastrointestinal bariatric surgery is effective, it can have serious adverse consequences [97]. To this end, pharmaceutical treatments targeting the amylin system may hold promise for treating obesity. Amylin receptor agonists reduce food intake and body weight in both humans and animal models

Conclusion

While the AP undoubtedly mediates some of the food intake-suppressive effects of amylin, the distributed nature of the control of energy balance [9], [10] and the multitude of CNS sites that bind amylin point to the likelihood that amylin can also reduce feeding through actions at other CNS nuclei. The recent data from our laboratory showing that amylin receptor signaling in the VTA is physiologically relevant for the control of food intake [16] are among the first published data indicating a

Acknowledgments

This manuscript is based in part on work presented during the 2013 Annual Meeting of the Society for the Study of Ingestive Behavior, July 30–August 3, 2013. This work was supported by NIH grants DK097954 (E.G.M.-B.) and DK096139 (M.R.H.).

References (122)

  • N.S. Narayanan et al.

    Metabolic hormones, dopamine circuits, and feeding

    Front Neuroendocrinol

    (2010)
  • K. Albrandt et al.

    Molecular cloning of two receptors from rat brain with high affinity for salmon calcitonin

    FEBS Lett

    (1993)
  • A. Young

    Receptor pharmacology

    Adv Pharmacol

    (2005)
  • T.J. Martin et al.

    Heterogeneity of the calcitonin receptor: functional aspects in osteoclasts and other sites

    J Nutr

    (1995)
  • P.M. Sexton et al.

    In vitro autoradiographic localization of amylin binding sites in rat brain

    Neuroscience

    (1994)
  • S.W. Barth et al.

    Peripheral amylin activates circumventricular organs expressing calcitonin receptor a/b subtypes and receptor-activity modifying proteins in the rat

    Brain Res

    (2004)
  • T.A. Lutz et al.

    Subdiaphragmatic vagotomy does not influence the anorectic effect of amylin

    Peptides

    (1995)
  • A. Mollet et al.

    Infusion of the amylin antagonist AC 187 into the area postrema increases food intake in rats

    Physiol Behav

    (2004)
  • N.E. Rowland et al.

    Comparison of Fos induced in rat brain by GLP-1 and amylin

    Regul Pept

    (1997)
  • T.A. Lutz et al.

    Lesion of the area postrema/nucleus of the solitary tract (AP/NTS) attenuates the anorectic effects of amylin and calcitonin gene-related peptide (CGRP) in rats

    Peptides

    (1998)
  • I.J. Llewellyn-Smith et al.

    Preproglucagon neurons project widely to autonomic control areas in the mouse brain

    Neuroscience

    (2011)
  • M. Funahashi et al.

    Variety of morphological and electrophysiological properties of area postrema neurons in adult rat brain slices

    Neurosci Res

    (2006)
  • Y. Cai et al.

    Synaptic connections and interactions between area postrema and nucleus tractus solitarius

    Brain Res

    (1996)
  • H.J. Grill et al.

    Hindbrain neurons as an essential hub in the neuroanatomically distributed control of energy balance

    Cell Metab

    (2012)
  • W.T. Chance et al.

    Tests of adipsia and conditioned taste aversion following the intrahypothalamic injection of amylin

    Peptides

    (1992)
  • S.W. Barth et al.

    Differential effects of amylin and salmon calcitonin on neuropeptide gene expression in the lateral hypothalamic area and the arcuate nucleus of the rat

    Neurosci Lett

    (2003)
  • C.S. Potes et al.

    Identification of central projections from amylin-activated neurons to the lateral hypothalamus

    Brain Res

    (2010)
  • C. Becskei et al.

    Immunohistochemical mapping of calcitonin receptors in the adult rat brain

    Brain Res

    (2004)
  • S. Fulton et al.

    Leptin regulation of the mesoaccumbens dopamine pathway

    Neuron

    (2006)
  • J.D. Hommel et al.

    Leptin receptor signaling in midbrain dopamine neurons regulates feeding

    Neuron

    (2006)
  • A.M. Naleid et al.

    Ghrelin induces feeding in the mesolimbic reward pathway between the ventral tegmental area and the nucleus accumbens

    Peptides

    (2005)
  • T.A. Lutz et al.

