1
|
Bodnar RJ, Castillo A, Carata I, Bochner Y, Sarker J, Rayman N, Narine S, Pines R, Limbu B, Sclafani A. Role of glutamatergic signaling in the acquisition and expression of learned sugar preferences in C57BL/6 mice. Physiol Behav 2025; 290:114748. [PMID: 39547434 DOI: 10.1016/j.physbeh.2024.114748] [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: 08/21/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
C57BL/6 (B6) mice learn to prefer glucose or sucrose to initially isopreferred or even more preferred nonnutritive sweeteners due to the postoral appetite stimulating (appetition) actions of glucose. Recent evidence indicates that specific duodenal neuropod cells transmit the glucose appetition signal to the brain via glutamatergic synaptic connections with vagal afferents. The present study found that intraperitoneal pretreatment with a glutamatergic receptor antagonist cocktail (kynurenic acid (KA)/D-2-amino-3-phosphonopentanoic acid (AP3)) in B6 mice did not block the expression of their learned preference for 8% glucose solution over an initially-preferred 0.1% sucralose + 0.1% saccharin solution. However, acquisition of the glucose preference was blocked by drug treatment during 1-h training sessions with the two sweeteners. Systemic KA/AP3 injections also did not block the expression of the learned preference for a 10.6% sucrose solution over a 0.6% sucralose solution. Drug effects on the acquisition of the sucrose preference were not determined because sucrose, unlike glucose conditioning, required 24-h training trials. The findings that the 1-h training regimen conditioned 8% glucose, but not 10.6% sucrose, preferences suggest that glucose has more potent appetition actions. This was confirmed by the finding that B6 mice learned to prefer 10.6% glucose to 10.6% sucrose after 1-h or 24-h training despite an initial strong sucrose preference. This action can be explained by 10.6% sucrose's digestion in the gut to glucose and fructose with only glucose activating the gut-brain appetition pathway.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA; Psychology Doctoral Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Alexander Castillo
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Ion Carata
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Yerachmiel Bochner
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Joymin Sarker
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Nareesa Rayman
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Shania Narine
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Rachel Pines
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Brian Limbu
- Department of Psychology, Queens College, City University of New York, Queens, NY, USA
| | - Anthony Sclafani
- Department of Psychology, Brooklyn College, City University of New York, Brooklyn, NY, USA; Psychology Doctoral Program, The Graduate Center, City University of New York, New York, NY, USA.
| |
Collapse
|
2
|
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 regulates 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
|
3
|
Wang H, Lou R, Wang Y, Hao L, Wang Q, Li R, Su J, Liu S, Zhou X, Gao X, Hao Q, Chen Z, Xu Y, Wu C, Zheng Y, Guo Q, Bai L. Parallel gut-to-brain pathways orchestrate feeding behaviors. Nat Neurosci 2025; 28:320-335. [PMID: 39627537 DOI: 10.1038/s41593-024-01828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/29/2024] [Indexed: 02/08/2025]
Abstract
The caudal nucleus of the solitary tract (cNTS) in the brainstem serves as a hub for integrating interoceptive cues from diverse sensory pathways. However, the mechanisms by which cNTS neurons transform these signals into behaviors remain debated. We analyzed 18 cNTS-Cre mouse lines and cataloged the dynamics of nine cNTS cell types during feeding. We show that Th+ cNTS neurons encode esophageal mechanical distension and transient gulp size via vagal afferent inputs, providing quick feedback regulation of ingestion speed. By contrast, Gcg+ cNTS neurons monitor intestinal nutrients and cumulative ingested calories and have long-term effects on food satiation and preference. These nutritive signals are conveyed through a portal vein-spinal ascending pathway rather than vagal sensory neurons. Our findings underscore distinctions among cNTS subtypes marked by differences in temporal dynamics, sensory modalities, associated visceral organs and ascending sensory pathways, all of which contribute to specific functions in coordinated feeding regulation.
