1
|
Hankir MK, Lutz TA. Novel neural pathways targeted by GLP-1R agonists and bariatric surgery. Pflugers Arch 2025; 477:171-185. [PMID: 39644359 PMCID: PMC11761532 DOI: 10.1007/s00424-024-03047-3] [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: 10/17/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
The glucagon-like peptide 1 receptor (GLP-1R) agonist semaglutide has revolutionized the treatment of obesity, with other gut hormone-based drugs lined up that show even greater weight-lowering ability in obese patients. Nevertheless, bariatric surgery remains the mainstay treatment for severe obesity and achieves unparalleled weight loss that generally stands the test of time. While their underlying mechanisms of action remain incompletely understood, it is clear that the common denominator between GLP-1R agonists and bariatric surgery is that they suppress food intake by targeting the brain. In this Review, we highlight recent preclinical studies using contemporary neuroscientific techniques that provide novel concepts in the neural control of food intake and body weight with reference to endogenous GLP-1, GLP-1R agonists, and bariatric surgery. We start in the periphery with vagal, intestinofugal, and spinal sensory nerves and then progress through the brainstem up to the hypothalamus and finish at non-canonical brain feeding centers such as the zona incerta and lateral septum. Further defining the commonalities and differences between GLP-1R agonists and bariatric surgery in terms of how they target the brain may not only help bridge the gap between pharmacological and surgical interventions for weight loss but also provide a neural basis for their combined use when each individually fails.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Yang K, Wu YT, He Y, Dai JX, Luo YL, Xie JH, Ding WJ. GLP-1 and IL-6 regulates obesity in the gut and brain. Life Sci 2025; 362:123339. [PMID: 39730038 DOI: 10.1016/j.lfs.2024.123339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Obesity is a chronic metabolic disease characterized by excessive nutrient intake leading to increased subcutaneous or visceral fat, resulting in pathological and physiological changes. The incidence rate of obesity, an important form of metabolic syndrome, is increasing worldwide. Excess appetite is a key pathogenesis of obesity, and the inflammatory response induced by obesity has received increasing attention. This review focuses on the role of appetite-regulating factor (Glucogan-like peptide 1) and inflammatory factor (Interleukin-6) in the gut and brain in individuals with obesity and draws insights from the current literature.
Collapse
Affiliation(s)
- Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ting Wu
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin-Xiu Dai
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yu-Lu Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing-Hui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei-Jun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
3
|
Turcano P, Savica R, Benarroch E. What Is the Role of Glucagon-Like Peptide 1 Signaling in the Nervous System and Its Potential Neuroprotective Effects? Neurology 2024; 103:e209781. [PMID: 39079072 DOI: 10.1212/wnl.0000000000209781] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 01/24/2025] Open
|
4
|
Nakashima M, Suga N, Yoshikawa S, Matsuda S. Caveolae with GLP-1 and NMDA Receptors as Crossfire Points for the Innovative Treatment of Cognitive Dysfunction Associated with Neurodegenerative Diseases. Molecules 2024; 29:3922. [PMID: 39203005 PMCID: PMC11357136 DOI: 10.3390/molecules29163922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Some neurodegenerative diseases may be characterized by continuing behavioral and cognitive dysfunction that encompasses memory loss and/or apathy. Alzheimer's disease is the most typical type of such neurodegenerative diseases that are characterized by deficits of cognition and alterations of behavior. Despite the huge efforts against Alzheimer's disease, there has yet been no successful treatment for this disease. Interestingly, several possible risk genes for cognitive dysfunction are frequently expressed within brain cells, which may also be linked to cholesterol metabolism, lipid transport, exosomes, and/or caveolae formation, suggesting that caveolae may be a therapeutic target for cognitive dysfunctions. Interestingly, the modulation of autophagy/mitophagy with the alteration of glucagon-like peptide-1 (GLP-1) and N-methyl-d-aspartate (NMDA) receptor signaling may offer a novel approach to preventing and alleviating cognitive dysfunction. A paradigm showing that both GLP-1 and NMDA receptors at caveolae sites may be promising and crucial targets for the treatment of cognitive dysfunctions has been presented here, which may also be able to modify the progression of Alzheimer's disease. This research direction may create the potential to move clinical care toward disease-modifying treatment strategies with maximal benefits for patients without detrimental adverse events for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan (N.S.)
| |
Collapse
|
5
|
Ferrario CR, Münzberg-Gruening H, Rinaman L, Betley JN, Borgland SL, Dus M, Fadool DA, Medler KF, Morton GJ, Sandoval DA, de La Serre CB, Stanley SA, Townsend KL, Watts AG, Maruvada P, Cummings D, Cooke BM. Obesity- and diet-induced plasticity in systems that control eating and energy balance. Obesity (Silver Spring) 2024; 32:1425-1440. [PMID: 39010249 PMCID: PMC11269035 DOI: 10.1002/oby.24060] [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: 01/30/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 07/17/2024]
Abstract
In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.
Collapse
Grants
- P30 DK048520 NIDDK NIH HHS
- NSF1949989 National Science Foundation
- T32 DC000044 NIDCD NIH HHS
- R01 DK133464 NIDDK NIH HHS
- R01 DK089056 NIDDK NIH HHS
- R01 DK130246 NIDDK NIH HHS
- R01 DK124801 NIDDK NIH HHS
- R01 DK100685 NIDDK NIH HHS
- R01 DK124238 NIDDK NIH HHS
- R01 DK130875 NIDDK NIH HHS
- R01 DK125890 NIDDK NIH HHS
- Z99 DK999999 Intramural NIH HHS
- R01 DK124461 NIDDK NIH HHS
- K26 DK138368 NIDDK NIH HHS
- R01 DK121995 NIDDK NIH HHS
- R01 DK121531 NIDDK NIH HHS
- P30 DK089503 NIDDK NIH HHS
- P01 DK119130 NIDDK NIH HHS
- R01 DK118910 NIDDK NIH HHS
- R01 AT011683 NCCIH NIH HHS
- Reported research was supported by DK130246, DK092587, AT011683, MH059911, DK100685, DK119130, DK124801, DK133399, AG079877, DK133464, T32DC000044, F31DC016817, NSF1949989, DK089056, DK124238, DK138368, DK121995, DK125890, DK118910, DK121531, DK124461, DK130875; Canada Research Chair: 950-232211, CIHRFDN148473, CIHRPJT185886; USDA Predoctoral Fellowship; Endowment from the Robinson Family and Tallahassee Memorial Hospital; Department of Defense W81XWH-20-1-0345 and HT9425-23-1-0244; American Diabetes Association #1-17-ACE-31; W.M. Keck Foundation Award; National Science Foundation CAREER 1941822
- R01 DK133399 NIDDK NIH HHS
- HT9425-23-1-0244 Department of Defense
- R01 DK092587 NIDDK NIH HHS
- W81XWH-20-1-0345 Department of Defense
- 1941822 National Science Foundation
- R01 MH059911 NIMH NIH HHS
- F31 DC016817 NIDCD NIH HHS
- R01 AG079877 NIA NIH HHS
- P30 DK017047 NIDDK NIH HHS
Collapse
Affiliation(s)
- Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Heike Münzberg-Gruening
- Laboratory of Central Leptin Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Debra A Fadool
- Department of Biological Science, Program in Neuroscience, Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Kathryn F Medler
- School of Animal Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute at South Lake Union, Seattle, Washington, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Alan G Watts
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Padma Maruvada
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diana Cummings
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Bradley M Cooke
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
7
|
Geisler CE, Décarie-Spain L, Loh MK, Trumbauer W, Gaisinsky J, Klug ME, Pelletier C, Davis JF, Schmidt HD, Roitman MF, Kanoski SE, Hayes MR. Amylin Modulates a Ventral Tegmental Area-to-Medial Prefrontal Cortex Circuit to Suppress Food Intake and Impulsive Food-Directed Behavior. Biol Psychiatry 2024; 95:938-950. [PMID: 37517705 DOI: 10.1016/j.biopsych.2023.07.011] [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: 02/14/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND A better understanding of the neural mechanisms regulating impaired satiety to palatable foods is essential to treat hyperphagia linked with obesity. The satiation hormone amylin signals centrally at multiple nuclei including the ventral tegmental area (VTA). VTA-to-medial prefrontal cortex (mPFC) projections encode food reward information to influence behaviors including impulsivity. We hypothesized that modulation of VTA-to-mPFC neurons underlies amylin-mediated decreases in palatable food-motivated behaviors. METHODS We used a variety of pharmacological, behavioral, genetic, and viral approaches (n = 4-16/experiment) to investigate the anatomical and functional circuitry of amylin-controlled VTA-to-mPFC signaling in rats. RESULTS To first establish that VTA amylin receptor (calcitonin receptor) activation can modulate mPFC activity, we showed that intra-VTA amylin decreased food-evoked mPFC cFos. VTA amylin delivery also attenuated food-directed impulsive behavior, implicating VTA amylin signaling as a regulator of mPFC functions. Palatable food activates VTA dopamine and mPFC neurons. Accordingly, dopamine receptor agonism in the mPFC blocked the hypophagic effect of intra-VTA amylin, and VTA amylin injection reduced food-evoked phasic dopamine levels in the mPFC, supporting the idea that VTA calcitonin receptor activation decreases dopamine release in the mPFC. Surprisingly, calcitonin receptor expression was not found on VTA-to-mPFC projecting neurons but was instead found on GABAergic (gamma-aminobutyric acidergic) interneurons in the VTA that provide monosynaptic inputs to this pathway. Blocking intra-VTA GABA signaling, through GABA receptor antagonists and DREADD (designer receptor exclusively activated by designer drugs)-mediated GABAergic neuronal silencing, attenuated intra-VTA amylin-induced hypophagia. CONCLUSIONS These results indicate that VTA amylin signaling stimulates GABA-mediated inhibition of dopaminergic projections to the mPFC to mitigate impulsive consumption of palatable foods.
