1
|
Yamada S, Natsubori A, Harada K, Tsuboi T, Monai H. Immediate glucose signaling transmitted via the vagus nerve in gut-brain neural communication. iScience 2025; 28:112439. [PMID: 40491956 PMCID: PMC12146620 DOI: 10.1016/j.isci.2025.112439] [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: 06/07/2024] [Revised: 11/10/2024] [Accepted: 04/10/2025] [Indexed: 06/11/2025] Open
Abstract
Sucrose consumption is influenced by certain gut-brain signaling mechanisms. One possible pathway could be the interaction between the vagus nerve and the central nervous system, mediated by neuropod cells forming synaptic connections with vagus nerves, which immediately activate the central dopaminergic pathways. In this study, we demonstrated that intestinal glucose administration activates the frontal cortex via the vagus nerve and central dopamine signaling. The immediate activation of both the vagus nerve and the frontal cortex was mediated by the sodium-glucose cotransporter 1 (SGLT1). Furthermore, Ca2+ signal activation in both astrocytes and neurons in the frontal cortex was mediated by D2 and D1 receptors, respectively. Finally, we showed that psychological stress, which causes a sucrose preference reduction, significantly diminished the activation levels of both the vagus nerve and the frontal cortex. These findings highlight the role of a comprehensive gut-brain network via vagus nerves in modulating sucrose preference.
Collapse
Affiliation(s)
- Serika Yamada
- Department of Biology, Faculty of Science, Ochanomizu University, 2-1-1 Ohtsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Hiromu Monai
- Department of Biology, Faculty of Science, Ochanomizu University, 2-1-1 Ohtsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| |
Collapse
|
2
|
Darcey VL, Guo J, Chi M, Chung ST, Courville AB, Gallagher I, Herscovitch P, Joseph PV, Howard R, La Noire M, Milley L, Schick A, Stagliano M, Turner S, Urbanski N, Yang S, Zhai N, Zhou MS, Hall KD. Brain dopamine responses to ultra-processed milkshakes are highly variable and not significantly related to adiposity in humans. Cell Metab 2025; 37:616-628.e5. [PMID: 40043691 DOI: 10.1016/j.cmet.2025.02.002] [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: 06/30/2024] [Revised: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 03/12/2025]
Abstract
Ultra-processed foods high in fat and sugar have been theorized to be addictive due to their purported ability to induce an exaggerated post-ingestive brain dopamine response akin to drugs of abuse. Using [11C]raclopride positron emission tomography (PET) displacement methods used to measure brain dopamine responses to addictive drugs, we measured striatal dopamine responses beginning 30 min after ingesting an ultra-processed milkshake high in fat and sugar in 50 young, healthy adults over a wide body mass index (BMI) range (20-45 kg/m2). Surprisingly, milkshake consumption did not result in a significant post-ingestive dopamine response in the striatum (p = 0.62) nor in any striatal subregion (p > 0.33), and the highly variable interindividual responses were not significantly related to adiposity (BMI: r = 0.076, p = 0.51; % body fat: r = 0.16, p = 0.28). Thus, post-ingestive striatal dopamine responses to an ultra-processed milkshake were likely substantially smaller than for many addictive drugs and below the limits of detection using standard PET methods.
Collapse
Affiliation(s)
- Valerie L Darcey
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Juen Guo
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Meible Chi
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber B Courville
- Human Energy and Body Weight Regulation Core, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Gallagher
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Paule V Joseph
- Section of Sensory Science and Metabolism, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA; National Institute of Nursing Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA; National Institute on Deafness and Other Communication Disorders, Smell and Taste Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Rebecca Howard
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melissa La Noire
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Milley
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Schick
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Stagliano
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara Turner
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Urbanski
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nan Zhai
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan S Zhou
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Çakır R, Büyükgüdük İ, Bilim P, Erdinç A, Veldhuizen MG. Transcutaneous Vagus Nerve Stimulation Enhances Probabilistic Learning. Psychophysiology 2025; 62:e70037. [PMID: 40059064 PMCID: PMC11891121 DOI: 10.1111/psyp.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 05/13/2025]
Abstract
tVNS enhances various memory and learning mechanisms, but there is inconclusive evidence on whether probabilistic learning can be enhanced by tVNS. Here, we tested a simplified version of the probabilistic learning task with monetary rewards in a between-participants design with left and right-sided cymba conchae and tragus stimulation (compared to sham stimulation) in a sample of healthy individuals (n = 80, 64 women, on average 26.38 years old). tVNS enhances overall accuracy significantly (p = 4.09 x 10-04) and reduces response times (p = 1.1006 x 10-49) in the probabilistic learning phase. Reinforcement learning modelling of the data revealed that the tVNS group uses a riskier strategy, dedicates more time to stimulus encoding and motor processes and exhibits greater reward sensitivity relative to the sham group. The learning advantage for tVNS relative to sham persists (p = 0.005 for accuracy and p = 9.2501 × 10-27 for response times) during an immediate extinction phase with continued stimulation in which feedback and reward were omitted. Our observations are in line with the proposal that tVNS enhances reinforcement learning in healthy individuals. This suggests that tVNS may be useful in contexts where fast learning and learning persistence in the absence of a reward is an advantage, for example, in the case of learning new habits.
Collapse
Affiliation(s)
- Resul Çakır
- Department of Psychology, Economics, Administrative and Social SciencesToros UniversityTürkiye
- Department of Psychology, Science and LettersMersin UniversityTürkiye
| | - İlkim Büyükgüdük
- Department of Psychology, Science and LettersMersin UniversityTürkiye
| | - Petek Bilim
- Department of Psychology, Economics, Administrative and Social SciencesKTO Karatay UniversityTürkiye
| | - Ataberk Erdinç
- Department of Psychology, Economics, Administrative and Social SciencesToros UniversityTürkiye
| | - Maria Geraldine Veldhuizen
- Department of Psychology, Science and LettersMersin UniversityTürkiye
- Department of Anatomy, MedicineMersin UniversityTürkiye
- National Magnetic Resonance Research Center (UMRAM)Bilkent UniversityTürkiye
| |
Collapse
|
4
|
Ribeiro G, Schellekens H, Cuesta-Marti C, Maneschy I, Ismael S, Cuevas-Sierra A, Martínez JA, Silvestre MP, Marques C, Moreira-Rosário A, Faria A, Moreno LA, Calhau C. A menu for microbes: unraveling appetite regulation and weight dynamics through the microbiota-brain connection across the lifespan. Am J Physiol Gastrointest Liver Physiol 2025; 328:G206-G228. [PMID: 39811913 DOI: 10.1152/ajpgi.00227.2024] [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: 07/26/2024] [Revised: 08/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
Appetite, as the internal drive for food intake, is often dysregulated in a broad spectrum of conditions associated with over- and under-nutrition across the lifespan. Appetite regulation is a complex, integrative process comprising psychological and behavioral events, peripheral and metabolic inputs, and central neurotransmitter and metabolic interactions. The microbiota-gut-brain axis has emerged as a critical mediator of multiple physiological processes, including energy metabolism, brain function, and behavior. Therefore, the role of the microbiota-gut-brain axis in appetite and obesity is receiving increased attention. Omics approaches such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics in appetite and weight regulation offer new opportunities for featuring obesity phenotypes. Furthermore, gut-microbiota-targeted approaches such as pre-, pro-, post-, and synbiotic, personalized nutrition, and fecal microbiota transplantation are novel avenues for precision treatments. The aim of this narrative review is 1) to provide an overview of the role of the microbiota-gut-brain axis in appetite regulation across the lifespan and 2) to discuss the potential of omics and gut microbiota-targeted approaches to deepen understanding of appetite regulation and obesity.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Harriët Schellekens
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Cristina Cuesta-Marti
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Ivie Maneschy
- Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Shámila Ismael
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Amanda Cuevas-Sierra
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, Madrid, Spain
| | - J Alfredo Martínez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, Madrid, Spain
| | - Marta P Silvestre
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cláudia Marques
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - André Moreira-Rosário
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Faria
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Luis A Moreno
- Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Conceição Calhau
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
5
|
Jameson KG, Kazmi SA, Ohara TE, Son C, Yu KB, Mazdeyasnan D, Leshan E, Vuong HE, Paramo J, Lopez-Romero A, Yang L, Schweizer FE, Hsiao EY. Select microbial metabolites in the small intestinal lumen regulate vagal activity via receptor-mediated signaling. iScience 2025; 28:111699. [PMID: 39877906 PMCID: PMC11772968 DOI: 10.1016/j.isci.2024.111699] [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: 07/12/2024] [Revised: 09/22/2024] [Accepted: 12/24/2024] [Indexed: 01/31/2025] Open
Abstract
The vagus nerve is proposed to enable communication between the gut microbiome and the brain, but activity-based evidence is lacking. We find that mice reared germ-free exhibit decreased vagal tone relative to colonized controls, which is reversed via microbiota restoration. Perfusing antibiotics into the small intestines of conventional mice, but not germ-free mice, acutely decreases vagal activity which is restored upon re-perfusion with intestinal filtrates from conventional, but not germ-free, mice. Microbiome-dependent short-chain fatty acids, bile acids, and 3-indoxyl sulfate indirectly stimulate vagal activity in a receptor-dependent manner. Serial perfusion of each metabolite class activates both shared and distinct neuronal subsets with varied response kinetics. Metabolite-induced and receptor-dependent increases in vagal activity correspond with the activation of brainstem neurons. Results from this study reveal that the gut microbiome regulates select metabolites in the intestinal lumen that differentially activate vagal afferent neurons, thereby enabling the microbial modulation of chemosensory signals for gut-brain communication.
Collapse
Affiliation(s)
- Kelly G. Jameson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sabeen A. Kazmi
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Takahiro E. Ohara
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Celine Son
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kristie B. Yu
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Donya Mazdeyasnan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Emma Leshan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Helen E. Vuong
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Paramo
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Arlene Lopez-Romero
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Long Yang
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Felix E. Schweizer
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elaine Y. Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
6
|
Thurn L, Schulz C, Borgmann D, Klaus J, Ellinger S, Walter M, Kroemer NB. Altered food liking in depression is driven by macronutrient composition. Psychol Med 2025; 55:e20. [PMID: 39905823 PMCID: PMC12017361 DOI: 10.1017/s0033291724003581] [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: 09/13/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025]
Abstract
Major depressive disorder (MDD) is characterized by changes in appetite and body weight as well as blunted reward sensitivity (‘anhedonia’). However, it is not well understood which mechanisms are driving changes in reward sensitivity, specifically regarding food. Here, we used a sample of 117 participants (54 patients with MDD and 63 healthy control participants [HCPs]) who completed a food cue reactivity task with ratings of wanting and liking for 60 food and 20 non-food items. To evaluate which components of the food may contribute to altered ratings in depression, we tested for associations with macronutrients of the depicted items. In line with previous studies, we found reduced ratings of food wanting (p = .003) but not liking (p = .23) in patients with MDD compared to matched HCPs. Adding macronutrient composition to the models of wanting and liking substantially improved their fit (ps < .001). Compared to carbohydrate-rich foods, patients with MDD reported lower liking and wanting ratings for high-fat and high-protein foods. Moreover, patients with MDD showed weaker correlations in their preferences for carbohydrate- versus fat- or protein-rich foods (ps < .001), pointing to potential disturbances in metabolic signaling. To conclude, our results suggest that depression-related alterations in food reward ratings are more specific to the macronutrient composition of the food than previously anticipated, hinting at disturbances in gut–brain signaling. These findings raise the intriguing question of whether interventions targeting the gut could help normalize aberrant reward signals for foods rich in fat or protein.
