1
|
Hankir MK, Le Foll C. Central nervous system pathways targeted by amylin in the regulation of food intake. Biochimie 2024:S0300-9084(24)00238-4. [PMID: 39426704 DOI: 10.1016/j.biochi.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Amylin is a peptide hormone co-released with insulin from pancreatic β-cells during a meal and primarily serves to promote satiation. While the caudal hindbrain was originally implicated as a major site of action in this regard, it is becoming increasingly clear that amylin recruits numerous central nervous system pathways to exert multifaceted effects on food intake. In this Review, we discuss the evidence derived from preclinical studies showing that amylin and the related peptide salmon calcitonin (sCT) directly or indirectly target genetically distinct neurons in the caudal hindbrain (nucleus tractus solitarius and area postrema), rostral hindbrain (lateral parabrachial nucleus), midbrain (lateral dorsal tegmentum and ventral tegmental area) and hypothalamus (arcuate nucleus and parasubthalamic nucleus) via activation of amylin and/or calcitonin receptors. Given that the stable amylin analogue cagrilintide is under clinical development for the treatment of obesity, it is important to determine whether this drug recruits overlapping or distinct central nervous system pathways to that of amylin and sCT with implications for minimising any aversive effects it potentially causes. Such insight will also be important to understand how amylin and sCT analogues synergize with other molecules as part of dual or triple agonist therapies for obesity, especially the glucagon-like peptide 1 receptor (GLP-1R) agonist semaglutide, which has been shown to synergistically lower body weight with cagrilintide (CagriSema) in clinical trials.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Department of Veterinary Physiology, University of Zurich, Zurich, Switzerland; School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
| | - Christelle Le Foll
- Department of Veterinary Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Kurachi T, Shinozuka K, Yoshihara C, Yano-Nashimoto S, Murayama AY, Hata J, Haga Y, Okano H, Kuroda KO. Distinct roles of amylin and oxytocin signaling in intrafamilial social behaviors at the medial preoptic area of common marmosets. Commun Biol 2023; 6:1231. [PMID: 38052969 PMCID: PMC10698028 DOI: 10.1038/s42003-023-05593-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Calcitonin receptor (Calcr) and its brain ligand amylin in the medial preoptic area (MPOA) are found to be critically involved in infant care and social contact behaviors in mice. In primates, however, the evidence is limited to an excitotoxic lesion study of the Calcr-expressing MPOA subregion (cMPOA) in a family-living primate species, the common marmoset. The present study utilized pharmacological manipulations of the cMPOA and shows that reversible inactivation of the cMPOA abolishes infant-care behaviors in sibling marmosets without affecting other social or non-social behaviors. Amylin-expressing neurons in the marmoset MPOA are distributed in the vicinity of oxytocin neurons in the anterior paraventricular nucleus of the hypothalamus. While amylin infusion facilitates infant carrying selectively, an oxytocin's inverse agonist, atosiban, reduces physical contact with non-infant family members without grossly affecting infant care. These data suggest that the amylin and oxytocin signaling mediate intrafamilial social interactions in a complementary manner in marmosets.
Collapse
Affiliation(s)
- Takuma Kurachi
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, Japan
- Department of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kazutaka Shinozuka
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, Japan
| | - Chihiro Yoshihara
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Saori Yano-Nashimoto
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, Japan
- Laboratory of Physiology, Department of Basic Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Ayako Y Murayama
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Junichi Hata
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Saitama, Japan
- Graduate School of Human Health Sciences, Tokyo Metropolitan University, Arakawa, Tokyo, Japan
| | - Yawara Haga
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Saitama, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Saitama, Japan
| | - Kumi O Kuroda
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, Japan.
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan.
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan.
| |
Collapse
|
3
|
Haspula D, Cui Z. Neurochemical Basis of Inter-Organ Crosstalk in Health and Obesity: Focus on the Hypothalamus and the Brainstem. Cells 2023; 12:1801. [PMID: 37443835 PMCID: PMC10341274 DOI: 10.3390/cells12131801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Precise neural regulation is required for maintenance of energy homeostasis. Essential to this are the hypothalamic and brainstem nuclei which are located adjacent and supra-adjacent to the circumventricular organs. They comprise multiple distinct neuronal populations which receive inputs not only from other brain regions, but also from circulating signals such as hormones, nutrients, metabolites and postprandial signals. Hence, they are ideally placed to exert a multi-tier control over metabolism. The neuronal sub-populations present in these key metabolically relevant nuclei regulate various facets of energy balance which includes appetite/satiety control, substrate utilization by peripheral organs and glucose homeostasis. In situations of heightened energy demand or excess, they maintain energy homeostasis by restoring the balance between energy intake and expenditure. While research on the metabolic role of the central nervous system has progressed rapidly, the neural circuitry and molecular mechanisms involved in regulating distinct metabolic functions have only gained traction in the last few decades. The focus of this review is to provide an updated summary of the mechanisms by which the various neuronal subpopulations, mainly located in the hypothalamus and the brainstem, regulate key metabolic functions.
Collapse
Affiliation(s)
- Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Zhenzhong Cui
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
| |
Collapse
|
4
|
Romano A, Friuli M, Eramo B, Gallelli CA, Koczwara JB, Azari EK, Paquot A, Arnold M, Langhans W, Muccioli GG, Lutz TA, Gaetani S. "To brain or not to brain": evaluating the possible direct effects of the satiety factor oleoylethanolamide in the central nervous system. Front Endocrinol (Lausanne) 2023; 14:1158287. [PMID: 37234803 PMCID: PMC10206109 DOI: 10.3389/fendo.2023.1158287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
Introduction Oleoylethanolamide (OEA), an endogenous N-acylethanolamine acting as a gut-to-brain signal to control food intake and metabolism, has been attracting attention as a target for novel therapies against obesity and eating disorders. Numerous observations suggested that the OEA effects might be peripherally mediated, although they involve central pathways including noradrenergic, histaminergic and oxytocinergic systems of the brainstem and the hypothalamus. Whether these pathways are activated directly by OEA or whether they are downstream of afferent nerves is still highly debated. Some early studies suggested vagal afferent fibers as the main route, but our previous observations have contradicted this idea and led us to consider the blood circulation as an alternative way for OEA's central actions. Methods To test this hypothesis, we first investigated the impact of subdiaphragmatic vagal deafferentation (SDA) on the OEA-induced activation of selected brain nuclei. Then, we analyzed the pattern of OEA distribution in plasma and brain at different time points after intraperitoneal administration in addition to measuring food intake. Results Confirming and extending our previous findings that subdiaphragmatic vagal afferents are not necessary for the eating-inhibitory effect of exogenous OEA, our present results demonstrate that vagal sensory fibers are also not necessary for the neurochemical effects of OEA. Rather, within a few minutes after intraperitoneal administration, we found an increased concentration of intact OEA in different brain areas, associated with the inhibition of food intake. Conclusion Our results support that systemic OEA rapidly reaches the brain via the circulation and inhibits eating by acting directly on selected brain nuclei.
Collapse
Affiliation(s)
- Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Marzia Friuli
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Barbara Eramo
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Cristina Anna Gallelli
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Justyna Barbara Koczwara
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | | | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, UCLouvain, Brussels, Belgium
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Zurich, Switzerland
| | | | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, UCLouvain, Brussels, Belgium
| | - Thomas Alexander Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Vagena E, Crneta J, Engström P, He L, Yulyaningsih E, Korpel NL, Cheang RT, Bachor TP, Huang A, Michel G, Attal K, Berrios DI, Valdearcos M, Koliwad SK, Olson DP, Yi CX, Xu AW. ASB4 modulates central melanocortinergic neurons and calcitonin signaling to control satiety and glucose homeostasis. Sci Signal 2022; 15:eabj8204. [PMID: 35536884 DOI: 10.1126/scisignal.abj8204] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Variants in the gene encoding ankyrin repeat and SOCS box-containing 4 (ASB4) are linked to human obesity. Here, we characterized the pathways underlying the metabolic functions of ASB4. Hypothalamic Asb4 expression was suppressed by fasting in wild-type mice but not in mice deficient in AgRP, which encodes Agouti-related protein (AgRP), an appetite-stimulating hormone, suggesting that ASB4 is a negative target of AgRP. Many ASB4 neurons in the brain were adjacent to AgRP terminals, and feeding induced by AgRP neuronal activation was disrupted in Asb4-deficient mice. Acute knockdown of Asb4 in the brain caused marked hyperphagia due to increased meal size, and Asb4 deficiency led to increased meal size and food intake at the onset of refeeding, when very large meals were consumed. Asb4-deficient mice were resistant to the meal-terminating effects of exogenously administered calcitonin and showed decreased neuronal expression of Calcr, which encodes the calcitonin receptor. Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus in mice are involved in glucose homeostasis, and Asb4 deficiency specifically in POMC neurons resulted in glucose intolerance that was independent of obesity. Furthermore, individuals with type 2 diabetes showed reduced ASB4 abundance in the infundibular nuclei, the human equivalent of the arcuate nucleus. Together, our results indicate that ASB4 acts in the brain to improve glucose homeostasis and to induce satiety after substantial meals, particularly those after food deprivation.
Collapse
Affiliation(s)
- Eirini Vagena
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jasmina Crneta
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Pauline Engström
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Li He
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ernie Yulyaningsih
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nikita L Korpel
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Rachel T Cheang
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tomas P Bachor
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alyssa Huang
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Guillermina Michel
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kush Attal
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David I Berrios
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Suneil K Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David P Olson
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Allison W Xu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
6
|
Mediators of Amylin Action in Metabolic Control. J Clin Med 2022; 11:jcm11082207. [PMID: 35456307 PMCID: PMC9025724 DOI: 10.3390/jcm11082207] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Amylin (also called islet amyloid polypeptide (IAPP)) is a pancreatic beta-cell hormone that is co-secreted with insulin in response to nutrient stimuli. The last 35 years of intensive research have shown that amylin exerts important physiological effects on metabolic control. Most importantly, amylin is a physiological control of meal-ending satiation, and it limits the rate of gastric emptying and reduces the secretion of pancreatic glucagon, in particular in postprandial states. The physiological effects of amylin and its analogs are mediated by direct brain activation, with the caudal hindbrain playing the most prominent role. The clarification of the structure of amylin receptors, consisting of the calcitonin core receptor plus receptor-activity modifying proteins, aided in the development of amylin analogs with a broad pharmacological profile. The general interest in amylin physiology and pharmacology was boosted by the finding that amylin is a sensitizer to the catabolic actions of leptin. Today, amylin derived analogs are considered to be among the most promising approaches for the pharmacotherapy against obesity. At least in conjunction with insulin, amylin analogs are also considered important treatment options in diabetic patients, so that new drugs may soon be added to the only currently approved compound pramlintide (Symlin®). This review provides a brief summary of the physiology of amylin’s mode of actions and its role in the control of the metabolism, in particular energy intake and glucose metabolism.