Collapse
Affiliation(s)
- Caroline E Geisler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Léa Décarie-Spain
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, California
| | - Maxine K Loh
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois
| | - Wolf Trumbauer
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jane Gaisinsky
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Molly E Klug
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, California
| | - Caitlyn Pelletier
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jon F Davis
- Novo Nordisk Research Center Seattle, Seattle, Washington
| | - Heath D Schmidt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mitchell F Roitman
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, California
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
8
|
Valyear MD, Brown A, Deyab G, Villaruel FR, Lahlou S, Caporicci-Dinucci N, Chaudhri N. Augmenting glutamatergic, but not dopaminergic, activity in the nucleus accumbens shell disrupts responding to a discrete alcohol cue in an alcohol context. Eur J Neurosci 2024; 59:1500-1518. [PMID: 38185906 DOI: 10.1111/ejn.16231] [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: 09/08/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024]
Abstract
Discrete alcohol cues and contexts are relapse triggers for people with alcohol use disorder exerting particularly powerful control over behaviour when they co-occur. Here, we investigated the neural substrates subserving the capacity for alcohol-associated contexts to elevate responding to an alcohol-predictive conditioned stimulus (CS). Specifically, rats were trained in a distinct 'alcohol context' to respond by entering a fluid port during a discrete auditory CS that predicted the delivery of alcohol and were familiarized with a 'neutral context' wherein alcohol was never available. When conditioned CS responding was tested by presenting the CS without alcohol, we found that augmenting glutamatergic activity in the nucleus accumbens (NAc) shell by microinfusing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) reduced responding to an alcohol CS in an alcohol, but not neutral, context. Further, AMPA microinfusion robustly affected behaviour, attenuating the number, duration and latency of CS responses selectively in the alcohol context. Although dopaminergic inputs to the NAc shell were previously shown to be necessary for CS responding in an alcohol context, here, chemogenetic excitation of ventral tegmental area (VTA) dopamine neurons and their inputs to the NAc shell did not affect CS responding. Critically, chemogenetic excitation of VTA dopamine neurons affected feeding behaviour and elevated c-fos immunoreactivity in the VTA and NAc shell, validating the chemogenetic approach. These findings enrich our understanding of the substrates underlying Pavlovian responding for alcohol and reveal that the capacity for contexts to modulate responding to discrete alcohol cues is delicately underpinned by the NAc shell.
Collapse
Affiliation(s)
- Milan D Valyear
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Alexa Brown
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Ghislaine Deyab
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Franz R Villaruel
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Soraya Lahlou
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nina Caporicci-Dinucci
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Aoun L, Almardini S, Saliba F, Haddadin F, Mourad O, Jdaidani J, Morcos Z, Al Saidi I, Bou Sanayeh E, Saliba S, Almardini M, Zaidan J. GLP-1 receptor agonists: A novel pharmacotherapy for binge eating (Binge eating disorder and bulimia nervosa)? A systematic review. J Clin Transl Endocrinol 2024; 35:100333. [PMID: 38449772 PMCID: PMC10915596 DOI: 10.1016/j.jcte.2024.100333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/04/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Objective Systematically review evidence on using GLP-1RAs for reducing BEB in BED and BN. Methods Comprehensive literature search (PubMed and Google Scholar) conducted for studies evaluating GLP-1Ras for BEB. Extracted data on study characteristics, efficacy, and safety. Results Studies show that GLP-1RAs (liraglutide and dulaglutide) reduce BE frequency and comorbidities in addition to favorable psychiatric side effect profile compared to current options. However, large-scale, blinded placebo-controlled trials are lacking. Conclusion Early findings suggest promising effects of GLP-1RAs on BEB. However, rigorous clinical trials are needed to firmly establish efficacy, dosing, safety, and comparative effectiveness before considering GLP-1RAs a viable novel approach.
Collapse
Affiliation(s)
- Laurence Aoun
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Shaza Almardini
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Fares Saliba
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Fadi Haddadin
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Omar Mourad
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Jennifer Jdaidani
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Zeina Morcos
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Ibrahim Al Saidi
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Elie Bou Sanayeh
- Department of Internal Medicine, Staten Island University Hospital, United States
| | - Saliba Saliba
- Department of Physiology, Mcgill University, United States
| | - Michel Almardini
- Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Julie Zaidan
- Endocrinology, Diabetes and Metabolism, Staten Island University Hospital, United States
| |
Collapse
|
10
|
Sun HZ, Shen FS, Li XX, Liu C, Xue Y, Han XH, Chen XY, Chen L. Exendin-4 increases the firing activity of hippocampal CA1 neurons through TRPC4/5 channels. Neurosci Res 2024; 199:48-56. [PMID: 37595875 DOI: 10.1016/j.neures.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
The central neuropeptide GLP-1 is synthesized by preproglucagon (PPG) neurons in the brain. GLP-1 receptors are widely distributed in central nervous system. Hippocampus is a key component of the limbic system which is involved in learning, memory, and cognition. Previous studies have shown that overexpression of GLP-1 receptors in the hippocampus could improve the process of learning and memory. However, up to now, the direct electrophysiological effects and possible molecular mechanisms of GLP-1 in hippocampal CAl neurons remain unexplored. The present study aims to evaluate the effects and mechanisms of GLP-1 on the spontaneous firing activity of hippocampal CAl neurons. Employing multibarrel single-unit extracellular recordings, the present study showed that micro-pressure administration of GLP-1 receptor agonist, exendin-4, significantly increased the spontaneous firing rate of hippocampal CA1 neurons in rats. Furthermore, application of the specific GLP-1 receptor antagonist, exendin(9-39), alone significantly decreased the firing rate of CA1 neurons, suggesting that endogenous GLP-1 modulates the firing activity of CA1 neurons. Co-application of exendin(9-39) completely blocked exendin-4-induced excitation of hippocampal CA1 neurons. Finally, the present study demonstrated for the first time that the transient receptor potential canonical 4 (TRPC4)/TRPC5 channels may be involved in exendin-4-induced excitation. The present studies may provide a rationale for further investigation of the modulation of GLP-1 on learning and memory as well as its possible involvement in Alzheimer's disease.
Collapse
Affiliation(s)
- Hui-Zhe Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fang-Shuai Shen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiao-Xue Li
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Cui Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Xue
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiao-Hua Han
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xin-Yi Chen
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
11
|
Bakoyiannis I, Ducourneau EG, Parkes SL, Ferreira G. Pathway specific interventions reveal the multiple roles of ventral hippocampus projections in cognitive functions. Rev Neurosci 2023; 34:825-838. [PMID: 37192533 DOI: 10.1515/revneuro-2023-0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/28/2023] [Indexed: 05/18/2023]
Abstract
Since the 1950s study of Scoville and Milner on the case H.M., the hippocampus has attracted neuroscientists' attention. The hippocampus has been traditionally divided into dorsal and ventral parts, each of which projects to different brain structures and mediates various functions. Despite a predominant interest in its dorsal part in animal models, especially regarding episodic-like and spatial cognition, recent data highlight the role of the ventral hippocampus (vHPC), as the main hippocampal output, in cognitive processes. Here, we review recent studies conducted in rodents that have used advanced in vivo functional techniques to specifically monitor and manipulate vHPC efferent pathways and delineate the roles of these specific projections in learning and memory processes. Results highlight that vHPC projections to basal amygdala are implicated in emotional memory, to nucleus accumbens in social memory and instrumental actions and to prefrontal cortex in all the above as well as in object-based memory. Some of these hippocampal projections also modulate feeding and anxiety-like behaviours providing further evidence that the "one pathway-one function" view is outdated and future directions are proposed to better understand the role of hippocampal pathways and shed further light on its connectivity and function.
Collapse
Affiliation(s)
- Ioannis Bakoyiannis
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| | - Eva-Gunnel Ducourneau
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| | - Shauna L Parkes
- University of Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Guillaume Ferreira
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| |
Collapse
|
12
|
Huang J, Wang C, Zhang HB, Zheng H, Huang T, Di JZ. Neuroimaging and neuroendocrine insights into food cravings and appetite interventions in obesity. PSYCHORADIOLOGY 2023; 3:kkad023. [PMID: 38666104 PMCID: PMC10917384 DOI: 10.1093/psyrad/kkad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 04/28/2024]
Abstract
This article reviews the previous studies on the distinction between food cravings and appetite, and how they are regulated by hormones and reflected in brain activity. Based on existing research, food cravings are defined as individual preferences influenced by hormones and psychological factors, which differ from appetite, as they are not necessarily related to hunger or nutritional needs. The article also evaluates the neuroimaging findings about food cravings, and interventions to reduce food cravings, such as mindfulness training, alternative sweeteners, non-invasive brain stimulation techniques, cognitive-behavioral therapy, and imaginal retraining, and points out their advantages, disadvantages, and limitations. Furthermore, the article delves into the potential future directions in the field, emphasizing the need for a neuroendocrine perspective, considerations for associated psychiatric disorders, innovative clinical interventions, and emerging therapeutic frontiers in obesity management. The article outlines the neuro-endocrine basis of food cravings, including ghrelin, leptin, melanocortin, oxytocin, glucagon-like peptide-1, baclofen, and other hormones and their brain regions of action. The article argues that food cravings are an important target for obesity, and more research is needed to explore their complex characteristics and mechanisms, and how to effectively interact with their neuro-endocrine pathways. The article provides a new perspective and approach to the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Jin Huang
- Department of Metabolic & Bariatric Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chen Wang
- Department of Metabolic & Bariatric Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Hang-Bin Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Centre for Mental Disorders, Shanghai Mental Health Centre, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Hui Zheng
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Centre for Mental Disorders, Shanghai Mental Health Centre, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Tao Huang
- Xuhui Health Care Commission, Shanghai 200030, China
| | - Jian-Zhong Di
- Department of Metabolic & Bariatric Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
13
|
Décarie-Spain L, Gu C, Lauer LT, Subramanian KS, Chehimi SN, Kao AE, Deng I, Bashaw AG, Klug ME, Galbokke AH, Donohue KN, Yang M, de Lartigue G, Myers KP, Crist RC, Reiner BC, Hayes MR, Kanoski SE. Ventral hippocampus neurons encode meal-related memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561731. [PMID: 37873229 PMCID: PMC10592790 DOI: 10.1101/2023.10.10.561731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The ability to encode and retrieve meal-related information is critical to efficiently guide energy acquisition and consumption, yet the underlying neural processes remain elusive. Here we reveal that ventral hippocampus (HPCv) neuronal activity dynamically elevates during meal consumption and this response is highly predictive of subsequent performance in a foraging-related spatial memory task. Targeted recombination-mediated ablation of HPCv meal-responsive neurons impairs foraging-related spatial memory without influencing food motivation, anxiety-like behavior, or escape-mediated spatial memory. These HPCv meal-responsive neurons project to the lateral hypothalamic area (LHA) and single-nucleus RNA sequencing and in situ hybridization analyses indicate they are enriched in serotonin 2a receptors (5HT2aR). Either chemogenetic silencing of HPCv-to-LHA projections or intra-HPCv 5HT2aR antagonist yielded foraging-related spatial memory deficits, as well as alterations in caloric intake and the temporal sequence of spontaneous meal consumption. Collective results identify a population of HPCv neurons that dynamically respond to eating to encode meal-related memories.