Collapse
Affiliation(s)
- Hongyun Wang
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Runxiang Lou
- Chinese Institute for Brain Research, Beijing, China
| | - Yunfeng Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Liufang Hao
- Chinese Institute for Brain Research, Beijing, China
| | - Qiushi Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Rui Li
- Chinese Institute for Brain Research, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Leaning, Beijing Normal University, Beijing, China
| | - Jiayi Su
- Chinese Institute for Brain Research, Beijing, China
| | - Shuhan Liu
- Chinese Institute for Brain Research, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Leaning, Beijing Normal University, Beijing, China
| | - Xiangyu Zhou
- Chinese Institute for Brain Research, Beijing, China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing, China
| | - Qianxi Hao
- Chinese Institute for Brain Research, Beijing, China
| | - Zihe Chen
- Chinese Institute for Brain Research, Beijing, China
| | - Yibo Xu
- Chinese Institute for Brain Research, Beijing, China
| | - Chongwei Wu
- Chinese Institute for Brain Research, Beijing, China
| | - Yang Zheng
- Chinese Institute for Brain Research, Beijing, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, China
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Ling Bai
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
4
|
Sampson TR, Tansey MG, West AB, Liddle RA. Lewy body diseases and the gut. Mol Neurodegener 2025; 20:14. [PMID: 39885558 PMCID: PMC11783828 DOI: 10.1186/s13024-025-00804-5] [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] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Gastrointestinal (GI) involvement in Lewy body diseases (LBDs) has been observed since the initial descriptions of patients by James Parkinson. Recent experimental and human observational studies raise the possibility that pathogenic alpha-synuclein (⍺-syn) might develop in the GI tract and subsequently spread to susceptible brain regions. The cellular and mechanistic origins of ⍺-syn propagation in disease are under intense investigation. Experimental LBD models have implicated important contributions from the intrinsic gut microbiome, the intestinal immune system, and environmental toxicants, acting as triggers and modifiers to GI pathologies. Here, we review the primary clinical observations that link GI dysfunctions to LBDs. We first provide an overview of GI anatomy and the cellular repertoire relevant for disease, with a focus on luminal-sensing cells of the intestinal epithelium including enteroendocrine cells that express ⍺-syn and make direct contact with nerves. We describe interactions within the GI tract with resident microbes and exogenous toxicants, and how these may directly contribute to ⍺-syn pathology along with related metabolic and immunological responses. Finally, critical knowledge gaps in the field are highlighted, focusing on pivotal questions that remain some 200 years after the first descriptions of GI tract dysfunction in LBDs. We predict that a better understanding of how pathophysiologies in the gut influence disease risk and progression will accelerate discoveries that will lead to a deeper overall mechanistic understanding of disease and potential therapeutic strategies targeting the gut-brain axis to delay, arrest, or prevent disease progression.
Collapse
Affiliation(s)
- Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30329, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Malú Gámez Tansey
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Normal Fixel Institute of Neurological Diseases, Gainesville, FL, 32608, USA
| | - Andrew B West
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Center for Neurodegeneration and Neurotherapeutic Research, Department of Pharmacology and Cancer Biology, Durham, NC, 27710, USA.
| | - Rodger A Liddle
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA.
- Department of Medicine, Duke University and Department of Veterans Affairs Health Care System, Durham, NC, 27710, USA.
| |
Collapse
|
5
|
Ohara TE, Hsiao EY. Microbiota-neuroepithelial signalling across the gut-brain axis. Nat Rev Microbiol 2025:10.1038/s41579-024-01136-9. [PMID: 39743581 DOI: 10.1038/s41579-024-01136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Research over the past two decades has established a remarkable ability of the gut microbiota to modulate brain activity and behaviour. Conversely, signals from the brain can influence the composition and function of the gut microbiota. This bidirectional communication across the gut microbiota-brain axis, involving multiple biochemical and cellular mediators, is recognized as a major brain-body network that integrates cues from the environment and the body's internal state. Central to this network is the gut sensory system, formed by intimate connections between chemosensory epithelial cells and sensory nerve fibres, that conveys interoceptive signals to the central nervous system. In this Review, we provide a broad overview of the pathways that connect the gut and the brain, and explore the complex dialogue between microorganisms and neurons at this emerging intestinal neuroepithelial interface. We highlight relevant microbial factors, endocrine cells and neural mechanisms that govern gut microbiota-brain interactions and their implications for gastrointestinal and neuropsychiatric health.
Collapse
Affiliation(s)
- Takahiro E Ohara
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA.
- UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
6
|
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
|
7
|
Yang C, Liu L, Cui C, Cai H, Dai Q, Chen G, McClements DJ, Hou R. Towards healthier low-sugar and low-fat beverages: Design, production, and characterization. Food Res Int 2025; 200:115457. [PMID: 39779115 DOI: 10.1016/j.foodres.2024.115457] [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: 08/14/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Many consumers are adopting low-sugar and low-fat beverages to avoid excessive calories and the negative impact of high trans- and/or saturated fat on health and wellbeing. This article reviews strategies to reduce sugar, fat, and high trans- and/or saturated fat content in beverages while maintaining their desirable physicochemical and sensory attributes. It assesses the impact of various sugar and fat replacers on the aroma, taste, texture, appearance, and nutritional profile of beverages. Combinations of natural sugar replacers and protein or polysaccharide-based fat replacers have shown partial success in mimicking the qualities of sucrose and fat. Future strategies for designing low-sugar and low-fat beverages include developing novel replacers and using odorants to enhance sensory profiles. The article also highlights methods for flavor detection and oral tribology methods, emphasizing their role in development of low-sugar and low-fat beverages. The information presented in this review article is intended to stimulate research into the design of healthier low-sugar and low-fat beverages in the future.
Collapse
Affiliation(s)
- Chen Yang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Lianliang Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315832, Zhejiang Province, China
| | - Chuanjian Cui
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Huimei Cai
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Qianying Dai
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, China
| | | | - Ruyan Hou
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, China; Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, New-style Industrial Tea Beverage Green Manufacturing Joint Laboratory of Anhui Province, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
8
|
Glendinning JI, Williams N. Fighting Fire with Fire: Impact of Sugary Diets on Metabolically Deranged Mice. Nutrients 2024; 17:100. [PMID: 39796534 PMCID: PMC11722652 DOI: 10.3390/nu17010100] [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: 11/15/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: There is controversy about the health risks of sugary diets. A recent study reported that chronic consumption of 11% sugar solutions improved glycemic control in lean mice. Based on this finding, we hypothesized that chronic consumption of the same 11% sugar solutions would also improve glycemic control in metabolically deranged mice. Methods: We exposed mice to a high-fat/high-sugar diet for 12 weeks. Then, we switched the mice to a control (i.e., standard chow) or one of four experimental diets for 8 weeks. The experimental diets contained standard chow plus an 11% solution of glucose or high-fructose syrup. The sugar syrups were derived from corn or cellulose. We included the cellulosic syrups because they contain polyphenols, which are thought to promote glycemic control. We measured body weight, adiposity, glucose tolerance, insulinemia, insulin sensitivity, body composition, and avidity for sweeteners. Results: Mice switched to the control diet lost weight, whereas mice switched to the experimental diets remained obese and hyperinsulinemic. Thus, the experimental diets did not cause the mice to regain normal metabolic health. Nevertheless, we observed (i) improvements in glucose tolerance in mice on both the control and experimental diets; (ii) reduced insulinemia and enhanced insulin sensitivity in mice offered the cellulosic syrups; (iii) elevations in cephalic-phase insulin responses in mice on the experimental diets; and (iv) increased avidity for sweeteners in mice on the control but not the experimental diets. Conclusions: Switching metabolically deranged mice to the experimental diets, particularly those with cellulosic sugars, improved glucose tolerance and insulin sensitivity.
Collapse
Affiliation(s)
- John I. Glendinning
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA;
| | - Niki Williams
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA;
| |
Collapse
|
9
|
Snyder JR, Ahmed M, Bhave S, Hotta R, Koppes RA, Goldstein AM, Koppes AN. Enteroendocrine Cells Sense Sucrose and Alter Enteric Neuron Excitability via Insulin Signaling. Adv Biol (Weinh) 2024:e2300566. [PMID: 39703141 DOI: 10.1002/adbi.202300566] [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: 07/19/2024] [Revised: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Neurosensory circuits of the gastrointestinal tract sense microbial and nutrient changes in the gut; however, studying these circuits in vivo is hindered by invasive techniques and ethical concerns. Here, an in vitro model of enteroendocrine cells (EECs) and calcium reporting enteric neurons (ENs) is established and validated for functional signaling. Both mechanical and sucrose stimulation of co-cultures increased the percentage of neurons undergoing a calcium flux, indicating an action potential. Neuronal activation is blocked with either a piezo or insulin receptor blocker. At baseline, a flow only stimulus elicited 51.9% of neurons to activate in co-culture, which is decreased to 15.1% with a piezo blocker. Piezo blocked and sucrose stimulated EECs increased neuronal activation to 43.9%, and an insulin blocker reduced response to 12.4%. Since a cell line is used to model the EEC in the previous experiments, primary rat duodenal epithelium enriched for EECs are also stimulated and found to produced measurable insulin. This work shows the ability of EECs to produce insulin and for ENs to sense insulin. These results inspire further work on how insulin production outside the pancreas effects diabetes, insulin as a neurotransmitter, and exploration of additional nutritional and microbiotic stimuli on enteroendocrine-to-neuronal signaling.