Collapse
Affiliation(s)
- Lilly Thurn
- Section of Medical Psychology, Department of Psychiatry & Psychotherapy, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Corinna Schulz
- Section of Medical Psychology, Department of Psychiatry & Psychotherapy, Faculty of Medicine, University of Bonn, Bonn, Germany
- Department of Psychiatry & Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
| | - Diba Borgmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Johannes Klaus
- Department of Psychiatry & Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
| | - Sabine Ellinger
- Institute of Nutritional and Food Sciences, Human Nutrition, University of Bonn, Bonn, Germany
| | - Martin Walter
- Department of Psychiatry & Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- Department of Psychiatry & Psychotherapy, University Hospital Jena, Jena, Germany
- Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- German Center for Mental Health (DZPG), partner site Jena-Magdeburg-Halle
| | - Nils B. Kroemer
- Section of Medical Psychology, Department of Psychiatry & Psychotherapy, Faculty of Medicine, University of Bonn, Bonn, Germany
- Department of Psychiatry & Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), partner site Tübingen
| |
Collapse
|
7
|
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
|
8
|
Gruber T, Lechner F, Krieger JP, García-Cáceres C. Neuroendocrine gut-brain signaling in obesity. Trends Endocrinol Metab 2025; 36:42-54. [PMID: 38821753 DOI: 10.1016/j.tem.2024.05.002] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 06/02/2024]
Abstract
The past decades have witnessed the rise and fall of several, largely unsuccessful, therapeutic attempts to bring the escalating obesity pandemic to a halt. Looking back to look ahead, the field has now put its highest hopes in translating insights from how the gastrointestinal (GI) tract communicates with the brain to calibrate behavior, physiology, and metabolism. A major focus of this review is to summarize the latest advances in comprehending the neuroendocrine aspects of this so-called 'gut-brain axis' and to explore novel concepts, cutting-edge technologies, and recent paradigm-shifting experiments. These exciting insights continue to refine our understanding of gut-brain crosstalk and are poised to promote the development of additional therapeutic avenues at the dawn of a new era of antiobesity therapeutics.
Collapse
Affiliation(s)
- Tim Gruber
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| | - Franziska Lechner
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Jean-Philippe Krieger
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Institute of Neuroscience and Physiology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Cristina García-Cáceres
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
9
|
Ribeiro G, Fernandes AB, Oliveira FPM, Duarte JS, Oliveira M, Limbert C, Costa RM, Costa DC, Oliveira-Maia AJ. Postingestive reward acts through behavioral reinforcement and is conserved in obesity and after bariatric surgery. PLoS Biol 2024; 22:e3002936. [PMID: 39689052 DOI: 10.1371/journal.pbio.3002936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024] Open
Abstract
Postingestive nutrient stimulation conditions food preferences through striatal dopamine and may be associated with blunted brain responses in obesity. In a cross-sectional study, we tested flavor-nutrient conditioning (FNC) with maltodextrin-enriched yogurt, with maltodextrin previously optimized for concentration and dextrose equivalents (n = 57), and to mask texture cues (n = 102). After conditioning, healthy volunteers (n = 52) increased preference for maltodextrin-paired (+102 kcal, CS+), relative to control (+1.8 kcal, CS-) flavors, as assessed according to intake, but not pleasantness. In a clinical study (n = 61), behavioral conditioning without effects on pleasantness was confirmed across pre-bariatric candidates with obesity, weight-stable post-surgery patients, and healthy controls, without significant differences between groups. Striatal dopamine D2-like receptor (DD2lR) availability, assessed with [123I]IBZM SPECT, was reduced in the obesity group and strongly correlated with conditioning strength and a measure of restrained eating in patients with gastric bypass. These results show that postingestive nutrient stimulation influences human food choices through behavioral reinforcement, and is conserved in obesity and after bariatric surgery. Trial Registration: ISRCTN17965026: Dopaminergic neurotransmission in dietary learning and obesity.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
- Lisbon Academic Medical Centre PhD Program, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Ana B Fernandes
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, Lisboa, Portugal
| | - Francisco P M Oliveira
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
| | - João S Duarte
- Department of Endocrinology, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, Lisboa, Portugal
| | - Manuela Oliveira
- Department of Endocrinology, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, Lisboa, Portugal
| | - Clotilde Limbert
- Department of Endocrinology, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, Lisboa, Portugal
| | - Rui M Costa
- Department of Neuroscience and Neurology, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Allen Institute, Seattle, Washington State, United States of America
| | - Durval C Costa
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
| | - Albino J Oliveira-Maia
- Champalimaud Research & Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, Lisboa, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, Lisboa, Portugal
| |
Collapse
|
10
|
Jia W, Peng J, Zhang Y, Zhu J, Qiang X, Zhang R, Shi L. Amelioration impact of gut-brain communication on obesity control by regulating gut microbiota composition through the ingestion of animal-plant-derived peptides and dietary fiber: can food reward effect as a hidden regulator? Crit Rev Food Sci Nutr 2024; 64:11575-11589. [PMID: 37526310 DOI: 10.1080/10408398.2023.2241078] [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] [Indexed: 08/02/2023]
Abstract
Various roles of intestinal flora in the gut-brain axis response pathway have received enormous attention because of their unique position in intestinal flora-derived metabolites regulating hormones, inducing appetite, and modulating energy metabolism. Reward pathways in the brain play a crucial role in gut-brain communications, but the mechanisms have not been methodically understood. This review outlined the mechanisms by which leptin, ghrelin, and insulin are influenced by intestinal flora-derived metabolites to regulate appetite and body weight, focused on the significance of the paraventricular nucleus and ventromedial prefrontal cortex in food reward. The vagus nerve and mitochondria are essential pathways of the intestinal flora involved in the modulation of neurotransmitters, neural signaling, and neurotransmission in gut-brain communications. The dynamic response to nutrient intake and changes in the characteristics of feeding activity requires the participation of the vagus nerve to transmit messages to be completed. SCFAs, Bas, BCAAs, and induced hormones mediate the sensory information and reward signaling of the host in the complex regulatory mechanism of food selection, and the composition of the intestinal flora significantly impacts this process. Food reward in the process of obesity based on gut-brain communications expands new ideas for the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Wei Jia
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an, China
- Shaanxi Sky Pet Biotechnology Co., Ltd, Xi'an, China
| | - Jian Peng
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Yan Zhang
- Inspection and Testing Center of Fuping County (Shaanxi Goat Milk Product Quality Supervision and Inspection Center), Wei nan, China
| | - Jiying Zhu
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Xin Qiang
- Inspection and Testing Center of Fuping County (Shaanxi Goat Milk Product Quality Supervision and Inspection Center), Wei nan, China
| | - Rong Zhang
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Lin Shi
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| |
Collapse
|
11
|
Petersson M, Uvnäs-Moberg K. Interactions of Oxytocin and Dopamine-Effects on Behavior in Health and Disease. Biomedicines 2024; 12:2440. [PMID: 39595007 PMCID: PMC11591571 DOI: 10.3390/biomedicines12112440] [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: 09/18/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 11/28/2024] Open
Abstract
The hypothalamic neuropeptide and hormone oxytocin are of fundamental importance for maternal, social, and sexual behavior. Deviations in oxytocin levels have also been associated with anxiety, autism spectrum disorders (ASD), depression, ADHD (attention deficit hyperactivity disorder), and schizophrenia. Both oxytocin and dopamine are often considered reward- and feel-good hormones, and dopamine is associated with the above-mentioned behaviors and, and dopamine is also associated with the above-mentioned behaviors and disorders. Although being structurally totally different, oxytocin, a peptide, and dopamine, a monoamine, they have a number of similar effects. They are synthesized both in the brain and in the periphery, and they affect each other's release and receptors. In addition, oxytocin and dopamine are released in response to, for example, social interaction, sex, feeding, and massage. This review discusses interactions between oxytocin and dopamine with a specific focus on behavioral effects and possible roles of oxytocin and dopamine in various mental disorders and functional diversities.
Collapse
Affiliation(s)
- Maria Petersson
- Department of Endocrinology, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Kerstin Uvnäs-Moberg
- Department of Applied Animal Science and Welfare, Swedish University of Agricultural Sciences, 532 31 Skara, Sweden
| |
Collapse
|
12
|
Ito K, Hosoki H, Kasai Y, Sasaki H, Haraguchi A, Shibata S, Nozaki C. A Cellulose-Rich Diet Disrupts Gut Homeostasis and Leads to Anxiety through the Gut-Brain Axis. ACS Pharmacol Transl Sci 2024; 7:3071-3085. [PMID: 39416961 PMCID: PMC11475280 DOI: 10.1021/acsptsci.4c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
It is widely said that a healthy intestinal environment plays an essential role in better mental condition. One known dietary nutrient that maintains the intestinal environment is dietary fiber. A recent study showed that maintaining the intestinal environment with dietary fiber alleviated symptoms of psychiatric disorders in animals. However, such effects have only been reported with soluble fiber, which is highly fermentable and promotes short-chain fatty acid (SCFA) production, and not with insoluble fiber. Therefore, we aimed to verify whether insoluble fiber, such as cellulose, can alter emotion via changes in the gut. We divided mice into two groups and fed either a standard diet (SD, which contains both insoluble and soluble dietary fibers) or a cellulose-rich diet (CRD, which contains cellulose alone as the dietary fibers). We found that CRD-fed mice display increased anxiety-like behavior. CRD-fed animals also showed decreased intestinal SCFA levels along with increased intestinal permeability, dysmotility, and hypersensitivity. This behavioral and physiological effect of CRD has been completely abolished in vagotomized mice, indicating the direct link between intestinal environment exacerbation to the emotion through the gut-brain axis. Additionally, we found that amygdalar dopamine signaling has been modified in CRD-fed animals, and the opioid antagonist abolished this dopaminergic modification as well as CRD-induced anxiety. Altogether, our findings indicate that consumption of cellulose alone as the dietary fiber may evoke intestinal abnormalities, which fire the vagus nerve, then the opioidergic system, and amygdalar dopamine upregulation, resulting in the enhancement of anxiety.