Collapse
|
7
|
Lutz TA. Creating the amylin story. Appetite 2022; 172:105965. [DOI: 10.1016/j.appet.2022.105965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023]
|
8
|
Tufvesson-Alm M, Shevchouk OT, Jerlhag E. Insight into the role of the gut-brain axis in alcohol-related responses: Emphasis on GLP-1, amylin, and ghrelin. Front Psychiatry 2022; 13:1092828. [PMID: 36699502 PMCID: PMC9868418 DOI: 10.3389/fpsyt.2022.1092828] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol's rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Stein LM, McGrath LE, Lhamo R, Koch-Laskowski K, Fortin SM, Skarbaliene J, Baader-Pagler T, Just R, Hayes MR, Mietlicki-Baase EG. The long-acting amylin/calcitonin receptor agonist ZP5461 suppresses food intake and body weight in male rats. Am J Physiol Regul Integr Comp Physiol 2021; 321:R250-R259. [PMID: 34259025 PMCID: PMC8409915 DOI: 10.1152/ajpregu.00337.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 06/30/2021] [Accepted: 07/04/2021] [Indexed: 01/18/2023]
Abstract
The peptide hormone amylin reduces food intake and body weight and is an attractive candidate target for novel pharmacotherapies to treat obesity. However, the short half-life of native amylin and amylin analogs like pramlintide limits these compounds' potential utility in promoting sustained negative energy balance. Here, we evaluate the ability of the novel long-acting amylin/calcitonin receptor agonist ZP5461 to reduce feeding and body weight in rats, and also test the role of calcitonin receptors (CTRs) in the dorsal vagal complex (DVC) of the hindbrain in the energy balance effects of chronic ZP5461 administration. Acute dose-response studies indicate that systemic ZP5461 (0.5-3 nmol/kg) robustly suppresses energy intake and body weight gain in chow- and high-fat diet (HFD)-fed rats. When HFD-fed rats received chronic systemic administration of ZP5461 (1-2 nmol/kg), the compound initially produced reductions in energy intake and weight gain but failed to produce sustained suppression of intake and body weight. Using virally mediated knockdown of DVC CTRs, the ability of chronic systemic ZP5461 to promote early reductions in intake and body weight gain was determined to be mediated in part by activation of DVC CTRs, implicating the DVC as a central site of action for ZP5461. Future studies should address other dosing regimens of ZP5461 to determine whether an alternative dose/frequency of administration would produce more sustained body weight suppression.
Collapse
Affiliation(s)
- Lauren M Stein
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lauren E McGrath
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rinzin Lhamo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kieran Koch-Laskowski
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samantha M Fortin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth G Mietlicki-Baase
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, New York
| |
Collapse
|
10
|
Arrigoni S, Le Foll C, Cabak A, Lundh S, Raun K, John LM, Lutz TA. A selective role for receptor activity-modifying proteins in subchronic action of the amylin selective receptor agonist NN1213 compared with salmon calcitonin on body weight and food intake in male mice. Eur J Neurosci 2021; 54:4863-4876. [PMID: 34189795 PMCID: PMC8457108 DOI: 10.1111/ejn.15376] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
The role of receptor activity‐modifying proteins (RAMPs) in modulating the pharmacological effects of an amylin receptor selective agonist (NN1213) or the dual amylin–calcitonin receptor agonist (DACRA), salmon calcitonin (sCT), was tested in three RAMP KO mouse models, RAMP1, RAMP3 and RAMP1/3 KO. Male wild‐type (WT) and knockout (KO) littermate mice were fed a 45% high‐fat diet for 20 weeks prior to the 3‐week treatment period. A decrease in body weight after NN1213 was observed in all WT mice, whereas sCT had no effect. The absence of RAMP1 had no significant effect on NN1213 efficacy, and sCT was still inactive. However, the absence of RAMP3 impeded NN1213 efficacy but improved sCT efficacy. Similar results were observed in RAMP1/3 KO suggesting that the amylin receptor 3 (AMY3 = CTR + RAMP3) is necessary for NN1213's maximal action on body weight and food intake and that the lack of AMY3 allowed sCT to be active. These results suggest that the chronic use of DACRA such as sCT can have unfavourable effect on body weight loss in mice (which differs from the situation in rats), whereas the use of the amylin receptor selective agonist does not. AMY3 seems to play a crucial role in modulating the action of these two compounds, but in opposite directions. The assessment of a long‐term effect of amylin and DACRA in different rodent models is necessary to understand potential physiological beneficial and unfavourable effects on weight loss before its transition to clinical trials.
Collapse
Affiliation(s)
- Soraya Arrigoni
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Andrea Cabak
- Global Research, Novo Nordisk AS, Måløv, Denmark
| | - Sofia Lundh
- Global Research, Novo Nordisk AS, Måløv, Denmark
| | - Kirsten Raun
- Global Research, Novo Nordisk AS, Måløv, Denmark
| | - Linu M John
- Global Research, Novo Nordisk AS, Måløv, Denmark
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Honegger M, Lutz TA, Boyle CN. Hypoglycemia attenuates acute amylin-induced reduction of food intake in male rats. Physiol Behav 2021; 237:113435. [PMID: 33933418 DOI: 10.1016/j.physbeh.2021.113435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/09/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022]
Abstract
The ability of amylin to inhibit gastric emptying and glucagon secretion in rats is reduced under hypoglycemic conditions. These effects are considered part of a fail-safe mechanism that prevents amylin from further decreasing nutrient supply when blood glucose levels are low. Because these actions and amylin-induced satiation are mediated by the area postrema (AP), it is plausible that these phenomena are based on the co-sensitivity of AP neurons to amylin and glucose. Using hyperinsulinemic glucose clamps in unrestrained and freely-feeding rats, we investigated whether amylin's ability to inhibit food intake is also reduced by hypoglycemia (HYPO). Following an 18 h fast, rats were infused with insulin and glucose for 45 min to clamp blood glucose at baseline levels (between 90 and 100 mg/dL). HYPO (approximately 55 mg/dL) was induced between 45 and 60 min and then maintained for the remainder of the clamp. Rats were injected with amylin (20 µg/kg) or saline and offered normal chow at 85 min. Food intake was measured at 30 and 60 min after amylin. Control hyperinsulinemic/euglycemic (EU) rats were maintained at approximately 150 mg/dL (which is a physiological periprandial glucose level) before and after amylin injection. Terminal experiments tested the effect of amylin to induce the phosphorylation of ERK, a marker of amylin action in the AP, in EU and HYPO conditions. Amylin significantly reduced 30- and 60-min food intake in EU rats, but the effect at 60-min was attenuated in HYPO rats. Interestingly, glucose infusion rate had to be dramatically reduced at meal onset in saline-treated, but not in amylin-treated, EU or HYPO rats; this suggests that meal-related glucose appearance in the blood was inhibited by amylin under both EU and HYPO. Finally, amylin induced a similar pERK response in the AP in EU and HYPO rats. We conclude that amylin's action to decrease eating is blunted in hypoglycemia, and this effect seems to be downstream from amylin-induced pERK in AP neurons. These data allow us to extend the idea of a hypoglycemic brake on amylin's actions to its food intake-reducing effect, but also demonstrate that amylin can buffer meal-induced glucose appearance at EU and HYPO levels.
Collapse
Affiliation(s)
- Miriam Honegger
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland; Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Christina N Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland.
| |
Collapse
|
12
|
Ludwig MQ, Cheng W, Gordian D, Lee J, Paulsen SJ, Hansen SN, Egerod KL, Barkholt P, Rhodes CJ, Secher A, Knudsen LB, Pyke C, Myers MG, Pers TH. A genetic map of the mouse dorsal vagal complex and its role in obesity. Nat Metab 2021; 3:530-545. [PMID: 33767443 DOI: 10.1038/s42255-021-00363-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
The brainstem dorsal vagal complex (DVC) is known to regulate energy balance and is the target of appetite-suppressing hormones, such as glucagon-like peptide 1 (GLP-1). Here we provide a comprehensive genetic map of the DVC and identify neuronal populations that control feeding. Combining bulk and single-nucleus gene expression and chromatin profiling of DVC cells, we reveal 25 neuronal populations with unique transcriptional and chromatin accessibility landscapes and peptide receptor expression profiles. GLP-1 receptor (GLP-1R) agonist administration induces gene expression alterations specific to two distinct sets of Glp1r neurons-one population in the area postrema and one in the nucleus of the solitary tract that also expresses calcitonin receptor (Calcr). Transcripts and regions of accessible chromatin near obesity-associated genetic variants are enriched in the area postrema and the nucleus of the solitary tract neurons that express Glp1r and/or Calcr, and activating several of these neuronal populations decreases feeding in rodents. Thus, DVC neuronal populations associated with obesity predisposition suppress feeding and may represent therapeutic targets for obesity.
Collapse
Affiliation(s)
- Mette Q Ludwig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Wenwen Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Desiree Gordian
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Julie Lee
- Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anna Secher
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | | | - Charles Pyke
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
13
|
Sonne N, Karsdal MA, Henriksen K. Mono and dual agonists of the amylin, calcitonin, and CGRP receptors and their potential in metabolic diseases. Mol Metab 2021; 46:101109. [PMID: 33166741 PMCID: PMC8085567 DOI: 10.1016/j.molmet.2020.101109] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Therapies for metabolic diseases are numerous, yet improving insulin sensitivity beyond that induced by weight loss remains challenging. Therefore, search continues for novel treatment candidates that can stimulate insulin sensitivity and increase weight loss efficacy in combination with current treatment options. Calcitonin gene-related peptide (CGRP) and amylin belong to the same peptide family and have been explored as treatments for metabolic diseases. However, their full potential remains controversial. SCOPE OF REVIEW In this article, we introduce this rather complex peptide family and its corresponding receptors. We discuss the physiology of the peptides with a focus on metabolism and insulin sensitivity. We also thoroughly review the pharmacological potential of amylin, calcitonin, CGRP, and peptide derivatives as treatments for metabolic diseases, emphasizing their ability to increase insulin sensitivity based on preclinical and clinical studies. MAJOR CONCLUSIONS Amylin receptor agonists and dual amylin and calcitonin receptor agonists are relevant treatment candidates, especially because they increase insulin sensitivity while also assisting weight loss, and their unique mode of action complements incretin-based therapies. However, CGRP and its derivatives seem to have only modest if any metabolic effects and are no longer of interest as therapies for metabolic diseases.