Collapse
Affiliation(s)
- Léa Décarie-Spain
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Cindy Gu
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Logan Tierno Lauer
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Keshav S. Subramanian
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States
| | - Samar N. Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Alicia E. Kao
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Iris Deng
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Alexander G. Bashaw
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States
| | - Molly E. Klug
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Ashyah Hewage Galbokke
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Kristen N. Donohue
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Mingxin Yang
- Monell Chemical Sense Center, Philadelphia, Pennsylvania, United States
| | | | - Kevin P. Myers
- Bucknell University, Lewisburg, Philadelphia, Pennsylvania, United States
| | - Richard C. Crist
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Benjamin C. Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Matthew R. Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Scott E. Kanoski
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States
| |
Collapse
|
14
|
Schoenfeld TJ, Rhee D, Smith JA, Padmanaban V, Brockett AT, Jacobs HN, Cameron HA. Rewarded Maze Training Increases Approach Behavior in Rats Through Neurogenesis-Dependent Growth of Ventral Hippocampus-Prelimbic Circuits. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:725-733. [PMID: 37881563 PMCID: PMC10593943 DOI: 10.1016/j.bpsgos.2023.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 10/27/2023] Open
Abstract
Background Learning complex navigation routes increases hippocampal volume in humans, but it is not clear whether this growth impacts behaviors outside the learning situation or what cellular mechanisms are involved. Methods We trained rats with pharmacogenetic suppression of adult neurogenesis and littermate controls in 3 mazes over 3 weeks and tested novelty approach behavior several days after maze exposure. We then measured hippocampus and prelimbic cortex volumes using magnetic resonance imaging and assessed neuronal and astrocyte morphology. Finally, we investigated the activation and behavioral role of the ventral CA1 (vCA1)-to-prelimbic pathway using immediate-early genes and DREADDs (designer receptors exclusively activated by designer drugs). Results Maze training led to volume increase of both the vCA1 region of the hippocampus and the prelimbic region of the neocortex compared with rats that followed fixed paths. Growth was also apparent in individual neurons and astrocytes in these 2 regions, and behavioral testing showed increased novelty approach in maze-trained rats in 2 different tests. Suppressing adult neurogenesis prevented the effects on structure and approach behavior after maze training without affecting maze learning itself. The vCA1 neurons projecting to the prelimbic area were more activated by novelty in maze-trained animals, and suppression of this pathway decreased approach behavior. Conclusions Rewarded navigational learning experiences induce volumetric and morphologic growth in the vCA1 and prelimbic cortex and enhance activation of the circuit connecting these 2 regions. Both the structural and behavioral effects of maze training require ongoing adult neurogenesis, suggesting a role for new neurons in experience-driven increases in novelty exploration.
Collapse
Affiliation(s)
- Timothy J. Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, Tennessee
| | - Diane Rhee
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jesse A. Smith
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Varun Padmanaban
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Adam T. Brockett
- Department of Psychology and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Hannah N. Jacobs
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Heather A. Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
15
|
Cerroni C, Steiner A, Seanez L, Kwon S, Lewis AS. Effects of repeated developmental GLP-1R agonist exposure on young adult behavior and hippocampal structure in mice. Neurosci Lett 2023; 808:137299. [PMID: 37196974 PMCID: PMC10330515 DOI: 10.1016/j.neulet.2023.137299] [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: 04/20/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/19/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are common type 2 diabetes medications that have been repurposed for adult chronic weight management. Clinical trials suggest this class may also be beneficial for obesity in pediatric populations. Since several GLP-1R agonists cross the blood-brain barrier, it is important to understand how postnatal developmental exposure to GLP-1R agonists might affect brain structure and function later in life. Toward that end, we systemically treated male and female C57BL/6 mice with the GLP-1R agonist exendin-4 (0.5 mg/kg, twice daily) or saline from postnatal day 14 to 21, then allowed uninterrupted development to young adulthood. Beginning at 7 weeks of age, we performed open field and marble burying tests to assess motor behavior and the spontaneous location recognition (SLR) task to assess hippocampal-dependent pattern separation and memory. Mice were sacrificed, and we counted ventral hippocampal mossy cells, as we have recently shown that most murine hippocampal neuronal GLP-1R is expressed in this cell population. We found that GLP-1R agonist treatment did not alter P14-P21 weight gain, but modestly reduced young adult open field distance traveled and marble burying. Despite these motor changes, there was no effect on SLR memory performance or time spent investigating objects. Finally, we did not detect any changes in ventral mossy cell number using two different markers. These data suggest developmental exposure to GLP-1R agonists might have specific rather than global effects on behavior later in life and that extensive additional study is necessary to clarify how drug timing and dose affect distinct constellations of behavior in young adulthood.
Collapse
Affiliation(s)
- Catherine Cerroni
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alex Steiner
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leann Seanez
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sam Kwon
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan S Lewis
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
16
|
Díaz-Megido C, Thomsen M. Sex-dependent divergence in the effects of GLP-1 agonist exendin-4 on alcohol reinforcement and reinstatement in C57BL/6J mice. Psychopharmacology (Berl) 2023; 240:1287-1298. [PMID: 37106129 PMCID: PMC10172234 DOI: 10.1007/s00213-023-06367-x] [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: 10/04/2022] [Accepted: 04/10/2023] [Indexed: 04/29/2023]
Abstract
RATIONALE Alcohol use disorder remains a leading cause of preventable deaths, and current treatments have limited efficacy. Glucagon-like peptide 1 (GLP-1) receptor agonists can reduce alcohol drinking in preclinical studies, but mechanisms are still not fully understood, and data in female subjects are scarce. OBJECTIVES To assess whether the GLP-1 receptor agonist exendin-4 could decrease alcohol-seeking behavior in the absence of alcohol consumption or intoxication, to compare the potency and efficacy of exendin-4 in the reduction of alcohol seeking vs. alcohol taking, and to compare effects between male and female mice. METHODS Male and female C57BL/6J mice were trained to self-administer 20% alcohol under an FR 1 schedule of reinforcement. After extinction, systemic exendin-4 (saline, 1.8, and 3.2 μg/kg) was tested in cue-induced reinstatement of alcohol seeking. Effects of exendin-4 on alcohol self-administration were tested in a separate group. RESULTS Exendin-4 suppressed reinstatement of alcohol seeking to extinction levels, at both doses, in the male mice, but had no effect in the female mice. Both doses of exendin-4 also significantly decreased alcohol self-administration in male mice; females again showed less pronounced effects. CONCLUSIONS In male mice, exendin-4 appeared more effective at suppressing alcohol seeking in the absence of alcohol relative to alcohol self-administration, consistent with modulation of alcohol reward or inhibitory control, rather than satiety or aversive effects of alcohol. We saw marked sex differences with less effect of exendin-4 in female mice, and it will be important to include both sexes in further investigations into GLP-1 receptor agonists.
Collapse
Affiliation(s)
- Claudia Díaz-Megido
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark.
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
17
|
Cerroni C, Steiner A, Seanez L, Kwon S, Lewis AS. Effects of repeated developmental GLP-1R agonist exposure on adult behavior and hippocampal structure in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537724. [PMID: 37131808 PMCID: PMC10153236 DOI: 10.1101/2023.04.21.537724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are common type 2 diabetes medications that have been repurposed for adult chronic weight management. Clinical trials suggest this class may also be beneficial for obesity in pediatric populations. Since several GLP-1R agonists cross the blood-brain barrier, it is important to understand how postnatal developmental exposure to GLP-1R agonists might affect brain structure and function in adulthood. Toward that end, we systemically treated male and female C57BL/6 mice with the GLP-1R agonist exendin-4 (0.5 mg/kg, twice daily) or saline from postnatal day 14 to 21, then allowed uninterrupted development to adulthood. Beginning at 7 weeks of age, we performed open field and marble burying tests to assess motor behavior and the spontaneous location recognition (SLR) task to assess hippocampal-dependent pattern separation and memory. Mice were sacrificed, and we counted ventral hippocampal mossy cells, as we have recently shown that most murine hippocampal neuronal GLP-1R is expressed in this cell population. We found that GLP-1R agonist treatment did not alter P14-P21 weight gain, but modestly reduced adult open field distance traveled and marble burying. Despite these motor changes, there was no effect on SLR memory performance or time spent investigating objects. Finally, we did not detect any changes in ventral mossy cell number using two different markers. These data suggest developmental exposure to GLP-1R agonists might have specific rather than global effects on behavior later in life and that extensive additional study is necessary to clarify how drug timing and dose affect distinct constellations of behavior in adulthood.
Collapse
|
18
|
Singh U, Saito K, Khan MZ, Jiang J, Toth BA, Rodeghiero SR, Dickey JE, Deng Y, Deng G, Kim YC, Cui H. Collateralizing ventral subiculum melanocortin 4 receptor circuits regulate energy balance and food motivation. Physiol Behav 2023; 262:114105. [PMID: 36736416 PMCID: PMC9981473 DOI: 10.1016/j.physbeh.2023.114105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Hippocampal dysfunction is associated with major depressive disorder, a serious mental illness characterized by not only depressed mood but also appetite disturbance and dysregulated body weight. However, the underlying mechanisms by which hippocampal circuits regulate metabolic homeostasis remain incompletely understood. Here we show that collateralizing melanocortin 4 receptor (MC4R) circuits in the ventral subiculum (vSUB), one of the major output structures of the hippocampal formation, affect food motivation and energy balance. Viral-mediated cell type- and projection-specific input-output circuit mapping revealed that the nucleus accumbens shell (NAcSh)-projecting vSUBMC4R+ neurons send extensive collateral projections of to various hypothalamic nuclei known to be important for energy balance, including the arcuate, ventromedial and dorsomedial nuclei, and receive monosynaptic inputs mainly from the ventral CA1 and the anterior paraventricular nucleus of thalamus. Chemogenetic activation of NAcSh-projecting vSUBMC4R+neurons lead to increase in motivation to obtain palatable food without noticeable effect on homeostatic feeding. Viral-mediated restoration of MC4R signaling in the vSUB partially restores obesity in MC4R-null mice without affecting anxiety- and depression-like behaviors. Collectively, these results delineate vSUBMC4R+ circuits to the unprecedented level of precision and identify the vSUBMC4R signaling as a novel regulator of food reward and energy balance.