Collapse
Affiliation(s)
- Kaede Ito
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
| | - Haruka Hosoki
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
| | - Yuya Kasai
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
| | - Hiroyuki Sasaki
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
| | - Atsushi Haraguchi
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
| | - Shigenobu Shibata
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
- Graduate
School of Biomedical and Health Science, Hiroshima University, Hiroshima 734-8553, Japan
| | - Chihiro Nozaki
- School
of Advanced Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
- Global
Center for Science and Engineering, Waseda
University, Tokyo 162-0056, Japan
| |
Collapse
|
13
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585262. [PMID: 38559190 PMCID: PMC10980014 DOI: 10.1101/2024.03.17.585262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, the most common neurodegenerative disorder, remain incompletely understood. To define relevant pathways, we integrated the results of an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila with human and Drosophila Alzheimer's disease-associated multi-omics. We measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's disease human genetic variants that modify expression in disease-vulnerable neurons. We used a network optimization approach to integrate these data with previously published Alzheimer's disease multi-omic data. We computationally predicted and experimentally demonstrated how HNRNPA2B1 and MEPCE enhance tau-mediated neurotoxicity. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human iPSC-derived neural progenitor cells. Our work identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: Stephen and Denise Adams Center of Yale School of Medicine, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Lead contact
| |
Collapse
|
14
|
Qu S, Yu Z, Zhou Y, Wang S, Jia M, Chen T, Zhang X. Gut microbiota modulates neurotransmitter and gut-brain signaling. Microbiol Res 2024; 287:127858. [PMID: 39106786 DOI: 10.1016/j.micres.2024.127858] [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: 02/08/2024] [Revised: 06/16/2024] [Accepted: 07/22/2024] [Indexed: 08/09/2024]
Abstract
Neurotransmitters, including 5-hydroxytryptamine (5-HT), dopamine (DA), gamma-aminobutyric acid (GABA), and glutamate, are essential transductors in the Gut-Brain Axis (GBA), playing critical roles both peripherally and centrally. Accumulating evidence suggests that the gut microbiota modulates intestinal neurotransmitter metabolism and gut-to-brain signaling, shedding light on the crucial role of the gut microbiota in brain function and the pathogenesis of various neuropsychiatric diseases, such as major depression disorder (MDD), anxiety, addiction and Parkinson's disease (PD). Despite the exciting findings, the mechanisms underlying the modulation of neurotransmitter metabolism and function by the gut microbiota are still being elucidated. In this review, we aim to provide a comprehensive overview of the existing knowledge about the role of the gut microbiota in neurotransmitter metabolism and function in animal and clinical experiments. Moreover, we will discuss the potential mechanisms through which gut microbiota-derived neurotransmitters contribute to the pathogenesis of neuropsychiatric diseases, thus highlighting a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Shiyan Qu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Zijin Yu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yaxuan Zhou
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Shiyi Wang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Minqi Jia
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Ti Chen
- Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China.
| |
Collapse
|
15
|
Zimmerman CA, Bolkan SS, Pan-Vazquez A, Wu B, Keppler EF, Meares-Garcia JB, Guthman EM, Fetcho RN, McMannon B, Lee J, Hoag AT, Lynch LA, Janarthanan SR, López Luna JF, Bondy AG, Falkner AL, Wang SSH, Witten IB. A neural mechanism for learning from delayed postingestive feedback. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.06.561214. [PMID: 37873112 PMCID: PMC10592633 DOI: 10.1101/2023.10.06.561214] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Animals learn the value of foods based on their postingestive effects and thereby develop aversions to foods that are toxic1-6 and preferences to those that are nutritious7-14. However, it remains unclear how the brain is able to assign credit to flavors experienced during a meal with postingestive feedback signals that can arise after a substantial delay. Here, we reveal an unexpected role for postingestive reactivation of neural flavor representations in this temporal credit assignment process. To begin, we leverage the fact that mice learn to associate novel15-18, but not familiar, flavors with delayed gastric malaise signals to investigate how the brain represents flavors that support aversive postingestive learning. Surveying cellular resolution brainwide activation patterns reveals that a network of amygdala regions is unique in being preferentially activated by novel flavors across every stage of the learning process: the initial meal, delayed malaise, and memory retrieval. By combining high-density recordings in the amygdala with optogenetic stimulation of genetically defined hindbrain malaise cells, we find that postingestive malaise signals potently and specifically reactivate amygdalar novel flavor representations from a recent meal. The degree of malaise-driven reactivation of individual neurons predicts strengthening of flavor responses upon memory retrieval, leading to stabilization of the population-level representation of the recently consumed flavor. In contrast, meals without postingestive consequences degrade neural flavor representations as flavors become familiar and safe. Thus, our findings demonstrate that interoceptive reactivation of amygdalar flavor representations provides a neural mechanism to resolve the temporal credit assignment problem inherent to postingestive learning.
Collapse
Affiliation(s)
| | - Scott S Bolkan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Bichan Wu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Emma F Keppler
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Eartha Mae Guthman
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Robert N Fetcho
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Junuk Lee
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Austin T Hoag
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Laura A Lynch
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Juan F López Luna
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Adrian G Bondy
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Annegret L Falkner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Samuel S-H Wang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Howard Hughes Medical Institute, Princeton University, Princeton, NJ, USA
| |
Collapse
|
16
|
Kim SQ, Spann RA, Khan MSH, Berthoud HR, Münzberg H, Albaugh VL, He Y, McDougal DH, Soto P, Yu S, Morrison CD. FGF21 as a mediator of adaptive changes in food intake and macronutrient preference in response to protein restriction. Neuropharmacology 2024; 255:110010. [PMID: 38797244 PMCID: PMC11156534 DOI: 10.1016/j.neuropharm.2024.110010] [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: 03/20/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Free-feeding animals navigate complex nutritional landscapes in which food availability, cost, and nutritional value can vary markedly. Animals have thus developed neural mechanisms that enable the detection of nutrient restriction, and these mechanisms engage adaptive physiological and behavioral responses that limit or reverse this nutrient restriction. This review focuses specifically on dietary protein as an essential and independently defended nutrient. Adequate protein intake is required for life, and ample evidence exists to support an active defense of protein that involves behavioral changes in food intake, food preference, and food motivation, likely mediated by neural changes that increase the reward value of protein foods. Available evidence also suggests that the circulating hormone fibroblast growth factor 21 (FGF21) acts in the brain to coordinate these adaptive changes in food intake, making it a unique endocrine signal that drives changes in macronutrient preference in the context of protein restriction. This article is part of the Special Issue on "Food intake and feeding states".
Collapse
Affiliation(s)
- Sora Q Kim
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Redin A Spann
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | | | | - Heike Münzberg
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Vance L Albaugh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - David H McDougal
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Paul Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, 70810, USA
| | - Sangho Yu
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | |
Collapse
|
17
|
Darcey VL, Guo J, Chi M, Chung ST, Courville AB, Gallagher I, Herscovitch P, Joseph PV, Howard R, LaNoire M, Milley L, Schick A, Stagliano M, Turner S, Urbanski N, Yang S, Zhai N, Zhou MS, Hall KD. Brain dopamine responses to ultra-processed milkshakes are highly variable and not significantly related to adiposity in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.24.24309440. [PMID: 39108535 PMCID: PMC11302720 DOI: 10.1101/2024.06.24.24309440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Ultra-processed foods high in fat and sugar may be addictive, in part, due to their purported ability to induce an exaggerated postingestive brain dopamine response akin to drugs of abuse. Using standard [11C]raclopride positron emission tomography (PET) displacement methods used to measure brain dopamine responses to addictive drugs, we measured postingestive striatal dopamine responses to an ultra-processed milkshake high in fat and sugar in 50 young, healthy adults over a wide body mass index range (BMI 20-45 kg/m2). Surprisingly, milkshake consumption did not result in significant postingestive dopamine response in the striatum (p=0.62) nor any striatal subregion (p>0.33) and the highly variable interindividual responses were not significantly related to adiposity (BMI: r=0.076, p=0.51; %body fat: r=0.16, p=0.28). Thus, postingestive striatal dopamine responses to an ultra-processed milkshake were likely substantially smaller than many addictive drugs and below the limits of detection using standard PET methods.
Collapse
Affiliation(s)
- Valerie L Darcey
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Juen Guo
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Meible Chi
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber B Courville
- Human Energy and Body Weight Regulation Core, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Gallagher
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Paule V Joseph
- Section of Sensory Science and Metabolism, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892, USA
- National Institute of Nursing Research, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892, USA
- National Institute on Deafness and Other Communication Disorders, Smell and Taste Center, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892, USA
| | - Rebecca Howard
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melissa LaNoire
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Milley
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Schick
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Stagliano
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara Turner
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Urbanski
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nan Zhai
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan S Zhou
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
McGovern DJ, Phillips A, Ly A, Prévost ED, Ward L, Siletti K, Kim YS, Fenno LE, Ramakrishnan C, Deisseroth K, Ford CP, Root DH. Salience signaling and stimulus scaling of ventral tegmental area glutamate neuron subtypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598688. [PMID: 38915564 PMCID: PMC11195246 DOI: 10.1101/2024.06.12.598688] [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] [Indexed: 06/26/2024]
Abstract
Ventral tegmental area (VTA) glutamatergic neurons participate in reward, aversion, drug-seeking, and stress. Subsets of VTA VGluT2+ neurons are capable of co-transmitting glutamate and GABA (VGluT2+VGaT+ neurons), transmitting glutamate without GABA (VGluT2+VGaT- neurons), or co-transmitting glutamate and dopamine (VGluT2+TH+ neurons), but whether these molecularly distinct subpopulations show behavior-related differences is not wholly understood. We identified that neuronal activity of each VGluT2+ subpopulation is sensitive to reward value but signaled this in different ways. The phasic maximum activity of VGluT2+VGaT+ neurons increased with sucrose concentration, whereas VGluT2+VGaT- neurons increased maximum and sustained activity with sucrose concentration, and VGluT2+TH+ neurons increased sustained but not maximum activity with sucrose concentration. Additionally, VGluT2+ subpopulations signaled consummatory preferences in different ways. VGluT2+VGaT- neurons and VGluT2+TH+ neurons showed a signaling preference for a behaviorally-preferred fat reward over sucrose, but in temporally-distinct ways. In contrast, VGluT2+VGaT+ neurons uniquely signaled a less behaviorally-preferred sucrose reward compared with fat. Further experiments suggested that VGluT2+VGaT+ consummatory reward-related activity was related to sweetness, partially modulated by hunger state, and not dependent on caloric content or behavioral preference. All VGluT2+ subtypes increased neuronal activity following aversive stimuli but VGluT2+VGaT+ neurons uniquely scaled their magnitude and sustained activity with footshock intensity. Optogenetic activation of VGluT2+VGaT+ neurons during low intensity footshock enhanced fear-related behavior without inducing place preference or aversion. We interpret these data such that VTA glutamatergic subpopulations signal different elements of rewarding and aversive experiences and highlight the unique role of VTA VGluT2+VGaT+ neurons in enhancing the salience of behavioral experiences.
Collapse
Affiliation(s)
- Dillon J. McGovern
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Alysabeth Phillips
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Annie Ly
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Emily D. Prévost
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Lucy Ward
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Kayla Siletti
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| | - Yoon Seok Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Lief E. Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Current address: Department of Neuroscience, Dell Medical School, The University of Texas at Austin 78712
| | - Charu Ramakrishnan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045
| | - David H. Root
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301
| |
Collapse
|
19
|
Teckentrup V, Kroemer NB. Mechanisms for survival: vagal control of goal-directed behavior. Trends Cogn Sci 2024; 28:237-251. [PMID: 38036309 DOI: 10.1016/j.tics.2023.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
Survival is a fundamental physiological drive, and neural circuits have evolved to prioritize actions that meet the energy demands of the body. This fine-tuning of goal-directed actions based on metabolic states ('allostasis') is deeply rooted in our brain, and hindbrain nuclei orchestrate the vital communication between the brain and body through the vagus nerve. Despite mounting evidence for vagal control of allostatic behavior in animals, its broader function in humans is still contested. Based on stimulation studies, we propose that the vagal afferent pathway supports transitions between survival modes by gating the integration of ascending bodily signals, thereby regulating reward-seeking. By reconceptualizing vagal signals as catalysts for goal-directed behavior, our perspective opens new avenues for theory-driven translational work in mental disorders.