Collapse
Affiliation(s)
- Nina Sonne
- Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Morten A Karsdal
- Nordic Bioscience Biomarkers and Research, Herlev, Denmark; KeyBioscience AG, Stans, Switzerland
| | - Kim Henriksen
- Nordic Bioscience Biomarkers and Research, Herlev, Denmark; KeyBioscience AG, Stans, Switzerland.
| |
Collapse
|
14
|
An amylin and calcitonin receptor agonist modulates alcohol behaviors by acting on reward-related areas in the brain. Prog Neurobiol 2020; 200:101969. [PMID: 33278524 DOI: 10.1016/j.pneurobio.2020.101969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/16/2020] [Accepted: 11/29/2020] [Indexed: 02/07/2023]
Abstract
Alcohol causes stimulatory behavioral responses by activating reward-processing brain areas including the laterodorsal (LDTg) and ventral tegmental areas (VTA) and the nucleus accumbens (NAc). Systemic administration of the amylin and calcitonin receptor agonist salmon calcitonin (sCT) attenuates alcohol-mediated behaviors, but the brain sites involved in this process remain unknown. Firstly, to identify potential sCT sites of action in the brain, we used immunohistochemistry after systemic administration of fluorescent-labeled sCT. We then performed behavioral experiments to explore how infused sCT into the aforementioned reward-processing brain areas affects acute alcohol-induced behaviors in mice and chronic alcohol consumption in rats. We show that peripheral sCT crosses the blood brain barrier and is detected in all the brain areas studied herein. sCT infused into the LDTg attenuates alcohol-evoked dopamine release in the NAc shell in mice and reduces alcohol intake in rats. sCT into the VTA blocks alcohol-induced locomotor stimulation and dopamine release in the NAc shell in mice and decreases alcohol intake in rats. Lastly, sCT into the NAc shell prevents alcohol-induced locomotor activity in mice. Our data suggest that central sCT modulates the ability of alcohol to activate reward-processing brain regions.
Collapse
|
15
|
Boccia L, Gamakharia S, Coester B, Whiting L, Lutz TA, Le Foll C. Amylin brain circuitry. Peptides 2020; 132:170366. [PMID: 32634450 DOI: 10.1016/j.peptides.2020.170366] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022]
Abstract
Amylin is a peptide hormone that is mainly known to be produced by pancreatic β-cells in response to a meal but amylin is also produced by brain cells in discrete brain areas albeit in a lesser amount. Amylin receptor (AMY) is composed of the calcitonin core-receptor (CTR) and one of the 3 receptor activity modifying protein (RAMP), thus forming AMY1-3; RAMP enhances amylin binding properties to the CTR. However, amylin receptor agonist such as salmon calcitonin is able to bind CTR alone. Peripheral amylin's main binding site is located in the area postrema (AP) which then propagate the signal to the nucleus of the solitary tract and lateral parabrachial nucleus (LPBN) and it is then transmitted to the forebrain areas such as central amygdala and bed nucleus of the stria terminalis. Amylin's activation of these different brain areas mediates eating and other metabolic pathways controlling energy expenditure and glucose homeostasis. Peripheral amylin can also bind in the arcuate nucleus of the hypothalamus where it acts independently of the AP to activate POMC and NPY neurons. Amylin activation of NPY neurons has been shown to be transmitted to LPBN neurons to act on eating while amylin POMC signaling affects energy expenditure and locomotor activity. While a large amount of experiments have already been conducted, future studies will have to further investigate how amylin is taken up by forebrain areas and deepen our understanding of amylin action on peripheral metabolism.
Collapse
Affiliation(s)
- Lavinia Boccia
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Salome Gamakharia
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Bernd Coester
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Lynda Whiting
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland.
| |
Collapse
|
16
|
Boccia L, Le Foll C, Lutz TA. Noradrenaline signaling in the LPBN mediates amylin's and salmon calcitonin's hypophagic effect in male rats. FASEB J 2020; 34:15448-15461. [PMID: 32985027 DOI: 10.1096/fj.202001456rrr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 11/11/2022]
Abstract
The LPBN (lateral parabrachial nucleus) plays an important role in feeding control. CGRP (calcitonin gene-related peptide) LPBN neurons activation mediates the anorectic effects of different gut-derived peptides, including amylin. Amylin and its long acting analog sCT (salmon calcitonin) exert their anorectic actions primarily by directly activating neurons located in the area postrema (AP). A large proportion of projections from the AP and the adjacent nucleus of the solitary tractNTS to the LPBN, are noradrenergic (NA), and amylin-activated NAAP neurons are critical in mediating amylin's hypophagic effects. Here, we determine the functional role of NAAP amylin activated neurons to activate CGRP and non-CGRP LPBN neurons. To this end, NA was specifically depleted in the rat LPBN through a stereotaxic microinfusion of 6-OHDA, a neurotoxic agent that destroys NA terminals. While amylin (50 μg/kg) and sCT (5 μg/kg) reduced eating in sham-lesioned rats, no reduction in feeding occurred in NA-depleted animals. Further, the amylin-induced c-Fos response in the LPBN and c-Fos/CGRP colocalization were reduced in NA-depleted animals compared to controls. We conclude that AP → LPBN NA signaling, through the activation of LPBN CGRP neurons, mediates part of amylin's hypophagic effect.
Collapse
Affiliation(s)
- Lavinia Boccia
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Foll CL, Lutz TA. Systemic and Central Amylin, Amylin Receptor Signaling, and Their Physiological and Pathophysiological Roles in Metabolism. Compr Physiol 2020; 10:811-837. [PMID: 32941692 DOI: 10.1002/cphy.c190034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This article in the Neural and Endocrine Section of Comprehensive Physiology discusses the physiology and pathophysiology of the pancreatic hormone amylin. Shortly after its discovery in 1986, amylin has been shown to reduce food intake as a satiation signal to limit meal size. Amylin also affects food reward, sensitizes the brain to the catabolic actions of leptin, and may also play a prominent role in the development of certain brain areas that are involved in metabolic control. Amylin may act at different sites in the brain in addition to the area postrema (AP) in the caudal hindbrain. In particular, the sensitizing effect of amylin on leptin action may depend on a direct interaction in the hypothalamus. The concept of central pathways mediating amylin action became more complex after the discovery that amylin is also synthesized in certain hypothalamic areas but the interaction between central and peripheral amylin signaling remains currently unexplored. Amylin may also play a dominant pathophysiological role that is associated with the aggregation of monomeric amylin into larger, cytotoxic molecular entities. This aggregation in certain species may contribute to the development of type 2 diabetes mellitus but also cardiovascular disease. Amylin receptor pharmacology is complex because several distinct amylin receptor subtypes have been described, because other neuropeptides [e.g., calcitonin gene-related peptide (CGRP)] can also bind to amylin receptors, and because some components of the functional amylin receptor are also used for other G-protein coupled receptor (GPCR) systems. © 2020 American Physiological Society. Compr Physiol 10:811-837, 2020.
Collapse
Affiliation(s)
- Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Zakariassen HL, John LM, Lutz TA. Central control of energy balance by amylin and calcitonin receptor agonists and their potential for treatment of metabolic diseases. Basic Clin Pharmacol Toxicol 2020; 127:163-177. [PMID: 32363722 DOI: 10.1111/bcpt.13427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
The prevalence of obesity and associated comorbidities such as type 2 diabetes and cardiovascular disease is increasing globally. Body-weight loss reduces the risk of morbidity and mortality in obese individuals, and thus, pharmacotherapies that induce weight loss can be of great value in improving the health and well-being of people living with obesity. Treatment with amylin and calcitonin receptor agonists reduces food intake and induces weight loss in several animal models, and a number of companies have started clinical testing for peptide analogues in the treatment of obesity and/or type 2 diabetes. Studies predominantly performed in rodent models show that amylin and the dual amylin/calcitonin receptor agonist salmon calcitonin achieve their metabolic effects by engaging areas in the brain associated with regulating homeostatic energy balance. In particular, signalling via neuronal circuits in the caudal hindbrain and the hypothalamus is implicated in mediating effects on food intake and energy expenditure. We review the current literature investigating the interaction of amylin/calcitonin receptor agonists with neurocircuits that induce the observed metabolic effects. Moreover, the status of drug development of amylin and calcitonin receptor agonists for the treatment of metabolic diseases is summarized.
Collapse
Affiliation(s)
- Hannah Louise Zakariassen
- Section of Experimental Animal Models, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark.,Obesity Pharmacology, Novo Nordisk A/S, Måløv, Denmark
| | | | | |
Collapse
|
19
|
Fortin SM, Lipsky RK, Lhamo R, Chen J, Kim E, Borner T, Schmidt HD, Hayes MR. GABA neurons in the nucleus tractus solitarius express GLP-1 receptors and mediate anorectic effects of liraglutide in rats. Sci Transl Med 2020; 12:eaay8071. [PMID: 32132220 PMCID: PMC7211411 DOI: 10.1126/scitranslmed.aay8071] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/22/2019] [Accepted: 02/13/2020] [Indexed: 01/04/2023]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) agonist liraglutide is approved for the treatment of obesity; however, there is still much to be learned regarding the neuronal sites of action that underlie its suppressive effects on food intake and body weight. Peripherally administered liraglutide in rats acts in part through central GLP-1Rs in both the hypothalamus and the hindbrain. Here, we extend findings supporting a role for hindbrain GLP-1Rs in mediating the anorectic effects of liraglutide in male rats. To dissociate the contribution of GLP-1Rs in the area postrema (AP) and the nucleus tractus solitarius (NTS), we examined the effects of liraglutide in both NTS AAV-shRNA-driven Glp1r knockdown and AP-lesioned animals. Knockdown of NTS GLP-1Rs, but not surgical lesioning of the AP, attenuated the anorectic and body weight-reducing effects of acutely delivered liraglutide. In addition, NTS c-Fos responses were maintained in AP-lesioned animals. Moreover, NTS Glp1r knockdown was sufficient to attenuate the intake- and body weight-reducing effects of chronic daily administered liraglutide over 3 weeks. Development of improved obesity pharmacotherapies requires an understanding of the cellular phenotypes targeted by GLP-1R agonists. Fluorescence in situ hybridization identified Glp1r transcripts in NTS GABAergic neurons, which when inhibited using chemogenetics, attenuated the food intake- and body weight-reducing effects of liraglutide. This work demonstrates the contribution of NTS GLP-1Rs to the anorectic potential of liraglutide and highlights a phenotypically distinct (GABAergic) population of neurons within the NTS that express the GLP-1R and are involved in the mediation of liraglutide signaling.
