1
|
McKnight AD, Alhadeff AL. Nutrient detection pathways for food reinforcement and satiation. Curr Opin Neurobiol 2025; 92:103040. [PMID: 40349609 DOI: 10.1016/j.conb.2025.103040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 05/14/2025]
Abstract
Ingested food is broken down into macronutrient components that are detected by gut-brain signaling pathways that play a vital role in feeding behavior. These specialized mechanisms both promote food intake (via appetition and food reinforcement pathways) and terminate food intake (via satiation pathways). Despite recent significant advances in our understanding of gut-brain signaling and behavior, questions remain about the distinct mechanisms mediating food reinforcement and satiation. Here, we review the receptors/transporters and gut-brain pathways that contribute to nutrient sensing and feeding behavior, and highlight key knowledge gaps that will guide future research on the complex gut-brain systems that influence food intake.
Collapse
Affiliation(s)
- Aaron D McKnight
- Monell Chemical Senses Center, Philadelphia, 19104, PA, United States; Department of Neuroscience, University of Pennsylvania, Philadelphia, 19104, PA, United States
| | - Amber L Alhadeff
- Monell Chemical Senses Center, Philadelphia, 19104, PA, United States; Department of Neuroscience, University of Pennsylvania, Philadelphia, 19104, PA, United States.
| |
Collapse
|
2
|
Østergaard S, Jessen C, Paulsson JF, Kasimova MA, Conde-Frieboes KW, Straarup EM, Skyggebjerg RB, Ynddal L, Sanfridson A, Wulff BS, Chambers AP. Variant screening of PYY 3-36 leads to potent long-acting PYY analogs with superior Y 2 receptor selectivity. Sci Transl Med 2025; 17:eadq6392. [PMID: 40138456 DOI: 10.1126/scitranslmed.adq6392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/27/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Peptide YY (PYY3-36) has attracted attention in diabetes and obesity research because of its involvement in food intake regulation and glucose homeostasis. Native PYY3-36 maintains high potency on the Y2 receptor with a loss of potency on the Y1, Y4, and Y5 receptors. However, PYY3-36 has a relatively short half-life, and the selectivity displayed by the native peptide may not be optimal if a long-acting analog is to be developed. We performed variant screening of PYY3-36 to identify key canonical amino acids that are pivotal to Y2 receptor selectivity, potency, and peptide stability. In combination with fatty diacid derivatization, this afforded highly selective long-acting analogs against the Y2 receptor, which improved glucose metabolism in diabetic db/db mice. When combined with a long-acting glucagon-like peptide 1 (GLP-1) receptor agonist, these analogs showed superior blood glucose lowering in diabetic ZSF1 rats and greater body weight loss in a high-fat diet-induced mouse model of obesity compared with treatment with the GLP-1 analog alone. One of the tested analogs, PYY1875, has progressed into clinical trials for obesity. Together, our results demonstrate the power of variant screening combined with fatty diacid derivatization in the development of a long-acting, highly efficacious PYY clinical candidate.
Collapse
Affiliation(s)
- Søren Østergaard
- Global Research Technologies, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Carsten Jessen
- Global Research Technologies, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Johan F Paulsson
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Marina A Kasimova
- Digital Science and Innovation, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | | | - Ellen Marie Straarup
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | | | - Lars Ynddal
- Global Research Technologies, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Annika Sanfridson
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Birgitte S Wulff
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| | - Adam P Chambers
- Global Drug Discovery, Novo Nordisk A/S, Novo Research Park, 2760 Måløv, Denmark
| |
Collapse
|
3
|
James-Okoro PP, Lewis JE, Gribble FM, Reimann F. The role of GIPR in food intake control. Front Endocrinol (Lausanne) 2025; 16:1532076. [PMID: 40166681 PMCID: PMC11955450 DOI: 10.3389/fendo.2025.1532076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is one of two incretin hormones playing key roles in the control of food intake, nutrient assimilation, insulin secretion and whole-body metabolism. Recent pharmacological advances and clinical trials show that unimolecular co-agonists that target the receptors for the incretins - GIP and glucagon-like peptide 1 (GLP-1) - offer more effective treatment strategies for obesity and type 2 diabetes mellitus (T2D) compared with GLP-1 receptor (GLP1R) agonists alone, suggesting previously underappreciated roles of GIP in regulating food intake and body weight. The mechanisms by which GIP regulates energy balance remain controversial as both agonism and antagonism of the GIP receptor (GIPR) produce weight loss and improve metabolic outcomes in preclinical models. Recent studies have shown that GIPR signalling in the central nervous system (CNS), especially in regions of the brain that regulate energy balance, is essential for its action on appetite regulation. This finding has sparked interest in understanding the mechanisms by which GIP engages brain circuits to reduce food intake and body weight. In this review, we present key knowledge around the actions of GIP on food intake regulation and the potential mechanisms by which GIPR and GIPR/GLP1R agonists may regulate energy balance.
Collapse
Affiliation(s)
| | | | - Fiona Mary Gribble
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Scott KA, Tan Y, Johnson DN, Elsaafien K, Baumer-Harrison C, Méndez-Hernández R, Kirchner MK, Eikenberry SA, Sa JM, Stern JE, de Lartigue G, de Kloet AD, Krause EG. Mechanosensation of the heart and gut elicits hypometabolism and vigilance in mice. Nat Metab 2025; 7:263-275. [PMID: 39824919 DOI: 10.1038/s42255-024-01205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/09/2024] [Indexed: 01/20/2025]
Abstract
Interoception broadly refers to awareness of one's internal milieu. Although the importance of the body-to-brain communication that underlies interoception is implicit, the vagal afferent signalling and corresponding brain circuits that shape perception of the viscera are not entirely clear. Here, we use mice to parse neural circuits subserving interoception of the heart and gut. We determine that vagal sensory neurons expressing the oxytocin receptor (Oxtr), referred to as NGOxtr, send projections to cardiovascular or gastrointestinal tissues and exhibit molecular and structural features indicative of mechanosensation. Chemogenetic excitation of NGOxtr decreases food and water consumption, and remarkably, produces a torpor-like phenotype characterized by reductions in cardiac output, body temperature and energy expenditure. Chemogenetic excitation of NGOxtr also creates patterns of brain activity associated with augmented hypothalamic-pituitary-adrenal axis activity and behavioural indices of vigilance. Recurrent excitation of NGOxtr suppresses food intake and lowers body mass, indicating that mechanosensation of the heart and gut can exert enduring effects on energy balance. These findings suggest that the sensation of vascular stretch and gastrointestinal distention may have profound effects on whole-body metabolism and, possibly, mental health.
Collapse
Affiliation(s)
- Karen A Scott
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Yalun Tan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Dominique N Johnson
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Khalid Elsaafien
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Caitlin Baumer-Harrison
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | - Matthew K Kirchner
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Sophia A Eikenberry
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jessica M Sa
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, PA, USA.
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Annette D de Kloet
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA.
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA.
| | - Eric G Krause
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA.
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
5
|
Dahir NS, Gui Y, Wu Y, Sweeney PR, Rouault AA, Williams SY, Gimenez LE, Sawyer TK, Joy ST, Mapp AK, Cone RD. Subthreshold activation of the melanocortin system causes generalized sensitization to anorectic agents in mice. J Clin Invest 2024; 134:e178250. [PMID: 39007271 PMCID: PMC11245150 DOI: 10.1172/jci178250] [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: 12/26/2023] [Accepted: 05/15/2024] [Indexed: 07/16/2024] Open
Abstract
The melanocortin-3 receptor (MC3R) regulates GABA release from agouti-related protein (AgRP) nerve terminals and thus tonically suppresses multiple circuits involved in feeding behavior and energy homeostasis. Here, we examined the role of the MC3R and the melanocortin system in regulating the response to various anorexigenic agents. The genetic deletion or pharmacological inhibition of the MC3R, or subthreshold doses of an MC4R agonist, improved the dose responsiveness to glucagon-like peptide 1 (GLP1) agonists, as assayed by inhibition of food intake and weight loss. An enhanced anorectic response to the acute satiety factors peptide YY (PYY3-36) and cholecystokinin (CCK) and the long-term adipostatic factor leptin demonstrated that increased sensitivity to anorectic agents was a generalized result of MC3R antagonism. We observed enhanced neuronal activation in multiple hypothalamic nuclei using Fos IHC following low-dose liraglutide in MC3R-KO mice (Mc3r-/-), supporting the hypothesis that the MC3R is a negative regulator of circuits that control multiple aspects of feeding behavior. The enhanced anorectic response in Mc3r-/- mice after administration of GLP1 analogs was also independent of the incretin effects and malaise induced by GLP1 receptor (GLP1R) analogs, suggesting that MC3R antagonists or MC4R agonists may have value in enhancing the dose-response range of obesity therapeutics.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Appetite Depressants/pharmacology
- Cholecystokinin/metabolism
- Eating/drug effects
- Glucagon-Like Peptide 1/metabolism
- Hypothalamus/metabolism
- Leptin/metabolism
- Liraglutide/pharmacology
- Mice, Inbred C57BL
- Mice, Knockout
- Peptide YY/metabolism
- Peptide YY/genetics
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 3/agonists
- Receptor, Melanocortin, Type 4/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/agonists
Collapse
Affiliation(s)
- Naima S. Dahir
- Life Sciences Institute
- Department of Molecular and Integrative Physiology, and
| | - Yijun Gui
- Life Sciences Institute
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yanan Wu
- Life Sciences Institute
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick R. Sweeney
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Illinois, USA
| | - Alix A.J. Rouault
- Life Sciences Institute
- Department of Molecular and Integrative Physiology, and
| | | | | | | | | | - Anna K. Mapp
- Life Sciences Institute
- Department of Chemistry, School of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan, USA
| | - Roger D. Cone
- Life Sciences Institute
- Department of Molecular and Integrative Physiology, and
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Alonso AM, Cork SC, Phuah P, Hansen B, Norton M, Cheng S, Xu X, Suba K, Ma Y, Dowsett GK, Tadross JA, Lam BY, Yeo GS, Herzog H, Bloom SR, Arnold M, Distaso W, Murphy KG, Salem V. The vagus nerve mediates the physiological but not pharmacological effects of PYY 3-36 on food intake. Mol Metab 2024; 81:101895. [PMID: 38340808 PMCID: PMC10877939 DOI: 10.1016/j.molmet.2024.101895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Peptide YY (PYY3-36) is a post-prandially released gut hormone with potent appetite-reducing activity, the mechanism of action of which is not fully understood. Unravelling how this system physiologically regulates food intake may help unlock its therapeutic potential, whilst minimising unwanted effects. Here we demonstrate that germline and post-natal targeted knockdown of the PYY3-36 preferring receptor (neuropeptide Y (NPY) Y2 receptor (Y2R)) in the afferent vagus nerve is required for the appetite inhibitory effects of physiologically-released PYY3-36, but not peripherally administered pharmacological doses. Post-natal knockdown of the Y2R results in a transient body weight phenotype that is not evident in the germline model. Loss of vagal Y2R signalling also results in altered meal patterning associated with accelerated gastric emptying. These results are important for the design of PYY-based anti-obesity agents.