Collapse
Affiliation(s)
- Jessica R Snyder
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Minhal Ahmed
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 185 Cambridge St, CPZN 6-215, Boston, MA, 02114, USA
| | - Abigail N Koppes
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| |
Collapse
|
10
|
Kelly AL, Baugh ME, Ahrens ML, Valle AN, Sullivan RM, Oster ME, Fowler ME, Carter BE, Davy BM, Hanlon AL, DiFeliceantonio AG. Neural and metabolic factors in carbohydrate reward: Rationale, design, and methods for a flavor-nutrient learning paradigm in humans. Contemp Clin Trials 2024; 147:107717. [PMID: 39413990 PMCID: PMC11688656 DOI: 10.1016/j.cct.2024.107717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/05/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Overconsumption of ultra-processed foods (UPFs), which are linked with adverse health outcomes, is a growing public health concern. UPFs deliver highly bioavailable calories rapidly, which may contribute to their reinforcing potential and drive overconsumption. Our primary aim is to test the role of speed of nutrient availability on reward learning. We hypothesize that brain activity in reward related areas and behavioral preferences will be greater to a flavored drink predicting rapidly available calories (CS + Fast) compared with a flavored drink predicting more slowly available (CS + Slow) or no (CS-) calories. Participants (n = 64, aged 18-45 years, will consume 3 novel flavored, isosweet beverages containing 110 kcal of sucrose (CS + Fast), 110 kcal of maltodextrin (CS + Slow), or 0-kcal sucralose (CS-) 6 times in randomized, crossover order. Blood metabolites and indirect calorimetry measures, including metabolic rate and carbohydrate oxidation, will be assessed before and for 1 h after beverage consumption. Behavioral preference for beverages will be assessed in a pre- and post-test. Brain response to each flavor without calories will be assessed via functional magnetic resonance imaging in a post-test. Findings from this study will contribute to the understanding of basic mechanisms that may drive overconsumption of UPFs. Trial registration:clinicaltrials.gov registration #NCT06053294.
Collapse
Affiliation(s)
- Amber L Kelly
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Center for Health Behaviors Research at Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Translational Biology, Medicine, and Health, Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - Mary Elizabeth Baugh
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Center for Health Behaviors Research at Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - Monica L Ahrens
- Center for Biostatistics and Health Data Science, Department of Statistics, Blacksburg, VA, USA
| | - Abigail N Valle
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | | | - Mary E Oster
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Center for Health Behaviors Research at Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - Mary E Fowler
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Center for Health Behaviors Research at Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - Bridget E Carter
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Center for Health Behaviors Research at Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - Brenda M Davy
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Alexandra L Hanlon
- Center for Biostatistics and Health Data Science, Department of Statistics, Blacksburg, VA, USA
| | - Alexandra G DiFeliceantonio
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Center for Health Behaviors Research at Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
11
|
Alcaino C, Reimann F, Gribble FM. Incretin hormones and obesity. J Physiol 2024:10.1113/JP286293. [PMID: 39576749 PMCID: PMC7617301 DOI: 10.1113/jp286293] [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: 08/28/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024] Open
Abstract
The incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) play critical roles in co-ordinating postprandial metabolism, including modulation of insulin secretion and food intake. They are secreted from enteroendocrine cells in the intestinal epithelium following food ingestion, and act at multiple target sites including pancreatic islets and the brain. With the recent development of agonists targeting GLP-1 and GIP receptors for the treatment of type 2 diabetes and obesity, and the ongoing development of new incretin-based drugs with improved efficacy, there is great interest in understanding the physiology and pharmacology of these hormones.