    Amylin decreases meal size in rats

    Physiol Behav

    (1995)
  • G. Clementi et al.

    Effects of centrally injected amylin on sexually behavior of male rats

    Peptides

    (1999)
  • W.A. Banks et al.

    Permeability of the blood–brain barrier to amylin

    Life Sci

    (1995)
  • H. Mulder et al.

    Islet amyloid polypeptide is expressed in endocrine cells of the gastric mucosa in the rat and mouse

    Gastroenterology

    (1994)
  • M.R. Hayes

    Neuronal and intracellular signaling pathways mediating GLP-1 energy balance and glycemic effects

    Physiol Behav

    (2012)
  • T.A. Lutz et al.

    The histaminergic, but not the serotoninergic, system mediates amylin's anorectic effect

    Peptides

    (1996)
  • R. Seth et al.

    Amylin-leptin coadministration stimulates central histaminergic signaling in rats

    Brain Res

    (2012)
  • C. De Mei et al.

    Getting specialized: presynaptic and postsynaptic dopamine D2 receptors

    Curr Opin Pharmacol

    (2009)
  • P.W. Kalivas

    Neurotransmitter regulation of dopamine neurons in the ventral tegmental area

    Brain Res Brain Res Rev

    (1993)
  • W.B. Inabnet et al.

    Early outcomes of bariatric surgery in patients with metabolic syndrome: an analysis of the bariatric outcomes longitudinal database

    J Am Coll Surg

    (2012)
  • A. Ogawa et al.

    Amylin secretion from the rat pancreas and its selective loss after streptozotocin treatment

    J Clin Invest

    (1990)
  • T.A. Lutz

    The role of amylin in the control of energy homeostasis

    Am J Physiol Regul Integr Comp Physiol

    (2010)
  • T.A. Lutz

    Roles of amylin in satiation, adiposity and brain development

    Forum Nutr

    (2010)
  • L.M. Younk et al.

    Pramlintide and the treatment of diabetes: a review of the data since its introduction

    Expert Opin Pharmacother

    (2011)
  • I. Chapman et al.

    Low-dose pramlintide reduced food intake and meal duration in healthy, normal-weight subjects

    Obesity

    (2007)
  • K. Beaumont et al.

    High affinity amylin binding sites in rat brain

    Mol Pharmacol

    (1993)
  • G. Skofitsch et al.

    Comparative immunohistochemical distribution of amylin-like and calcitonin gene related peptide like immunoreactivity in the rat central nervous system

    Can J Physiol Pharmacol

    (1995)
  • E.G. Mietlicki-Baase et al.

    Amylin receptor signaling in the ventral tegmental area is physiologically relevant for the control of food intake

    Neuropsychopharmacology

    (2013)
  • T. Riediger et al.

    The anorectic hormone amylin contributes to feeding-related changes of neuronal activity in key structures of the gut-brain axis

    Am J Physiol Regul Integr Comp Physiol

    (2004)
  • Cited by (43)

    • Creating the amylin story

      2022, Appetite
      Citation Excerpt :

      Various groups studied effects of amylin and its agonists on reward mediated actions. These studies have been elegantly summarized in recent reviews (Kern & Mietlicki-Baase, 2020; E.G.; Mietlicki-Baase & Hayes, 2014). Briefly, amylin activation of the VTA and the NAc seems to be linked to a decrease in the rewarding effect of food (Mietlicki-Baase et al., 2013) but also of alcohol intake (Kalafateli, Vallof, & Jerlhag, 2019).

    • Effects of pramlintide on energy intake and food preference in rats given a choice diet

      2021, Physiology and Behavior
      Citation Excerpt :

      We also assessed effects of centrally administered pramlintide on food intake and choice of chow or HFD, because peripherally administered pramlintide can cross the blood-brain barrier [37]. Further, as the feeding effects of amylin are mediated centrally (reviewed in [38]), we were interested in understanding how a more directed activation of central amylin receptors with pramlintide would impact feeding and weight gain. We hypothesized that pramlintide treatment would reduce total energy intake in rats via a preferential reduction in intake of HFD rather than chow intake.

    View all citing articles on Scopus
    View full text