Collapse
Affiliation(s)
- Aldara Martin Alonso
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Simon C Cork
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom; School of Medicine, Faculty of Health, Education, Medicine & Social Care, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Phyllis Phuah
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Benjamin Hansen
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mariana Norton
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Sijing Cheng
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Xiang Xu
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Kinga Suba
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Yue Ma
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Georgina Kc Dowsett
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - John A Tadross
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - Brian Yh Lam
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - Giles Sh Yeo
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, United Kingdom
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Stephen R Bloom
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Myrtha Arnold
- Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Walter Distaso
- Imperial College Business School, Imperial College London, United Kingdom
| | - Kevin G Murphy
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Victoria Salem
- Section of Investigative Medicine and Endocrinology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom; Department of Bioengineering, Imperial College London, London, United Kingdom.
| |
Collapse
|
7
|
Yu M, Yu B, Chen D. The effects of gut microbiota on appetite regulation and the underlying mechanisms. Gut Microbes 2024; 16:2414796. [PMID: 39501848 PMCID: PMC11542600 DOI: 10.1080/19490976.2024.2414796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 11/09/2024] Open
Abstract
Appetite, a crucial aspect regulated by both the central nervous system and peripheral hormones, is influenced by the composition and dynamics of the intestinal microbiota, as evidenced by recent research. This review highlights the role of intestinal microbiota in appetite regulation, elucidating the involvement of various pathways. Notably, the metabolites generated by intestinal microorganisms, including short-chain fatty acids, bile acids, and amino acid derivatives, play a pivotal role in this intricate process. Furthermore, intestinal microorganisms contribute to appetite regulation by modulating nutritional perception, neural signal transmission, and hormone secretion within the digestive system. Consequently, manipulating and modulating the intestinal microbiota represent innovative strategies for ameliorating appetite-related disorders. This paper provides a comprehensive review of the effects of gut microbes and their metabolites on the central nervous system and host appetite. By exploring their potential regulatory pathways and mechanisms, this study aims to enhance our understanding of how gut microbes influence appetite regulation in the host.
Collapse
Affiliation(s)
- Miao Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
- DadHank(Chengdu)Biotech Corp, Chengdu, Sichuan Province, China
| | - Bing Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
| |
Collapse
|
8
|
Dahir NS, Gui Y, Wu Y, Sweeney PR, Williams SY, Gimenez LE, Sawyer TK, Joy ST, Mapp AK, Cone RD. Inhibition of the melanocortin-3 receptor (MC3R) causes generalized sensitization to anorectic agents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570114. [PMID: 38106197 PMCID: PMC10723368 DOI: 10.1101/2023.12.05.570114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The melanocortin-3 receptor (MC3R) acts presynaptically to regulate GABA release from agouti-related protein (AgRP) nerve terminals and thus may be a negative regulator of multiple circuits involved in feeding behavior and energy homeostasis. Here, we examined the role of MC3R in regulating the response to various anorexigenic agents. Our findings reveal that genetic deletion or pharmacological inhibition of MC3R improves the dose responsiveness to Glucagon-like peptide 1 (GLP1) agonists, as assayed by inhibition of food intake and weight loss. An enhanced anorectic response to other agents, including the acute satiety factors peptide YY (PYY3-36) and cholecystokinin (CCK) and the long-term adipostatic factor, leptin, demonstrated that increased sensitivity to anorectic agents is a generalized result of MC3R antagonism. Enhanced neuronal activation in multiple nuclei, including ARH, VMH, and DMH, was observed using Fos immunohistochemistry following low-dose liraglutide in MC3R knockout mice (Mc3r-/-), supporting the hypothesis that the MC3R is a negative regulator of circuits regulating multiple aspects of feeding behavior. The enhanced anorectic response in Mc3r -/- mice after administration of GLP1 analogs was also independent of the incretin effects and malaise induced by GLP1R analogs, suggesting that MC3R antagonists may have value in enhancing the dose-response range of obesity therapeutics.
Collapse
Affiliation(s)
- Naima S. Dahir
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Yijun Gui
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Yanan Wu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Patrick R. Sweeney
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, IL
| | | | - Luis E. Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Tomi K. Sawyer
- Courage Therapeutics, 64 Homer Street, Newton, Massachusetts 02459, United States
| | - Stephen T. Joy
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Anna K. Mapp
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Department of Chemistry, School of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roger D. Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
- Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
9
|
Caffrey A, Lavecchia E, Merkel R, Zhang Y, Chichura KS, Hayes MR, Doyle RP, Schmidt HD. PYY 3-36 infused systemically or directly into the VTA attenuates fentanyl seeking in male rats. Neuropharmacology 2023; 239:109686. [PMID: 37572954 PMCID: PMC10528880 DOI: 10.1016/j.neuropharm.2023.109686] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
More effective treatments for fentanyl use disorder are urgently needed. An emerging literature indicates that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate voluntary opioid taking and seeking in rodents. However, GLP-1R agonists produce adverse malaise-like effects that may limit patient compliance. Recently, we developed a dual agonist of GLP-1Rs and neuropeptide Y2 receptors (Y2Rs) that attenuates fentanyl taking and seeking at doses that do not produce malaise-like effects in opioid-experienced rats. Whether activating Y2Rs alone is sufficient to reduce opioid taking and seeking, however, is not known. Here, we investigated the efficacy of the Y2R ligand PYY3-36 to reduce fentanyl self-administration and the reinstatement of fentanyl-seeking behavior, a model of relapse in humans. Male rats were allowed to self-administer fentanyl (2.5 μg/kg, i.v.) for 21 days on a fixed-ratio 5 (FR5) schedule of reinforcement. Rats were then pretreated with vehicle or PYY3-36 (50 μg/kg s.c.; 0.1 and 1.0 μg/100 nL intra-VTA) prior to fentanyl self-administration test sessions. There were no effects of systemic or intra-VTA PYY3-36 on intravenous fentanyl self-administration. Opioid taking was then extinguished. Prior to subsequent reinstatement test sessions, rats were pretreated with vehicle or PYY3-36 (50 μg/kg s.c.; 0.1 and 1.0 μg/100 nL intra-VTA). Both systemic and intra-VTA administration of PYY3-36 attenuated fentanyl reinstatement in male rats at doses that did not affect food intake or produce adverse malaise-like effects. These findings indicate that Y2R agonism alone is sufficient to decrease fentanyl-seeking behavior during abstinence in opioid-experienced rats and further support strategies aimed at targeting Y2Rs for treating opioid use disorders.
Collapse
Affiliation(s)
- A Caffrey
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - E Lavecchia
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Merkel
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Y Zhang
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - K S Chichura
- Department of Chemistry, Syracuse University, NY, 13244, USA
| | - M R Hayes
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R P Doyle
- Department of Chemistry, Syracuse University, NY, 13244, USA; Departments of Medicine and Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - H D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
10
|
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
|
11
|
Scott KA, Tan Y, Johnson DN, Elsaafien K, Baumer-Harrison C, Eikenberry SA, Sa JM, de Lartigue G, de Kloet AD, Krause EG. Mechanosensation of the heart and gut elicits hypometabolism and vigilance in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547073. [PMID: 37425814 PMCID: PMC10327188 DOI: 10.1101/2023.06.29.547073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Interoception broadly refers to awareness of one's internal milieu. Vagal sensory afferents monitor the internal milieu and maintain homeostasis by engaging brain circuits that alter physiology and behavior. While the importance of the body-to-brain communication that underlies interoception is implicit, the vagal afferents and corresponding brain circuits that shape perception of the viscera are largely unknown. Here, we use mice to parse neural circuits subserving interoception of the heart and gut. We determine vagal sensory afferents expressing the oxytocin receptor, hereafter referred to as NDGOxtr, send projections to the aortic arch or stomach and duodenum with molecular and structural features indicative of mechanosensation. Chemogenetic excitation of NDGOxtr significantly decreases food and water consumption, and remarkably, produces a torpor-like phenotype characterized by reductions in cardiac output, body temperature, and energy expenditure. Chemogenetic excitation of NDGOxtr also creates patterns of brain activity associated with augmented hypothalamic-pituitary-adrenal axis activity and behavioral indices of vigilance. Recurrent excitation of NDGOxtr suppresses food intake and lowers body mass, indicating that mechanosensation of the heart and gut can exert enduring effects on energy balance. These findings suggest that the sensation of vascular stretch and gastrointestinal distention may have profound effects on whole body metabolism and mental health.
Collapse
Affiliation(s)
- Karen A. Scott
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Yalun Tan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Dominique N. Johnson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Khalid Elsaafien
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Caitlin Baumer-Harrison
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Sophia A. Eikenberry
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Jessica M. Sa
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
| | | | - Annette D. de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Eric G. Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL 32611, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
12
|
Wei B, Xiao H, Xu B, Kuca K, Qin Z, Guo X, Wu W, Wu Q. Emesis to trichothecene deoxynivalenol and its congeners correspond to secretion of peptide YY and 5-HT. Food Chem Toxicol 2023:113874. [PMID: 37286030 DOI: 10.1016/j.fct.2023.113874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023]
Abstract
The type B trichothecenes pollute food crops and have been associated to alimentary toxicosis resulted in emetic reaction in human and animal. This group of mycotoxins consists deoxynivalenol (DON) and four structurally related congeners: 3-acetyl-deoxynivalenol (3-ADON), 15-acetyl deoxynivalenol (15-ADON), nivalenol (NIV) and 4-acetyl-nivalenol (fusarenon X, FX). While emesis induced by intraperitoneally dosed to DON in the mink has been related to plasma up-grading of 5-hydroxytryptamine (5-HT) and neurotransmitters peptide YY (PYY), the impact of oral dosing with DON or its four congeners on secretion of these chemical substances have not been established. The aim of this work was to contraste emetic influence to type B trichothecene mycotoxins by orally dosing and involve these influence to PYY and 5-HT. All five toxins attracted marked emetic reaction that are relevant to elevated PYY and 5-HT. The reduction in vomiting induced by the five toxins and PYY was due to blocking of the neuropeptide Y2 receptor. The inhibition of the induced vomiting response by 5-HT and all five toxins is regulated by the 5-HT3 receptor inhibitor granisetron. In a word, our results indicate that PYY and 5-HT take a key role in the emetic reaction evoked by type B trichothecenes.