Collapse
Affiliation(s)
- Vanessa Teckentrup
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; School of Psychology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Nils B Kroemer
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; Section of Medical Psychology, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Bonn, 53127 Bonn, Germany; German Center for Mental Health (DZPG), 72076 Tübingen, Germany.
| |
Collapse
|
20
|
Yu KB, Son C, Chandra A, Paramo J, Novoselov A, Özcan E, Kazmi SA, Lum GR, Lopez-Romero A, Lynch JB, Hsiao EY. Complex carbohydrate utilization by gut bacteria modulates host food preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580152. [PMID: 38405943 PMCID: PMC10888876 DOI: 10.1101/2024.02.13.580152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The gut microbiota interacts directly with dietary nutrients and has the ability to modify host feeding behavior, but the underlying mechanisms remain poorly understood. Select gut bacteria digest complex carbohydrates that are non-digestible by the host and liberate metabolites that serve as additional energy sources and pleiotropic signaling molecules. Here we use a gnotobiotic mouse model to examine how differential fructose polysaccharide metabolism by commensal gut bacteria influences host preference for diets containing these carbohydrates. Bacteroides thetaiotaomicron and Bacteroides ovatus selectively ferment fructans with different glycosidic linkages: B. thetaiotaomicron ferments levan with β2-6 linkages, whereas B. ovatus ferments inulin with β2-1 linkages. Since inulin and levan are both fructose polymers, inulin and levan diet have similar perceptual salience to mice. We find that mice colonized with B. thetaiotaomicron prefer the non-fermentable inulin diet, while mice colonized with B. ovatus prefer the non-fermentable levan diet. Knockout of bacterial fructan utilization genes abrogates this preference, whereas swapping the fermentation ability of B. thetaiotaomicron to inulin confers host preference for the levan diet. Bacterial fructan fermentation and host behavioral preference for the non-fermentable fructan are associated with increased neuronal activation in the arcuate nucleus of the hypothalamus, a key brain region for appetite regulation. These results reveal that selective nutrient metabolism by gut bacteria contributes to host associative learning of dietary preference, and further informs fundamental understanding of the biological determinants of food choice.
Collapse
Affiliation(s)
- Kristie B Yu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Celine Son
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Anisha Chandra
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jorge Paramo
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Anna Novoselov
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Ezgi Özcan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Sabeen A Kazmi
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Gregory R Lum
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Arlene Lopez-Romero
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Jonathan B Lynch
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Current address: Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Valente EEL, Klotz JL, Markmann RC, Trotta RJ, Edwards JL, May JB, Harmon DL. Levodopa attenuates the feed intake reduction caused by ergot alkaloids in cattle. J Anim Sci 2024; 102:skae078. [PMID: 38502533 PMCID: PMC11003531 DOI: 10.1093/jas/skae078] [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: 01/26/2024] [Accepted: 03/18/2024] [Indexed: 03/21/2024] Open
Abstract
Consumption of ergot alkaloids from endophyte-infected tall fescue results in losses to the livestock industry in many countries and a means to mitigate these losses is needed. The objective of this study was to evaluate intra-abomasal infusion of the dopamine precursor, levodopa (L-DOPA), on dopamine metabolism, feed intake, and serum metabolites of steers exposed to ergot alkaloids. Twelve Holstein steers (344.9 ± 9.48 kg) fitted with ruminal cannula were housed with a cycle of heat challenge during the daytime (32 °C) and thermoneutral at night (25 °C). The steers received a basal diet of alfalfa cubes containing equal amounts of tall fescue seed composed of a mixture of endophyte-free (E-) or endophyte-infected tall fescue seeds (E+) equivalent to 15 µg ergovaline/kg body weight (BW) for 9 d followed by intra-abomasal infusion of water (L-DOPA-) or levodopa (L-DOPA+; 2 mg/kg BW) for an additional 9 d. Afterward, the steers were pair-fed for 5 d to conduct a glucose tolerance test. The E+ treatment decreased (P = 0.005) prolactin by approximately 50%. However, prolactin increased (P = 0.050) with L-DOPA+. Steers receiving E+ decreased (P < 0.001) dry matter intake (DMI); however, when supplemented with L-DOPA+ the decrease in DMI was less severe (L-DOPA × E, P = 0.003). Also, L-DOPA+ infusion increased eating duration (L-DOPA × E, P = 0.012) when steers were receiving E+. The number of meals, meal duration, and intake rate were not affected (P > 0.05) by E+ or L-DOPA+. The L-DOPA+ infusion increased (P < 0.05) free L-DOPA, free dopamine, total L-DOPA, and total dopamine. Conversely, free epinephrine and free norepinephrine decreased (P < 0.05) with L-DOPA+. Total epinephrine and total norepinephrine were not affected (P > 0.05) by L-DOPA+. Ergot alkaloids did not affect (P > 0.05) circulating free or total L-DOPA, dopamine, or epinephrine. However, free and total norepinephrine decreased (P = 0.046) with E+. Glucose clearance rates at 15 to 30 min after glucose infusion increased with L-DOPA+ (P < 0.001), but not with E+ (P = 0.280). Administration of L-DOPA as an agonist therapy to treat fescue toxicosis provided a moderate increase in DMI and eating time and increased plasma glucose clearance for cattle dosed with E+ seed.
Collapse
Affiliation(s)
- Eriton E L Valente
- Animal Science Department, State University of Western Parana, Marechal Cândido Rondon, PR, Brazil
| | - James L Klotz
- Forage-Animal Production Research Unit, USDA-ARS, Lexington, KY, USA
| | - Ryana C Markmann
- Animal Science Department, State University of Western Parana, Marechal Cândido Rondon, PR, Brazil
| | - Ronald J Trotta
- Department of Animal and Food Science, University of Kentucky, Lexington, KY, USA
| | - J Lannett Edwards
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - John B May
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
| | - David L Harmon
- Department of Animal and Food Science, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
22
|
Jameson KG, Kazmi SA, Son C, Mazdeyasnan D, Leshan E, Vuong HE, Paramo J, Lopez-Romero A, Yang L, Schweizer FE, Hsiao EY. Vagal interoception of microbial metabolites from the small intestinal lumen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572257. [PMID: 38187610 PMCID: PMC10769238 DOI: 10.1101/2023.12.18.572257] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The vagus nerve is proposed to enable communication between the gut microbiome and brain, but activity-based evidence is lacking. Herein, we assess the extent of gut microbial influences on afferent vagal activity and metabolite signaling mechanisms involved. We find that mice reared without microbiota (germ-free, GF) exhibit decreased vagal afferent tone relative to conventionally colonized mice (specific pathogen-free, SPF), which is reversed by colonization with SPF microbiota. Perfusing non-absorbable antibiotics (ABX) into the small intestine of SPF mice, but not GF mice, acutely decreases vagal activity, which is restored upon re-perfusion with bulk lumenal contents or sterile filtrates from the small intestine and cecum of SPF, but not GF, mice. Of several candidates identified by metabolomic profiling, microbiome-dependent short-chain fatty acids, bile acids, and 3-indoxyl sulfate stimulate vagal activity with varied response kinetics, which is blocked by co-perfusion of pharmacological antagonists of FFAR2, TGR5, and TRPA1, respectively, into the small intestine. At the single-unit level, serial perfusion of each metabolite class elicits more singly responsive neurons than dually responsive neurons, suggesting distinct neuronal detection of different microbiome- and macronutrient-dependent metabolites. Finally, microbial metabolite-induced increases in vagal activity correspond with activation of neurons in the nucleus of the solitary tract, which is also blocked by co-administration of their respective receptor antagonists. Results from this study reveal that the gut microbiome regulates select metabolites in the intestinal lumen that differentially activate chemosensory vagal afferent neurons, thereby enabling microbial modulation of interoceptive signals for gut-brain communication. HIGHLIGHTS Microbiota colonization status modulates afferent vagal nerve activityGut microbes differentially regulate metabolites in the small intestine and cecumSelect microbial metabolites stimulate vagal afferents with varied response kineticsSelect microbial metabolites activate vagal afferent neurons and brainstem neurons via receptor-dependent signaling.
Collapse
|
23
|
Nyema NT, McKnight AD, Vargas-Elvira AG, Schneps HM, Gold EG, Myers KP, Alhadeff AL. AgRP neuron activity promotes associations between sensory and nutritive signals to guide flavor preference. Mol Metab 2023; 78:101833. [PMID: 37925021 PMCID: PMC10665654 DOI: 10.1016/j.molmet.2023.101833] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023] Open
Abstract
OBJECTIVE The learned associations between sensory cues (e.g., taste, smell) and nutritive value (e.g., calories, post-ingestive signaling) of foods powerfully influences our eating behavior [1], but the neural circuits that mediate these associations are not well understood. Here, we examined the role of agouti-related protein (AgRP)-expressing neurons - neurons which are critical drivers of feeding behavior [2; 3] - in mediating flavor-nutrient learning (FNL). METHODS Because mice prefer flavors associated with AgRP neuron activity suppression [4], we examined how optogenetic stimulation of AgRP neurons during intake influences FNL, and used fiber photometry to determine how endogenous AgRP neuron activity tracks associations between flavors and nutrients. RESULTS We unexpectedly found that tonic activity in AgRP neurons during FNL potentiated, rather than prevented, the development of flavor preferences. There were notable sex differences in the mechanisms for this potentiation. Specifically, in male mice, AgRP neuron activity increased flavor consumption during FNL training, thereby strengthening the association between flavors and nutrients. In female mice, AgRP neuron activity enhanced flavor-nutrient preferences independently of consumption during training, suggesting that AgRP neuron activity enhances the reward value of the nutrient-paired flavor. Finally, in vivo neural activity analyses demonstrated that acute AgRP neuron dynamics track the association between flavors and nutrients in both sexes. CONCLUSIONS Overall, these data (1) demonstrate that AgRP neuron activity enhances associations between flavors and nutrients in a sex-dependent manner and (2) reveal that AgRP neurons track and rapidly update these associations. Taken together, our findings provide new insight into the role of AgRP neurons in assimilating sensory and nutritive signals for food reinforcement.
Collapse
Affiliation(s)
- Nathaniel T Nyema
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron D McKnight
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Heather M Schneps
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | - Amber L Alhadeff
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in mouse VTA dopamine neurons. Neuron 2023; 111:3541-3553.e8. [PMID: 37657441 PMCID: PMC11672631 DOI: 10.1016/j.neuron.2023.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 05/17/2023] [Accepted: 08/03/2023] [Indexed: 09/03/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTADA) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear whether the same or different VTADA neurons encode these different stimuli. To address this question, we performed two-photon calcium imaging in mice presented with food and conspecifics and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that increasing motivation for one stimulus increases overlap. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone-related genes in individual VTADA neurons. Taken together, our functional and transcriptional data suggest overlapping VTADA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Adelaide R Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Faculty of Medicine, Technion, Haifa 3525433, Israel.