Collapse
Affiliation(s)
- Samantha M Fortin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachele K Lipsky
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rinzin Lhamo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jack Chen
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eun Kim
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tito Borner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heath D Schmidt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Larsen AT, Sonne N, Andreassen KV, Karsdal MA, Henriksen K. Dose Frequency Optimization of the Dual Amylin and Calcitonin Receptor Agonist KBP-088: Long-Lasting Improvement in Food Preference and Body Weight Loss. J Pharmacol Exp Ther 2020; 373:269-278. [DOI: 10.1124/jpet.119.263400] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
|
21
|
Zakariassen HL, John LM, Lykkesfeldt J, Raun K, Glendorf T, Schaffer L, Lundh S, Secher A, Lutz TA, Le Foll C. Salmon calcitonin distributes into the arcuate nucleus to a subset of NPY neurons in mice. Neuropharmacology 2020; 167:107987. [PMID: 32035146 DOI: 10.1016/j.neuropharm.2020.107987] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 01/28/2020] [Accepted: 02/02/2020] [Indexed: 01/07/2023]
Abstract
The amylin receptor (AMY) and calcitonin receptor (CTR) agonists induce acute suppression of food intake in rodents by binding to receptors in the area postrema (AP) and potentially by targeting arcuate (ARC) neurons directly. Salmon calcitonin (sCT) induces more potent, longer lasting anorectic effects compared to amylin. We thus aimed to investigate whether AMY/CTR agonists target key neuronal populations in the ARC, and whether differing brain distribution patterns could mediate the observed differences in efficacy with sCT and amylin treatment. Brains were examined by whole brain 3D imaging and confocal microscopy following subcutaneous administration of fluorescently labelled peptides to mice. We found that sCT, but not amylin, internalizes into a subset of ARC NPY neurons, along with an unknown subset of ARC, AP and dorsal vagal motor nucleus cells. ARC POMC neurons were not targeted. Furthermore, amylin and sCT displayed similar distribution patterns binding to receptors in the AP, the organum vasculosum of the lamina terminalis (OVLT) and the ARC. Amylin distributed within the median eminence with only specs of sCT being present in this region, however amylin was only detectable 10 minutes after injection while sCT displayed a residence time of up to 2 hours post injection. We conclude that AMY/CTR agonists bind to receptors in a subset of ARC NPY neurons and in circumventricular organs. Furthermore, the more sustained and greater anorectic efficacy of sCT compared to rat amylin is not attributable to differences in brain distribution patterns but may more likely be explained by greater potency at both the CTR and AMY.
Collapse
Affiliation(s)
- Hannah Louise Zakariassen
- Section of Experimental Animal Models, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1871, Frederiksberg C, Denmark; Obesity Pharmacology, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Linu Mary John
- Obesity Pharmacology, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Jens Lykkesfeldt
- Section of Experimental Animal Models, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1871, Frederiksberg C, Denmark
| | - Kirsten Raun
- Obesity Pharmacology, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Tine Glendorf
- Diabetes Pharmacology 2, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Lauge Schaffer
- Research Chemistry, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Sofia Lundh
- Pathology and Imaging, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Anna Secher
- Diabetes Pharmacology 2, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Thomas Alexander Lutz
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland.
| |
Collapse
|
22
|
Cheng W, Gonzalez I, Pan W, Tsang AH, Adams J, Ndoka E, Gordian D, Khoury B, Roelofs K, Evers SS, MacKinnon A, Wu S, Frikke-Schmidt H, Flak JN, Trevaskis JL, Rhodes CJ, Fukada SI, Seeley RJ, Sandoval DA, Olson DP, Blouet C, Myers MG. Calcitonin Receptor Neurons in the Mouse Nucleus Tractus Solitarius Control Energy Balance via the Non-aversive Suppression of Feeding. Cell Metab 2020; 31:301-312.e5. [PMID: 31955990 PMCID: PMC7104375 DOI: 10.1016/j.cmet.2019.12.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 08/29/2019] [Accepted: 12/20/2019] [Indexed: 02/02/2023]
Abstract
To understand hindbrain pathways involved in the control of food intake, we examined roles for calcitonin receptor (CALCR)-containing neurons in the NTS. Ablation of NTS Calcr abrogated the long-term suppression of food intake, but not aversive responses, by CALCR agonists. Similarly, activating CalcrNTS neurons decreased food intake and body weight but (unlike neighboring CckNTS cells) failed to promote aversion, revealing that CalcrNTS neurons mediate a non-aversive suppression of food intake. While both CalcrNTS and CckNTS neurons decreased feeding via projections to the PBN, CckNTS cells activated aversive CGRPPBN cells while CalcrNTS cells activated distinct non-CGRP PBN cells. Hence, CalcrNTS cells suppress feeding via non-aversive, non-CGRP PBN targets. Additionally, silencing CalcrNTS cells blunted food intake suppression by gut peptides and nutrients, increasing food intake and promoting obesity. Hence, CalcrNTS neurons define a hindbrain system that participates in physiological energy balance and suppresses food intake without activating aversive systems.
Collapse
Affiliation(s)
- Wenwen Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - Ian Gonzalez
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48105, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Warren Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA; Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Anthony H Tsang
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jessica Adams
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48105, USA
| | - Ermelinda Ndoka
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - Desiree Gordian
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - Basma Khoury
- Department of Surgery, University of Michigan, Ann Arbor, MI 48105, USA
| | - Karen Roelofs
- Department of Surgery, University of Michigan, Ann Arbor, MI 48105, USA
| | - Simon S Evers
- Department of Surgery, University of Michigan, Ann Arbor, MI 48105, USA
| | - Andrew MacKinnon
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - Shuangcheng Wu
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA
| | | | - Jonathan N Flak
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - James L Trevaskis
- Cardiovascular, Renal and Metabolic Diseases, AstraZenica LLC, Gaithersburg, MD 20878, USA
| | - Christopher J Rhodes
- Cardiovascular, Renal and Metabolic Diseases, AstraZenica LLC, Gaithersburg, MD 20878, USA
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Osaka University, Osaka 565-0871, Japan
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI 48105, USA
| | - Darleen A Sandoval
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA; Department of Surgery, University of Michigan, Ann Arbor, MI 48105, USA
| | - David P Olson
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48105, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Clemence Blouet
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA; Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48105, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA; Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
23
|
Sonne N, Larsen AT, Andreassen KV, Karsdal MA, Henriksen K. The Dual Amylin and Calcitonin Receptor Agonist, KBP-066, Induces an Equally Potent Weight Loss Across a Broad Dose Range While Higher Doses May Further Improve Insulin Action. J Pharmacol Exp Ther 2020; 373:92-102. [PMID: 31992608 DOI: 10.1124/jpet.119.263723] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/21/2020] [Indexed: 12/27/2022] Open
Abstract
Pharmacological treatment with dual amylin and calcitonin receptor agonists (DACRAs) cause significant weight loss and improvement of glucose homeostasis. In this study, the maximally efficacious dose of the novel DACRA, KeyBiosciencePeptide (KBP)-066, was investigated. Two different rat models were used: high-fat diet (HFD)-fed male Sprague-Dawley rats and male Zucker diabetic fatty (ZDF, fa/fa) rats to determine the maximum weight loss and glucose homeostatic effect, respectively. One acute study and one chronic study was performed in HFD rats. Two chronic studies were performed in ZDF rats: a preventive and an interventive. All studies covered a dose range of 5, 50, and 500 µg/kg KBP-066 delivered by subcutaneous injection. Treatment with KBP-066 resulted in a significant weight reduction of 13%-16% and improved glucose tolerance in HFD rats, which was independent of dose concentration. Dosing with 50 and 500 µg/kg led to a transient but significant increase in blood glucose, both in the acute and the chronic study in HFD rats. All doses of KBP-066 significantly improved glucose homeostasis in ZDF rats, both in the preventive and interventive study. Moreover, dosing with 50 and 500 µg/kg preserved insulin secretion to a greater extent than 5 µg/kg when compared with ZDF vehicle rats. Taken together, these results show that maximum weight loss is achieved with 5 µg/kg, which is within the range of previously reported DACRA dosing, whereas increasing dosing concentration to 50 and 500 µg/kg may further improve preservation of insulin secretion compared with 5 µg/kg in diabetic ZDF rats. SIGNIFICANCE STATEMENT: Here we show that KeyBiosciencePeptide (KBP)-066 induces an equally potent body weight loss across a broad dose range in obese rats. However, higher dosing of KBP-066 may improve insulin action in diabetic rats both as preventive and interventive treatment.
Collapse
Affiliation(s)
- Nina Sonne
- Nordic Bioscience Biomarkers and Research, Department of Endocrinology, Herlev, Denmark
| | - Anna Thorsø Larsen
- Nordic Bioscience Biomarkers and Research, Department of Endocrinology, Herlev, Denmark
| | - Kim Vietz Andreassen
- Nordic Bioscience Biomarkers and Research, Department of Endocrinology, Herlev, Denmark
| | - Morten Asser Karsdal
- Nordic Bioscience Biomarkers and Research, Department of Endocrinology, Herlev, Denmark
| | - Kim Henriksen
- Nordic Bioscience Biomarkers and Research, Department of Endocrinology, Herlev, Denmark
| |
Collapse
|
24
|
Kalafateli AL, Aranäs C, Jerlhag E. Effects of sub-chronic amylin receptor activation on alcohol-induced locomotor stimulation and monoamine levels in mice. Psychopharmacology (Berl) 2020; 237:3249-3257. [PMID: 32651639 PMCID: PMC7561575 DOI: 10.1007/s00213-020-05607-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 07/03/2020] [Indexed: 12/16/2022]
Abstract
RATIONALE Amylin receptors consist of the calcitonin receptor (CTR) and one of three receptor activity-modifying proteins (RAMPs). The identification of amylin receptors in areas processing reward, namely laterodorsal tegmental area (LDTg), ventral tegmental area (VTA), and nucleus accumbens (NAc), has attributed them a role as reward regulators. Indeed, acute activation of amylin receptors by the amylin receptor agonist salmon calcitonin (sCT) attenuates alcohol-induced behaviours in rodents. OBJECTIVES The effects of long-term administration of sCT on alcohol-related behaviours and the molecular mechanisms underlying these processes are not yet elucidated. To fill this knowledge gap, we investigated the effects of sub-chronic sCT treatment on the locomotor stimulatory responses to alcohol in mice and the molecular pathways involved. METHODS We assessed the behavioural effects of sub-chronic sCT treatment by means of locomotor activity experiments in mice. We used western blot to identify changes of the CTR levels and ex vivo biochemical analysis to detect changes in monoamines and their metabolites. RESULTS After discontinuation for 5 days of sCT treatment, alcohol did not induce locomotor stimulation in mice pre-treated with sCT when compared with vehicle, without altering secondary behavioural parameters of the locomotor activity experiment or the protein levels of the CTR in reward-related areas in the same set of animals. Moreover, repeated sCT treatment altered monoaminergic neurotransmission in various brain areas, including increased serotonin and decreased dopamine turnover in the VTA. Lastly, we identified a differential effect of repeated sCT and acute alcohol administration on alcohol-induced locomotion in mice, where sCT initially attenuated and later increased this alcohol response. It was further found that this treatment combination did not affect secondary behavioural parameters measured in this locomotor activity experiments. CONCLUSIONS These data suggest that sub-chronic sCT treatment differentially alters the ability of alcohol to cause locomotor stimulation, possibly through molecular mechanisms involving various neurotransmitter systems and not the CTR levels per se.