Collapse
Affiliation(s)
- Ben Wei
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Huiping Xiao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Baocai Xu
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Zihui Qin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Xinyi Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic.
| | - Qinghua Wu
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic; College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
13
|
Behary P, Alessimii H, Miras AD, Tharakan G, Alexiadou K, Aldhwayan MM, Purkayastha S, Moorthy K, Ahmed AR, Bloom SR, Tan TM. Tripeptide gut hormone infusion does not alter food preferences or sweet taste function in volunteers with obesity and prediabetes/diabetes but promotes restraint eating: A secondary analysis of a randomized single-blind placebo-controlled study. Diabetes Obes Metab 2023; 25:1731-1739. [PMID: 36811311 PMCID: PMC11497251 DOI: 10.1111/dom.15028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/12/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
AIMS To investigate whether the elevation in postprandial concentrations of the gut hormones glucagon-like peptide-1 (GLP-1), oxyntomodulin (OXM) and peptide YY (PYY) accounts for the beneficial changes in food preferences, sweet taste function and eating behaviour after Roux-en-Y gastric bypass (RYGB). MATERIALS AND METHODS This was a secondary analysis of a randomized single-blind study in which we infused GLP-1, OXM, PYY (GOP) or 0.9% saline subcutaneously for 4 weeks in 24 subjects with obesity and prediabetes/diabetes, to replicate their peak postprandial concentrations, as measured at 1 month in a matched RYGB cohort (ClinicalTrials.gov NCT01945840). A 4-day food diary and validated eating behaviour questionnaires were completed. Sweet taste detection was measured using the method of constant stimuli. Correct sucrose identification (corrected hit rates) was recorded, and sweet taste detection thresholds (EC50s: half maximum effective concencration values) were derived from concentration curves. The intensity and consummatory reward value of sweet taste were assessed using the generalized Labelled Magnitude Scale. RESULTS Mean daily energy intake was reduced by 27% with GOP but no significant changes in food preferences were observed, whereas a reduction in fat and increase in protein intake were seen post-RYGB. There was no change in corrected hit rates or detection thresholds for sucrose detection following GOP infusion. Additionally, GOP did not alter the intensity or consummatory reward value of sweet taste. A significant reduction in restraint eating, comparable to the RYGB group was observed with GOP. CONCLUSION The elevation in plasma GOP concentrations after RYGB is unlikely to mediate changes in food preferences and sweet taste function after surgery but may promote restraint eating.
Collapse
Affiliation(s)
- Preeshila Behary
- Section of Endocrinology and Investigative Medicine, Imperial College LondonLondonUK
- Department of Endocrinology, Imperial College Healthcare NHS TrustLondonUK
| | - Haya Alessimii
- Clinical Nutrition Department, College of Applied Medical SciencesUmm Al Qura UniversityMeccaSaudi Arabia
| | - Alexander D. Miras
- Section of Endocrinology and Investigative Medicine, Imperial College LondonLondonUK
- Department of Endocrinology, Imperial College Healthcare NHS TrustLondonUK
- School of MedicineUlster UniversityLondonderryUK
| | - George Tharakan
- Section of Endocrinology and Investigative Medicine, Imperial College LondonLondonUK
- Department of Endocrinology, Imperial College Healthcare NHS TrustLondonUK
| | - Kleopatra Alexiadou
- Section of Endocrinology and Investigative Medicine, Imperial College LondonLondonUK
- Department of Endocrinology, Imperial College Healthcare NHS TrustLondonUK
| | - Madhawi M. Aldhwayan
- Community Health Sciences, College of Applied Medical SciencesKing Saud UniversityRiyadhSaudi Arabia
| | - Sanjay Purkayastha
- Department of Surgery and Cancer, Imperial College Healthcare National Health Service TrustLondonUK
| | - Krishna Moorthy
- Department of Surgery and Cancer, Imperial College Healthcare National Health Service TrustLondonUK
| | - Ahmed R. Ahmed
- Department of Surgery and Cancer, Imperial College Healthcare National Health Service TrustLondonUK
| | - Stephen R. Bloom
- Section of Endocrinology and Investigative Medicine, Imperial College LondonLondonUK
| | - Tricia M. Tan
- Section of Endocrinology and Investigative Medicine, Imperial College LondonLondonUK
- Department of Endocrinology, Imperial College Healthcare NHS TrustLondonUK
| |
Collapse
|
14
|
Feris F, McRae A, Kellogg TA, McKenzie T, Ghanem O, Acosta A. Mucosal and hormonal adaptations after Roux-en-Y gastric bypass. Surg Obes Relat Dis 2023; 19:37-49. [PMID: 36243547 PMCID: PMC9797451 DOI: 10.1016/j.soard.2022.08.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/12/2023]
Abstract
The aim of this study was to perform a comprehensive literature review regarding the relevant hormonal and histologic changes observed after Roux-en-Y gastric bypass (RYGB). We aimed to describe the relevant hormonal (glucagon-like peptides 1 and 2 [GLP-1 and GLP-2], peptide YY [PYY], oxyntomodulin [OXM], bile acids [BA], cholecystokinin [CCK], ghrelin, glucagon, gastric inhibitory polypeptide [GIP], and amylin) profiles, as well as the histologic (mucosal cellular) adaptations happening after patients undergo RYGB. Our review compiles the current evidence and furthers the understanding of the rationale behind the food intake regulatory adaptations occurring after RYGB surgery. We identify gaps in the literature where the potential for future investigations and therapeutics may lie. We performed a comprehensive database search without language restrictions looking for RYGB bariatric surgery outcomes in patients with pre- and postoperative blood work hormonal profiling and/or gut mucosal biopsies. We gathered the relevant study results and describe them in this review. Where human findings were lacking, we included animal model studies. The amalgamation of physiologic, metabolic, and cellular adaptations following RYGB is yet to be fully characterized. This constitutes a fundamental aspiration for enhancing and individualizing obesity therapy.
Collapse
Affiliation(s)
- Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alison McRae
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Todd A Kellogg
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Travis McKenzie
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Omar Ghanem
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
15
|
Bai L, Sivakumar N, Yu S, Mesgarzadeh S, Ding T, Ly T, Corpuz TV, Grove JCR, Jarvie BC, Knight ZA. Enteroendocrine cell types that drive food reward and aversion. eLife 2022; 11:74964. [PMID: 35913117 PMCID: PMC9363118 DOI: 10.7554/elife.74964] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 07/27/2022] [Indexed: 11/26/2022] Open
Abstract
Animals must learn through experience which foods are nutritious and should be consumed, and which are toxic and should be avoided. Enteroendocrine cells (EECs) are the principal chemosensors in the GI tract, but investigation of their role in behavior has been limited by the difficulty of selectively targeting these cells in vivo. Here, we describe an intersectional genetic approach for manipulating EEC subtypes in behaving mice. We show that multiple EEC subtypes inhibit food intake but have different effects on learning. Conditioned flavor preference is driven by release of cholecystokinin whereas conditioned taste aversion is mediated by serotonin and substance P. These positive and negative valence signals are transmitted by vagal and spinal afferents, respectively. These findings establish a cellular basis for how chemosensing in the gut drives learning about food.
Collapse
Affiliation(s)
- Ling Bai
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Nilla Sivakumar
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Shenliang Yu
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Sheyda Mesgarzadeh
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Tom Ding
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Truong Ly
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Timothy V Corpuz
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - James C R Grove
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Brooke C Jarvie
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Zachary A Knight
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
16
|
Samms RJ, Cosgrove R, Snider BM, Furber EC, Droz BA, Briere DA, Dunbar J, Dogra M, Alsina-Fernandez J, Borner T, De Jonghe BC, Hayes MR, Coskun T, Sloop KW, Emmerson PJ, Ai M. GIPR Agonism Inhibits PYY-Induced Nausea-Like Behavior. Diabetes 2022; 71:1410-1423. [PMID: 35499381 PMCID: PMC9233244 DOI: 10.2337/db21-0848] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/29/2022] [Indexed: 12/01/2022]
Abstract
The induction of nausea and emesis is a major barrier to maximizing the weight loss profile of obesity medications, and therefore, identifying mechanisms that improve tolerability could result in added therapeutic benefit. The development of peptide YY (PYY)-based approaches to treat obesity are no exception, as PYY receptor agonism is often accompanied by nausea and vomiting. Here, we sought to determine whether glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) agonism reduces PYY-induced nausea-like behavior in mice. We found that central and peripheral administration of a GIPR agonist reduced conditioned taste avoidance (CTA) without affecting hypophagia mediated by a PYY analog. The receptors for GIP and PYY (Gipr and Npy2r) were found to be expressed by the same neurons in the area postrema (AP), a brainstem nucleus involved in detecting aversive stimuli. Peripheral administration of a GIPR agonist induced neuronal activation (cFos) in the AP. Further, whole-brain cFos analyses indicated that PYY-induced CTA was associated with augmented neuronal activity in the parabrachial nucleus (PBN), a brainstem nucleus that relays aversive/emetic signals to brain regions that control feeding behavior. Importantly, GIPR agonism reduced PYY-mediated neuronal activity in the PBN, providing a potential mechanistic explanation for how GIPR agonist treatment reduces PYY-induced nausea-like behavior. Together, the results of our study indicate a novel mechanism by which GIP-based therapeutics may have benefit in improving the tolerability of weight loss agents.
Collapse
Affiliation(s)
- Ricardo J. Samms
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
- Corresponding authors: Ricardo J. Samms, , and Minrong Ai,
| | - Richard Cosgrove
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Brandy M. Snider
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Ellen C. Furber
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Brian A. Droz
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Daniel A. Briere
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - James Dunbar
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Mridula Dogra
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | | | - Tito Borner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Bart C. De Jonghe
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Matthew R. Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Tamer Coskun
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Kyle W. Sloop
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Paul J. Emmerson
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | - Minrong Ai
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
- Corresponding authors: Ricardo J. Samms, , and Minrong Ai,
| |
Collapse
|
17
|
Lee RHC, Wu CYC, Citadin CT, Couto E Silva A, Possoit HE, Clemons GA, Acosta CH, de la Llama VA, Neumann JT, Lin HW. Activation of Neuropeptide Y2 Receptor Can Inhibit Global Cerebral Ischemia-Induced Brain Injury. Neuromolecular Med 2022; 24:97-112. [PMID: 34019239 PMCID: PMC8606017 DOI: 10.1007/s12017-021-08665-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.
Collapse
Affiliation(s)
- Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Cristiane T Citadin
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alexandre Couto E Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Harlee E Possoit
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Garrett A Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Christina H Acosta
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Victoria A de la Llama
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Jake T Neumann
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA.