Collapse
Affiliation(s)
- Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Michael Z. Khan
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A. Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Samuel R. Rodeghiero
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jacob E. Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Young-Cho Kim
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
19
|
Murphy S, Collis Glynn M, Dixon TN, Grill HJ, McNally GP, Ong ZY. Nucleus of the solitary tract A2 neurons control feeding behaviors via projections to the paraventricular hypothalamus. Neuropsychopharmacology 2023; 48:351-361. [PMID: 36114285 PMCID: PMC9751294 DOI: 10.1038/s41386-022-01448-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 12/26/2022]
Abstract
Hindbrain NTS neurons are highly attuned to internal physiological and external environmental factors that contribute to the control of food intake but the relevant neural phenotypes and pathways remain elusive. Here, we investigated the role of NTS A2 neurons and their projections in the control of feeding behaviors. In male TH Cre rats, we first confirmed selective targeting of NTS A2 neurons and showed that chemogenetic stimulation of these neurons significantly suppressed dark cycle food intake, deprivation re-feed and high fat diet intake. Despite reducing intake, activation of NTS A2 neurons had no effect on food approach, anxiety-like behaviors, locomotor activity, blood glucose levels nor did it induce nausea/malaise, thus revealing a selective role for these neurons in the consummatory aspect of food intake control. Pathway-specific mapping and manipulation of NTS A2 neurons showed that these effects were mediated by NTS A2 neurons projecting to the paraventricular nucleus of the hypothalamus (PVH) because chemogenetic activation of these projections, but not projections to bed nucleus of the stria terminalis (BNST), reduced food intake. Cell-type specific analyses demonstrated that activation of NTS A2 neurons recruited both PVH oxytocin (OT)- and corticotropin-releasing factor (CRF)-expressing neurons, and plasma analyses showed increased plasma corticosterone following NTS A2 stimulation. While we also showed that chemogenetic inhibition of NTS A2 neurons attenuated the intake inhibitory effects of CCK, the specificity of transgene expression was low. Together, these findings showed that NTS A2 neurons are sufficient to control the consummatory aspects of feeding, regardless of energy status or food palatability and identified their projections to PVH, but not BNST, in food intake control.
Collapse
Affiliation(s)
- Stephanie Murphy
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Metika Collis Glynn
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Tiarani N Dixon
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gavan P McNally
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia
| | - Zhi Yi Ong
- School of Psychology, University of New South Wales, UNSW, Sydney, NSW, 2052, Australia.
| |
Collapse
|
20
|
Steiner A, Owen BM, Bauer JP, Seanez L, Kwon S, Biddinger JE, Huffman R, Ayala JE, Nobis WP, Lewis AS. Glucagon-like peptide-1 receptor differentially controls mossy cell activity across the dentate gyrus longitudinal axis. Hippocampus 2022; 32:797-807. [PMID: 36063105 PMCID: PMC9675713 DOI: 10.1002/hipo.23469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/27/2022] [Accepted: 08/18/2022] [Indexed: 01/07/2023]
Abstract
Understanding the role of dentate gyrus (DG) mossy cells (MCs) in learning and memory has rapidly evolved due to increasingly precise methods for targeting MCs and for in vivo recording and activity manipulation in rodents. These studies have shown MCs are highly active in vivo, strongly remap to contextual manipulation, and that their inhibition or hyperactivation impairs pattern separation and location or context discrimination. Less well understood is how MC activity is modulated by neurohormonal mechanisms, which might differentially control the participation of MCs in cognitive functions during discrete states, such as hunger or satiety. In this study, we demonstrate that glucagon-like peptide-1 (GLP-1), a neuropeptide produced in the gut and the brain that regulates food consumption and hippocampal-dependent mnemonic function, might regulate MC function through expression of its receptor, GLP-1R. RNA-seq demonstrated that most, though not all, Glp1r in hippocampal principal neurons is expressed in MCs, and in situ hybridization revealed strong expression of Glp1r in hilar neurons. Glp1r-ires-Cre mice crossed with Ai14D reporter mice followed by co-labeling for the MC marker GluR2/3 revealed that almost all MCs in the ventral DG expressed Glp1r and that almost all Glp1r-expressing hilar neurons were MCs. However, only ~60% of dorsal DG MCs expressed Glp1r, and Glp1r was also expressed in small hilar neurons that were not MCs. Consistent with this expression pattern, peripheral administration of the GLP-1R agonist exendin-4 (5 μg/kg) increased cFos expression in ventral but not dorsal DG hilar neurons. Finally, whole-cell patch-clamp recordings from ventral MCs showed that bath application of exendin-4 (200 nM) depolarized MCs and increased action potential firing. Taken together, this study adds to known MC activity modulators a neurohormonal mechanism that may preferentially affect ventral DG physiology and may potentially be targetable by several GLP-1R pharmacotherapies already in clinical use.
Collapse
Affiliation(s)
- Alex Steiner
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin M. Owen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James P. Bauer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leann Seanez
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sam Kwon
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jessica E. Biddinger
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ragan Huffman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julio E. Ayala
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - William P. Nobis
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan S. Lewis
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
21
|
Décarie-Spain L, Liu CM, Lauer LT, Subramanian K, Bashaw AG, Klug ME, Gianatiempo IH, Suarez AN, Noble EE, Donohue KN, Cortella AM, Hahn JD, Davis EA, Kanoski SE. Ventral hippocampus-lateral septum circuitry promotes foraging-related memory. Cell Rep 2022; 40:111402. [PMID: 36170832 PMCID: PMC9605732 DOI: 10.1016/j.celrep.2022.111402] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/27/2022] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Remembering the location of a food or water source is essential for survival. Here, we reveal that spatial memory for food location is reflected in ventral hippocampus (HPCv) neuron activity and is impaired by HPCv lesion. HPCv mediation of foraging-related memory involves communication to the lateral septum (LS), as either reversible or chronic disconnection of HPCv-to-LS signaling impairs spatial memory retention for food or water location. This neural pathway selectively encodes appetitive spatial memory, as HPCv-LS disconnection does not affect spatial memory for escape location in a negative reinforcement procedure, food intake, or social and olfactory-based appetitive learning. Neural pathway tracing and functional mapping analyses reveal that LS neurons recruited during the appetitive spatial memory procedure are primarily GABAergic neurons that project to the lateral hypothalamus. Collective results emphasize that the neural substrates controlling spatial memory are outcome specific based on reinforcer modality.
Collapse
Affiliation(s)
- Léa Décarie-Spain
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Clarissa M Liu
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA
| | - Logan Tierno Lauer
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Keshav Subramanian
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA
| | - Alexander G Bashaw
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA
| | - Molly E Klug
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Isabella H Gianatiempo
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Andrea N Suarez
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, 305 Sanford Drive, Athens, GA 30602, USA
| | - Kristen N Donohue
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Alyssa M Cortella
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Joel D Hahn
- Neurobiology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Elizabeth A Davis
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA
| | - Scott E Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, 3616 Trousdale Pkwy, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, University of Southern California, 3641Watt Way, Los Angeles, CA 90089, USA.
| |
Collapse
|
22
|
Abstract
The modern obesogenic environment contains an abundance of food cues (e.g., sight, smell of food) as well cues that are associated with food through learning and memory processes. Food cue exposure can lead to food seeking and excessive consumption in otherwise food-sated individuals, and a high level of food cue responsivity is a risk factor for overweight and obesity. Similar food cue responses are observed in experimental rodent models, and these models are therefore useful for mechanistically identifying the neural circuits mediating food cue responsivity. This review draws from both experimental rodent models and human data to characterize the behavioral and biological processes through which food-associated stimuli contribute to overeating and weight gain. Two rodent models are emphasized - cue-potentiated feeding and Pavlovian-instrumental transfer - that provide insight in the neural circuits and peptide systems underlying food cue responsivity. Data from humans are highlighted that reveal physiological, psychological, and neural mechanisms that connect food cue responsivity with overeating and weight gain. The collective literature identifies connections between heightened food cue responsivity and obesity in both rodents and humans, and identifies underlying brain regions (nucleus accumbens, amygdala, orbitofrontal cortex, hippocampus) and endocrine systems (ghrelin) that regulate food cue responsivity in both species. These species similarities are encouraging for the possibility of mechanistic rodent model research and further human research leading to novel treatments for excessive food cue responsivity in humans.
Collapse
Affiliation(s)
- Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Kerri N Boutelle
- Department of Pediatrics, Herbert Wertheim School of Public Health and Human Longevity Science, and Psychiatry, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
23
|
Azevedo EP, Ivan VJ, Friedman JM, Stern SA. Higher-Order Inputs Involved in Appetite Control. Biol Psychiatry 2022; 91:869-878. [PMID: 34593204 PMCID: PMC9704062 DOI: 10.1016/j.biopsych.2021.07.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 01/01/2023]
Abstract
The understanding of the neural control of appetite sheds light on the pathogenesis of eating disorders such as anorexia nervosa and obesity. Both diseases are a result of maladaptive eating behaviors (overeating or undereating) and are associated with life-threatening health problems. The fine regulation of appetite involves genetic, physiological, and environmental factors, which are detected and integrated in the brain by specific neuronal populations. For centuries, the hypothalamus has been the center of attention in the scientific community as a key regulator of appetite. The hypothalamus receives and sends axonal projections to several other brain regions that are important for the integration of sensory and emotional information. These connections ensure that appropriate behavioral decisions are made depending on the individual's emotional state and environment. Thus, the mechanisms by which higher-order brain regions integrate exteroceptive information to coordinate feeding is of great importance. In this review, we will focus on the functional and anatomical projections connecting the hypothalamus to the limbic system and higher-order brain centers in the cortex. We will also address the mechanisms by which specific neuronal populations located in higher-order centers regulate appetite and how maladaptive eating behaviors might arise from altered connections among cortical and subcortical areas with the hypothalamus.
Collapse
Affiliation(s)
- Estefania P Azevedo
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York.
| | - Violet J Ivan
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York; Howard Hughes Medical Institute, New York, New York
| | - Sarah A Stern
- Integrative Neural Circuits and Behavior Research Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida.
| |
Collapse
|
24
|
Hones VI, Mizumori SJY. Response Flexibility: The Role of the Lateral Habenula. Front Behav Neurosci 2022; 16:852235. [PMID: 35444521 PMCID: PMC9014270 DOI: 10.3389/fnbeh.2022.852235] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/01/2022] [Indexed: 01/13/2023] Open
Abstract
The ability to make appropriate decisions that result in an optimal outcome is critical for survival. This process involves assessing the environment as well as integrating prior knowledge about the environment with information about one's current internal state. There are many neural structures that play critical roles in mediating these processes, but it is not yet known how such information coalesces to influence behavioral output. The lateral habenula (LHb) has often been cited as a structure critical for adaptive and flexible responding when environmental contexts and internal state changes. A challenge, however, has been understanding how LHb promotes response flexibility. In this review, we hypothesize that the LHb enables flexible responding following the integration of context memory and internal state information by signaling downstream brainstem structures known to drive hippocampal theta. In this way, animals respond more flexibly in a task situation not because the LHb selects a particular action, but rather because LHb enhances a hippocampal neural state that is often associated with greater attention, arousal, and exploration. In freely navigating animals, these are essential conditions that are needed to discover and implement appropriate alternative choices and behaviors. As a corollary to our hypothesis, we describe short- and intermediate-term functions of the LHb. Finally, we discuss the effects on the behavior of LHb dysfunction in short- and intermediate-timescales, and then suggest that new therapies may act on the LHb to alleviate the behavioral impairments following long-term LHb disruption.