Collapse
Affiliation(s)
- Constanza Alcaino
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, CambridgeCB2 0QQ, UK
| | - Frank Reimann
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, CambridgeCB2 0QQ, UK
| | - Fiona M Gribble
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Addenbrooke’s Hospital, CambridgeCB2 0QQ, UK
| |
Collapse
|
12
|
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
|
13
|
Asim M, Wang H, Waris A, Qianqian G, Chen X. Cholecystokinin neurotransmission in the central nervous system: Insights into its role in health and disease. Biofactors 2024; 50:1060-1075. [PMID: 38777339 PMCID: PMC11627476 DOI: 10.1002/biof.2081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Cholecystokinin (CCK) plays a key role in various brain functions, including both health and disease states. Despite the extensive research conducted on CCK, there remain several important questions regarding its specific role in the brain. As a result, the existing body of literature on the subject is complex and sometimes conflicting. The primary objective of this review article is to provide a comprehensive overview of recent advancements in understanding the central nervous system role of CCK, with a specific emphasis on elucidating CCK's mechanisms for neuroplasticity, exploring its interactions with other neurotransmitters, and discussing its significant involvement in neurological disorders. Studies demonstrate that CCK mediates both inhibitory long-term potentiation (iLTP) and excitatory long-term potentiation (eLTP) in the brain. Activation of the GPR173 receptor could facilitate iLTP, while the Cholecystokinin B receptor (CCKBR) facilitates eLTP. CCK receptors' expression on different neurons regulates activity, neurotransmitter release, and plasticity, emphasizing CCK's role in modulating brain function. Furthermore, CCK plays a pivotal role in modulating emotional states, Alzheimer's disease, addiction, schizophrenia, and epileptic conditions. Targeting CCK cell types and circuits holds promise as a therapeutic strategy for alleviating these brain disorders.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
- Department of Biomedical ScienceCity University of Hong KongKowloon TongHong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of SciencesPak Shek KokHong Kong
| | - Huajie Wang
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
| | - Abdul Waris
- Department of Biomedical ScienceCity University of Hong KongKowloon TongHong Kong
| | - Gao Qianqian
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
| | - Xi Chen
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
- Department of Biomedical ScienceCity University of Hong KongKowloon TongHong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of SciencesPak Shek KokHong Kong
| |
Collapse
|
14
|
Welch HF, Sankaranarayanan I, Hong VM, Mazhar K, Kolber BJ, Price TJ, Thorn CA. Lateralized nodose ganglia gene expression implicates cholecystokinin receptors in interoceptive reward signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621506. [PMID: 39554115 PMCID: PMC11565973 DOI: 10.1101/2024.11.01.621506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The vagus nerves are important carriers of sensory information from the viscera to the central nervous system. Emerging evidence suggests that sensory signaling through the right, but not the left, vagus nerve evokes striatal dopamine release and reinforces appetitive behaviors. However, the extent to which differential gene expression within vagal sensory neurons contributes to this asymmetric reward-related signaling remains unknown. Here, we use single-cell RNA sequencing to identify genes that are differentially expressed between the left and right nodose ganglia (NG) to identify candidate genes likely to contribute to vagus-mediated reward signaling. We find that a group of neurons expressing Chrna3 (nicotinic acetylcholine receptor subunit 3) and Cckar (cholecystokinin A receptor) is preferentially expressed in the right NG of both rats and mice. This result suggests that differential expression of gut-innervating nutrient sensors in NG neurons may contribute to asymmetric encoding of interoceptive rewards by the vagus nerves.
Collapse
|
15
|
Warner JO. Artificial food additives: hazardous to long-term health? Arch Dis Child 2024; 109:882-885. [PMID: 38423749 DOI: 10.1136/archdischild-2023-326565] [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: 11/02/2023] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
Many additives, some of which have no nutritional value, can be legally used in processed foods. They intensify colour, thicken, increase shelf life and enhance flavour. Regulatory authorities issue approvals as safe within acceptable quantitative limits. Ultra-processed foods (UPFs) contain combinations of all these additives and are particularly attractive to children.Many publications suggest that artificial colourants, benzoate preservatives, non-caloric sweeteners, emulsifiers and their degradation derivatives have adverse effects by increasing risks of mental health disorders, attention deficit hyperactivity disorder, cardiovascular disease, metabolic syndrome and potential carcinogenic effects.A systematic review has established that artificial azo dye food colourants (AFCs) and sodium benzoate preservative cause disturbed behaviour in children. AFCs and benzoates in animal models have neurotoxic properties through gut microbial generation of toxic metabolites. Observational studies show associations between high emulsifier intake and cardiovascular disease. Animal models and in vitro studies have highlighted neurotoxic, cytotoxic, genotoxic and carcinogenic effects. High intake of non-caloric sweeteners has been linked to cardiovascular disease and depression in adults and is linked to childhood obesity.Little research has focused on children who are the largest consumers of UPFs. Potentially, they are a ticking time bomb for adult obesity, metabolic syndrome, cardiovascular diseases, mental health disorders and cancers. Based on risk/benefit analysis, azo dye AFCs should be banned. Benzoates, emulsifiers and sweeteners require assessment of quantitative limits and cumulative effects of combinations. Consumers purchasing UPFs require information through ingredient health warnings and recommendations to use natural unprocessed foods which have well-described health-promoting properties.