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA.
| |
Collapse
|
18
|
Woodward ORM, Gribble FM, Reimann F, Lewis JE. Gut peptide regulation of food intake - evidence for the modulation of hedonic feeding. J Physiol 2022; 600:1053-1078. [PMID: 34152020 DOI: 10.1113/jp280581] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
The number of people living with obesity has tripled worldwide since 1975 with serious implications for public health, as obesity is linked to a significantly higher chance of early death from associated comorbidities (metabolic syndrome, type 2 diabetes, cardiovascular disease and cancer). As obesity is a consequence of food intake exceeding the demands of energy expenditure, efforts are being made to better understand the homeostatic and hedonic mechanisms governing food intake. Gastrointestinal peptides are secreted from enteroendocrine cells in response to nutrient and energy intake, and modulate food intake either via afferent nerves, including the vagus nerve, or directly within the central nervous system, predominantly gaining access at circumventricular organs. Enteroendocrine hormones modulate homeostatic control centres at hypothalamic nuclei and the dorso-vagal complex. Additional roles of these peptides in modulating hedonic food intake and/or preference via the neural systems of reward are starting to be elucidated, with both peripheral and central peptide sources potentially contributing to central receptor activation. Pharmacological interventions and gastric bypass surgery for the treatment of type 2 diabetes and obesity elevate enteroendocrine hormone levels and also alter food preference. Hence, understanding of the hedonic mechanisms mediated by gut peptide action could advance development of potential therapeutic strategies for the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Orla R M Woodward
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jo E Lewis
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| |
Collapse
|
19
|
Al-Alsheikh AS, Alabdulkader S, Johnson B, Goldstone AP, Miras AD. Effect of Obesity Surgery on Taste. Nutrients 2022; 14:866. [PMID: 35215515 PMCID: PMC8878262 DOI: 10.3390/nu14040866] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/03/2022] Open
Abstract
Obesity surgery is a highly efficacious treatment for obesity and its comorbidities. The underlying mechanisms of weight loss after obesity surgery are not yet fully understood. Changes to taste function could be a contributing factor. However, the pattern of change in different taste domains and among obesity surgery operations is not consistent in the literature. A systematic search was performed to identify all articles investigating gustation in human studies following bariatric procedures. A total of 3323 articles were identified after database searches, searching references and deduplication, and 17 articles were included. These articles provided evidence of changes in the sensory and reward domains of taste following obesity procedures. No study investigated the effect of obesity surgery on the physiological domain of taste. Taste detection sensitivity for sweetness increases shortly after Roux-en-Y gastric bypass. Additionally, patients have a reduced appetitive reward value to sweet stimuli. For the subgroup of patients who experience changes in their food preferences after Roux-en-Y gastric bypass or vertical sleeve gastrectomy, changes in taste function may be underlying mechanisms for changing food preferences which may lead to weight loss and its maintenance. However, data are heterogeneous; the potential effect dilutes over time and varies significantly between different procedures.
Collapse
Affiliation(s)
- Alhanouf S. Al-Alsheikh
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shahd Alabdulkader
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
- Department of Health Sciences, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Brett Johnson
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
| | - Anthony P. Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Alexander Dimitri Miras
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (A.S.A.-A.); (S.A.); (B.J.); (A.D.M.)
| |
Collapse
|
20
|
Abstract
The enteroendocrine system coordinates the physiological response to food intake by regulating rates of digestion, nutrient absorption, insulin secretion, satiation and satiety. Gut hormones with important anorexigenic and/or insulinotropic roles include glucagon-like peptide 1 (GLP-1), peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP). High BMI or obesogenic diets do not markedly disrupt this enteroendocrine system, which represents a critical target for inducing weight loss and treating co-morbidities in individuals with obesity.
Collapse
|
21
|
Bloise AMNDLG, Simões-Alves AC, Debora Santos A, Morio B, Costa-Silva JH. Cardiometabolic impacts of saturated fatty acids: are they all comparable? Int J Food Sci Nutr 2021; 73:1-14. [PMID: 34229557 DOI: 10.1080/09637486.2021.1940885] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In last decades, a phenomenon named nutrition transition has been observed in many countries around the world. It has been characterised by increased consumption of fat-rich diets, predisposing to cardiometabolic diseases and high prevalence of the obesity. In the dietary recommendations cited to prevent metabolic diseases, there is a consensus to decrease intake of saturated fatty acids (SFA) to less than 10% of total energy intake, as recommended by the Food Safety Authorities. However, fatty acids of different chain lengths may exhibit different cardiometabolic effects. Thus, our major aim was to review the cardiometabolic effects of different classes of SFA according to carbon chain length, i.e. short-, medium- and long-chains. The review emphasises that not all SFA may have harmful cardiometabolic effects since short- and medium-chain SFA can provide beneficial health effects and participate to the prevention of metabolic disorders.
Collapse
Affiliation(s)
- Aline Maria Nunes de Lira Gomes Bloise
- Department of Physical Education and Sport Sciences, Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, UFPE, Vitória de Santo Antão-PE, Brazil
| | - Aiany Cibelle Simões-Alves
- Department of Physical Education and Sport Sciences, Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, UFPE, Vitória de Santo Antão-PE, Brazil.,Laboratoire de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition (CarMeN), INSERM U1060, INRA U1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Alves Debora Santos
- Department of Physical Education and Sport Sciences, Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, UFPE, Vitória de Santo Antão-PE, Brazil
| | - Beatrice Morio
- Laboratoire de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition (CarMeN), INSERM U1060, INRA U1397, Université Claude Bernard Lyon 1, Lyon, France
| | - João Henrique Costa-Silva
- Department of Physical Education and Sport Sciences, Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, UFPE, Vitória de Santo Antão-PE, Brazil.,Laboratoire de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition (CarMeN), INSERM U1060, INRA U1397, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
22
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Klockars A, Levine AS, Head MA, Perez-Leighton CE, Kotz CM, Olszewski PK. Impact of Gut and Metabolic Hormones on Feeding Reward. Compr Physiol 2021; 11:1425-1447. [PMID: 33577129 DOI: 10.1002/cphy.c190042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ingestion of food activates a cascade of endocrine responses (thereby reflecting a contemporaneous feeding status) that include the release of hormones from the gastrointestinal (GI) tract, such as cholecystokinin (CCK), glucagonlike peptide YY (PYY), peptide PP, and oleoylethanolamide, as well as suppression of ghrelin secretion. The pancreas and adipose tissue, on the other hand, release hormones that serve as a measure of the current metabolic state or the long-term energy stores, that is, insulin, leptin, and adiponectin. It is well known and intuitively understandable that these hormones target either directly (by crossing the blood-brain barrier) or indirectly (e.g., via vagal input) the "homeostatic" brainstem-hypothalamic pathways involved in the regulation of appetite. The current article focuses on yet another target of the metabolic and GI hormones that is critical in inducing changes in food intake, namely, the reward system. We discuss the physiological basis of this functional interaction, its importance in the control of appetite, and the impact that disruption of this crosstalk has on energy intake in select physiological and pathophysiological states. We conclude that metabolic and GI hormones have a capacity to strengthen or weaken a response of the reward system to a given food, and thus, they are fundamental in ensuring that feeding reward is plastic and dependent on the energy status of the organism. © 2021 American Physiological Society. Compr Physiol 11:1425-1447, 2021.
Collapse
Affiliation(s)
- Anica Klockars
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | - Allen S Levine
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA
| | - Mitchell A Head
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | | | - Catherine M Kotz
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA.,Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand.,Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA.,Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
24
|
Wu Q, Guo D, Jia H, Nepovimova E, Wu W, Kuca K. The trichothecene neosolaniol stimulates an emetic response through neuropeptide Y2 and serotonin 3 receptors in mink. Toxicology 2021; 452:152718. [PMID: 33581213 DOI: 10.1016/j.tox.2021.152718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/31/2021] [Accepted: 02/06/2021] [Indexed: 01/05/2023]
Abstract
Type A trichothecene neosolaniol (NEO) is considered a potential risk to human and animal health by the European Food Safety Authority (EFSA). To date, available data do not allow making conclusions about the toxicological properties of this toxin. Trichothecenes have been previously demonstrated to induce emetic responses in mink, and this response has been associated with neurotransmitter peptide YY (PYY) and serotonin (5-hydroxytryptamine, 5-HT). The goal of this study was to compare emetic effects of NEO administered by intraperitoneal and oral routes and relate these effects to PYY and 5-HT. The effective doses resulting in emetic events in 50% of the animals following intraperitoneal and oral exposure to NEO were 0.4 and 0.09 mg/kg bw, respectively. This emetic response corresponded to elevated PYY and 5-HT levels. Blocking the neuropeptide Y2 receptor diminished emesis induction by PYY and NEO. The 5-HT3 receptor inhibitor granisetron completely restrained the induction of emesis by 5-HT and NEO. To summarize, our findings demonstrate that PYY and 5-HT play important roles in the NEO-induced emetic response.
Collapse
Affiliation(s)
- Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Dawei Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Hui Jia
- Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212499, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic.
| |
Collapse
|
25
|
Blanco AM, Calo J, Soengas JL. The gut-brain axis in vertebrates: implications for food intake regulation. J Exp Biol 2021; 224:jeb231571. [PMID: 33414256 DOI: 10.1242/jeb.231571] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
The gut and brain are constantly communicating and influencing each other through neural, endocrine and immune signals in an interaction referred to as the gut-brain axis. Within this communication system, the gastrointestinal tract, including the gut microbiota, sends information on energy status to the brain, which, after integrating these and other inputs, transmits feedback to the gastrointestinal tract. This allows the regulation of food intake and other physiological processes occurring in the gastrointestinal tract, including motility, secretion, digestion and absorption. Although extensive literature is available on the mechanisms governing the communication between the gut and the brain in mammals, studies on this axis in other vertebrates are scarce and often limited to a single species, which may not be representative for obtaining conclusions for an entire group. This Review aims to compile the available information on the gut-brain axis in birds, reptiles, amphibians and fish, with a special focus on its involvement in food intake regulation and, to a lesser extent, in digestive processes. Additionally, we will identify gaps of knowledge that need to be filled in order to better understand the functioning and physiological significance of such an axis in non-mammalian vertebrates.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, 36310 Vigo, Pontevedra, Spain
| | - Jessica Calo
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, 36310 Vigo, Pontevedra, Spain
| | - José Luis Soengas
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, 36310 Vigo, Pontevedra, Spain
| |
Collapse
|
26
|
Olson B, Marks DL, Grossberg AJ. Diverging metabolic programmes and behaviours during states of starvation, protein malnutrition, and cachexia. J Cachexia Sarcopenia Muscle 2020; 11:1429-1446. [PMID: 32985801 PMCID: PMC7749623 DOI: 10.1002/jcsm.12630] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Our evolutionary history is defined, in part, by our ability to survive times of nutrient scarcity. The outcomes of the metabolic and behavioural adaptations during starvation are highly efficient macronutrient allocation, minimization of energy expenditure, and maximized odds of finding food. However, in different contexts, caloric deprivation is met with vastly different physiologic and behavioural responses, which challenge the primacy of energy homeostasis. METHODS We conducted a literature review of scientific studies in humans, laboratory animals, and non-laboratory animals that evaluated the physiologic, metabolic, and behavioural responses to fasting, starvation, protein-deficient or essential amino acid-deficient diets, and cachexia. Studies that investigated the changes in ingestive behaviour, locomotor activity, resting metabolic rate, and tissue catabolism were selected as the focus of discussion. RESULTS Whereas starvation responses prioritize energy balance, both protein malnutrition and cachexia present existential threats that induce unique adaptive programmes, which can exacerbate the caloric insufficiency of undernutrition. We compare and contrast the behavioural and metabolic responses and elucidate the mechanistic pathways that drive state-dependent alterations in energy seeking and partitioning. CONCLUSIONS The evolution of energetically inefficient metabolic and behavioural responses to protein malnutrition and cachexia reveal a hierarchy of metabolic priorities governed by discrete regulatory networks.