Collapse
Affiliation(s)
- Victoria I. Hones
- Department of Psychology, University of Washington, Seattle, WA, United States
| | - Sheri J. Y. Mizumori
- Department of Psychology, University of Washington, Seattle, WA, United States
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| |
Collapse
|
25
|
Liao RM, Pattij T. Neural basis of operant behaviors maintained on the differential-reinforcement-of-low-rate (DRL) schedule in rodents. Brain Res Bull 2022; 185:1-17. [DOI: 10.1016/j.brainresbull.2022.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/02/2022] [Accepted: 04/08/2022] [Indexed: 12/20/2022]
|
26
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
27
|
Cui QN, Stein LM, Fortin SM, Hayes MR. The role of glia in the physiology and pharmacology of glucagon-like peptide-1: implications for obesity, diabetes, neurodegeneration and glaucoma. Br J Pharmacol 2022; 179:715-726. [PMID: 34519040 PMCID: PMC8820182 DOI: 10.1111/bph.15683] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 11/28/2022] Open
Abstract
The medical applications of glucagon-like peptide-1 receptor (GLP-1R) agonists is evergrowing in scope, highlighting the urgent need for a comprehensive understanding of the mechanisms through which GLP-1R activation impacts physiology and behaviour. A new area of research aims to elucidate the role GLP-1R signalling in glia, which play a role in regulating energy balance, glycemic control, neuroinflammation and oxidative stress. Once controversial, existing evidence now suggests that subsets of glia (e.g. microglia, tanycytes and astrocytes) and infiltrating macrophages express GLP-1Rs. In this review, we discuss the implications of these findings, with particular focus on the effectiveness of both clinically available and novel GLP-1R agonists for treating metabolic and neurodegenerative diseases, enhancing cognition and combating substance abuse. LINKED ARTICLES: This article is part of a themed issue on GLP1 receptor ligands (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.4/issuetoc.
Collapse
Affiliation(s)
- Qi N. Cui
- Scheie Eye InstitutePhiladelphiaPennsylvaniaUSA
| | - Lauren M. Stein
- Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Samantha M. Fortin
- Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Matthew R. Hayes
- Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
28
|
Weight loss did not modify macronutrient specific response of hormones and satiety in overweight and obese people without metabolic disease – results from a clinical trial. Clin Nutr 2022; 41:948-957. [DOI: 10.1016/j.clnu.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 11/18/2022]
|
29
|
Chen XY, Chen L, Yang W, Xie AM. GLP-1 Suppresses Feeding Behaviors and Modulates Neuronal Electrophysiological Properties in Multiple Brain Regions. Front Mol Neurosci 2022; 14:793004. [PMID: 34975402 PMCID: PMC8718614 DOI: 10.3389/fnmol.2021.793004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022] Open
Abstract
The glucagon-like peptide-1 (GLP-1) plays important roles in the regulation of food intake and energy metabolism. Peripheral or central GLP-1 suppresses food intake and reduces body weight. The electrophysiological properties of neurons in the mammalian central nervous system reflect the neuronal excitability and the functional organization of the brain. Recent studies focus on elucidating GLP-1-induced suppression of feeding behaviors and modulation of neuronal electrophysiological properties in several brain regions. Here, we summarize that activation of GLP-1 receptor (GLP-1R) suppresses food intake and induces postsynaptic depolarization of membrane potential and/or presynaptic modulation of glutamatergic or GABAergic neurotransmission in brain nuclei located within the medulla oblongata, pons, mesencephalon, diencephalon, and telencephalon. This review may provide a background to guide future research about the cellular mechanisms of GLP-1-induced feeding inhibition.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wu Yang
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| | - An-Mu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
30
|
Parent MB, Higgs S, Cheke LG, Kanoski SE. Memory and eating: A bidirectional relationship implicated in obesity. Neurosci Biobehav Rev 2022; 132:110-129. [PMID: 34813827 PMCID: PMC8816841 DOI: 10.1016/j.neubiorev.2021.10.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 01/03/2023]
Abstract
This paper reviews evidence demonstrating a bidirectional relationship between memory and eating in humans and rodents. In humans, amnesia is associated with impaired processing of hunger and satiety cues, disrupted memory of recent meals, and overconsumption. In healthy participants, meal-related memory limits subsequent ingestive behavior and obesity is associated with impaired memory and disturbances in the hippocampus. Evidence from rodents suggests that dorsal hippocampal neural activity contributes to the ability of meal-related memory to control future intake, that endocrine and neuropeptide systems act in the ventral hippocampus to provide cues regarding energy status and regulate learned aspects of eating, and that consumption of hypercaloric diets and obesity disrupt these processes. Collectively, this evidence indicates that diet-induced obesity may be caused and/or maintained, at least in part, by a vicious cycle wherein excess intake disrupts hippocampal functioning, which further increases intake. This perspective may advance our understanding of how the brain controls eating, the neural mechanisms that contribute to eating-related disorders, and identify how to treat diet-induced obesity.
Collapse
Affiliation(s)
- Marise B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, Box 5030, Atlanta, GA 30303-5030, United States.
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, BI5 2TT, United Kingdom.
| | - Lucy G Cheke
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB, United Kingdom.
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, 90089-0371, United States.
| |
Collapse
|
31
|
Smith KR, Moran TH. Gastrointestinal peptides in eating-related disorders. Physiol Behav 2021; 238:113456. [PMID: 33989649 PMCID: PMC8462672 DOI: 10.1016/j.physbeh.2021.113456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Food intake is tightly controlled by homeostatic signals sensitive to metabolic need for the regulation of body weight. This review focuses on the peripherally-secreted gastrointestinal peptides (i.e., ghrelin, cholecystokinin, glucagon-like peptide 1, and peptide tyrosine tyrosine) that contribute to the control of appetite and discusses how these peptides or the signals arising from their release are disrupted in eating-related disorders across the weight spectrum, namely anorexia nervosa, bulimia nervosa, and obesity, and whether they are normalized following weight restoration or weight loss treatment. Further, the role of gut peptides in the pathogenesis and treatment response in human weight conditions as identified by rodent models are discussed. Lastly, we review the incretin- and hormone-based pharmacotherapies available for the treatment of obesity and eating-related disorders.
Collapse
Affiliation(s)
- Kimberly R Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
32
|
Ruze R, Xu Q, Liu G, Li Y, Chen W, Cheng Z, Xiong Y, Liu S, Zhang G, Hu S, Yan Z. Central GLP-1 contributes to improved cognitive function and brain glucose uptake after duodenum-jejunum bypass on obese and diabetic rats. Am J Physiol Endocrinol Metab 2021; 321:E392-E409. [PMID: 34370593 DOI: 10.1152/ajpendo.00126.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
The improvement of cognitive function following bariatric surgery has been highlighted, yet its underlying mechanisms remain elusive. Finding the improved brain glucose uptake of patients after Roux-en-Y gastric bypass (RYGB), duodenum-jejunum bypass (DJB), and sham surgery (Sham) were performed on obese and diabetic Wistar rats, and intracerebroventricular (ICV) injection of glucagon-like peptide-1 (GLP-1) analog liraglutide (Lira), antagonist exendin-(9-39) (Exe-9), and the viral-mediated GLP-1 receptor (Glp-1r) knockdown (KD) were applied on both groups to elucidate the role of GLP-1 in mediating cognitive function and brain glucose uptake assessed with the Morris water maze (MWM) and positron emission tomography (PET). Insulin and GLP-1 in serum and cerebral spinal fluid (CSF) were measured, and the expression of glucose uptake-related proteins including glucose transporter 1 (GLUT-1), GLUT-4, phospho-Akt substrate of 160kDa (pAS160), AS160, Rab10, Myosin-Va as well as the c-fos marker in the brain were examined. Along with augmented glucose homeostasis following DJB, central GLP-1 was correlated with the improved cognitive function and ameliorated brain glucose uptake, which was further confirmed by the enhancive role of Lira on both groups whereas the Exe-9 and Glp-1r KD were opposite. Known to activate insulin-signaling pathways, central GLP-1 contributes to improved cognitive function and brain glucose uptake after DJB.NEW & NOTEWORTHY The improvement of cognitive function following bariatric surgery has been highlighted while its mechanisms remain elusive. The brain glucose uptake of patients was improved after RYGB, and the DJB and sham surgery performed on obese and diabetic Wistar rats revealed that the elevated central GLP-1 contributes to the dramatic improvement of cognitive function, brain glucose uptake, transport, glucose sensing, and neuronal activation.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qian Xu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, People's Republic of China
| | - Guoqin Liu
- Department of General Surgery, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Yuekai Li
- Department of Nuclear Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Weijie Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhiqiang Cheng
- Department of Colorectal Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Yacheng Xiong
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, People's Republic of China
| | - Shaozhuang Liu
- Department of Bariatric and Metabolic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China
| | - Sanyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China
| | - Zhibo Yan
- Department of Colorectal Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| |
Collapse
|
33
|
Guerrero-Hreins E, Goldstone AP, Brown RM, Sumithran P. The therapeutic potential of GLP-1 analogues for stress-related eating and role of GLP-1 in stress, emotion and mood: a review. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110303. [PMID: 33741445 DOI: 10.1016/j.pnpbp.2021.110303] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/19/2021] [Accepted: 03/09/2021] [Indexed: 01/11/2023]
Abstract
Stress and low mood are powerful triggers for compulsive overeating, a maladaptive form of eating leading to negative physical and mental health consequences. Stress-vulnerable individuals, such as people with obesity, are particularly prone to overconsumption of high energy foods and may use it as a coping mechanism for general life stressors. Recent advances in the treatment of obesity and related co-morbidities have focused on the therapeutic potential of anorexigenic gut hormones, such as glucagon-like peptide 1 (GLP-1), which acts both peripherally and centrally to reduce energy intake. Besides its appetite suppressing effect, GLP-1 acts on areas of the brain involved in stress response and emotion regulation. However, the role of GLP-1 in emotion and stress regulation, and whether it is a viable treatment for stress-induced compulsive overeating, has yet to be established. A thorough review of the pre-clinical literature measuring markers of stress, anxiety and mood after GLP-1 exposure points to potential divergent effects based on temporality. Specifically, acute GLP-1 injection consistently stimulates the physiological stress response in rodents whereas long-term exposure indicates anxiolytic and anti-depressive benefits. However, the limited clinical evidence is not as clear cut. While prolonged GLP-1 analogue treatment in people with type 2 diabetes improved measures of mood and general psychological wellbeing, the mechanisms underlying this may be confounded by associated weight loss and improved blood glucose control. There is a paucity of longitudinal clinical literature on mechanistic pathways by which stress influences eating behavior and how centrally-acting gut hormones such as GLP-1, can modify these. (250).