Collapse
Affiliation(s)
- John O Warner
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
16
|
Alway E, Reicher N, Bohórquez DV. Deciphering visceral instincts: a scientific quest to unravel food choices from molecules to mind. Genes Dev 2024; 38:798-801. [PMID: 39362782 PMCID: PMC11535160 DOI: 10.1101/gad.352279.124] [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] [Indexed: 10/05/2024]
Abstract
The study of biological mechanisms, while crucial, cannot fully explain complex phenomena like the instinct to eat. The mind-body connection, as exemplified by the concept of "voodoo death," highlights the profound influence of belief and cultural context on physiology. Indigenous knowledge systems further emphasize the interconnectedness of humans with their environment. Recent discoveries in gut-brain communication reveal the intricate neural circuits that drive our visceral desires, but a holistic approach that integrates both physiological mechanisms and the subjective experience of life, informed by diverse cultural perspectives, will be essential to truly understand what it means to be alive.
Collapse
Affiliation(s)
- Emily Alway
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, North Carolina 27710; USA;
- Department of Medicine, Duke University, Durham, North Carolina 27710; USA
- Department of Neurobiology, Duke University, Durham, North Carolina 27710; USA
| | - Naama Reicher
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, North Carolina 27710; USA
- Department of Medicine, Duke University, Durham, North Carolina 27710; USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, North Carolina 27710; USA
- Department of Medicine, Duke University, Durham, North Carolina 27710; USA
- Department of Neurobiology, Duke University, Durham, North Carolina 27710; USA
- Department of Pathology, Duke University, Durham, North Carolina 27710; USA
- Department of Cell Biology, Duke University, Durham, North Carolina 27710; USA
- Duke Institute for Brain Sciences, Duke University, Durham, North Carolina 27710; USA
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [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
|
20
|
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
|
21
|
Sclafani A, Ackroff K. Glucose appetition in C57BL/6J mice: Influence of nonnutritive sweetener experience, food deprivation state and sex differences. Physiol Behav 2024; 283:114596. [PMID: 38815713 PMCID: PMC11246822 DOI: 10.1016/j.physbeh.2024.114596] [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: 03/08/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
In addition to its sweet taste, glucose has potent and rapid postoral actions (appetition) that enhance its reward value. This has been demonstrated by the experience-induced preference for glucose over initially preferred nonnutritive sweetener solutions in 24-h choice tests. However, some sweetener solutions (e.g., 0.8% sucralose) have inhibitory postoral actions that may exaggerate glucose appetition whereas others (e.g., 0.1% sucralose + 0.1% saccharin, S+S) do not. Experiment 1 revealed that food-restricted (FR) male C57BL/6J mice displayed similar rapid glucose appetition effects (stimulation of glucose licking within minutes) and conditioned flavor preferences following 1-h experience with flavored 0.8% sucralose or 0.1% S+S and 8% glucose solutions. Thus, the inhibitory effects of 0.8% sucralose observed in 24-h tests were not apparent in 1-h tests. Experiment 2 evaluated the effects of food deprivation state and sweetener concentration on glucose appetition in female mice. Unlike FR mice tested with 0.1% S+S and 8% glucose, ad libitum (AL) fed mice displayed no stimulation of 8% glucose licking in the 1-h tests. A second ad libitum group (AL) tested with 0.2% S+S and 16% glucose solutions displayed stimulation of 16% glucose licking by the third 1-h test. Both AL groups, like the FR group, developed a preference for the glucose-paired flavor over the S+S paired flavor. Thus, food restriction promotes increased glucose licking but is not required for a conditioned preference. The FR male mice (Exp. 1) and FR female mice (Exp. 2) showed similar appetition responses (licking stimulation and flavor preference) to 8% glucose.
Collapse
Affiliation(s)
- Anthony Sclafani
- Department of Psychology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| | - Karen Ackroff
- Department of Psychology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| |
Collapse
|
22
|
Shekarabi A, Qureishy I, Puglisi CH, Dalseth M, Vuong HE. Host-microbe interactions: communication in the microbiota-gut-brain axis. Curr Opin Microbiol 2024; 80:102494. [PMID: 38824840 PMCID: PMC11323153 DOI: 10.1016/j.mib.2024.102494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/04/2024]
Abstract
Animals harbor a diverse array of symbiotic micro-organisms that coexist in communities across different body sites. These microbes maintain host homeostasis and respond to environmental insults to impact host physiological processes. Trillions of indigenous microbes reside in the gastrointestinal tract and engage with the host central nervous system (microbiota-gut-brain axis) by modulating immune responses, interacting with gut intrinsic and extrinsic nervous system, and regulating neuromodulators and biochemicals. These gut microbiota to brain signaling pathways are constantly informed by each other and are hypothesized to mediate brain health across the lifespan. In this review, we will examine the crosstalk of gut microbiota to brain communications in neurological pathologies, with an emphasis on microbial metabolites and neuromodulators, and provide a discussion of recent advances that help elucidate the microbiota as a therapeutic target for treating brain and behavioral disorders.