Collapse
Affiliation(s)
- Brennan Olson
- Medical Scientist Training ProgramOregon Health & Science UniversityPortlandORUSA
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
| | - Daniel L. Marks
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
| | - Aaron J. Grossberg
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
- Department of Radiation MedicineOregon Health & Science UniversityPortlandORUSA
- Cancer Early Detection Advanced Research CenterOregon Health & Science UniversityPortlandORUSA
| |
Collapse
|
27
|
Lear S, Pflimlin E, Zhou Z, Huang D, Weng S, Nguyen-Tran V, Joseph SB, Roller S, Peterson S, Li J, Tremblay M, Schultz PG, Shen W. Engineering of a Potent, Long-Acting NPY2R Agonist for Combination with a GLP-1R Agonist as a Multi-Hormonal Treatment for Obesity. J Med Chem 2020; 63:9660-9671. [PMID: 32844654 DOI: 10.1021/acs.jmedchem.0c00740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bariatric surgery results in increased intestinal secretion of hormones GLP-1 and anorexigenic PYY, which is believed to contribute to the clinical efficacy associated with the procedure. This observation raises the question whether combination treatment with gut hormone analogs might recapitulate the efficacy and mitigate the significant risks associated with surgery. Despite PYY demonstrating excellent efficacy and safety profiles with regard to food intake reduction, weight loss, and glucose control in preclinical animal models, PYY-based therapeutic development remains challenging given a low serum stability and half-life for the native peptide. Here, combined peptide stapling and PEG-fatty acid conjugation affords potent PYY analogs with >14 h rat half-lives, which are expected to translate into a human half-life suitable for once-weekly dosing. Excellent efficacy in glucose control, food intake reduction, and weight loss for lead candidate 22 in combination with our previously reported long-acting GLP-1 analog is demonstrated in a diet-induced obesity mouse model.
Collapse
Affiliation(s)
- Sam Lear
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Elsa Pflimlin
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Zhihong Zhou
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - David Huang
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Sharon Weng
- Intarcia Therapeutics, Inc., Research Triangle Park, 6 Davis Drive, Durham, North Carolina 27709, United States
| | - Van Nguyen-Tran
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Sean B Joseph
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Shane Roller
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Scott Peterson
- Intarcia Therapeutics, Inc., Research Triangle Park, 6 Davis Drive, Durham, North Carolina 27709, United States
| | - Jing Li
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Matthew Tremblay
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Peter G Schultz
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| | - Weijun Shen
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, 11119 North Torrey Pines Road, Suite 100, La Jolla, California 92037, United States
| |
Collapse
|
28
|
Wu Q, Kuca K, Nepovimova E, Wu W. Type A Trichothecene Diacetoxyscirpenol-Induced Emesis Corresponds to Secretion of Peptide YY and Serotonin in Mink. Toxins (Basel) 2020; 12:toxins12060419. [PMID: 32630472 PMCID: PMC7354585 DOI: 10.3390/toxins12060419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022] Open
Abstract
The trichothecene mycotoxins contaminate cereal grains and have been related to alimentary toxicosis resulted in emetic response. This family of mycotoxins comprises type A to D groups of toxic sesquiterpene chemicals. Diacetoxyscirpenol (DAS), one of the most toxic type A trichothecenes, is considered to be a potential risk for human and animal health by the European Food Safety Authority. Other type A trichothecenes, T-2 toxin and HT-2 toxin, as well as type B trichothecene deoxynivalenol (DON), have been previously demonstrated to induce emetic response in the mink, and this response has been associated with the plasma elevation of neurotransmitters peptide YY (PYY) and serotonin (5-hydroxytryptamine, 5-HT). However, it is found that not all the type A and type B trichothecenes have the capacity to induce PYY and 5-HT. It is necessary to identify the roles of these two emetogenic mediators on DAS-induced emesis. The goal of this study was to determine the emetic effect of DAS and relate this effect to PYY and 5-HT, using a mink bioassay. Briefly, minks were fasted one day before experiment and given DAS by intraperitoneally and orally dosing on the experiment day. Then, emetic episodes were calculated and blood collection was employed for PYY and 5-HT test. DAS elicited robust emetic responses that corresponded to upraised PYY and 5-HT. Blocking the neuropeptide Y2 receptor (NPY2R) diminished emesis induction by PYY and DAS. The serotonin 3 receptor (5-HT3R) inhibitor granisetron totally restrained the induction of emesis by serotonin and DAS. In conclusion, our findings demonstrate that PYY and 5-HT have critical roles in DAS-induced emetic response.
Collapse
Affiliation(s)
- Qinghua Wu
- College of Life Science, Yangtze University, Jinzhou 434025, China;
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
- Correspondence: (K.K.); (W.W.)
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
| | - Wenda Wu
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (K.K.); (W.W.)
| |
Collapse
|
29
|
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: 70] [Impact Index Per Article: 14.0] [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
|
30
|
Sanches E, Timmermans M, Topal B, Celik A, Sundbom M, Ribeiro R, Parmar C, Ugale S, Proczko M, Stepaniak PS, Pujol Rafols J, Mahawar K, Buise MP, Neimark A, Severin R, Pouwels S. Cardiac remodeling in obesity and after bariatric and metabolic surgery; is there a role for gastro-intestinal hormones? Expert Rev Cardiovasc Ther 2019; 17:771-790. [PMID: 31746657 DOI: 10.1080/14779072.2019.1690991] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Obesity is associated with various diseases such as type 2 diabetes, hypertension, obstructive sleep apnea syndrome (OSAS), metabolic syndrome, and cardiovascular diseases. It affects several organ systems, including the pulmonary and cardiac systems. Furthermore, it induces pulmonary and cardiac changes that can result in right and/or left heart failure.Areas covered: In this review, authors provide an overview of obesity and cardiovascular remodeling, the individual actions of the gut hormones (like GLP-1 and PYY), the effects after bariatric/metabolic surgery and its influence on cardiac remodeling. In this review, we focussed and searched for literature in Pubmed and The Cochrane library (from the earliest date until April 2019), regarding cardiac function changes before and after bariatric surgery and literature regarding changes in gastrointestinal hormones.Expert opinion: Regarding the surgical treatment of obesity and metabolic diseases there is recognition of the importance of both weight loss (bariatric surgery) and improvement in metabolic milieu (metabolic surgery). A growing body of evidence further suggests that bariatric surgical procedures [like the Sleeve Gastrectomy (SG), Roux-en Y Gastric Bypass (RYGB), or One Anastomosis Gastric Bypass (OAGB)] have can improve outcomes of patients suffering from a number of cardiovascular diseases, including heart failure.
Collapse
Affiliation(s)
- Elijah Sanches
- Department of Surgery, Haaglanden Medical Center, The Hague, The Netherlands
| | - Marieke Timmermans
- Department of Surgery, Haaglanden Medical Center, The Hague, The Netherlands
| | - Besir Topal
- Department of Cardiothoracic Surgery, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Alper Celik
- Department of Bariatric and Metabolic Surgery, Metabolic Surgery Clinic, Sisli, Turkey
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rui Ribeiro
- Centro Multidisciplinar da Doença Metabólica, Clínica de Santo António, Lisbon, Portugal
| | - Chetan Parmar
- Department of Surgery, Whittington Hospital, London, UK
| | - Surendra Ugale
- Bariatric & Metabolic Surgery Clinic, Kirloskar Hospital, Hyderabad, India
| | - Monika Proczko
- Department of General, Endocrine and Transplant Surgery, University Medical Center, Gdansk University, Gdansk, Poland
| | - Pieter S Stepaniak
- Department of Operating Rooms, Catharina Hospital, Eindhoven, The Netherlands
| | | | - Kamal Mahawar
- Bariatric Unit, Sunderland Royal Hospital, Sunderland, UK
| | - Marc P Buise
- Department of Anesthesiology, Intensive Care and Pain Medicine, Catharina Hospital, Eindhoven, The Netherlands
| | - Aleksandr Neimark
- Department of Surgery, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Rich Severin
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA.,Doctor of Physical Therapy Program, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA
| | - Sjaak Pouwels
- Department of Surgery, Haaglanden Medical Center, The Hague, The Netherlands
| |
Collapse
|
31
|
Jones ES, Nunn N, Chambers AP, Østergaard S, Wulff BS, Luckman SM. Modified Peptide YY Molecule Attenuates the Activity of NPY/AgRP Neurons and Reduces Food Intake in Male Mice. Endocrinology 2019; 160:2737-2747. [PMID: 31074796 PMCID: PMC6806261 DOI: 10.1210/en.2019-00100] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/06/2019] [Indexed: 01/15/2023]
Abstract
To study the effects of an analog of the gut-produced hormone peptide YY (PYY3-36), which has increased selectivity for the Y2 receptor; specifically, to record its effects on food intake and on hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neuron activity. NNC0165-1273, a modified form of the peptide hormone PYY3-36 with potent selectivity at Y2 receptor (>5000-fold over Y1, 1250-fold over Y4, and 650-fold over Y5 receptor), was tested in vivo and in vitro in mouse models. NNC0165-1273 has fivefold lower relative affinity for Y2 compared with PYY3-36, but >250-, 192-, and 400-fold higher selectivity, respectively, for the Y1, Y4, and Y5 receptors. NNC0165-1273 produced a reduction in nighttime feeding at a dose at which PYY3-36 loses efficacy. The normal behavioral satiety sequence observed suggests that NNC0165-1273 is not nauseating and, instead, reduces food intake by producing early satiety. Additionally, NNC0165-1273 blocked ghrelin-induced cFos expression in NPY/AgRP neurons. In vitro electrophysiological recordings showed that, opposite to ghrelin, NNC0165-1273 hyperpolarized NPY/AgRP neurons and reduced action potential frequency. Administration of NNC0165-1273 via subcutaneous osmotic minipump caused a dose-dependent decrease in body weight and fat mass in an obese mouse model. Finally, NNC0165-1273 attenuated the feeding response when NPY/AgRP neurons were activated using ghrelin or more selectively with designer receptors. NNC0165-1273 is nonnauseating and stimulates a satiety response through, at least in part, a direct action on hypothalamic NPY/AgRP neurons. Modification of PYY3-36 to produce compounds with increased affinity to Y2 receptors may be useful as antiobesity therapies in humans.