Collapse
Affiliation(s)
- Eva Guerrero-Hreins
- The Florey Institute of Neuroscience and Mental Health, Mental Health Theme, Parkville, Melbourne, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia; PsychoNeuroEndocrinology Research Group, Centre for Neuropsychopharmacology, Division of Psychiatry, and Computational, Cognitive and Clinical Neuroimaging Laboratory, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Centre for Neuropsychopharmacology, Division of Psychiatry, and Computational, Cognitive and Clinical Neuroimaging Laboratory, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Robyn M Brown
- The Florey Institute of Neuroscience and Mental Health, Mental Health Theme, Parkville, Melbourne, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Priya Sumithran
- Department of Medicine (St Vincent's), University of Melbourne, Victoria, Australia; Dept. of Endocrinology, Austin Health, Victoria, Australia.
| |
Collapse
|
34
|
Neural mechanisms underlying the role of fructose in overfeeding. Neurosci Biobehav Rev 2021; 128:346-357. [PMID: 34182019 DOI: 10.1016/j.neubiorev.2021.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022]
Abstract
Fructose consumption has been linked with metabolic syndrome and obesity. Fructose-based sweeteners like high fructose corn syrup taste sweeter, improve food palatability, and are increasingly prevalent in our diet. The increase in fructose consumption precedes the rise in obesity and is a contributing driver to the obesity epidemic worldwide. The role of dietary fructose in obesity can be multifactorial by promoting visceral adiposity, hypertension, and insulin resistance. Interestingly, one emergent finding from human and animal studies is that dietary fructose promotes overfeeding. As the brain is a critical regulator of food intake, we reviewed the evidence that fructose can act in the brain and elucidated the major brain systems underlying fructose-induced overfeeding. We found that fructose acts on multiple interdependent brain systems to increase orexigenic drive and the incentive salience of food while decreasing the latency between food bouts and reducing cognitive control to disinhibit feeding. We concluded that the collective actions of fructose may promote feeding behavior by producing a hunger-like state in the brain.
Collapse
|
35
|
Briggs SB, Hannapel R, Ramesh J, Parent MB. Inhibiting ventral hippocampal NMDA receptors and Arc increases energy intake in male rats. ACTA ACUST UNITED AC 2021; 28:187-194. [PMID: 34011515 PMCID: PMC8139633 DOI: 10.1101/lm.053215.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/02/2021] [Indexed: 11/24/2022]
Abstract
Research into the neural mechanisms that underlie higher-order cognitive control of eating behavior suggests that ventral hippocampal (vHC) neurons, which are critical for emotional memory, also inhibit energy intake. We showed previously that optogenetically inhibiting vHC glutamatergic neurons during the early postprandial period, when the memory of the meal would be undergoing consolidation, caused rats to eat their next meal sooner and to eat more during that next meal when the neurons were no longer inhibited. The present research determined whether manipulations known to interfere with synaptic plasticity and memory when given pretraining would increase energy intake when given prior to ingestion. Specifically, we tested the effects of blocking vHC glutamatergic N-methyl-D-aspartate receptors (NMDARs) and activity-regulated cytoskeleton-associated protein (Arc) on sucrose ingestion. The results showed that male rats consumed a larger sucrose meal on days when they were given vHC infusions of the NMDAR antagonist APV or Arc antisense oligodeoxynucleotides than on days when they were given control infusions. The rats did not accommodate for that increase by delaying the onset of their next sucrose meal (i.e., decreased satiety ratio) or by eating less during the next meal. These data suggest that vHC NMDARs and Arc limit meal size and inhibit meal initiation.
Collapse
Affiliation(s)
- Sherri B Briggs
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
| | - Reilly Hannapel
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
| | - Janavi Ramesh
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
| | - Marise B Parent
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA.,Department of Psychology, Georgia State University, Atlanta, Georgia 30303, USA
| |
Collapse
|
36
|
Williams DL. The diverse effects of brain glucagon-like peptide 1 receptors on ingestive behaviour. Br J Pharmacol 2021; 179:571-583. [PMID: 33990944 DOI: 10.1111/bph.15535] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/12/2021] [Accepted: 05/07/2021] [Indexed: 12/31/2022] Open
Abstract
Glucagon-like peptide 1 (GLP-1) is well known as a gut hormone and also acts as a neuropeptide, produced in a discrete population of caudal brainstem neurons that project widely throughout the brain. GLP-1 receptors are expressed in many brain areas of relevance to energy balance, and stimulation of these receptors at many of these sites potently suppresses food intake. This review surveys the current evidence for effects mediated by GLP-1 receptors on feeding behaviour at a wide array of brain sites and discusses behavioural and neurophysiological mechanisms for the effects identified thus far. Taken together, it is clear that GLP-1 receptor activity in the brain can influence feeding by diverse means, including mediation of gastrointestinal satiation and/or satiety signalling, suppression of motivation for food reward, induction of nausea and mediation of restraint stress-induced hypophagia, but many questions about the organization of this system remain.
Collapse
Affiliation(s)
- Diana L Williams
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
37
|
Vestlund J, Bergquist F, Licheri V, Adermark L, Jerlhag E. Activation of glucagon-like peptide-1 receptors and skilled reach foraging. Addict Biol 2021; 26:e12953. [PMID: 32770792 PMCID: PMC8244104 DOI: 10.1111/adb.12953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Glucagon‐like peptide‐1 receptor (GLP‐1R) agonists, such as exendin‐4 (Ex4), liraglutide and dulaglutide, regulate glucose homeostasis and are thus used to treat diabetes type II. GLP‐1 also contributes towards a variety of additional physiological functions, including suppression of reward and improvement of learning. Acute activation of GLP‐1R in the nucleus accumbens (NAc) shell, an area essential for motivation, reduces the motivation to consume sucrose or alcohol when assessed in a simple motor task. However, the effects of repeated administration of the different GLP‐1R agonists on behaviours in a more complex motor task are unknown. The aim was therefore to investigate the effects of repeated Ex4, liraglutide or dulaglutide on the motivation and learning of a complex motor tasks such as skilled reach foraging in the Montoya staircase test. To explore the neurophysiological correlates of the different GLP‐1R agonists on motivation, ex vivo electrophysiological recordings were conducted. In rats with an acquired skilled reach performance, Ex4 or liraglutide but not dulaglutide reduced the motivation of skilled reach foraging. In trained rats, Ex4 infusion into NAc shell decreased this motivated behaviour, and both Ex4 and liraglutide supressed the evoked field potentials in NAc shell. In rats without prior Montoya experience, dulaglutide but not Ex4 or liraglutide enhanced the learning of skilled reach foraging. Taken together, these findings indicate that the tested GLP‐1R agonists have different behavioural outcomes depending on the context.
Collapse
Affiliation(s)
- Jesper Vestlund
- Department of Pharmacology, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Filip Bergquist
- Department of Pharmacology, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Valentina Licheri
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| |
Collapse
|
38
|
Noble EE, Olson CA, Davis E, Tsan L, Chen YW, Schade R, Liu C, Suarez A, Jones RB, de La Serre C, Yang X, Hsiao EY, Kanoski SE. Gut microbial taxa elevated by dietary sugar disrupt memory function. Transl Psychiatry 2021; 11:194. [PMID: 33790226 PMCID: PMC8012713 DOI: 10.1038/s41398-021-01309-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 01/16/2023] Open
Abstract
Emerging evidence highlights a critical relationship between gut microbiota and neurocognitive development. Excessive consumption of sugar and other unhealthy dietary factors during early life developmental periods yields changes in the gut microbiome as well as neurocognitive impairments. However, it is unclear whether these two outcomes are functionally connected. Here we explore whether excessive early life consumption of added sugars negatively impacts memory function via the gut microbiome. Rats were given free access to a sugar-sweetened beverage (SSB) during the adolescent stage of development. Memory function and anxiety-like behavior were assessed during adulthood and gut bacterial and brain transcriptome analyses were conducted. Taxa-specific microbial enrichment experiments examined the functional relationship between sugar-induced microbiome changes and neurocognitive and brain transcriptome outcomes. Chronic early life sugar consumption impaired adult hippocampal-dependent memory function without affecting body weight or anxiety-like behavior. Adolescent SSB consumption during adolescence also altered the gut microbiome, including elevated abundance of two species in the genus Parabacteroides (P. distasonis and P. johnsonii) that were negatively correlated with hippocampal function. Transferred enrichment of these specific bacterial taxa in adolescent rats impaired hippocampal-dependent memory during adulthood. Hippocampus transcriptome analyses revealed that early life sugar consumption altered gene expression in intracellular kinase and synaptic neurotransmitter signaling pathways, whereas Parabacteroides microbial enrichment altered gene expression in pathways associated with metabolic function, neurodegenerative disease, and dopaminergic signaling. Collectively these results identify a role for microbiota "dysbiosis" in mediating the detrimental effects of early life unhealthy dietary factors on hippocampal-dependent memory function.
Collapse
Affiliation(s)
- Emily E. Noble
- grid.213876.90000 0004 1936 738XUniversity of Georgia, Athens, GA USA
| | - Christine A. Olson
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Elizabeth Davis
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Linda Tsan
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Yen-Wei Chen
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Ruth Schade
- grid.213876.90000 0004 1936 738XUniversity of Georgia, Athens, GA USA
| | - Clarissa Liu
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Andrea Suarez
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Roshonda B. Jones
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | | | - Xia Yang
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Elaine Y. Hsiao
- grid.19006.3e0000 0000 9632 6718University of California, Los Angeles, CA USA
| | - Scott E. Kanoski
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| |
Collapse
|
39
|
Decarie-Spain L, Kanoski SE. Ghrelin and Glucagon-Like Peptide-1: A Gut-Brain Axis Battle for Food Reward. Nutrients 2021; 13:977. [PMID: 33803053 PMCID: PMC8002922 DOI: 10.3390/nu13030977] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/14/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022] Open
Abstract
Eating behaviors are influenced by the reinforcing properties of foods that can favor decisions driven by reward incentives over metabolic needs. These food reward-motivated behaviors are modulated by gut-derived peptides such as ghrelin and glucagon-like peptide-1 (GLP-1) that are well-established to promote or reduce energy intake, respectively. In this review we highlight the antagonizing actions of ghrelin and GLP-1 on various behavioral constructs related to food reward/reinforcement, including reactivity to food cues, conditioned meal anticipation, effort-based food-motivated behaviors, and flavor-nutrient preference and aversion learning. We integrate physiological and behavioral neuroscience studies conducted in both rodents and human to illustrate translational findings of interest for the treatment of obesity or metabolic impairments. Collectively, the literature discussed herein highlights a model where ghrelin and GLP-1 regulate food reward-motivated behaviors via both competing and independent neurobiological and behavioral mechanisms.