Collapse
Affiliation(s)
- Aimilia Lydia Kalafateli
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, POB 431, SE-405 30 Gothenburg, Sweden
| | - Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, POB 431, SE-405 30 Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, POB 431, SE-405 30, Gothenburg, Sweden.
| |
Collapse
|
25
|
Coester B, Pence SW, Arrigoni S, Boyle CN, Le Foll C, Lutz TA. RAMP1 and RAMP3 Differentially Control Amylin's Effects on Food Intake, Glucose and Energy Balance in Male and Female Mice. Neuroscience 2019; 447:74-93. [PMID: 31881259 DOI: 10.1016/j.neuroscience.2019.11.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/31/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Amylin is a pancreatic peptide, which acts as a key controller of food intake and energy balance and predominately binds to three receptors (AMY 1-3). AMY 1-3 are composed of a calcitonin core receptor (CTR) and associated receptor-activity modifying proteins (RAMPs) 1-3. Using RAMP1, RAMP3 and RAMP1/3 global KO mice, this study aimed to determine whether the absence of one or two RAMP subunits affects food intake, glucose homeostasis and metabolism. Of all the RAMP-deficient mice, only high-fat diet fed RAMP1/3 KO mice had increased body weight. Chow-fed RAMP3 KO and high-fat diet fed 1/3 KO male mice were glucose intolerant. Fat depots were increased in RAMP1 KO male mice. No difference in energy expenditure was observed but the respiratory exchange ratio (RER) was elevated in RAMP1/3 KO. RAMP1 and 1/3 KO male mice displayed an increase in intermeal interval (IMI) and meal duration, whereas IMI was decreased in RAMP3 KO male and female mice. WT and RAMP1, RAMP3, and RAMP1/3 KO male and female littermates were then assessed for their food intake response to an acute intraperitoneal injection of amylin or its receptor agonist, salmon calcitonin (sCT). RAMP1/3 KO were insensitive to both, while RAMP3 KO were responsive to sCT only and RAMP1 KO to amylin only. While female mice generally weighed less than male mice, only RAMP1 KO showed a clear sex difference in meal pattern and food intake tests. Lastly, a decrease in CTR fibers did not consistently correlate with a decrease in amylin- induced c-Fos expression in the area postrema (AP). Ultimately, the results from this study provide evidence for a role of RAMP1 in mediation of fat utilization and a role for RAMP3 in glucose homeostasis and amylin's anorectic effect.
Collapse
Affiliation(s)
- Bernd Coester
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Sydney W Pence
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Soraya Arrigoni
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| |
Collapse
|
26
|
Boyle CN, Le Foll C. Amylin and Leptin interaction: Role During Pregnancy, Lactation and Neonatal Development. Neuroscience 2019; 447:136-147. [PMID: 31846753 DOI: 10.1016/j.neuroscience.2019.11.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/08/2019] [Accepted: 11/21/2019] [Indexed: 01/04/2023]
Abstract
Amylin is co-secreted with insulin by pancreatic β-cells in response to a meal and produced by neurons in discrete hypothalamic brain areas. Leptin is proportionally secreted by the adipose tissue. Both hormones control food intake and energy homeostasis post-weaning in rodents. While amylin's main site of action is located in the area postrema (AP) and leptin's is located in the mediobasal hypothalamus, both hormones can also influence the other's signaling pathway; amylin has been shown enhance hypothalamic leptin signaling, and amylin signaling in the AP may rely on functional leptin receptors to modulate its effects. These two hormones also play major roles during other life periods. During pregnancy, leptin levels rise as a result of an increase in fat depot resulting in gestational leptin-resistance to prepare the maternal body for the metabolic needs during fetal development. The role of amylin is far less studied during pregnancy and lactation, though amylin levels seem to be elevated during pregnancy relative to insulin. Whether amylin and leptin interact during pregnancy and lactation remains to be assessed. Lastly, during brain development, amylin and leptin are major regulators of cell birth during embryogenesis and act as neurotrophic factors in the neonatal period. This review will highlight the role of amylin and leptin, and their possible interaction, during these dynamic time periods of pregnancy, lactation, and early development.
Collapse
Affiliation(s)
- Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| |
Collapse
|
27
|
Schultz N, Janelidze S, Byman E, Minthon L, Nägga K, Hansson O, Wennström M. Levels of islet amyloid polypeptide in cerebrospinal fluid and plasma from patients with Alzheimer's disease. PLoS One 2019; 14:e0218561. [PMID: 31206565 PMCID: PMC6576764 DOI: 10.1371/journal.pone.0218561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
The biologically active pancreatic hormone peptide islet amyloid polypeptide (IAPP) regulates brain functions such as appetite and cognition. It also plays a role in clearance of amyloid beta (Aβ), a peptide implicated in the dementia disorder Alzheimer’s disease (AD). If IAPP becomes modified, it loses its biological activity and starts to aggregate. Such aggregations have been found in the AD brain and decreased plasma levels of the unmodified IAPP (uIAPP) have been shown in the same patients. In the current study, we analyze levels of uIAPP and total IAPP (unmodified and modified) in cerebrospinal fluid (CSF) to investigate its potential as a biomarker for AD. We found no differences in neither CSF nor plasma levels of uIAPP or total IAPP in AD patients compared to cognitive healthy individuals (NC). The levels of uIAPP in CSF of NC were positively correlated with uIAPP in plasma, Q-albumin and albumin levels in CSF, but negatively correlated with CSF levels of t-tau and p-tau. These findings were not seen in AD patients. Levels of total CSF IAPP correlated positively with total Q-albumin and albumin levels in CSF in both AD and NC. In addition, total plasma IAPP correlated positively with CSF t-tau and p-tau in NC and negatively with CSF Aβ42 in AD patients. To conclude, our studies did not find evidence supporting the use of CSF IAPP as an AD biomarker. However, our findings, indicating a compromised translocation of uIAPP in and out of the brain in AD patients as well as the correlations between total plasma IAPP and AD biomarkers, encourage further research on the role for IAPP in AD.
Collapse
Affiliation(s)
- Nina Schultz
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Elin Byman
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Lennart Minthon
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Department of Acute Internal Medicine and Geriatrics, Linköping University, Linköping, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Malin Wennström
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
28
|
Liberini CG, Koch-Laskowski K, Shaulson E, McGrath LE, Lipsky RK, Lhamo R, Ghidewon M, Ling T, Stein LM, Hayes MR. Combined Amylin/GLP-1 pharmacotherapy to promote and sustain long-lasting weight loss. Sci Rep 2019; 9:8447. [PMID: 31186439 PMCID: PMC6560126 DOI: 10.1038/s41598-019-44591-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/20/2019] [Indexed: 11/24/2022] Open
Abstract
A growing appreciation of the overlapping neuroendocrine mechanisms controlling energy balance has highlighted combination therapies as a promising strategy to enhance sustained weight loss. Here, we investigated whether amylin- and glucagon-like-peptide-1 (GLP-1)-based combination therapies produce greater food intake- and body weight-suppressive effects compared to monotherapies in both lean and diet-induced obese (DIO) rats. In chow-maintained rats, systemic amylin and GLP-1 combine to reduce meal size. Furthermore, the amylin and GLP-1 analogs salmon calcitonin (sCT) and liraglutide produce synergistic-like reductions in 24 hours energy intake and body weight. The administration of sCT with liraglutide also led to a significant enhancement in cFos-activation in the dorsal-vagal-complex (DVC) compared to mono-therapy, suggesting an activation of distinct, yet overlapping neural substrates in this critical energy balance hub. In DIO animals, long-term daily administration of this combination therapy, specifically in a stepwise manner, results in reduced energy intake and greater body weight loss over time when compared to chronic mono- and combined-treated groups, without affecting GLP-1 receptor, preproglucagon or amylin-receptor gene expression in the DVC.
Collapse
Affiliation(s)
- Claudia G Liberini
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Kieran Koch-Laskowski
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Evan Shaulson
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Lauren E McGrath
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Rachele K Lipsky
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Rinzin Lhamo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Misgana Ghidewon
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Tyler Ling
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Lauren M Stein
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Kalafateli AL, Vallöf D, Colombo G, Lorrai I, Maccioni P, Jerlhag E. An amylin analogue attenuates alcohol-related behaviours in various animal models of alcohol use disorder. Neuropsychopharmacology 2019; 44:1093-1102. [PMID: 30710109 PMCID: PMC6461824 DOI: 10.1038/s41386-019-0323-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
Recent findings have identified salmon calcitonin (sCT), an amylin receptor agonist and analogue of endogenous amylin, as a potential regulator of alcohol-induced activation of the mesolimbic dopamine system and alcohol consumption. Providing that the role of amylin signalling in alcohol-related behaviours remains unknown, the present experiments investigate the effect of sCT on these behaviours and the mechanisms involved. We showed that repeated sCT administration decreased alcohol and food intake in outbred rats. Moreover, single administration of the potent amylin receptor antagonist, AC187, increased short-term alcohol intake in outbred alcohol-consuming rats, but did not affect food intake. Acute administration of sCT prevented relapse-like drinking in the "alcohol deprivation effect" model in outbred alcohol-experienced rats. Additionally, acute sCT administration reduced operant oral alcohol self-administration (under the fixed ratio 4 schedule of reinforcement) in selectively bred Sardinian alcohol-preferring rats, while it did not alter operant self-administration (under the progressive ratio schedule of reinforcement) of a highly palatable chocolate-flavoured beverage in outbred rats. Lastly, we identified differential amylin receptor expression in high compared to low alcohol-consuming rats, as reflected by decreased calcitonin receptor and increased receptor activity modifying protein 1 expression in the nucleus accumbens (NAc) of high consumers. Collectively, our data suggest that amylin signalling, especially in the NAc, may contribute to reduction of various alcohol-related behaviours.