Collapse
Affiliation(s)
- Edward S Jones
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nicolas Nunn
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Adam P Chambers
- GLP-1 & T2D Pharmacology, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Søren Østergaard
- Research Chemistry 2, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Birgitte S Wulff
- Obesity Research, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Simon M Luckman
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
32
|
Chang MR, Ciesla A, Strutzenberg TS, Novick SJ, He Y, Garcia-Ordonez RD, Frkic RL, Bruning JB, Kamenecka TM, Griffin PR. Unique Polypharmacology Nuclear Receptor Modulator Blocks Inflammatory Signaling Pathways. ACS Chem Biol 2019; 14:1051-1062. [PMID: 30951276 DOI: 10.1021/acschembio.9b00236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity and rheumatic disease are mechanistically linked via chronic inflammation. The orphan receptor TREM-1 (triggering receptor expressed on myeloid cells-1) is a potent amplifier of proinflammatory and noninfectious immune responses. Here, we show that the pan modulator SR1903 effectively blocks TREM-1 activation. SR1903 emerged from a chemical series of potent RORγ inverse agonists, although unlike close structural analogues, it has modest agonist activity on LXR and weak repressive activity (inverse agonism) of PPARγ, three receptors that play essential roles in inflammation and metabolism. The anti-inflammatory and antidiabetic efficacy of this unique modulator in collagen-induced arthritis and diet-induced obesity mouse models is demonstrated. Interestingly, in the context of obesity, SR1903 aided in the maintenance of the thymic homeostasis unlike selective RORγ inverse agonists. SR1903 was well-tolerated following chronic administration, and combined, these data suggest that it may represent a viable strategy for treatment of both metabolic and inflammatory disease. More importantly, the ability of SR1903 to block LPS signaling suggests the potential utility of this unique polypharmacological modulator for treatment of innate immune response disorders.
Collapse
Affiliation(s)
- Mi Ra Chang
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Anthony Ciesla
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Timothy S. Strutzenberg
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Scott J. Novick
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Yuanjun He
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Ruben D. Garcia-Ordonez
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Rebecca L. Frkic
- Institute for Photonics & Advanced Sensing (IPAS), School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - John B. Bruning
- Institute for Photonics & Advanced Sensing (IPAS), School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Theodore M. Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Patrick R. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida 33458, United States
| |
Collapse
|
33
|
Al-Najim W, Docherty NG, le Roux CW. Food Intake and Eating Behavior After Bariatric Surgery. Physiol Rev 2018; 98:1113-1141. [PMID: 29717927 DOI: 10.1152/physrev.00021.2017] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Obesity is an escalating global chronic disease. Bariatric surgery is a very efficacious treatment for obesity and its comorbidities. Alterations to gastrointestinal anatomy during bariatric surgery result in neurological and physiological changes affecting hypothalamic signaling, gut hormones, bile acids, and gut microbiota, which coalesce to exert a profound influence on eating behavior. A thorough understanding of the mechanisms underlying eating behavior is essential in the management of patients after bariatric surgery. Studies investigating candidate mechanisms have expanded dramatically in the last decade. Herein we review the proposed mechanisms governing changes in eating behavior, food intake, and body weight after bariatric surgery. Additive or synergistic effects of both conditioned and unconditioned factors likely account for the complete picture of changes in eating behavior. Considered application of strategies designed to support the underlying principles governing changes in eating behavior holds promise as a means of optimizing responses to surgery and long-term outcomes.
Collapse
Affiliation(s)
- Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| |
Collapse
|
34
|
Cork SC. The role of the vagus nerve in appetite control: Implications for the pathogenesis of obesity. J Neuroendocrinol 2018; 30:e12643. [PMID: 30203877 DOI: 10.1111/jne.12643] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
The communication between the gut and the brain is important for the control of energy homeostasis. In response to food intake, enteroendocrine cells secrete gut hormones, which ultimately suppress appetite through centrally-mediated processes. Increasing evidence implicates the vagus nerve as an important conduit in transmitting these signals from the gastrointestinal tract to the brain. Studies have demonstrated that many of the gut hormones secreted from enteroendocrine cells signal through the vagus nerve, and the sensitivity of the vagus to these signals is regulated by feeding status. Furthermore, evidence suggests that a reduction in the ability of the vagus nerve to respond to the switch between a "fasted" and "fed" state, retaining sensitivity to orexigenic signals when fed or a reduced ability to respond to satiety hormones, may contribute to obesity. This review draws together the evidence that the vagus nerve is a crucial component of appetite regulation via the gut-brain axis, with a particular emphasis on experimental techniques and future developments.
Collapse
Affiliation(s)
- Simon C Cork
- Section of Endocrinology and Investigative Medicine, Division of Endocrinology, Diabetes and Metabolism, Imperial College London, London, UK
| |
Collapse
|
35
|
Kim KS, Seeley RJ, Sandoval DA. Signalling from the periphery to the brain that regulates energy homeostasis. Nat Rev Neurosci 2018; 19:185-196. [PMID: 29467468 PMCID: PMC9190118 DOI: 10.1038/nrn.2018.8] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The CNS regulates body weight; however, we still lack a clear understanding of what drives decisions about when, how much and what to eat. A vast array of peripheral signals provides information to the CNS regarding fluctuations in energy status. The CNS then integrates this information to influence acute feeding behaviour and long-term energy homeostasis. Previous paradigms have delegated the control of long-term energy homeostasis to the hypothalamus and short-term changes in feeding behaviour to the hindbrain. However, recent studies have identified target hindbrain neurocircuitry that integrates the orchestration of individual bouts of ingestion with the long-term regulation of energy balance.
Collapse
Affiliation(s)
- Ki-Suk Kim
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Randy J. Seeley
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Darleen A. Sandoval
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Abstract
Hypothalamic integration of gastrointestinal and adipose tissue-derived hormones serves as a key element of neuroendocrine control of food intake. Leptin, adiponectin, oleoylethanolamide, cholecystokinin, and ghrelin, to name a few, are in a constant "cross talk" with the feeding-related brain circuits that encompass hypothalamic populations synthesizing anorexigens (melanocortins, CART, oxytocin) and orexigens (Agouti-related protein, neuropeptide Y, orexins). While this integrated neuroendocrine circuit successfully ensures that enough energy is acquired, it does not seem to be equally efficient in preventing excessive energy intake, especially in the obesogenic environment in which highly caloric and palatable food is constantly available. The current review presents an overview of intricate mechanisms underlying hypothalamic integration of energy balance-related peripheral endocrine input. We discuss vulnerabilities and maladaptive neuroregulatory processes, including changes in hypothalamic neuronal plasticity that propel overeating despite negative consequences.
Collapse
|
37
|
Miller GD. Appetite Regulation: Hormones, Peptides, and Neurotransmitters and Their Role in Obesity. Am J Lifestyle Med 2017; 13:586-601. [PMID: 31662725 DOI: 10.1177/1559827617716376] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding body weight regulation will aid in the development of new strategies to combat obesity. This review examines energy homeostasis and food intake behaviors, specifically with regards to hormones, peptides, and neurotransmitters in the periphery and central nervous system, and their potential role in obesity. Dysfunction in feeding signals by the brain is a factor in obesity. The hypothalamic (arcuate nucleus) and brainstem (nucleus tractus solitaris) areas integrate behavioral, endocrine, and autonomic responses via afferent and efferent pathways from and to the brainstem and peripheral organs. Neurons present in the arcuate nucleus express pro-opiomelanocortin, Neuropeptide Y, and Agouti Related Peptide, with the former involved in lowering food intake, and the latter two acutely increasing feeding behaviors. Action of peripheral hormones from the gut, pancreas, adipose, and liver are also involved in energy homeostasis. Vagal afferent neurons are also important in regulating energy homeostasis. Peripheral signals respond to the level of stored and currently available fuel. By studying their actions, new agents maybe developed that disable orexigenic responses and enhance anorexigenic signals. Although there are relatively few medications currently available for obesity treatment, a number of agents are in development that work through these pathways.
Collapse
Affiliation(s)
- Gary D Miller
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
38
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 402] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
39
|
Systemic administration of anorexic gut peptide hormones impairs hedonic-driven sucrose consumption in mice. Physiol Behav 2016; 171:158-164. [PMID: 28040488 DOI: 10.1016/j.physbeh.2016.12.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 12/26/2016] [Accepted: 12/26/2016] [Indexed: 01/01/2023]
Abstract
A number of reports suggest that gut hormones such as cholecystokinin (CCK), glucagon-like peptide 1 (GLP-1), and peptide YY(3-36) (PYY3-36), which are released postprandially, suppress homeostatic food intake and result in satiety and the termination of feeding. However, it remains unclear whether these peptide hormones also suppress non-homeostatic consumption of palatable foods or fluids. To examine whether gut hormones reduce hedonically motivated sugar consumption, we assessed the effects of intraperitoneal administration of these gut hormones on the consumption of a highly palatable sucrose solution, using a mouse model we previously established for binge-like sucrose overconsumption (Yasoshima and Shimura, 2015). To reduce homeostatic hunger, chow was available at nighttime prior to testing. After a limited-access training procedure for 10days, during which access to both sucrose and chow were controlled, on the test day, control mice injected with saline consumed significantly more sucrose than during the pre-training period. In contrast, sucrose consumption on the test day in the mice injected with CCK-8 (2 and 4μg/kg), GLP-1 (500 and 1000nmol/kg), or PYY3-36 (12.5 and 25nmol/kg) was significantly less than that in saline-injected mice. In a separate cohort of mice, the higher doses of CCK-8 and GLP-1 and a greater dose of PYY3-36 (50nmol/kg) did not produce conditioned taste aversion to saccharin, suggesting that the doses of exogenous hormones in the present study do not cause aversive visceral distress. The present findings suggest that the systemic administration of these three gut hormones suppresses hedonic-driven sugar consumption due to the anorexic, but not aversive-visceral, effects of these hormones.