Collapse
Affiliation(s)
- Lea Decarie-Spain
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA;
| | - Scott E. Kanoski
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA;
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
40
|
McLean BA, Wong CK, Campbell JE, Hodson DJ, Trapp S, Drucker DJ. Revisiting the Complexity of GLP-1 Action from Sites of Synthesis to Receptor Activation. Endocr Rev 2021; 42:101-132. [PMID: 33320179 PMCID: PMC7958144 DOI: 10.1210/endrev/bnaa032] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) is produced in gut endocrine cells and in the brain, and acts through hormonal and neural pathways to regulate islet function, satiety, and gut motility, supporting development of GLP-1 receptor (GLP-1R) agonists for the treatment of diabetes and obesity. Classic notions of GLP-1 acting as a meal-stimulated hormone from the distal gut are challenged by data supporting production of GLP-1 in the endocrine pancreas, and by the importance of brain-derived GLP-1 in the control of neural activity. Moreover, attribution of direct vs indirect actions of GLP-1 is difficult, as many tissue and cellular targets of GLP-1 action do not exhibit robust or detectable GLP-1R expression. Furthermore, reliable detection of the GLP-1R is technically challenging, highly method dependent, and subject to misinterpretation. Here we revisit the actions of GLP-1, scrutinizing key concepts supporting gut vs extra-intestinal GLP-1 synthesis and secretion. We discuss new insights refining cellular localization of GLP-1R expression and integrate recent data to refine our understanding of how and where GLP-1 acts to control inflammation, cardiovascular function, islet hormone secretion, gastric emptying, appetite, and body weight. These findings update our knowledge of cell types and mechanisms linking endogenous vs pharmacological GLP-1 action to activation of the canonical GLP-1R, and the control of metabolic activity in multiple organs.
Collapse
Affiliation(s)
- Brent A McLean
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Chi Kin Wong
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Jonathan E Campbell
- The Department of Medicine, Division of Endocrinology, Department of Pharmacology and Cancer Biology, Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| |
Collapse
|
41
|
Browning KN, Carson KE. Central Neurocircuits Regulating Food Intake in Response to Gut Inputs-Preclinical Evidence. Nutrients 2021; 13:nu13030908. [PMID: 33799575 PMCID: PMC7998662 DOI: 10.3390/nu13030908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/02/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of energy balance requires the complex integration of homeostatic and hedonic pathways, but sensory inputs from the gastrointestinal (GI) tract are increasingly recognized as playing critical roles. The stomach and small intestine relay sensory information to the central nervous system (CNS) via the sensory afferent vagus nerve. This vast volume of complex sensory information is received by neurons of the nucleus of the tractus solitarius (NTS) and is integrated with responses to circulating factors as well as descending inputs from the brainstem, midbrain, and forebrain nuclei involved in autonomic regulation. The integrated signal is relayed to the adjacent dorsal motor nucleus of the vagus (DMV), which supplies the motor output response via the efferent vagus nerve to regulate and modulate gastric motility, tone, secretion, and emptying, as well as intestinal motility and transit; the precise coordination of these responses is essential for the control of meal size, meal termination, and nutrient absorption. The interconnectivity of the NTS implies that many other CNS areas are capable of modulating vagal efferent output, emphasized by the many CNS disorders associated with dysregulated GI functions including feeding. This review will summarize the role of major CNS centers to gut-related inputs in the regulation of gastric function with specific reference to the regulation of food intake.
Collapse
|
42
|
Neuropeptidergic Control of Feeding: Focus on the Galanin Family of Peptides. Int J Mol Sci 2021; 22:ijms22052544. [PMID: 33802616 PMCID: PMC7961366 DOI: 10.3390/ijms22052544] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity/overweight are important health problems due to metabolic complications. Dysregulation of peptides exerting orexigenic/anorexigenic effects must be investigated in-depth to understand the mechanisms involved in feeding behaviour. One of the most important and studied orexigenic peptides is galanin (GAL). The aim of this review is to update the mechanisms of action and physiological roles played by the GAL family of peptides (GAL, GAL-like peptide, GAL message-associated peptide, alarin) in the control of food intake and to review the involvement of these peptides in metabolic diseases and food intake disorders in experimental animal models and humans. The interaction between GAL and NPY in feeding and energy metabolism, the relationships between GAL and other substances involved in food intake mechanisms, the potential pharmacological strategies to treat food intake disorders and obesity and the possible clinical applications will be mentioned and discussed. Some research lines are suggested to be developed in the future, such as studies focused on GAL receptor/neuropeptide Y Y1 receptor interactions in hypothalamic and extra-hypothalamic nuclei and sexual differences regarding the expression of GAL in feeding behaviour. It is also important to study the possible GAL resistance in obese individuals to better understand the molecular mechanisms by which GAL regulates insulin/glucose metabolism. GAL does not exert a pivotal role in weight regulation and food intake, but this role is crucial in fat intake and also exerts an important action by regulating the activity of other key compounds under conditions of stress/altered diet.
Collapse
|
43
|
Lord MN, Subramanian K, Kanoski SE, Noble EE. Melanin-concentrating hormone and food intake control: Sites of action, peptide interactions, and appetition. Peptides 2021; 137:170476. [PMID: 33370567 PMCID: PMC8025943 DOI: 10.1016/j.peptides.2020.170476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
Given the increased prevalence of obesity and its associated comorbidities, understanding the mechanisms through which the brain regulates energy balance is of critical importance. The neuropeptide melanin-concentrating hormone (MCH) is produced in the lateral hypothalamic area and the adjacent incerto-hypothalamic area and promotes both food intake and energy conservation, overall contributing to body weight gain. Decades of research into this system has provided insight into the neural pathways and mechanisms (behavioral and neurobiological) through which MCH stimulates food intake. Recent technological advancements that allow for selective manipulation of MCH neuron activity have elucidated novel mechanisms of action for the hyperphagic effects of MCH, implicating neural "volume" transmission in the cerebrospinal fluid and sex-specific effects of MCH on food intake control as understudied areas for future investigation. Highlighted here are historical and recent findings that illuminate the neurobiological mechanisms through which MCH promotes food intake, including the identification of various specific neural signaling pathways and interactions with other peptide systems. We conclude with a framework that the hyperphagic effects of MCH signaling are predominantly mediated through enhancement of an "appetition" process in which early postoral prandial signals promote further caloric consumption.
Collapse
Affiliation(s)
- Magen N Lord
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30606, USA
| | - Keshav Subramanian
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA; Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30606, USA.
| |
Collapse
|
44
|
Smith NK, Grueter BA. Hunger-driven adaptive prioritization of behavior. FEBS J 2021; 289:922-936. [PMID: 33630426 DOI: 10.1111/febs.15791] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/11/2021] [Accepted: 02/24/2021] [Indexed: 12/19/2022]
Abstract
In order to survive, an animal must adapt its behavioral priorities to accommodate changing internal and external conditions. Hunger, a universally recognized interoceptive signal, promotes food intake though increasingly well-understood neural circuits. Less understood, is how hunger is integrated into the neural computations that guide nonfeeding behaviors. Within the brain, agouti-related peptide neurons in the arcuate nucleus of the hypothalamus have been found to powerfully stimulate feeding in addition to mediating other hunger-driven behavioral phenotypes. In this review, we compile the behavioral plasticity downstream of hunger and present identified or potential molecular and neural circuit mechanisms. We catalogue hunger's ability to increase exploration, decrease anxiety, and alter social behavior, among other phenotypes. Finally, we suggest paths forward for understanding hunger-driven behavioral adaptation and discuss the benefits of understanding state-dependent modulation of neural circuits controlling behavior.
Collapse
Affiliation(s)
- Nicholas K Smith
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA
| | - Brad A Grueter
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
45
|
Escartín Pérez RE, Mancilla Díaz JM, Cortés Salazar F, López Alonso VE, Florán Garduño B. CB1/5-HT/GABA interactions and food intake regulation. PROGRESS IN BRAIN RESEARCH 2021; 259:177-196. [PMID: 33541676 DOI: 10.1016/bs.pbr.2021.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite historically the serotonergic, GABAergic, and cannabinoid systems have been shown to play a crucial role in the central regulation of eating behavior, interest in the study of the interactions of these neurotransmission systems has only now been investigated. Current evidence suggests that serotonin may influence normal and pathological eating behavior in significantly more complex ways than was initially thought. This knowledge has opened the possibility of exploring the potential clinical utility of new therapeutic strategies more effective and safer than the current approaches to treat pathological eating behavior. Furthermore, the nature and complexity of the interactions between these neurotransmitter systems have provided a better understanding of the pathophysiological mechanisms not only of eating behavior and eating disorders but also of some of the comorbidities associated with modulation of cortical circuits, which are involved in high order cognitive processes. Accordingly, in the present chapter, the clinical and experimental findings of the interactions between serotonin, GABA, and cannabinoids are synthesized, emphasizing the pharmacological, neurophysiological, and neuroanatomical aspects that could potentially improve the current therapeutic approaches against pathological eating behavior.