Collapse
Affiliation(s)
- Aryan Shekarabi
- University of Minnesota Twin-Cities, Department of Pediatrics, Neonatology Division, USA
| | - Izhan Qureishy
- University of Minnesota Twin-Cities, Department of Pediatrics, Neonatology Division, USA
| | - Chloe H Puglisi
- University of Minnesota Twin-Cities, Department of Pediatrics, Neonatology Division, USA
| | - Marge Dalseth
- University of Minnesota Twin-Cities, Department of Pediatrics, Neonatology Division, USA
| | - Helen E Vuong
- University of Minnesota Twin-Cities, Department of Pediatrics, Neonatology Division, USA.
| |
Collapse
|
23
|
Chi ZC. Recent studies on gut-brain axis and irritable bowel syndrome. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:468-483. [DOI: 10.11569/wcjd.v32.i7.468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
|
24
|
Wang Y, Duan C, Du X, Zhu Y, Wang L, Hu J, Sun Y. Vagus Nerve and Gut-Brain Communication. Neuroscientist 2024:10738584241259702. [PMID: 39041416 DOI: 10.1177/10738584241259702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
The vagus nerve, as an important component of the gut-brain axis, plays a crucial role in the communication between the gut and brain. It influences food intake, fat metabolism, and emotion by regulating the gut-brain axis, which is closely associated with the development of gastrointestinal, psychiatric, and metabolism-related disorders. In recent years, significant progress has been made in understanding the vagus-mediated regulatory pathway, highlighting its profound implications in the development of many diseases. Here, we summarize the latest advancements in vagus-mediated gut-brain pathways and the novel interventions targeting the vagus nerve. This will provide valuable insights for future research on treatment of obesity and gastrointestinal and depressive disorders based on vagus nerve stimulation.
Collapse
Affiliation(s)
- Yiyang Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenxi Duan
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinyi Du
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| | - Lihua Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| | - Jun Hu
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Yanhong Sun
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
25
|
Glendinning JI, Archambeau A, Brouwer LR, Dennis A, Georgiou K, Ivanov J, Vayntrub R, Sclafani A. Mice Condition Cephalic-Phase Insulin Release to Flavors Associated with Postoral Actions of Concentrated Glucose. Nutrients 2024; 16:2250. [PMID: 39064693 PMCID: PMC11279997 DOI: 10.3390/nu16142250] [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: 06/19/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Rats can condition cephalic-phase insulin responses (CPIRs) to specific sounds or times of the day that predict food availability. The present study asked whether mice can condition a CPIR to the flavor of sapid solutions that produce postoral glucose stimulation. To this end, we subjected C57BL/6 mice to one of six experimental protocols. We varied both the duration of the five training sessions (i.e., 23 h or 1 h) and the nature of the training solution. In Experiment 1, consumption of a 0.61% saccharin solution was paired with IG co-infusion of a 16% glucose solution. In Experiments 2-6, the mice consumed a training solution containing a mixture of 0.61% saccharin + 16% glucose, 32% sucrose, 32% maltodextrin, flavored 32% maltodextrin, or 16% maltodextrin. We subsequently asked whether consumption of any of these fluids conditioned a CPIR to a test solution that produced a similar flavor, but which did not elicit a CPIR in naïve mice. The mice did condition a CPIR, but only to the solutions containing 32% maltodextrin. We attribute this conditioning to postoral actions of the concentrated maltodextrin solutions.
Collapse
Affiliation(s)
- John I. Glendinning
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA; (L.R.B.); (A.D.); (K.G.); (J.I.)