Collapse
|
40
|
Reidelberger R, Haver A, Anders K, Apenteng B, Lanio C. Effects of solid-phase extraction of plasma in measuring gut metabolic hormones in fasted and fed blood of lean and diet-induced obese rats. Physiol Rep 2016; 4:4/10/e12800. [PMID: 27207785 PMCID: PMC4886168 DOI: 10.14814/phy2.12800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/26/2016] [Indexed: 12/25/2022] Open
Abstract
Glucagon‐like peptide‐1 (GLP‐1), peptide YY (3‐36) [PYY(3‐36)], amylin, ghrelin, insulin, and leptin are thought to act as hormonal signals from periphery to brain to control food intake. Here, we determined the effects of solid‐phase extraction of plasma in measuring these hormones in blood of lean and diet‐induced obese rats. Individual enzyme‐linked immunoassays and a multiplex assay were used to measure active GLP‐1, total PYY, active amylin, active ghrelin, insulin, leptin, and total GIP in response to (1) addition of known amounts of the peptides to lean and obese plasma, (2) a large meal in lean and obese rats, and (3) intravenous infusions of anorexigenic doses of GLP‐1, PYY(3‐36), amylin, and leptin in lean rats. Extraction of lean and obese plasma prior to assays produced consistent recoveries across assays for GLP‐1, PYY, amylin, ghrelin, and insulin, reflecting losses inherent to the extraction procedure. Plasma extraction prior to assays generally revealed larger meal‐induced changes in plasma GLP‐1, PYY, amylin, ghrelin, and insulin in lean and obese rats. Plasma extraction and the multiplex assay were used to compare plasma levels of GLP‐1, PYY, and amylin after a large meal with plasma levels produced by IV infusions of anorexigenic doses of GLP‐1, PYY(3‐36), and amylin. Infusions produced dose‐dependent increases in plasma peptide levels, which were well above their postprandial levels. These results do not support the hypothesis that postprandial plasma levels of GLP‐1, PYY(3‐36), and amylin are sufficient to decrease food intake by an endocrine mechanism.
Collapse
Affiliation(s)
- Roger Reidelberger
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska VA Research Service, VA Nebraska Western Iowa Health Care System, Omaha, Nebraska
| | - Alvin Haver
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska VA Research Service, VA Nebraska Western Iowa Health Care System, Omaha, Nebraska
| | - Krista Anders
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Bettye Apenteng
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Craig Lanio
- EMD Millipore Corp., Billerica, Massachusetts
| |
Collapse
|
41
|
Ratner C, Skov LJ, Raida Z, Bächler T, Bellmann-Sickert K, Le Foll C, Sivertsen B, Dalbøge LS, Hartmann B, Beck-Sickinger AG, Madsen AN, Jelsing J, Holst JJ, Lutz TA, Andrews ZB, Holst B. Effects of Peripheral Neurotensin on Appetite Regulation and Its Role in Gastric Bypass Surgery. Endocrinology 2016; 157:3482-92. [PMID: 27580810 DOI: 10.1210/en.2016-1329] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neurotensin (NT) is a peptide expressed in the brain and in the gastrointestinal tract. Brain NT inhibits food intake, but the effects of peripheral NT are less investigated. In this study, peripheral NT decreased food intake in both mice and rats, which was abolished by a NT antagonist. Using c-Fos immunohistochemistry, we found that peripheral NT activated brainstem and hypothalamic regions. The anorexigenic effect of NT was preserved in vagotomized mice but lasted shorter than in sham-operated mice. This in combination with a strong increase in c-Fos activation in area postrema after ip administration indicates that NT acts both through the blood circulation and the vagus. To improve the pharmacokinetics of NT, we developed a pegylated NT peptide, which presumably prolonged the half-life, and thus, the effect on feeding was extended compared with native NT. On a molecular level, the pegylated NT peptide increased proopiomelanocortin mRNA in the arcuate nucleus. We also investigated the importance of NT for the decreased food intake after gastric bypass surgery in a rat model of Roux-en-Y gastric bypass (RYGB). NT was increased in plasma and in the gastrointestinal tract in RYGB rats, and pharmacological antagonism of NT increased food intake transiently in RYGB rats. Taken together, our data suggest that NT is a metabolically active hormone, which contributes to the regulation of food intake.
Collapse
Affiliation(s)
- Cecilia Ratner
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Louise J Skov
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Zindy Raida
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Thomas Bächler
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Kathrin Bellmann-Sickert
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Christelle Le Foll
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Bjørn Sivertsen
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Louise S Dalbøge
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Bolette Hartmann
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Annette G Beck-Sickinger
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Andreas N Madsen
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Jacob Jelsing
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Jens J Holst
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Thomas A Lutz
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Zane B Andrews
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| | - Birgitte Holst
- Laboratory for Molecular Pharmacology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.), Department of Neuroscience and Pharmacology, and Department of Biomedical Sciences (B.Ha., J.J.H.), Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Section for Metabolic Receptology (C.R., L.J.S., Z.R., B.S., A.N.M., B.Ho.) and Section for Translational Metabolic Physiology (B.Ha., J.J.H.), the Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; Institute of Veterinary Physiology (T.B., C.L.F., T.A.L.), Vetsuisse Faculty, and Center for Integrative Human Physiology (T.A.L.), University of Zurich, CH-8057 Zurich, Switzerland; Institute of Biochemistry (K.B.-S., A.G.B.-S.), University of Leipzig, D-04103 Leipzig, Germany; Gubra ApS (L.S.D., J.J.), Hørsholm, DK-2970 Denmark; and Biomedicine Discovery Institute (Z.B.A.), Metabolic Disease and Obesity Program, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
42
|
Ferrer I, Garcia-Esparcia P, Carmona M, Carro E, Aronica E, Kovacs GG, Grison A, Gustincich S. Olfactory Receptors in Non-Chemosensory Organs: The Nervous System in Health and Disease. Front Aging Neurosci 2016; 8:163. [PMID: 27458372 PMCID: PMC4932117 DOI: 10.3389/fnagi.2016.00163] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/21/2016] [Indexed: 12/22/2022] Open
Abstract
Olfactory receptors (ORs) and down-stream functional signaling molecules adenylyl cyclase 3 (AC3), olfactory G protein α subunit (Gαolf), OR transporters receptor transporter proteins 1 and 2 (RTP1 and RTP2), receptor expression enhancing protein 1 (REEP1), and UDP-glucuronosyltransferases (UGTs) are expressed in neurons of the human and murine central nervous system (CNS). In vitro studies have shown that these receptors react to external stimuli and therefore are equipped to be functional. However, ORs are not directly related to the detection of odors. Several molecules delivered from the blood, cerebrospinal fluid, neighboring local neurons and glial cells, distant cells through the extracellular space, and the cells’ own self-regulating internal homeostasis can be postulated as possible ligands. Moreover, a single neuron outside the olfactory epithelium expresses more than one receptor, and the mechanism of transcriptional regulation may be different in olfactory epithelia and brain neurons. OR gene expression is altered in several neurodegenerative diseases including Parkinson’s disease (PD), Alzheimer’s disease (AD), progressive supranuclear palsy (PSP) and sporadic Creutzfeldt-Jakob disease (sCJD) subtypes MM1 and VV2 with disease-, region- and subtype-specific patterns. Altered gene expression is also observed in the prefrontal cortex in schizophrenia with a major but not total influence of chlorpromazine treatment. Preliminary parallel observations have also shown the presence of taste receptors (TASRs), mainly of the bitter taste family, in the mammalian brain, whose function is not related to taste. TASRs in brain are also abnormally regulated in neurodegenerative diseases. These seminal observations point to the need for further studies on ORs and TASRs chemoreceptors in the mammalian brain.
Collapse
Affiliation(s)
- Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Hospitalet de Llobregat, University of BarcelonaBarcelona, Spain; Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED)Madrid, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de LlobregatBarcelona, Spain
| | - Paula Garcia-Esparcia
- Institute of Neuropathology, Bellvitge University Hospital, Hospitalet de Llobregat, University of BarcelonaBarcelona, Spain; Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED)Madrid, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de LlobregatBarcelona, Spain
| | - Margarita Carmona
- Institute of Neuropathology, Bellvitge University Hospital, Hospitalet de Llobregat, University of BarcelonaBarcelona, Spain; Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED)Madrid, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de LlobregatBarcelona, Spain
| | - Eva Carro
- Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED)Madrid, Spain; Neuroscience Group, Research Institute HospitalMadrid, Spain
| | - Eleonora Aronica
- Department of Neuropathology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna Vienna, Austria
| | - Alice Grison
- Scuola Internazionale Superiore di Studi Avanzati (SISSA), Area of Neuroscience Trieste, Italy
| | - Stefano Gustincich
- Scuola Internazionale Superiore di Studi Avanzati (SISSA), Area of Neuroscience Trieste, Italy
| |
Collapse
|
43
|
Latorre R, Sternini C, De Giorgio R, Greenwood-Van Meerveld B. Enteroendocrine cells: a review of their role in brain-gut communication. Neurogastroenterol Motil 2016; 28:620-30. [PMID: 26691223 PMCID: PMC4842178 DOI: 10.1111/nmo.12754] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/17/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Specialized endoderm-derived epithelial cells, that is, enteroendocrine cells (EECs), are widely distributed throughout the gastrointestinal (GI) tract. Enteroendocrine cells form the largest endocrine organ in the body and play a key role in the control of GI secretion and motility, the regulation of food intake, postprandial glucose levels and metabolism. EECs sense luminal content and release signaling molecules that can enter the circulation to act as classic hormones on distant targets, act locally on neighboring cells and on distinct neuronal pathways including enteric and extrinsic neurons. Recent studies have shed light on EEC sensory transmission by showing direct connections between EECs and the nervous system via axon-like processes that form a well-defined neuroepithelial circuits through which EECs can directly communicate with the neurons innervating the GI tract to initiate appropriate functional responses. PURPOSE This review will highlight the role played by the EECs in the complex and integrated sensory information responses, and discuss the new findings regarding EECs in the brain-gut axis bidirectional communication.