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Nirja Oak
- Faculty of Medicine, Technion, Haifa 3525433, Israel
| | - Stephan Y Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Catherine J Peña
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
25
|
Nyema NT, McKnight AD, Vargas-Elvira AG, Schneps HM, Gold EG, Myers KP, Alhadeff AL. AgRP neuron activity promotes associations between sensory and nutritive signals to guide flavor preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558483. [PMID: 37786670 PMCID: PMC10541598 DOI: 10.1101/2023.09.19.558483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Objective The learned associations between sensory cues (e.g., taste, smell) and nutritive value (e.g., calories, post-ingestive signaling) of foods powerfully influences our eating behavior [1], but the neural circuits that mediate these associations are not well understood. Here, we examined the role of agouti-related protein (AgRP)-expressing neurons - neurons which are critical drivers of feeding behavior [2; 3] - in mediating flavor-nutrient learning (FNL). Methods Because mice prefer flavors associated with AgRP neuron activity suppression [4], we examined how optogenetic stimulation of AgRP neurons during intake influences FNL, and used fiber photometry to determine how endogenous AgRP neuron activity tracks associations between flavors and nutrients. Results We unexpectedly found that tonic activity in AgRP neurons during FNL potentiated, rather than prevented, the development of flavor preferences. There were notable sex differences in the mechanisms for this potentiation. Specifically, in male mice, AgRP neuron activity increased flavor consumption during FNL training, thereby strengthening the association between flavors and nutrients. In female mice, AgRP neuron activity enhanced flavor-nutrient preferences independently of consumption during training, suggesting that AgRP neuron activity enhances the reward value of the nutrient-paired flavor. Finally, in vivo neural activity analyses demonstrated that acute AgRP neuron dynamics track the association between flavors and nutrients in both sexes. Conclusions Overall, these data (1) demonstrate that AgRP neuron activity enhances associations between flavors and nutrients in a sex-dependent manner and (2) reveal that AgRP neurons track and update these associations on fast timescales. Taken together, our findings provide new insight into the role of AgRP neurons in assimilating sensory and nutritive signals for food reinforcement.
Collapse
Affiliation(s)
- Nathaniel T. Nyema
- Monell Chemical Senses Center, Philadelphia PA 19104, USA
- University of Pennsylvania, Philadelphia PA 19104, USA
| | - Aaron D. McKnight
- Monell Chemical Senses Center, Philadelphia PA 19104, USA
- University of Pennsylvania, Philadelphia PA 19104, USA
| | | | - Heather M. Schneps
- Monell Chemical Senses Center, Philadelphia PA 19104, USA
- University of Pennsylvania, Philadelphia PA 19104, USA
| | | | | | - Amber L. Alhadeff
- Monell Chemical Senses Center, Philadelphia PA 19104, USA
- University of Pennsylvania, Philadelphia PA 19104, USA
| |
Collapse
|
26
|
Berthoud HR. Reduced postabsorptive interoception of nutrients in obesity: cause or consequence? Am J Clin Nutr 2023; 118:496-497. [PMID: 37661103 DOI: 10.1016/j.ajcnut.2023.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 09/05/2023] Open
Affiliation(s)
- Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States.
| |
Collapse
|
27
|
Hanssen R, Rigoux L, Kuzmanovic B, Iglesias S, Kretschmer AC, Schlamann M, Albus K, Edwin Thanarajah S, Sitnikow T, Melzer C, Cornely OA, Brüning JC, Tittgemeyer M. Liraglutide restores impaired associative learning in individuals with obesity. Nat Metab 2023; 5:1352-1363. [PMID: 37592007 PMCID: PMC10447249 DOI: 10.1038/s42255-023-00859-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 07/07/2023] [Indexed: 08/19/2023]
Abstract
Survival under selective pressure is driven by the ability of our brain to use sensory information to our advantage to control physiological needs. To that end, neural circuits receive and integrate external environmental cues and internal metabolic signals to form learned sensory associations, consequently motivating and adapting our behaviour. The dopaminergic midbrain plays a crucial role in learning adaptive behaviour and is particularly sensitive to peripheral metabolic signals, including intestinal peptides, such as glucagon-like peptide 1 (GLP-1). In a single-blinded, randomized, controlled, crossover basic human functional magnetic resonance imaging study relying on a computational model of the adaptive learning process underlying behavioural responses, we show that adaptive learning is reduced when metabolic sensing is impaired in obesity, as indexed by reduced insulin sensitivity (participants: N = 30 with normal insulin sensitivity; N = 24 with impaired insulin sensitivity). Treatment with the GLP-1 receptor agonist liraglutide normalizes impaired learning of sensory associations in men and women with obesity. Collectively, our findings reveal that GLP-1 receptor activation modulates associative learning in people with obesity via its central effects within the mesoaccumbens pathway. These findings provide evidence for how metabolic signals can act as neuromodulators to adapt our behaviour to our body's internal state and how GLP-1 receptor agonists work in clinics.
Collapse
Affiliation(s)
- Ruth Hanssen
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Cologne, Germany
| | - Lionel Rigoux
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | | | - Sandra Iglesias
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich and Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Alina C Kretschmer
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, Germany
| | - Marc Schlamann
- Faculty of Medicine and University Hospital Cologne, Institute for Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - Kerstin Albus
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sharmili Edwin Thanarajah
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tamara Sitnikow
- Faculty of Medicine and University Hospital Cologne, Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Cologne, Germany
| | - Corina Melzer
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Oliver A Cornely
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
28
|
Shaik Mohamed Sayed UF, Moshawih S, Goh HP, Kifli N, Gupta G, Singh SK, Chellappan DK, Dua K, Hermansyah A, Ser HL, Ming LC, Goh BH. Natural products as novel anti-obesity agents: insights into mechanisms of action and potential for therapeutic management. Front Pharmacol 2023; 14:1182937. [PMID: 37408757 PMCID: PMC10318930 DOI: 10.3389/fphar.2023.1182937] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 07/07/2023] Open
Abstract
Obesity affects more than 10% of the adult population globally. Despite the introduction of diverse medications aimed at combating fat accumulation and obesity, a significant number of these pharmaceutical interventions are linked to substantial occurrences of severe adverse events, occasionally leading to their withdrawal from the market. Natural products serve as attractive sources for anti-obesity agents as many of them can alter the host metabolic processes and maintain glucose homeostasis via metabolic and thermogenic stimulation, appetite regulation, pancreatic lipase and amylase inhibition, insulin sensitivity enhancing, adipogenesis inhibition and adipocyte apoptosis induction. In this review, we shed light on the biological processes that control energy balance and thermogenesis as well as metabolic pathways in white adipose tissue browning, we also highlight the anti-obesity potential of natural products with their mechanism of action. Based on previous findings, the crucial proteins and molecular pathways involved in adipose tissue browning and lipolysis induction are uncoupling protein-1, PR domain containing 16, and peroxisome proliferator-activated receptor-γ in addition to Sirtuin-1 and AMP-activated protein kinase pathway. Given that some phytochemicals can also lower proinflammatory substances like TNF-α, IL-6, and IL-1 secreted from adipose tissue and change the production of adipokines like leptin and adiponectin, which are important regulators of body weight, natural products represent a treasure trove for anti-obesity agents. In conclusion, conducting comprehensive research on natural products holds the potential to accelerate the development of an improved obesity management strategy characterized by heightened efficacy and reduced incidence of side effects.
Collapse
Affiliation(s)
| | - Said Moshawih
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Hui Poh Goh
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Nurolaini Kifli
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Andi Hermansyah
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
| | - Hooi Leng Ser
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in VTA dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541104. [PMID: 37293057 PMCID: PMC10245666 DOI: 10.1101/2023.05.17.541104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTA DA ) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear if the same or different VTA DA neurons encode these different stimuli. To address this question, we performed 2-photon calcium imaging in mice presented with food and conspecifics, and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that modifying motivation for one stimulus affects responses to both stimuli. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone related genes in individual VTA DA neurons. Taken together, our functional and transcriptional data suggest overlapping VTA DA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Adelaide R. Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Nirja Oak
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Stephan Y. Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Catherine J. Peña
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ilana B. Witten
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| |
Collapse
|
30
|
Titos I, Juginović A, Vaccaro A, Nambara K, Gorelik P, Mazor O, Rogulja D. A gut-secreted peptide suppresses arousability from sleep. Cell 2023; 186:1382-1397.e21. [PMID: 36958331 PMCID: PMC10216829 DOI: 10.1016/j.cell.2023.02.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 08/26/2022] [Accepted: 02/16/2023] [Indexed: 03/25/2023]
Abstract
Suppressing sensory arousal is critical for sleep, with deeper sleep requiring stronger sensory suppression. The mechanisms that enable sleeping animals to largely ignore their surroundings are not well understood. We show that the responsiveness of sleeping flies and mice to mechanical vibrations is better suppressed when the diet is protein rich. In flies, we describe a signaling pathway through which information about ingested proteins is conveyed from the gut to the brain to help suppress arousability. Higher protein concentration in the gut leads to increased activity of enteroendocrine cells that release the peptide CCHa1. CCHa1 signals to a small group of dopamine neurons in the brain to modulate their activity; the dopaminergic activity regulates the behavioral responsiveness of animals to vibrations. The CCHa1 pathway and dietary proteins do not influence responsiveness to all sensory inputs, showing that during sleep, different information streams can be gated through independent mechanisms.
Collapse
Affiliation(s)
- Iris Titos
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alen Juginović
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra Vaccaro
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Keishi Nambara
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pavel Gorelik
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Mazor
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Dragana Rogulja
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Valente EEL, Klotz JL, Egert-McLean AM, Costa GW, May JB, Harmon DL. Influence of intra-abomasal administration of L-DOPA on circulating catecholamines and feed intake in cattle. FRONTIERS IN ANIMAL SCIENCE 2023. [DOI: 10.3389/fanim.2023.1127575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Dopamine has multiple physiological functions including feed intake control in which it can act as an anorectic or orexigenic agent. This study had the objective to evaluate intra-abomasal administration of L-DOPA (levodopa; L-3,4-dihydroxyphenylalanine) from -Mucuna pruriens on circulating catecholamines, indicators of energy metabolism and feed intake in cattle. Eight Holstein steers (340 ± 20 kg) fitted with ruminal cannula were used in a replicated 4 x 4 Latin Square design experiment. Intra-abomasal infusion of L-DOPA at 0, 0.5, 1 and 2 mg/kg BW was carried out for seven days and blood samples were collected at 0, 30, 60, 120, 240 and 480 min from L-DOPA infusion on day 7. The area under the curve (AUC) of plasma L-DOPA and free dopamine increased quadratically with the administration of L-DOPA. However, the AUC of plasma total dopamine had a positive linear response with the increase of L-DOPA. Conversely, the serum 5-hydroxytriptophan (5-HTP), plasma serotonin, serum serotonin, serum tyrosine, plasma glucose and plasma free fatty acids were not affected by the intra-abomasal infusion of L-DOPA. The circulating concentration of the epinephrine, norepinephrine, serotonin, glucose and free fatty acids did not change with L-DOPA infusion. It can be concluded that intra-abomasal L-DOPA administration produced a strong increase in circulating dopamine with no change in energy metabolites and feed intake in cattle.