Collapse
Affiliation(s)
- Aimilia Lydia Kalafateli
- 0000 0000 9919 9582grid.8761.8Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Daniel Vallöf
- 0000 0000 9919 9582grid.8761.8Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Giancarlo Colombo
- 0000 0001 1940 4177grid.5326.2Neuroscience Institute, Section of Cagliari, National Research Council of Italy, Monserrato, CA Italy
| | - Irene Lorrai
- 0000 0001 1940 4177grid.5326.2Neuroscience Institute, Section of Cagliari, National Research Council of Italy, Monserrato, CA Italy
| | - Paola Maccioni
- 0000 0001 1940 4177grid.5326.2Neuroscience Institute, Section of Cagliari, National Research Council of Italy, Monserrato, CA Italy
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
30
|
Sarnat HB, Flores-Sarnat L, Boltshauser E. Area Postrema: Fetal Maturation, Tumors, Vomiting Center, Growth, Role in Neuromyelitis Optica. Pediatr Neurol 2019; 94:21-31. [PMID: 30797593 DOI: 10.1016/j.pediatrneurol.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The area postrema in the caudal fourth ventricular floor is highly vascular without blood-brain or blood-cerebrospinal fluid barrier. In addition to its function as vomiting center, several others are part of the circumventricular organs for vasomotor/angiotensin II regulation, role in neuromyelitis optica related to aquaporin-4, and somatic growth and appetite regulation. Functions are immature at birth. The purpose was to demonstrate neuronal, synaptic, glial, or ependymal maturation in the area postrema of normal fetuses. We describe three area postrema tumors. METHODS Sections of caudal fourth ventricle of 12 normal human fetal brains at autopsy aged six to 40 weeks and three infants aged three to 18 months were examined. Immunocytochemical neuronal and glial markers were applied to paraffin sections. Two infants with area postrema tumors and another with neurocutaneous melanocytosis and pernicious vomiting also studied. RESULTS Area postrema neurons exhibited cytologic maturity and synaptic circuitry by 14 weeks'. Astrocytes coexpressed vimentin, glial fibrillary acidic protein, and S-100β protein. The ependyma is thin over area postrema, with fetal ependymocytic basal processes. A glial layer separates area postrema from medullary tegmentum. Melanocytes infiltrated area postrema in the toddler with pernicious vomiting; two children had primary area postrema pilocytic astrocytomas. CONCLUSIONS Although area postrema is cytologically mature by 14 weeks, growth increases and functions mature during postnatal months. We recommend neuroimaging for patients with unexplained vomiting and that area postrema neuropathology includes synaptophysin and microtubule-associated protein-2 in patients with suspected dysfunction.
Collapse
Affiliation(s)
- Harvey B Sarnat
- Departments of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Pathology (Neuropathology), University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.
| | - Laura Flores-Sarnat
- Departments of Paediatrics, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada; Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Eugen Boltshauser
- Department of Paediatric Neurology, Children's University Hospital, Zürich, Switzerland
| |
Collapse
|
31
|
Kalafateli AL, Vallöf D, Jerlhag E. Activation of amylin receptors attenuates alcohol-mediated behaviours in rodents. Addict Biol 2019; 24:388-402. [PMID: 29405517 PMCID: PMC6585842 DOI: 10.1111/adb.12603] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/14/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023]
Abstract
Alcohol expresses its reinforcing properties by activating areas of the mesolimbic dopamine system, which consists of dopaminergic neurons projecting from the ventral tegmental area to the nucleus accumbens. The findings that reward induced by food and addictive drugs involve common mechanisms raise the possibility that gut-brain hormones, which control appetite, such as amylin, could be involved in reward regulation. Amylin decreases food intake, and despite its implication in the regulation of natural rewards, tenuous evidence support amylinergic mediation of artificial rewards, such as alcohol. Therefore, the present experiments were designed to investigate the effect of salmon calcitonin (sCT), an amylin receptor agonist and analogue of endogenous amylin, on various alcohol-related behaviours in rodents. We showed that acute sCT administration attenuated the established effects of alcohol on the mesolimbic dopamine system, particularly alcohol-induced locomotor stimulation and accumbal dopamine release. Using the conditioned place preference model, we demonstrated that repeated sCT administration prevented the expression of alcohol's rewarding properties and that acute sCT administration blocked the reward-dependent memory consolidation. In addition, sCT pre-treatment attenuated alcohol intake in low alcohol-consuming rats, with a more evident decrease in high alcohol consumers in the intermittent alcohol access model. Lastly, sCT did not alter peanut butter intake, blood alcohol concentration and plasma corticosterone levels in mice. Taken together, the present data support that amylin signalling is involved in the expression of alcohol reinforcement and that amylin receptor agonists could be considered for the treatment of alcohol use disorder in humans.
Collapse
Affiliation(s)
- Aimilia Lydia Kalafateli
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Daniel Vallöf
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| |
Collapse
|
32
|
Lutz TA, Le Foll C. Endogenous amylin contributes to birth of microglial cells in arcuate nucleus of hypothalamus and area postrema during fetal development. Am J Physiol Regul Integr Comp Physiol 2019; 316:R791-R801. [PMID: 30943041 DOI: 10.1152/ajpregu.00004.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Amylin acts in the area postrema (AP) and arcuate nucleus (ARC) to control food intake. Amylin also increases axonal fiber outgrowth from the AP→nucleus tractus solitarius and from ARC→hypothalamic paraventricular nucleus. More recently, exogenous amylin infusion for 4 wk was shown to increase neurogenesis in adult rats in the AP. Furthermore, amylin has been shown to enhance leptin signaling in the ARC and ventromedial nucleus of the hypothalamus (VMN). Thus, we hypothesized that endogenous amylin could be a critical factor in regulating cell birth in the ARC and AP and that amylin could also be involved in the birth of leptin-sensitive neurons. Amylin+/- dams were injected with BrdU at embryonic day 12 and at postnatal day 2; BrdU+ cells were quantified in wild-type (WT) and amylin knockout (KO) mice. The number of BrdU+HuC/D+ neurons was similar in ARC and AP, but the number of BrdU+Iba1+ microglia was significantly decreased in both nuclei. Five-week-old WT and KO littermates were injected with leptin to test whether amylin is involved in the birth of leptin-sensitive neurons. Although there was no difference in the number of BrdU+c-Fos+ neurons in the ARC and dorsomedial nucleus, an increase in BrdU+c-Fos+ neurons was seen in VMN and lateral hypothalamus (LH) in amylin KO mice. In conclusion, these data suggest that during fetal development, endogenous amylin favors the birth of microglial cells in the ARC and AP and that it decreases the birth of leptin-sensitive neurons in the VMN and LH.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich , Zurich , Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich , Zurich , Switzerland
| |
Collapse
|
33
|
Jerlhag E. Gut-brain axis and addictive disorders: A review with focus on alcohol and drugs of abuse. Pharmacol Ther 2018; 196:1-14. [PMID: 30439457 DOI: 10.1016/j.pharmthera.2018.11.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to the limited efficacy of existing medications for addictive disorders including alcohol use disorder (AUD), the need for additional medications is substantial. Potential new medications for addiction can be identified through investigation of the neurochemical substrates mediating the ability of drugs of abuse such as alcohol to activate the mesolimbic dopamine system. Interestingly, recent studies implicate neuropeptides of the gut-brain axis as modulators of reward and addiction processes. The present review therefore summarizes the current studies investigating the ability of the gut-brain peptides ghrelin, glucagon-like peptide-1 (GLP-1), amylin and neuromedin U (NMU) to modulate alcohol- and drug-related behaviors in rodents and humans. Extensive literature demonstrates that ghrelin, the only known orexigenic neuropeptide to date, enhances reward as well as the intake of alcohol, and other drugs of abuse, while ghrelin receptor antagonism has the opposite effects. On the other hand, the anorexigenic peptides GLP-1, amylin and NMU independently inhibits reward from alcohol and drugs of abuse in rodents. Collectively, these rodent and human studies imply that central ghrelin, GLP-1, amylin and NMU signaling may contribute to addiction processes. Therefore, the need for randomized clinical trials investigating the effects of agents targeting these aforementioned systems on drug/alcohol use is substantial.
Collapse
Affiliation(s)
- Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
34
|
Duffy S, Lutz TA, Boyle CN. Rodent models of leptin receptor deficiency are less sensitive to amylin. Am J Physiol Regul Integr Comp Physiol 2018; 315:R856-R865. [DOI: 10.1152/ajpregu.00179.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pancreatic hormone amylin is released from beta cells following nutrient ingestion and contributes to the control of body weight and glucose homeostasis. Amylin reduces food intake by activating neurons in the area postrema (AP). Amylin was also shown to synergize with the adipokine leptin, with combination therapy producing greater weight loss and food intake reduction than either hormone alone. Although amylin and leptin were initially thought to interact downstream of the AP in the hypothalamus, recent findings show that the two hormones can act on the same AP neurons, suggesting a more direct relationship. The objective of this study was to determine whether amylin action depends on functional leptin signaling. We tested the ability of amylin to induce satiation and to activate its primary target neurons in the AP in two rodent models of LepR deficiency, the db/db mouse and the Zucker diabetic fatty (ZDF) rat. When compared with wild-type (WT) mice, db/db mice exhibited reduced amylin-induced satiation, reduced amylin-induced Fos in the AP, and a lower expression of calcitonin receptor (CTR) protein, the core component of all amylin receptors. ZDF rats also showed no reduction in food intake following amylin treatment; however, unlike the db/db mice, levels of amylin-induced Fos and CTR in the AP were no different than WT rats. Our results suggest that LepR expression is required for the full anorexic effect of amylin; however, the neuronal activation in the AP seems to depend on the type of LepR mutation.