Collapse
Affiliation(s)
- R Latorre
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - C Sternini
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Departments of Medicine and Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - R De Giorgio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - B Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Physiology, Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
44
|
Latorre R, Huynh J, Mazzoni M, Gupta A, Bonora E, Clavenzani P, Chang L, Mayer EA, De Giorgio R, Sternini C. Expression of the Bitter Taste Receptor, T2R38, in Enteroendocrine Cells of the Colonic Mucosa of Overweight/Obese vs. Lean Subjects. PLoS One 2016; 11:e0147468. [PMID: 26866366 PMCID: PMC4750998 DOI: 10.1371/journal.pone.0147468] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/03/2016] [Indexed: 12/12/2022] Open
Abstract
Bitter taste receptors (T2Rs) are expressed in the mammalian gastrointestinal mucosa. In the mouse colon, T2R138 is localized to enteroendocrine cells and is upregulated by long-term high fat diet that induces obesity. The aims of this study were to test whether T2R38 expression is altered in overweight/obese (OW/OB) compared to normal weight (NW) subjects and characterize the cell types expressing T2R38, the human counterpart of mouse T2R138, in human colon. Colonic mucosal biopsies were obtained during colonoscopy from 35 healthy subjects (20 OW/OB and 15 NW) and processed for quantitative RT-PCR and immunohistochemistry using antibodies to T2R38, chromogranin A (CgA), glucagon like peptide-1 (GLP-1), cholecystokinin (CCK), or peptide YY (PYY). T2R38 mRNA levels in the colonic mucosa of OW/OB were increased (> 2 fold) compared to NW subjects but did not reach statistical significance (P = 0.06). However, the number of T2R38 immunoreactive (IR) cells was significantly increased in OW/OB vs. NW subjects (P = 0.01) and was significantly correlated with BMI values (r = 0.7557; P = 0.001). In both OW/OB and NW individuals, all T2R38-IR cells contained CgA-IR supporting they are enteroendocrine. In both groups, T2R38-IR colocalized with CCK-, GLP1- or PYY-IR. The overall CgA-IR cell population was comparable in OW/OB and NW individuals. This study shows that T2R38 is expressed in distinct populations of enteroendocrine cells in the human colonic mucosa and supports T2R38 upregulation in OW/OB subjects. T2R38 might mediate host functional responses to increased energy balance and intraluminal changes occurring in obesity, which could involve peptide release from enteroendocrine cells.
Collapse
Affiliation(s)
- Rocco Latorre
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Huynh
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maurizio Mazzoni
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - Arpana Gupta
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Elena Bonora
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Clavenzani
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - Lin Chang
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Emeran A. Mayer
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Roberto De Giorgio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Catia Sternini
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
45
|
Bauer PV, Hamr SC, Duca FA. Regulation of energy balance by a gut-brain axis and involvement of the gut microbiota. Cell Mol Life Sci 2016; 73:737-55. [PMID: 26542800 PMCID: PMC11108299 DOI: 10.1007/s00018-015-2083-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 12/11/2022]
Abstract
Despite significant progress in understanding the homeostatic regulation of energy balance, successful therapeutic options for curbing obesity remain elusive. One potential target for the treatment of obesity is via manipulation of the gut-brain axis, a complex bidirectional communication system that is crucial in maintaining energy homeostasis. Indeed, ingested nutrients induce secretion of gut peptides that act either via paracrine signaling through vagal and non-vagal neuronal relays, or in an endocrine fashion via entry into circulation, to ultimately signal to the central nervous system where appropriate responses are generated. We review here the current hypotheses of nutrient sensing mechanisms of enteroendocrine cells, including the release of gut peptides, mainly cholecystokinin, glucagon-like peptide-1, and peptide YY, and subsequent gut-to-brain signaling pathways promoting a reduction of food intake and an increase in energy expenditure. Furthermore, this review highlights recent research suggesting this energy regulating gut-brain axis can be influenced by gut microbiota, potentially contributing to the development of obesity.
Collapse
Affiliation(s)
- Paige V Bauer
- Department of Medicine, Toronto General Research Institute, UHN, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sophie C Hamr
- Department of Medicine, Toronto General Research Institute, UHN, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frank A Duca
- Department of Medicine, Toronto General Research Institute, UHN, Toronto, ON, M5G 1L7, Canada.
- MaRS Centre, Toronto Medical Discovery Tower, Room 10-701H, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
46
|
Spreckley E, Murphy KG. The L-Cell in Nutritional Sensing and the Regulation of Appetite. Front Nutr 2015; 2:23. [PMID: 26258126 PMCID: PMC4507148 DOI: 10.3389/fnut.2015.00023] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/06/2015] [Indexed: 12/25/2022] Open
Abstract
The gastrointestinal (GI) tract senses the ingestion of food and responds by signaling to the brain to promote satiation and satiety. Representing an important part of the gut-brain axis, enteroendocrine L-cells secrete the anorectic peptide hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) in response to the ingestion of food. The release of GLP-1 has multiple effects, including the secretion of insulin from pancreatic β-cells, decreased gastric emptying, and increased satiation. PYY also slows GI motility and reduces food intake. At least part of the gut-brain response seems to be due to direct sensing of macronutrients by L-cells, by mechanisms including specific nutrient-sensing receptors. Such receptors may represent possible pathways to target to decrease appetite and increase energy expenditure. Designing drugs or functional foods to exploit the machinery of these nutrient-sensing mechanisms may offer a potential approach for agents to treat obesity and metabolic disease.
Collapse
Affiliation(s)
- Eleanor Spreckley
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital , London , UK
| | - Kevin Graeme Murphy
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital , London , UK
| |
Collapse
|
47
|
Abstract
The gastrointestinal hormone peptide tyrosine tyrosine 3-36 (PYY(3-36)) has attained broad recognition with respect to its involvement in energy homeostasis and the control of food intake. It is mainly secreted by distal intestinal enteroendocrine L-cells in response to eating and exerts neurally mediated, paracrine and endocrine effects on various target organs. In addition to its gastrointestinal effects, PYY(3-36) has long been known to inhibit food intake. Recent closer examination of the effects of PYY(3-36) revealed that this gut-derived peptide also influences a wide spectrum of behavioral and cognitive functions that are pivotal for basic processes of perception and judgment, including central information processing, salience learning, working memory, and behavioral responding to novelty. Here, we review the effects of PYY(3-36) that go beyond food intake and provide a conceptual framework suggesting that several apparently unrelated behavioral actions of PYY(3-36) may actually reflect different manifestations of modulating the central dopamine system.
Collapse
|
48
|
Henry KE, Elfers CT, Burke RM, Chepurny OG, Holz GG, Blevins JE, Roth CL, Doyle RP. Vitamin B12 conjugation of peptide-YY(3-36) decreases food intake compared to native peptide-YY(3-36) upon subcutaneous administration in male rats. Endocrinology 2015; 156:1739-49. [PMID: 25658456 PMCID: PMC4398759 DOI: 10.1210/en.2014-1825] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Challenges to peptide-based therapies include rapid clearance, ready degradation by hydrolysis/proteolysis, and poor intestinal uptake and/or a need for blood brain barrier transport. This work evaluates the efficacy of conjugation of vitamin B12 (B12) on sc administered peptide tyrosine tyrosine (PYY)(3-36) function. In the current experiments, a B12-PYY(3-36) conjugate was tested against native PYY(3-36), and an inactive conjugate B12-PYYC36 (null control) in vitro and in vivo. In vitro experiments demonstrated similar agonism for the neuropeptide Y2 receptor by the B12-PYY(3-36) conjugate (EC50 26.5 nM) compared with native PYY(3-36) (EC50 16.0 nM), with the null control having an EC50 of 1.8 μM. In vivo experiments were performed in young adult male Sprague Dawley rats (9 wk). Daily treatments were delivered sc in five 1-hour pulses, each pulse delivering 5-10 nmol/kg, by implanted microinfusion pumps. Increases in hindbrain Fos expression were comparable 90 minutes after B12-PYY(3-36) or PYY3-36 injection relative to saline or B12-PYYC36. Food intake was reduced during a 5-day treatment for both B12-PYY(3-36)- (24%, P = .001) and PYY(3-36)-(13%, P = .008) treated groups relative to baseline. In addition, reduction of food intake after the three dark cycle treatment pulses was more consistent with B12-PYY(3-36) treatment (-26%, -29%, -27%) compared with the PYY(3-36) treatment (-3%, -21%, -16%), and B12-PYY(3-36) generated a significantly longer inhibition of food intake vs. PYY(3-36) treatment after the first two pulses (P = .041 and P = .036, respectively). These findings demonstrate a stronger, more consistent, and longer inhibition of food intake after the pulses of B12-PYY(3-36) conjugate compared with the native PYY(3-36).
Collapse
Affiliation(s)
- Kelly E Henry
- Department of Chemistry (K.E.H., R.M.B., R.P.D.), Center for Science and Technology, Syracuse University, Syracuse, New York 13244; Center for Integrative Brain Research (C.T.E., C.L.R.), Division of Endocrinology, Seattle Children's Research Institute, Seattle, Washington 98101; Departments of Medicine (O.G.C., G.G.H., R.P.D.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Research and Development Service (J.E.B.), Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108; Department of Medicine (J.E.B.), Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, Washington 98195; and Division of Endocrinology (C.L.R.), Department of Pediatrics, University of Washington, Seattle, Washington 98105
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Loh K, Herzog H, Shi YC. Regulation of energy homeostasis by the NPY system. Trends Endocrinol Metab 2015; 26:125-35. [PMID: 25662369 DOI: 10.1016/j.tem.2015.01.003] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 01/01/2023]
Abstract
Obesity develops when energy intake exceeds energy expenditure over time. Numerous neurotransmitters, hormones, and factors have been implicated to coordinately control energy homeostasis, centrally and peripherally. However, the neuropeptide Y (NPY) system has emerged as the one with the most critical functions in this process. While NPY centrally promotes feeding and reduces energy expenditure, peptide YY (PYY) and pancreatic polypeptide (PP), the other family members, mediate satiety. Importantly, recent research has uncovered additional functions for these peptides that go beyond the simple feeding/satiety circuits and indicate a more extensive function in controlling energy homeostasis. In this review, we will discuss the actions of the NPY system in the regulation of energy balance, with a particular focus on energy expenditure.
Collapse
Affiliation(s)
- Kim Loh
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia.
| | - Yan-Chuan Shi
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia.
| |
Collapse
|
50
|
Park HK, Ahima RS. Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism 2015; 64:24-34. [PMID: 25199978 PMCID: PMC4267898 DOI: 10.1016/j.metabol.2014.08.004] [Citation(s) in RCA: 428] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/01/2014] [Accepted: 08/08/2014] [Indexed: 12/24/2022]
Abstract
Leptin is secreted by adipose tissue and regulates energy homeostasis, neuroendocrine function, metabolism, immune function and other systems through its effects on the central nervous system and peripheral tissues. Leptin administration has been shown to restore metabolic and neuroendocrine abnormalities in individuals with leptin-deficient states, including hypothalamic amenorrhea and lipoatrophy. In contrast, obese individuals are resistant to leptin. Recombinant leptin is beneficial in patients with congenital leptin deficiency or generalized lipodystrophy. However, further research on molecular mediators of leptin resistance is needed for the development of targeted leptin sensitizing therapies for obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Hyeong-Kyu Park
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Republic of Korea
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes and Metabolism, and the Institute for Diabetes, Obesity and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|