Collapse
|
32
|
Zhang XL, Chen L, Yang J, Zhao SS, Jin S, Ao N, Yang J, Liu HX, Du J. Vitamin D alleviates non-alcoholic fatty liver disease via restoring gut microbiota and metabolism. Front Microbiol 2023; 14:1117644. [PMID: 36819064 PMCID: PMC9932697 DOI: 10.3389/fmicb.2023.1117644] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) represents a severe public health problem. Dysbiosis of gut microbiome has been identified as one of the key environmental factors contributing to NAFLD. As an essential nutrition, Vitamin D (VD) plays an important role in regulating gut microbiota based on its receptor (Vitamin D Receptor, VDR) which is highly expressed in the gastrointestinal tract. Methods Rats were fed with HFD (high-fat diet) for 12 weeks. And the rats were treated with VD two times a week by intraperitoneal injection for 12 weeks. H&E staining combined with plasma biochemical index was performed to characterize pathological changes and function of the liver. Fecal microbiota 16S rRNA gene sequencing and metabolomics were taken to reveal the altered gut microbiota and metabolites. Result The VD alleviates the HFD-induced lipid accumulation in the liver as well as decreases the levels of amlodipine besylate (ALT) and amlodipine aspartate (AST). VD supplement decreased the ratio of phylum Firmicutes/Bacteroidetes (F/B) but increased alpha diversity. In addition, the VD treatment improved the HFD-induced gut microbiota by increasing the Prevotella and Porphyromonadaceae and decreasing Mucispirillum, Acetatifactor, Desulfovibrio, and Oscillospira abundance. Furthermore, the capability of tyrosine metabolism, tryptophan metabolism, arginine biosynthesis, and sphingolipid metabolism was enhanced after VD treatment. Consistently, Prevotella positively correlated with tryptophan metabolism and sphingolipid metabolism. Importantly, the Prevotella abundance was positively associated with serotonin, melatonin, tryptamine, L-arginine, and 3-dehydrosphinganine which synthesize from tryptophan, tyrosine, arginosuccinate, and serine, respectively. Conclusion VD treatment inhibited HFD-induced NAFLD accompany by dysbiosis gut microbiota and metabolites, suggesting that VD supplement could be a potential intervention used for NAFLD treatment by targeting the specific microbiota.
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lei Chen
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Jiang Yang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shan-Shan Zhao
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Shi Jin
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Ao
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Yang
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Hui-Xin Liu
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China,*Correspondence: Hui-Xin Liu,
| | - Jian Du
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,Jian Du,
| |
Collapse
|
33
|
Tavares G, Rosendo-Silva D, Simões F, Eickhoff H, Marques D, Sacramento JF, Capucho AM, Seiça R, Conde SV, Matafome P. Circulating Dopamine Is Regulated by Dietary Glucose and Controls Glucagon-like 1 Peptide Action in White Adipose Tissue. Int J Mol Sci 2023; 24:ijms24032464. [PMID: 36768789 PMCID: PMC9916853 DOI: 10.3390/ijms24032464] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Dopamine directly acts in the liver and white adipose tissue (WAT) to regulate insulin signaling, glucose uptake, and catabolic activity. Given that dopamine is secreted by the gut and regulates insulin secretion in the pancreas, we aimed to determine its regulation by nutritional cues and its role in regulating glucagon-like peptide 1 (GLP-1) action in WAT. Solutions with different nutrients were administered to Wistar rats and postprandial dopamine levels showed elevations following a mixed meal and glucose intake. In high-fat diet-fed diabetic Goto-Kakizaki rats, sleeve gastrectomy upregulated dopaminergic machinery, showing the role of the gut in dopamine signaling in WAT. Bromocriptine treatment in the same model increased GLP-1R in WAT, showing the role of dopamine in regulating GLP-1R. By contrast, treatment with the GLP-1 receptor agonist Liraglutide had no impact on dopamine receptors. GLP-1 and dopamine crosstalk was shown in rat WAT explants, since dopamine upregulated GLP-1-induced AMPK activity in mesenteric WAT in the presence of the D2R and D3R inhibitor Domperidone. In human WAT, dopamine receptor 1 (D1DR) and GLP-1R expression were correlated. Our results point out a dietary and gut regulation of plasma dopamine, acting in the WAT to regulate GLP-1 action. Together with the known dopamine action in the pancreas, such results may identify new therapeutic opportunities to improve metabolic control in metabolic disorders.
Collapse
Affiliation(s)
- Gabriela Tavares
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Daniela Rosendo-Silva
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
| | - Flávia Simões
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Hans Eickhoff
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Marques
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Joana F. Sacramento
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Adriana M. Capucho
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Raquel Seiça
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
| | - Sílvia V. Conde
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Paulo Matafome
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School, 3046-854 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
34
|
Ribeiro G, Torres S, Fernandes AB, Camacho M, Branco TL, Martins SS, Raimundo A, Oliveira-Maia AJ, Food Reward in Bariatric Surgery Portuguese Study Group. Enhanced sweet taste perception in obesity: Joint analysis of gustatory data from multiple studies. Front Nutr 2022; 9:1028261. [PMID: 36606228 PMCID: PMC9807659 DOI: 10.3389/fnut.2022.1028261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction While sweet taste perception is a potential determinant of feeding behavior in obesity, the supporting evidence is inconsistent and is typically associated with methodological limitations. Notably, possible associations between sweet taste perception and measures of food reward remain undetermined. Materials and methods We conducted a cross-sectional analysis comparing 246 individuals with severe obesity and 174 healthy volunteers using a validated method for taste perception assessment. We included gustatory variables, namely intensity and pleasantness ratings of sour, salt, sweet, and bitter tastants, and taste thresholds assessed by electrogustometry. Reward-related feeding behavior, including hedonic hunger, food addiction, feeding behavior traits, and acceptance of foods and alcohol, was evaluated using self-rated scales for comparison with gustatory measures. Result In logistic regressions adjusted for age, gender, educational level, and research center, we found that a greater likelihood of belonging to the obesity group was associated with higher sweet intensity ratings (OR = 1.4, P = 0.01), hedonic hunger, food addiction symptoms, restrained and emotional eating (1.7 < OR ≤ 4.6, all P ≤ 0.001), and lower alcohol acceptance (OR = 0.6, P = 0.0002). Using principal component analysis, we found that while hedonic hunger, food addiction, and emotional eating were strongly interrelated, they were not associated with sweet intensity perception that, in turn, had a closer relationship with alcohol acceptance and restrained eating. Conclusion We found that individuals with obesity report higher sweet taste intensity ratings than healthy controls. Furthermore, while psychological measures of reward-related feeding behavior assess a common construct, sweet intensity perception may represent a different obesity-related dimension.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Lisbon, Portugal,Lisbon Academic Medical Centre PhD Program, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Sandra Torres
- Faculdade de Psicologia e de Ciências da Educação, Universidade do Porto, Porto, Portugal,Centro de Psicologia da Universidade do Porto, Porto, Portugal
| | - Ana B. Fernandes
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Lisbon, Portugal,NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Marta Camacho
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Lisbon, Portugal
| | - Teresa L. Branco
- Exercise and Health Laboratory, CIPER, Faculdade de Medicina da Universidade de Lisboa, Cruz Quebrada, Portugal
| | - Sandra S. Martins
- Universidade Europeia, Lisbon, Portugal,Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Armando Raimundo
- Departamento de Desporto e Saúde, Escola de Saúde e Desenvolvimento Humano, Universidade de Évora, Évora, Portugal,Comprehensive Health Research Centre (CHRC), Universidade de Évora, Évora, Portugal
| | - Albino J. Oliveira-Maia
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Lisbon, Portugal,NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal,*Correspondence: Albino J. Oliveira-Maia,
| | | |
Collapse
|
35
|
Hanßen R, Schiweck C, Aichholzer M, Reif A, Edwin Thanarajah S. Food reward and its aberrations in obesity. Curr Opin Behav Sci 2022. [DOI: 10.1016/j.cobeha.2022.101224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
36
|
Akiti K, Tsutsui-Kimura I, Xie Y, Mathis A, Markowitz JE, Anyoha R, Datta SR, Mathis MW, Uchida N, Watabe-Uchida M. Striatal dopamine explains novelty-induced behavioral dynamics and individual variability in threat prediction. Neuron 2022; 110:3789-3804.e9. [PMID: 36130595 PMCID: PMC9671833 DOI: 10.1016/j.neuron.2022.08.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/03/2022] [Accepted: 08/18/2022] [Indexed: 12/15/2022]
Abstract
Animals both explore and avoid novel objects in the environment, but the neural mechanisms that underlie these behaviors and their dynamics remain uncharacterized. Here, we used multi-point tracking (DeepLabCut) and behavioral segmentation (MoSeq) to characterize the behavior of mice freely interacting with a novel object. Novelty elicits a characteristic sequence of behavior, starting with investigatory approach and culminating in object engagement or avoidance. Dopamine in the tail of the striatum (TS) suppresses engagement, and dopamine responses were predictive of individual variability in behavior. Behavioral dynamics and individual variability are explained by a reinforcement-learning (RL) model of threat prediction in which behavior arises from a novelty-induced initial threat prediction (akin to "shaping bonus") and a threat prediction that is learned through dopamine-mediated threat prediction errors. These results uncover an algorithmic similarity between reward- and threat-related dopamine sub-systems.
Collapse
Affiliation(s)
- Korleki Akiti
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Iku Tsutsui-Kimura
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yudi Xie
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander Mathis
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; The Rowland Institute at Harvard, Harvard University, Cambridge, MA 02138, USA; Swiss Federal Institute of Technology Lausanne, Geneve 1202, Switzerland
| | - Jeffrey E Markowitz
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Wallace H. Coulter Department of Biomedical Engineering, Emory School of Medicine, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Rockwell Anyoha
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Mackenzie Weygandt Mathis
- The Rowland Institute at Harvard, Harvard University, Cambridge, MA 02138, USA; Swiss Federal Institute of Technology Lausanne, Geneve 1202, Switzerland
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
37
|
Kelly AL, Baugh ME, Oster ME, DiFeliceantonio AG. The impact of caloric availability on eating behavior and ultra-processed food reward. Appetite 2022; 178:106274. [PMID: 35963586 PMCID: PMC9749763 DOI: 10.1016/j.appet.2022.106274] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/07/2022] [Accepted: 08/07/2022] [Indexed: 12/19/2022]
Abstract
The food environment has changed rapidly and dramatically in the last 50 years. While industrial food processing has increased the safety and stability of the food supply, a rapid expansion in the scope and scale of food processing in the 1980's has resulted in a market dominated by ultra-processed foods. Here, we use the NOVA definition of category 4 ultra-processed foods (UPFs) as they make up around 58% of total calories consumed in the US and 66% of calories in US children. UPFs are formulated from ingredients with no or infrequent culinary use, contain additives, and have a long shelf-life, spending long periods in contact with packaging materials, allowing for the absorption of compounds from those materials. The full implications of this dietary shift to UPFs on human health and disease outcomes are difficult, if not impossible, to quantify. However, UPF consumption is linked with various forms of cancer, increased cardiovascular disease, and increased all-cause mortality. Understanding food choice is, therefore, a critical problem in health research. Although many factors influence food choice, here we focus on the properties of the foods themselves. UPFs are generally treated as food, not as the highly refined, industrialized substances that they are, whose properties and components must be studied. Here, we examine one property of UPFs, that they deliver useable calories rapidly as a potential factor driving UPF overconsumption. First, we explore evidence that UPFs deliver calories more rapidly. Next, we examine the role of the gut-brain axis and its interplay with canonical reward systems, and last, we describe how speed affects both basic learning processes and drugs of abuse.