Collapse
Affiliation(s)
- Sonya Duffy
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
| | - Thomas A. Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
- Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Christina N. Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Lutz TA, Coester B, Whiting L, Dunn-Meynell AA, Boyle CN, Bouret SG, Levin BE, Le Foll C. Amylin Selectively Signals Onto POMC Neurons in the Arcuate Nucleus of the Hypothalamus. Diabetes 2018; 67:805-817. [PMID: 29467172 PMCID: PMC5910000 DOI: 10.2337/db17-1347] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/09/2018] [Indexed: 02/05/2023]
Abstract
Amylin phosphorylates ERK (p-ERK) in the area postrema to reduce eating and synergizes with leptin to phosphorylate STAT3 in the arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei to reduce food intake and body weight. The current studies assessed potential amylin and amylin-leptin ARC/VMN interactions on ERK signaling and their roles in postnatal hypothalamic pathway development. In amylin knockout mice, the density of agouti-related protein (AgRP)-immunoreactive (IR) fibers in the hypothalamic paraventricular nucleus (PVN) was increased, while the density of α-melanocyte-stimulating hormone (αMSH) fibers was decreased. In mice deficient of the amylin receptor components RAMP1/3, both AgRP and αMSH-IR fiber densities were decreased, while only αMSH-IR fiber density was decreased in rats injected neonatally in the ARC/VMN with an adeno-associated virus short hairpin RNA against the amylin core receptor. Amylin induced p-ERK in ARC neurons, 60% of which was present in POMC-expressing neurons, with none in NPY neurons. An amylin-leptin interaction was shown by an additive effect on ARC ERK signaling in neonatal rats and a 44% decrease in amylin-induced p-ERK in the ARC of leptin receptor-deficient and of ob/ob mice. Together, these results suggest that amylin directly acts, through a p-ERK-mediated process, on POMC neurons to enhance ARC-PVN αMSH pathway development.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Bernd Coester
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Lynda Whiting
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | | | - Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Sebastien G Bouret
- Developmental Neuroscience Program, The Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, CA
- INSERM U1172, Jean-Pierre Aubert Research Center, Lille, France
| | - Barry E Levin
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Boyle CN, Lutz TA, Le Foll C. Amylin - Its role in the homeostatic and hedonic control of eating and recent developments of amylin analogs to treat obesity. Mol Metab 2017; 8:203-210. [PMID: 29203236 PMCID: PMC5985014 DOI: 10.1016/j.molmet.2017.11.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Amylin is a pancreatic β-cell hormone that produces effects in several different organ systems. One of its best-characterized effects is the reduction in eating and body weight seen in preclinical and clinical studies. Amylin activates specific receptors, a portion of which it shares with calcitonin gene-related peptide (CGRP). Amylin's role in the control of energy metabolism relates to its satiating effect, but recent data indicate that amylin may also affect hedonic aspects in the control of eating, including a reduction of the rewarding value of food. Recently, several amylin-based peptides have been characterized. Pramlintide (Symlin®) is currently the only one being used clinically to treat type 1 and type 2 diabetes. However other amylin analogs with improved pharmacokinetic properties are being considered as anti-obesity treatment strategies. Several other studies in obesity have shown that amylin agonists could also be useful for weight loss, especially in combination with other agents. SCOPE OF REVIEW This review will briefly summarize amylin physiology and pharmacology and then focus on amylin's role in food reward and the effects of amylin analogs in pre-clinical testing for anti-obesity drugs. CONCLUSION We propose here that the effects of amylin may be homeostatic and hedonic in nature.
Collapse
Affiliation(s)
- Christina Neuner Boyle
- Institute of Veterinary Physiology and Zurich Centre for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas Alexander Lutz
- Institute of Veterinary Physiology and Zurich Centre for Integrative Human Physiology, University of Zurich, Switzerland.
| | - Christelle Le Foll
- Institute of Veterinary Physiology and Zurich Centre for Integrative Human Physiology, University of Zurich, Switzerland
| |
Collapse
|
37
|
Amylin Receptor: A Potential Therapeutic Target for Alzheimer's Disease. Trends Mol Med 2017; 23:709-720. [PMID: 28694141 DOI: 10.1016/j.molmed.2017.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/08/2017] [Accepted: 06/14/2017] [Indexed: 01/29/2023]
Abstract
Alzheimer'sdisease (AD) is a progressive neurodegenerative disorder, characterized by senile plaques constituting extracellular deposits of β-amyloid (Aβ) fibrils. Since Aβ accumulation in the brain is considered an early event preceding, by decades, cognitive dysfunction, disease-modifying treatments are aimed at facilitating clearance of this protein from the brain or ameliorating its toxic effects. Recent studies have identified the amylin receptor as a capable mediator of the deleterious actions of Aβ and furthermore, administration of amylin receptor-based peptides has been shown to improve spatial memory and learning in transgenic mouse models of AD. Here, by discussing available evidence, we posit that the amylin receptor could be considered a potential therapeutic target for AD, and present the rationale for using amylin receptor antagonists to treat this debilitating condition.
Collapse
|
38
|
Borner T, Arnold M, Ruud J, Breit SN, Langhans W, Lutz TA, Blomqvist A, Riediger T. Anorexia-cachexia syndrome in hepatoma tumour-bearing rats requires the area postrema but not vagal afferents and is paralleled by increased MIC-1/GDF15. J Cachexia Sarcopenia Muscle 2017; 8:417-427. [PMID: 28025863 PMCID: PMC5476861 DOI: 10.1002/jcsm.12169] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/26/2016] [Accepted: 10/28/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The cancer-anorexia-cachexia syndrome (CACS) negatively affects survival and therapy success in cancer patients. Inflammatory mediators and tumour-derived factors are thought to play an important role in the aetiology of CACS. However, the central and peripheral mechanisms contributing to CACS are insufficiently understood. The area postrema (AP) and the nucleus tractus solitarii are two important brainstem centres for the control of eating during acute sickness conditions. Recently, the tumour-derived macrophage inhibitory cytokine-1 (MIC-1) emerged as a possible mediator of cancer anorexia because lesions of these brainstem areas attenuated the anorectic effect of exogenous MIC-1 in mice. METHODS Using a rat hepatoma tumour model, we examined the roles of the AP and of vagal afferents in the mediation of CACS. Specifically, we investigated whether a lesion of the AP (APX) or subdiaphragmatic vagal deafferentation (SDA) attenuate anorexia, body weight, muscle, and fat loss. Moreover, we analysed MIC-1 levels in this tumour model and their correlation with tumour size and the severity of the anorectic response. RESULTS In tumour-bearing sham-operated animals mean daily food intake significantly decreased. The anorectic response was paralleled by a significant loss of body weight and muscle mass. APX rats were protected against anorexia, body weight loss, and muscle atrophy after tumour induction. In contrast, subdiaphragmatic vagal deafferentation did not attenuate cancer-induced anorexia or body weight loss. Tumour-bearing rats had substantially increased MIC-1 levels, which positively correlated with tumour size and cancer progression and negatively correlated with food intake. CONCLUSIONS These findings demonstrate the importance of the AP in the mediation of cancer-dependent anorexia and body weight loss and support a pathological role of MIC-1 as a tumour-derived factor mediating CACS, possibly via an AP-dependent action.
Collapse
Affiliation(s)
- Tito Borner
- Vetsuisse Faculty, Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Johan Ruud
- Department of Clinical and Experimental Medicine, University of Linköping, Linköping, Sweden
| | - Samuel N Breit
- St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, University of New South Wales, Sydney, Australia
| | - Wolfgang Langhans
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Thomas A Lutz
- Vetsuisse Faculty, Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Anders Blomqvist
- Department of Clinical and Experimental Medicine, University of Linköping, Linköping, Sweden
| | - Thomas Riediger
- Vetsuisse Faculty, Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Role of the area postrema in the hypophagic effects of oleoylethanolamide. Pharmacol Res 2017; 122:20-34. [PMID: 28535974 DOI: 10.1016/j.phrs.2017.05.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/10/2017] [Accepted: 05/16/2017] [Indexed: 12/25/2022]
Abstract
The satiety-promoting action of oleoylethanolamide (OEA) has been associated to the indirect activation of selected brain areas, such as the nucleus of the solitary tract (NST) in the brainstem and the tuberomammillary (TMN) and paraventricular (PVN) nuclei in the hypothalamus, where noradrenergic, histaminergic and oxytocinergic neurons play a necessary role. Visceral ascending fibers were hypothesized to mediate such effects. However, our previous findings demonstrated that the hypophagic action of peripherally administered OEA does not require intact vagal afferents and is associated to a strong activation of the area postrema (AP). Therefore, we hypothesized that OEA may exert its central effects through the direct activation of this circumventricular organ. To test this hypothesis, we subjected rats to the surgical ablation of the AP (APX rats) and evaluated the effects of OEA (10mgkg-1 i.p.) on food intake, Fos expression, hypothalamic oxytocin (OXY) immunoreactivity and on the expression of dopamine beta hydroxylase (DBH) in the brainstem and hypothalamus. We found that the AP lesion completely prevented OEA's behavioral and neurochemical effects in the brainstem and the hypothalamus. Moreover OEA increased DBH expression in AP and NST neurons of SHAM rats while the effect in the NST was absent in APX rats, thus suggesting the possible involvement of noradrenergic AP neurons. These results support the hypothesis of a necessary role of the AP in mediating OEA's central effects that sustain its pro-satiety action.
Collapse
|
40
|
Whiting L, McCutcheon JE, Boyle CN, Roitman MF, Lutz TA. The area postrema (AP) and the parabrachial nucleus (PBN) are important sites for salmon calcitonin (sCT) to decrease evoked phasic dopamine release in the nucleus accumbens (NAc). Physiol Behav 2017; 176:9-16. [PMID: 28342771 DOI: 10.1016/j.physbeh.2017.03.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 12/26/2022]
Abstract
The pancreatic hormone amylin and its agonist salmon calcitonin (sCT) act via the area postrema (AP) and the lateral parabrachial nucleus (PBN) to reduce food intake. Investigations of amylin and sCT signaling in the ventral tegmental area (VTA) and nucleus accumbens (NAc) suggest that the eating inhibitory effect of amylin is, in part, mediated through the mesolimbic 'reward' pathway. Indeed, administration of the sCT directly to the VTA decreased phasic dopamine release (DA) in the NAc. However, it is not known if peripheral amylin modulates the mesolimbic system directly or whether this occurs via the AP and PBN. To determine whether and how peripheral amylin or sCT affect mesolimbic reward circuitry we utilized fast scan cyclic voltammetry under anesthesia to measure phasic DA release in the NAc evoked by electrical stimulation of the VTA in intact, AP lesioned and bilaterally PBN lesioned rats. Amylin (50μg/kg i.p.) did not change phasic DA responses compared to saline control rats. However, sCT (50μg/kg i.p.) decreased evoked DA release to VTA-stimulation over 1h compared to saline treated control rats. Further investigations determined that AP and bilateral PBN lesions abolished the ability of sCT to suppress evoked phasic DA responses to VTA-stimulation. These findings implicate the AP and the PBN as important sites for peripheral sCT to decrease evoked DA release in the NAc and suggest that these nuclei may influence hedonic and motivational processes to modulate food intake.