Collapse
Affiliation(s)
- Rodrigo Erick Escartín Pérez
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México.
| | - Juan Manuel Mancilla Díaz
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| | - Felipe Cortés Salazar
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| | - Verónica Elsa López Alonso
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| | - Benjamín Florán Garduño
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| |
Collapse
|
46
|
Eren-Yazicioglu CY, Yigit A, Dogruoz RE, Yapici-Eser H. Can GLP-1 Be a Target for Reward System Related Disorders? A Qualitative Synthesis and Systematic Review Analysis of Studies on Palatable Food, Drugs of Abuse, and Alcohol. Front Behav Neurosci 2021; 14:614884. [PMID: 33536884 PMCID: PMC7848227 DOI: 10.3389/fnbeh.2020.614884] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/15/2020] [Indexed: 11/15/2022] Open
Abstract
The role of glucagon-like peptide 1 (GLP-1) in insulin-dependent signaling is well-known; GLP-1 enhances glucose-dependent insulin secretion and lowers blood glucose in diabetes. GLP-1 receptors (GLP-1R) are also widely expressed in the brain, and in addition to its role in neuroprotection, it affects reward pathways. This systematic review aimed to analyze the studies on GLP-1 and reward pathways and its currently identified mechanisms. Methods: “Web of Science” and “Pubmed” were searched to identify relevant studies using GLP-1 as the keyword. Among the identified 26,539 studies, 30 clinical, and 71 preclinical studies were included. Data is presented by grouping rodent studies on palatable food intake, drugs of abuse, and studies on humans focusing on GLP-1 and reward systems. Results: GLP-1Rs are located in reward-related areas, and GLP-1, its agonists, and DPP-IV inhibitors are effective in decreasing palatable food intake, along with reducing cocaine, amphetamine, alcohol, and nicotine use in animals. GLP-1 modulates dopamine levels and glutamatergic neurotransmission, which results in observed behavioral changes. In humans, GLP-1 alters palatable food intake and improves activity deficits in the insula, hypothalamus, and orbitofrontal cortex (OFC). GLP-1 reduces food cravings partially by decreasing activity to the anticipation of food in the left insula of obese patients with diabetes and may inhibit overeating by increasing activity to the consumption of food in the right OFC of obese and left insula of obese with diabetes. Conclusion: Current preclinical studies support the view that GLP-1 can be a target for reward system related disorders. More translational research is needed to evaluate its efficacy on human reward system related disorders.
Collapse
Affiliation(s)
| | - Arya Yigit
- School of Medicine, Koç University, Istanbul, Turkey
| | - Ramazan Efe Dogruoz
- Department of Neuroscience, University of Chicago, Chicago, IL, United States
| | - Hale Yapici-Eser
- Koç University, Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Department of Psychiatry, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
47
|
Clasen MM, Riley AL, Davidson TL. Hippocampal-Dependent Inhibitory Learning and Memory Processes in the Control of Eating and Drug Taking. Curr Pharm Des 2020; 26:2334-2352. [PMID: 32026771 DOI: 10.2174/1381612826666200206091447] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
As manifestations of excessive and uncontrolled intake, obesity and drug addiction have generated much research aimed at identifying common neuroadaptations that could underlie both disorders. Much work has focused on changes in brain reward and motivational circuitry that can overexcite eating and drug-taking behaviors. We suggest that the regulation of both behaviors depends on balancing excitation produced by stimuli associated with food and drug rewards with the behavioral inhibition produced by physiological "satiety" and other stimuli that signal when those rewards are unavailable. Our main hypothesis is that dysregulated eating and drug use are consequences of diet- and drug-induced degradations in this inhibitory power. We first outline a learning and memory mechanism that could underlie the inhibition of both food and drug-intake, and we describe data that identifies the hippocampus as a brain substrate for this mechanism. We then present evidence that obesitypromoting western diets (WD) impair the operation of this process and generate pathophysiologies that disrupt hippocampal functioning. Next, we present parallel evidence that drugs of abuse also impair this same learning and memory process and generate similar hippocampal pathophysiologies. We also describe recent findings that prior WD intake elevates drug self-administration, and the implications of using drugs (i.e., glucagon-like peptide- 1 agonists) that enhance hippocampal functioning to treat both obesity and addiction are also considered. We conclude with a description of how both WD and drugs of abuse could initiate a "vicious-cycle" of hippocampal pathophysiology and impaired hippocampal-dependent behavioral inhibition.
Collapse
Affiliation(s)
- Matthew M Clasen
- Department of Psychology, Program in Neuroscience, Williams College, Williamstown, MA 01267, United States
| | - Anthony L Riley
- Department of Neuroscience, Center for Behavioral Neuroscience, American University, Washington, DC 20016, United States
| | - Terry L Davidson
- Department of Neuroscience, Center for Behavioral Neuroscience, American University, Washington, DC 20016, United States
| |
Collapse
|
48
|
Colvin KJ, Killen HS, Kanter MJ, Halperin MC, Engel L, Currie PJ. Brain Site-Specific Inhibitory Effects of the GLP-1 Analogue Exendin-4 on Alcohol Intake and Operant Responding for Palatable Food. Int J Mol Sci 2020; 21:ijms21249710. [PMID: 33352692 PMCID: PMC7766977 DOI: 10.3390/ijms21249710] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Approximately 14.4 million Americans are experiencing alcohol use disorder (AUD) and about two-thirds of people who experience drug addiction will relapse, highlighting the need to develop novel and effective treatments. Glucagon-like peptide-1 (GLP-1) is a peptide hormone implicated in the mesocorticolimbic reward system and has become a peptide of interest with respect to its putative inhibitory effects on drug reward. In order to further develop treatments for those diagnosed with AUD, the interplay between GLP-1 receptor signaling and ethanol consumption must be elucidated. In the present study, we investigated the ability of the GLP-1 analogue, exendin-4 (Ex-4), to alter alcohol intake and operant responding for sucrose pellets in order to further understand the role of this compound in mediating reward. We selected multiple sites throughout the prosencephalic and mesencephalic regions of the brain, where we directly administered various doses of Ex-4 to male Sprague Dawley rats. In alcohol investigations, we utilized a two-bottle choice intermittent access protocol. In separate groups of rats, we adopted an operant paradigm in order to examine the effect of Ex-4 on motivated responding for palatable food. Results indicated that GLP-1 receptor signaling effectively suppressed voluntary alcohol intake when injected into the ventral tegmental area (VTA), the accumbens core (NAcC) and shell (NAcS), the dorsomedial hippocampus (DMHipp), and the lateral hypothalamus (LH), which are all structures linked to brain reward mechanisms. The arcuate nucleus (ARcN) and the paraventricular nucleus (PVN) of the hypothalamus were unresponsive, as was the basolateral amygdala (BLA). However, Ex-4 treatment into the ArcN and PVN suppressed operant responding for sucrose pellets. In fact, the VTA, NAcC, NAcS, LH, and the DMHipp all showed comparable suppression of sucrose responding. Overall, our findings suggest that these central structures are implicated in brain reward circuitry, including alcohol and appetitive motivation, which may be mediated by GLP-1 receptor mechanisms. GLP-1, therefore, may play a critical role in modifying addictive behaviors via activation of multiple GLP-1 systems throughout the brain.
Collapse
|
49
|
Shinohara K, Yasoshima Y. Inactivation of the ventral hippocampus facilitates the attenuation of odor neophobia in rats. Behav Brain Res 2020; 401:113077. [PMID: 33345825 DOI: 10.1016/j.bbr.2020.113077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 11/20/2022]
Abstract
Food neophobia is a behavior observed in rodents involving reduced consumption of a novel food or drink. In the absence of negative post-ingestive consequences, consumption increases with exposure (attenuation of neophobia), which is seen as an associative safe memory. Olfaction and gustation are sensory modalities essential for the development of a food preference. However, little is known about the neural mechanisms underlying neophobia to a food-related odor stimulus. In the present study, we examined the effect of pharmacological inactivation of the ventral hippocampus (vHPC) on neophobia to orally consumed solutions in rats using muscimol, a gamma aminobutyric acid type A receptor agonist. Two different types of solutions, almond odor (benzaldehyde) and sweet taste (saccharin), were prepared. In the results, microinjections of muscimol into the bilateral vHPC before the first odor and taste exposures did not alter the neophobic reactions of the rats to each stimulus. However, in the second odor, but not taste, exposure, the muscimol-injected rats showed higher consumption in comparison to that observed in the control rats, suggesting that the vHPC inactivation facilitates the attenuation of odor neophobia. On the other hand, intra-vHPC muscimol microinjections after the first odor and taste exposures did not facilitate consumption at the second exposures. These results indicate that neural activations within vHPC during orally consuming a novel odor, but not taste, solution play an inhibitory role in the subsequent attenuation of neophobia.
Collapse
Affiliation(s)
- Keisuke Shinohara
- Division of Behavioral Physiology, Department of Behavioral Sciences, Graduate School of Human Sciences, Osaka University, 1-2 Yamadaoka, Suita, Osaka, Japan
| | - Yasunobu Yasoshima
- Division of Behavioral Physiology, Department of Behavioral Sciences, Graduate School of Human Sciences, Osaka University, 1-2 Yamadaoka, Suita, Osaka, Japan.
| |
Collapse
|
50
|
Naneix F, Bakoyiannis I, Santoyo-Zedillo M, Bosch-Bouju C, Pacheco-Lopez G, Coutureau E, Ferreira G. Chemogenetic silencing of hippocampus and amygdala reveals a double dissociation in periadolescent obesogenic diet-induced memory alterations. Neurobiol Learn Mem 2020; 178:107354. [PMID: 33276069 DOI: 10.1016/j.nlm.2020.107354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 11/30/2022]
Abstract
In addition to numerous metabolic comorbidities, obesity is associated with several adverse neurobiological outcomes, especially learning and memory alterations. Obesity prevalence is rising dramatically in youth and is persisting in adulthood. This is especially worrying since adolescence is a crucial period for the maturation of certain brain regions playing a central role in memory processes such as the hippocampus and the amygdala. We previously showed that periadolescent, but not adult, exposure to obesogenic high-fat diet (HFD) had opposite effects on hippocampus- and amygdala-dependent memory, impairing the former and enhancing the latter. However, the causal role of these two brain regions in periadolescent HFD-induced memory alterations remains unclear. Here, we first showed that periadolescent HFD induced long-term, but not short-term, object recognition memory deficits, specifically when rats were exposed to a novel context. Using chemogenetic approaches to inhibit targeted brain regions, we then demonstrated that recognition memory deficits are dependent on the activity of the ventral hippocampus, but not the basolateral amygdala. On the contrary, the HFD- induced enhancement of conditioned odor aversion specifically requires amygdala activity. Taken together, these findings suggest that HFD consumption throughout adolescence impairs long-term object recognition memory through alterations of ventral hippocampal activity during memory acquisition. Moreover, these results further highlight the bidirectional effects of adolescent HFD on hippocampal and amygdala functions.
Collapse
Affiliation(s)
- Fabien Naneix
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077, Bordeaux, France; Univ. Bordeaux, CNRS, INCIA, UMR 5287, 33077 Bordeaux, France
| | - Ioannis Bakoyiannis
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077, Bordeaux, France
| | - Marianela Santoyo-Zedillo
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077, Bordeaux, France; Department of Health Sciences, Metropolitan Autonomous University (UAM), Campus Lerma, Mexico
| | | | - Gustavo Pacheco-Lopez
- Department of Health Sciences, Metropolitan Autonomous University (UAM), Campus Lerma, Mexico
| | | | - Guillaume Ferreira
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077, Bordeaux, France.
| | | |
Collapse
|