Collapse
Affiliation(s)
- Amber L Kelly
- Fralin Biomedical Research Institute at Virginia Tech Carilion, USA
| | | | - Mary E Oster
- Fralin Biomedical Research Institute at Virginia Tech Carilion, USA
| | - Alexandra G DiFeliceantonio
- Fralin Biomedical Research Institute at Virginia Tech Carilion, USA; Center for Health Behaviors Research; Department of Human Nutrition Foods and Exercise at Virginia Tech, USA.
| |
Collapse
|
38
|
Abstract
When it comes to food, one tempting substance is sugar. Although sweetness is detected by the tongue, the desire to consume sugar arises from the gut. Even when sweet taste is impaired, animals can distinguish sugars from non-nutritive sweeteners guided by sensory cues arising from the gut epithelium. Here, we review the molecular receptors, cells, circuits and behavioural consequences associated with sugar sensing in the gut. Recent work demonstrates that some duodenal cells, termed neuropod cells, can detect glucose using sodium-glucose co-transporter 1 and release glutamate onto vagal afferent neurons. Based on these and other data, we propose a model in which specific populations of vagal neurons relay these sensory cues to distinct sets of neurons in the brain, including neurons in the caudal nucleus of the solitary tract, dopaminergic reward circuits in the basal ganglia and homeostatic feeding circuits in the hypothalamus, that alter current and future sugar consumption. This emerging model highlights the critical role of the gut in sensing the chemical properties of ingested nutrients to guide appetitive decisions.
Collapse
Affiliation(s)
- Winston W Liu
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
39
|
Vagus nerve stimulation increases stomach-brain coupling via a vagal afferent pathway. Brain Stimul 2022; 15:1279-1289. [PMID: 36067977 DOI: 10.1016/j.brs.2022.08.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Maintaining energy homeostasis is vital and supported by vagal signaling between digestive organs and the brain. Previous research has established a gastric network in the brain that is phase synchronized with the rhythm of the stomach, but tools to perturb its function were lacking. OBJECTIVE To evaluate whether stomach-brain coupling can be acutely increased by non-invasively stimulating vagal afferent projections to the brain. METHODS Using a single-blind randomized crossover design, we investigated the effect of acute right-sided transcutaneous auricular vagus nerve stimulation (taVNS) versus sham stimulation on stomach-brain coupling. RESULTS In line with preclinical research, taVNS increased stomach-brain coupling in the nucleus of the solitary tract (NTS) and the midbrain while boosting coupling across the brain. Crucially, in the cortex, taVNS-induced changes in coupling occurred primarily in transmodal regions and were associated with changes in hunger ratings as indicators of the subjective metabolic state. CONCLUSIONS taVNS increases stomach-brain coupling via an NTS-midbrain pathway that signals gut-induced reward, indicating that communication between the brain and the body is effectively modulated by vago-vagal signaling. Such insights may help us better understand the role of vagal afferents in orchestrating the recruitment of the gastric network which could pave the way for novel neuromodulatory treatments.
Collapse
|
40
|
Grove JCR, Gray LA, La Santa Medina N, Sivakumar N, Ahn JS, Corpuz TV, Berke JD, Kreitzer AC, Knight ZA. Dopamine subsystems that track internal states. Nature 2022; 608:374-380. [PMID: 35831501 PMCID: PMC9365689 DOI: 10.1038/s41586-022-04954-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/08/2022] [Indexed: 12/11/2022]
Abstract
Food and water are rewarding in part because they satisfy our internal needs1,2. Dopaminergic neurons in the ventral tegmental area (VTA) are activated by gustatory rewards3-5, but how animals learn to associate these oral cues with the delayed physiological effects of ingestion is unknown. Here we show that individual dopaminergic neurons in the VTA respond to detection of nutrients or water at specific stages of ingestion. A major subset of dopaminergic neurons tracks changes in systemic hydration that occur tens of minutes after thirsty mice drink water, whereas different dopaminergic neurons respond to nutrients in the gastrointestinal tract. We show that information about fluid balance is transmitted to the VTA by a hypothalamic pathway and then re-routed to downstream circuits that track the oral, gastrointestinal and post-absorptive stages of ingestion. To investigate the function of these signals, we used a paradigm in which a fluid's oral and post-absorptive effects can be independently manipulated and temporally separated. We show that mice rapidly learn to prefer one fluid over another based solely on its rehydrating ability and that this post-ingestive learning is prevented if dopaminergic neurons in the VTA are selectively silenced after consumption. These findings reveal that the midbrain dopamine system contains subsystems that track different modalities and stages of ingestion, on timescales from seconds to tens of minutes, and that this information is used to drive learning about the consequences of ingestion.
Collapse
Affiliation(s)
- James C R Grove
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | - Jamie S Ahn
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | | | - Joshua D Berke
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Anatol C Kreitzer
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Gladstone Institutes, San Francisco, CA, USA
| | - Zachary A Knight
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
41
|
Obray JD, Small CA, Baldwin EK, Jang EY, Lee JG, Yang CH, Yorgason JT, Steffensen SC. Dopamine D2-Subtype Receptors Outside the Blood-Brain Barrier Mediate Enhancement of Mesolimbic Dopamine Release and Conditioned Place Preference by Intravenous Dopamine. Front Cell Neurosci 2022; 16:944243. [PMID: 35903367 PMCID: PMC9314669 DOI: 10.3389/fncel.2022.944243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/17/2022] [Indexed: 11/26/2022] Open
Abstract
Dopamine (DA) is a cell-signaling molecule that does not readily cross the blood-brain barrier. Despite this, peripherally administered DA enhances DA levels in the nucleus accumbens and alters DA-related behaviors. This study was designed to investigate whether DA subtype-2 receptors are involved in the enhancement of nucleus accumbens (NAc) DA levels elicited by intravenous DA administration. This was accomplished by using microdialysis in the NAc and extracellular single unit recordings of putative DA neurons in the ventral tegmental area (VTA). Additionally, the reinforcing properties of intravenous DA were investigated using a place conditioning paradigm and the effects of intravenous DA on ultrasonic vocalizations were assessed. Following administration of intravenous dopamine, the firing rate of putative DA neurons in the VTA displayed a biphasic response and DA levels in the nucleus accumbens were enhanced. Pretreatment with domperidone, a peripheral-only DA D2 receptor (D2R) antagonist, reduced intravenous DA mediated increases in VTA DA neuron activity and NAc DA levels. Pretreatment with phentolamine, a peripheral α-adrenergic receptor antagonist, did not alter the effects of IV DA on mesolimbic DA neurotransmission. These results provide evidence for peripheral D2R mediation of the effects of intravenous DA on mesolimbic DA signaling.
Collapse
Affiliation(s)
- J. Daniel Obray
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, United States
| | - Christina A. Small
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, United States
| | - Emily K. Baldwin
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, United States
| | - Eun Young Jang
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, United States
- Research Center for Convergence Toxicology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Jin Gyeom Lee
- College of Korean Medicine, Daegu Haany University, Daegu, South Korea
| | - Chae Ha Yang
- College of Korean Medicine, Daegu Haany University, Daegu, South Korea
| | - Jordan T. Yorgason
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, United States
| | - Scott C. Steffensen
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, United States
| |
Collapse
|
42
|
Rodenkirch C, Carmel JB, Wang Q. Rapid Effects of Vagus Nerve Stimulation on Sensory Processing Through Activation of Neuromodulatory Systems. Front Neurosci 2022; 16:922424. [PMID: 35864985 PMCID: PMC9294458 DOI: 10.3389/fnins.2022.922424] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
After sensory information is encoded into neural signals at the periphery, it is processed through multiple brain regions before perception occurs (i.e., sensory processing). Recent work has begun to tease apart how neuromodulatory systems influence sensory processing. Vagus nerve stimulation (VNS) is well-known as an effective and safe method of activating neuromodulatory systems. There is a growing body of studies confirming VNS has immediate effects on sensory processing across multiple sensory modalities. These immediate effects of VNS on sensory processing are distinct from the more well-documented method of inducing lasting neuroplastic changes to the sensory pathways through repeatedly delivering a brief VNS burst paired with a sensory stimulus. Immediate effects occur upon VNS onset, often disappear upon VNS offset, and the modulation is present for all sensory stimuli. Conversely, the neuroplastic effect of pairing sub-second bursts of VNS with a sensory stimulus alters sensory processing only after multiple pairing sessions, this alteration remains after cessation of pairing sessions, and the alteration selectively affects the response properties of neurons encoding the specific paired sensory stimulus. Here, we call attention to the immediate effects VNS has on sensory processing. This review discusses existing studies on this topic, provides an overview of the underlying neuromodulatory systems that likely play a role, and briefly explores the potential translational applications of using VNS to rapidly regulate sensory processing.
Collapse
Affiliation(s)
- Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
- Jacobs Technion-Cornell Institute, Cornell Tech, New York, NY, United States
- *Correspondence: Charles Rodenkirch,
| | - Jason B. Carmel
- Department of Neurology and Orthopedics, Columbia University Medical Center, New York, NY, United States
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
- Qi Wang,
| |
Collapse
|
43
|
Non-invasive vagus nerve stimulation in epilepsy patients enhances cooperative behavior in the prisoner's dilemma task. Sci Rep 2022; 12:10255. [PMID: 35715460 PMCID: PMC9205877 DOI: 10.1038/s41598-022-14237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/03/2022] [Indexed: 11/24/2022] Open
Abstract
The vagus nerve constitutes a key link between the autonomic and the central nervous system. Previous studies provide evidence for the impact of vagal activity on distinct cognitive processes including functions related to social cognition. Recent studies in animals and humans show that vagus nerve stimulation is associated with enhanced reward-seeking and dopamine-release in the brain. Social interaction recruits similar brain circuits to reward processing. We hypothesize that vagus nerve stimulation (VNS) boosts rewarding aspects of social behavior and compare the impact of transcutaneous VNS (tVNS) and sham stimulation on social interaction in 19 epilepsy patients in a double-blind pseudo-randomized study with cross-over design. Using a well-established paradigm, i.e., the prisoner’s dilemma, we investigate effects of stimulation on cooperative behavior, as well as interactions of stimulation effects with patient characteristics. A repeated-measures ANOVA and a linear mixed-effects model provide converging evidence that tVNS boosts cooperation. Post-hoc correlations reveal that this effect varies as a function of neuroticism, a personality trait linked to the dopaminergic system. Behavioral modeling indicates that tVNS induces a behavioral starting bias towards cooperation, which is independent of the decision process. This study provides evidence for the causal influence of vagus nerve activity on social interaction.