Collapse
Affiliation(s)
- Lynda Whiting
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - James E McCutcheon
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, England
| | - Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Mitchell F Roitman
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Centre of Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
41
|
Lutz TA. Gut hormones such as amylin and GLP-1 in the control of eating and energy expenditure. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2016; 6:S15-S21. [PMID: 28685025 DOI: 10.1038/ijosup.2016.4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The control of meal size is the best studied aspect of the control of energy balance, and manipulation of this system constitutes a promising target to treat obesity. A major part of this control system is based on gastrointestinal hormones such as glucagon-like peptide-1 (GLP-1) or amylin, which are released in response to a meal and which limit the size of an ongoing meal. Both amylin and GLP-1 have also been shown to increase energy expenditure in experimental rodents, but mechanistically we know much less how this effect may be mediated, which brain sites may be involved, and what the physiological relevance of these findings may be. Most studies indicate that the effect of peripheral amylin is centrally mediated via the area postrema, but other brain areas, such as the ventral tegmental area, may also be involved. GLP-1's effect on eating seems to be mainly mediated by vagal afferents projecting to the caudal hindbrain. Chronic exposure to amylin, GLP-1 or their analogs decrease food intake and body weight gain. Next to the induction of satiation, amylin may also constitute an adiposity signal and in fact interact with the adiposity signal leptin. Amylin analogs are under clinical consideration for their effect to reduce food intake and body weight in humans, and similar to rodents, amylin analogs seem to be particularly active when combined with leptin analogs.
Collapse
Affiliation(s)
- T A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.,Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
42
|
Liberini CG, Borner T, Boyle CN, Lutz TA. The satiating hormone amylin enhances neurogenesis in the area postrema of adult rats. Mol Metab 2016; 5:834-843. [PMID: 27688997 PMCID: PMC5034493 DOI: 10.1016/j.molmet.2016.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Adult neurogenesis in the subgranular zone and subventricular zone is generally accepted, but its existence in other brain areas is still controversial. Circumventricular organs, such as the area postrema (AP) have recently been described as potential neurogenic niches in the adult brain. The AP is the major site of action of the satiating hormone amylin. Amylin has been shown to promote the formation of neuronal projections originating from the AP in neonatal rodents but the role of amylin in adult neurogenesis remains unknown. METHODS To test this, we first performed an RNA-sequencing of the AP of adult rats acutely injected with either amylin (20 μg/kg), amylin plus the amylin receptor antagonist AC187 (500 μg/kg) or vehicle. Second, animals were subcutaneously equipped with minipumps releasing either amylin (50 μg/kg/day) or vehicle for 3 weeks to assess cell proliferation and differentiation with the 5'-bromo-2-deoxyuridine (BrdU) technique. RESULTS Acute amylin injections affected genes involved in pathways and processes that control adult neurogenesis. Amylin consistently upregulated NeuroD1 transcript and protein in the adult AP, and this effect was blocked by the co-administration of AC187. Further, chronic amylin treatment increased the number of newly proliferated AP-cells and significantly promoted their differentiation into neurons rather than astrocytes. CONCLUSION Our findings revealed a novel role of the satiating hormone amylin in promoting neurogenesis in the AP of adult rats.
Collapse
Key Words
- AP, area postrema
- Adult neurogenesis
- Amylin
- Area postrema
- BrdU
- BrdU, 5′-bromo-2-deoxyuridine
- CR, calretinin
- CTR, calcitonin receptor
- CVO, circumventricular organs
- Circumventricular organs
- ERK1/2, extracellular signal-regulated kinase 1 and 2
- EphRs, ephrin receptors
- FDR, false discovery rate
- GO, gene ontology
- ME, median eminence
- NGS, next generation sequencing
- NSC, neural stem cells
- NeuroD, neuronal differentiation
- NeuroD1, neuronal differentiation-1
- RAMP, receptor activity-modifying protein
- Wnt, Wingless-Type MMTV Integration Site Family
- bHLH, basic helix-loop-helix
Collapse
Affiliation(s)
- Claudia G Liberini
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland; Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland; Zurich Centre for Clinical Studies, Vetsuisse Faculty University of Zurich, 8057 Zurich, Switzerland
| | - Tito Borner
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland; Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Christina N Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland.
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich (UZH), 8057 Zurich, Switzerland; Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
43
|
Amylin-mediated control of glycemia, energy balance, and cognition. Physiol Behav 2016; 162:130-40. [PMID: 26922873 DOI: 10.1016/j.physbeh.2016.02.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/20/2016] [Accepted: 02/22/2016] [Indexed: 12/26/2022]
Abstract
Amylin, a peptide hormone produced in the pancreas and in the brain, has well-established physiological roles in glycemic regulation and energy balance control. It improves postprandial blood glucose levels by suppressing gastric emptying and glucagon secretion; these beneficial effects have led to the FDA-approved use of the amylin analog pramlintide in the treatment of diabetes mellitus. Amylin also acts centrally as a satiation signal, reducing food intake and body weight. The ability of amylin to promote negative energy balance, along with its unique capacity to cooperatively facilitate or enhance the intake- and body weight-suppressive effects of other neuroendocrine signals like leptin, have made amylin a leading target for the development of novel pharmacotherapies for the treatment of obesity. In addition to these more widely studied effects, a growing body of literature suggests that amylin may play a role in processes related to cognition, including the neurodegeneration and cognitive deficits associated with Alzheimer's disease (AD). Although the function of amylin in AD is still unclear, intriguing recent reports indicate that amylin may improve cognitive ability and reduce hallmarks of neurodegeneration in the brain. The frequent comorbidity of diabetes mellitus and obesity, as well as the increased risk for and occurrence of AD associated with these metabolic diseases, suggests that amylin-based pharmaceutical strategies may provide multiple therapeutic benefits. This review will discuss the known effects of amylin on glycemic regulation, energy balance control, and cognitive/motivational processes. Particular focus will be devoted to the current and/or potential future clinical use of amylin pharmacotherapies for the treatment of diseases in each of these realms.
Collapse
|
44
|
Lutz TA, Meyer U. Amylin at the interface between metabolic and neurodegenerative disorders. Front Neurosci 2015; 9:216. [PMID: 26136651 PMCID: PMC4468610 DOI: 10.3389/fnins.2015.00216] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 05/29/2015] [Indexed: 12/14/2022] Open
Abstract
The pancreatic peptide amylin is best known for its role as a satiation hormone in the control of food intake and as the major component of islet amyloid deposits in the pancreatic islets of patients with type 2 diabetes mellitus (T2DM). Epidemiological studies have established a clear association between metabolic and neurodegenerative disorders in general, and between T2DM and Alzheimer's disease (AD) in particular. Here, we discuss that amylin may be an important player acting at the interface between these metabolic and neurodegenerative disorders. Abnormal amylin production is a hallmark peripheral pathology both in the early (pre-diabetic) and late phases of T2DM, where hyperamylinemic (early phase) and hypoamylinemic (late phase) conditions coincide with hyper- and hypo-insulinemia, respectively. Moreover, there are notable biochemical similarities between amylin and β-amyloids (Aβ), which are both prone to amyloid plaque formation and to cytotoxic effects. Amylin's propensity to form amyloid plaques is not restricted to pancreatic islet cells, but readily extends to the CNS, where it has been found to co-localize with Aβ plaques in at least a subset of AD patients. Hence, amylin may constitute a “second amyloid” in neurodegenerative disorders such as AD. We further argue that hyperamylinemic conditions may be more relevant for the early processes of amyloid formation in the CNS, whereas hypoamylinemic conditions may be more strongly associated with late stages of central amyloid pathologies. Advancing our understanding of these temporal relationships may help to establish amylin-based interventions in the treatment of AD and other neurodegenerative disorders with metabolic comorbidities.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich Zurich, Switzerland ; Zurich Center of Integrative Human Physiology, University of Zurich Zurich, Switzerland
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich Zurich, Switzerland
| |
Collapse
|
45
|
Le Foll C, Johnson MD, Dunn-Meynell AA, Boyle CN, Lutz TA, Levin BE. Amylin-induced central IL-6 production enhances ventromedial hypothalamic leptin signaling. Diabetes 2015; 64:1621-31. [PMID: 25409701 PMCID: PMC4407855 DOI: 10.2337/db14-0645] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 11/14/2014] [Indexed: 02/05/2023]
Abstract
Amylin acts acutely via the area postrema to reduce food intake and body weight, but it also interacts with leptin over longer periods of time, possibly via the ventromedial hypothalamus (VMH), to increase leptin signaling and phosphorylation of STAT3. We postulated that amylin enhances VMH leptin signaling by inducing interleukin (IL)-6, which then interacts with its gp130 receptor to activate STAT3 signaling and gene transcription downstream of the leptin receptor. We found that components of the amylin receptor (RAMPs1-3, CTR1a,b) are expressed in cultured VMH astrocytes, neurons, and microglia, as well as in micropunches of arcuate and ventromedial hypothalamic nuclei (VMN). Amylin exposure for 5 days increased IL-6 mRNA expression in VMH explants and microglia by two- to threefold, respectively, as well as protein abundance in culture supernatants by five- and twofold, respectively. Amylin had no similar effects on cultured astrocytes or neurons. In rats, 5 days of amylin treatment decreased body weight gain and/or food intake and increased IL-6 mRNA expression in the VMN. Similar 5-day amylin treatment increased VMN leptin-induced phosphorylation of STAT3 expression in wild-type mice and rats infused with lateral ventricular IgG but not in IL-6 knockout mice or rats infused with ventricular IL-6 antibody. Lateral ventricular infusion of IL-6 antibody also prevented the amylin-induced decrease of body weight gain. These results show that amylin-induced VMH microglial IL-6 production is the likely mechanism by which amylin treatment interacts with VMH leptin signaling to increase its effect on weight loss.
Collapse
Affiliation(s)
- Christelle Le Foll
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ
| | - Miranda D Johnson
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ Rutgers Graduate School of Biomedical Sciences at New Jersey Medical School and Rutgers School of Dental Medicine, Newark, NJ
| | | | - Christina N Boyle
- Zurich Center for Integrative Human Physiology, Zurich, Switzerland Institute of Veterinary Physiology, Zurich, Switzerland
| | - Thomas A Lutz
- Zurich Center for Integrative Human Physiology, Zurich, Switzerland Institute of Veterinary Physiology, Zurich, Switzerland Institute of Laboratory Animal Sciences, Zurich, Switzerland
| | - Barry E Levin
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ VA Medical Center, East Orange, NJ
| |
Collapse
|