Collapse
|
44
|
Uchida Y, Hikida T, Yamashita Y. Computational Mechanisms of Osmoregulation: A Reinforcement Learning Model for Sodium Appetite. Front Neurosci 2022; 16:857009. [PMID: 35663557 PMCID: PMC9160331 DOI: 10.3389/fnins.2022.857009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/15/2022] [Indexed: 11/30/2022] Open
Abstract
Homeostatic control with oral nutrient intake is a vital complex system involving the orderly interactions between the external and internal senses, behavioral control, reward learning, and decision-making. Sodium appetite is a representative system and has been intensively investigated in animal models of homeostatic systems and oral nutrient intake. However, the system-level mechanisms for regulating sodium intake behavior and homeostatic control remain unclear. In the current study, we attempted to provide a mechanistic understanding of sodium appetite behavior by using a computational model, the homeostatic reinforcement learning model, in which homeostatic behaviors are interpreted as reinforcement learning processes. Through simulation experiments, we confirmed that our homeostatic reinforcement learning model successfully reproduced homeostatic behaviors by regulating sodium appetite. These behaviors include the approach and avoidance behaviors to sodium according to the internal states of individuals. In addition, based on the assumption that the sense of taste is a predictor of changes in the internal state, the homeostatic reinforcement learning model successfully reproduced the previous paradoxical observations of the intragastric infusion test, which cannot be explained by the classical drive reduction theory. Moreover, we extended the homeostatic reinforcement learning model to multimodal data, and successfully reproduced the behavioral tests in which water and sodium appetite were mediated by each other. Finally, through an experimental simulation of chemical manipulation in a specific neural population in the brain stem, we proposed a testable hypothesis for the function of neural circuits involving sodium appetite behavior. The study results support the idea that osmoregulation via sodium appetitive behavior can be understood as a reinforcement learning process, and provide a mechanistic explanation for the underlying neural mechanisms of decision-making related to sodium appetite and homeostatic behavior.
Collapse
Affiliation(s)
- Yuuki Uchida
- Department of Information Medicine, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Medical and Dental Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
- *Correspondence: Takatoshi Hikida,
| | - Yuichi Yamashita
- Department of Information Medicine, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Yuichi Yamashita,
| |
Collapse
|
45
|
Luquet S, Gangarossa G. Dopamine drives food craving during pregnancy. Nat Metab 2022; 4:410-411. [PMID: 35379971 DOI: 10.1038/s42255-022-00555-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Serge Luquet
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France.
| | - Giuseppe Gangarossa
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France.
| |
Collapse
|
46
|
Berland C, Castel J, Terrasi R, Montalban E, Foppen E, Martin C, Muccioli GG, Luquet S, Gangarossa G. Identification of an endocannabinoid gut-brain vagal mechanism controlling food reward and energy homeostasis. Mol Psychiatry 2022; 27:2340-2354. [PMID: 35075269 DOI: 10.1038/s41380-021-01428-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 12/07/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022]
Abstract
The regulation of food intake, a sine qua non requirement for survival, thoroughly shapes feeding and energy balance by integrating both homeostatic and hedonic values of food. Unfortunately, the widespread access to palatable food has led to the development of feeding habits that are independent from metabolic needs. Among these, binge eating (BE) is characterized by uncontrolled voracious eating. While reward deficit seems to be a major contributor of BE, the physiological and molecular underpinnings of BE establishment remain elusive. Here, we combined a physiologically relevant BE mouse model with multiscale in vivo approaches to explore the functional connection between the gut-brain axis and the reward and homeostatic brain structures. Our results show that BE elicits compensatory adaptations requiring the gut-to-brain axis which, through the vagus nerve, relies on the permissive actions of peripheral endocannabinoids (eCBs) signaling. Selective inhibition of peripheral CB1 receptors resulted in a vagus-dependent increased hypothalamic activity, modified metabolic efficiency, and dampened activity of mesolimbic dopamine circuit, altogether leading to the suppression of palatable eating. We provide compelling evidence for a yet unappreciated physiological integrative mechanism by which variations of peripheral eCBs control the activity of the vagus nerve, thereby in turn gating the additive responses of both homeostatic and hedonic brain circuits which govern homeostatic and reward-driven feeding. In conclusion, we reveal that vagus-mediated eCBs/CB1R functions represent an interesting and innovative target to modulate energy balance and counteract food-reward disorders.
Collapse
Affiliation(s)
- Chloé Berland
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Julien Castel
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Romano Terrasi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200, Brussels, Belgium
| | - Enrica Montalban
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Ewout Foppen
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Claire Martin
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200, Brussels, Belgium
| | - Serge Luquet
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France
| | - Giuseppe Gangarossa
- Université de Paris, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013, Paris, France.
| |
Collapse
|
47
|
Brain circuits for promoting homeostatic and non-homeostatic appetites. Exp Mol Med 2022; 54:349-357. [PMID: 35474340 PMCID: PMC9076862 DOI: 10.1038/s12276-022-00758-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 11/28/2022] Open
Abstract
As the principal means of acquiring nutrients, feeding behavior is indispensable to the survival and well-being of animals. In response to energy or nutrient deficits, animals seek and consume food to maintain energy homeostasis. On the other hand, even when animals are calorically replete, non-homeostatic factors, such as the sight, smell, and taste of palatable food, or environmental cues that predict food, can stimulate feeding behavior. These homeostatic and non-homeostatic factors have traditionally been investigated separately, but a growing body of literature highlights that these factors work synergistically to promote feeding behavior. Furthermore, recent breakthroughs in cell type-specific and circuit-specific labeling, recording, and manipulation techniques have markedly accelerated the discovery of well-defined neural populations underlying homeostatic and non-homeostatic appetite control, as well as overlapping circuits that contribute to both types of appetite. This review aims to provide an update on our understanding of the neural circuit mechanisms for promoting homeostatic and non-homeostatic appetites, focusing on the function of recently identified, genetically defined cell types. Research on the neural circuit mechanisms underlying feeding behaviors is critical to identifying therapeutic targets for food-related disorders like obesity and anorexia. Sung-Yon Kim and colleagues at Seoul National University, South Korea, reviewed the current understanding of neural circuits promoting feeding behavior, which is regulated by homeostatic and non-homeostatic appetites. In response to deficits in energy (caloric) or nutrients, specific populations of neurons sensitive to hormones leptin and ghrelin generate homeostatic appetite and promote feeding. In addition, diverse neural populations stimulate non-homeostatic appetite in the absence of immediate internal needs and are thought to drive overconsumption in the modern obesogenic environment. These appetites extensively interact through overlapping neural circuits to jointly promote feeding behaviors.
Collapse
|
48
|
Obray JD, Jang EY, Klomp AM, Small CA, Richardson AP, LeBaron JJ, Lee JG, Yorgason JT, Yang CH, Steffensen SC. The peripheral dopamine 2 receptor antagonist domperidone attenuates ethanol enhancement of dopamine levels in the nucleus accumbens. Alcohol Clin Exp Res 2022; 46:396-409. [PMID: 35040146 PMCID: PMC8920780 DOI: 10.1111/acer.14775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 12/28/2021] [Accepted: 01/08/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Dopamine neuron firing in the ventral tegmental area (VTA) and dopamine release in the nucleus accumbens have been implicated in reward learning. Ethanol is known to increase both dopamine neuron firing in the VTA and dopamine levels in the nucleus accumbens. Despite this, some discrepancies exist between the dose of ethanol required to enhance firing in vivo and ex vivo. In the present study we investigated the effects of peripheral dopamine 2 subtype receptor antagonism on ethanol's effects on dopamine neurotransmission. METHODS Plasma catecholamine levels were assessed following ethanol administration across four different doses of EtOH. Microdialysis and voltammetry were used to assess the effects of domperidone pretreatment on ethanol-mediated increases in dopamine release in the nucleus accumbens. A place conditioning paradigm was used to assess conditioned preference for ethanol and whether domperidone pretreatment altered this preference. Open-field and loss-of-righting reflex paradigms were used to assess the effects of domperidone on ethanol-induced sedation. A rotarod apparatus was used to assess the effects of domperidone on ethanol-induced motor impairment. RESULTS Domperidone attenuated ethanol's enhancement of mesolimbic dopamine release under non-physiological conditions at intermediate (1.0 and 2.0 g/kg) doses of ethanol. Domperidone also decreased EtOH-induced sedation at 2.0 g/kg. Domperidone did not alter ethanol conditioned place preference nor did it affect ethanol-induced motor impairment. CONCLUSIONS These results show that peripheral dopamine 2 receptors mediate some of the effects of ethanol on nonphysiological dopamine neurotransmission, although these effects are not related to the rewarding properties of ethanol.
Collapse
Affiliation(s)
- James Daniel Obray
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA,Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Eun Young Jang
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA,Research Center for Convergence Toxicology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Anneke M. Klomp
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Christina A. Small
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Aaron P. Richardson
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Joshua J. LeBaron
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Jin Gyeom Lee
- College of Korean Medicine, Daegu Haany University, Daegu, South Korea
| | - Jordan T. Yorgason
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Chae Ha Yang
- College of Korean Medicine, Daegu Haany University, Daegu, South Korea
| | - Scott C. Steffensen
- Department of Psychology, Center for Neuroscience, Brigham Young University, Provo, Utah, USA
| |
Collapse
|
49
|
Hanssen R, Thanarajah SE, Tittgemeyer M, Brüning JC. Obesity - A Matter of Motivation? Exp Clin Endocrinol Diabetes 2022; 130:290-295. [PMID: 35181879 PMCID: PMC9286865 DOI: 10.1055/a-1749-4852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Excessive food intake and reduced physical activity have long been established as
primary causes of obesity. However, the underlying mechanisms causing this
unhealthy behavior characterized by heightened motivation for food but not for
physical effort are unclear. Despite the common unjustified stigmatization that
obesity is a result of laziness and lack of discipline, it is becoming
increasingly clear that high-fat diet feeding and obesity cause alterations in
brain circuits that are critical for the control of motivational behavior. In this mini-review, we provide a comprehensive overview of incentive motivation,
its neural encoding in the dopaminergic mesolimbic system as well as its
metabolic modulation with a focus on derangements of incentive motivation in
obesity. We further discuss the emerging field of metabolic interventions to
counteract motivational deficits and their potential clinical implications.
Collapse
Affiliation(s)
- Ruth Hanssen
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sharmili E Thanarajah
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
50
|
Prescott SL, Liberles SD. Internal senses of the vagus nerve. Neuron 2022; 110:579-599. [PMID: 35051375 PMCID: PMC8857038 DOI: 10.1016/j.neuron.2021.12.020] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/30/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022]
Abstract
The vagus nerve is an indispensable body-brain connection that controls vital aspects of autonomic physiology like breathing, heart rate, blood pressure, and gut motility, reflexes like coughing and swallowing, and survival behaviors like feeding, drinking, and sickness responses. Classical physiological studies and recent molecular/genetic approaches have revealed a tremendous diversity of vagal sensory neuron types that innervate different internal organs, with many cell types remaining poorly understood. Here, we review the state of knowledge related to vagal sensory neurons that innervate the respiratory, cardiovascular, and digestive systems. We focus on cell types and their response properties, physiological/behavioral roles, engaged neural circuits and, when possible, sensory receptors. We are only beginning to understand the signal transduction mechanisms used by vagal sensory neurons and upstream sentinel cells, and future studies are needed to advance the field of interoception to the level of mechanistic understanding previously achieved for our external senses.
Collapse
|