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA; (A.A.); (R.V.)
| | - Alix Archambeau
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA; (A.A.); (R.V.)
| | - Lillian R. Brouwer
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA; (L.R.B.); (A.D.); (K.G.); (J.I.)
| | - Alyson Dennis
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA; (L.R.B.); (A.D.); (K.G.); (J.I.)
| | - Kiriaki Georgiou
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA; (L.R.B.); (A.D.); (K.G.); (J.I.)
| | - Jessica Ivanov
- Department of Biology, Barnard College, Columbia University, New York, NY 10027, USA; (L.R.B.); (A.D.); (K.G.); (J.I.)
| | - Rochelle Vayntrub
- Department of Neuroscience & Behavior, Barnard College, Columbia University, New York, NY 10027, USA; (A.A.); (R.V.)
| | - Anthony Sclafani
- Department of Psychology, Brooklyn College of City University of New York, Brooklyn, NY 11210, USA;
| |
Collapse
|
26
|
Yoshida R, Ninomiya Y. Mechanisms and Functions of Sweet Reception in Oral and Extraoral Organs. Int J Mol Sci 2024; 25:7398. [PMID: 39000505 PMCID: PMC11242429 DOI: 10.3390/ijms25137398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The oral detection of sugars relies on two types of receptor systems. The first is the G-protein-coupled receptor TAS1R2/TAS1R3. When activated, this receptor triggers a downstream signaling cascade involving gustducin, phospholipase Cβ2 (PLCβ2), and transient receptor potential channel M5 (TRPM5). The second type of receptor is the glucose transporter. When glucose enters the cell via this transporter, it is metabolized to produce ATP. This ATP inhibits the opening of KATP channels, leading to cell depolarization. Beside these receptor systems, sweet-sensitive taste cells have mechanisms to regulate their sensitivity to sweet substances based on internal and external states of the body. Sweet taste receptors are not limited to the oral cavity; they are also present in extraoral organs such as the gastrointestinal tract, pancreas, and brain. These extraoral sweet receptors are involved in various functions, including glucose absorption, insulin release, sugar preference, and food intake, contributing to the maintenance of energy homeostasis. Additionally, sweet receptors may have unique roles in certain organs like the trachea and bone. This review summarizes past and recent studies on sweet receptor systems, exploring the molecular mechanisms and physiological functions of sweet (sugar) detection in both oral and extraoral organs.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Yuzo Ninomiya
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| |
Collapse
|
27
|
Sahasrabudhe A, Rupprecht LE, Orguc S, Khudiyev T, Tanaka T, Sands J, Zhu W, Tabet A, Manthey M, Allen H, Loke G, Antonini MJ, Rosenfeld D, Park J, Garwood IC, Yan W, Niroui F, Fink Y, Chandrakasan A, Bohórquez DV, Anikeeva P. Multifunctional microelectronic fibers enable wireless modulation of gut and brain neural circuits. Nat Biotechnol 2024; 42:892-904. [PMID: 37349522 PMCID: PMC11180606 DOI: 10.1038/s41587-023-01833-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Progress in understanding brain-viscera interoceptive signaling is hindered by a dearth of implantable devices suitable for probing both brain and peripheral organ neurophysiology during behavior. Here we describe multifunctional neural interfaces that combine the scalability and mechanical versatility of thermally drawn polymer-based fibers with the sophistication of microelectronic chips for organs as diverse as the brain and the gut. Our approach uses meters-long continuous fibers that can integrate light sources, electrodes, thermal sensors and microfluidic channels in a miniature footprint. Paired with custom-fabricated control modules, the fibers wirelessly deliver light for optogenetics and transfer data for physiological recording. We validate this technology by modulating the mesolimbic reward pathway in the mouse brain. We then apply the fibers in the anatomically challenging intestinal lumen and demonstrate wireless control of sensory epithelial cells that guide feeding behaviors. Finally, we show that optogenetic stimulation of vagal afferents from the intestinal lumen is sufficient to evoke a reward phenotype in untethered mice.
Collapse
Affiliation(s)
- Atharva Sahasrabudhe
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura E Rupprecht
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Sirma Orguc
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tural Khudiyev
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tomo Tanaka
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Secure System Platform Research Laboratories, NEC Corporation, Kawasaki, Japan
| | - Joanna Sands
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Weikun Zhu
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie Manthey
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harrison Allen
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gabriel Loke
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc-Joseph Antonini
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Dekel Rosenfeld
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jimin Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Indie C Garwood
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Wei Yan
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Farnaz Niroui
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoel Fink
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anantha Chandrakasan
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 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
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
28
|
McKay DM, Defaye M, Rajeev S, MacNaughton WK, Nasser Y, Sharkey KA. Neuroimmunophysiology of the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2024; 326:G712-G725. [PMID: 38626403 PMCID: PMC11376980 DOI: 10.1152/ajpgi.00075.2024] [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: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/18/2024]
Abstract
Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.
Collapse
Affiliation(s)
- Derek M McKay
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sruthi Rajeev
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
29
|
Bharadiya V, Rong Y, Zhang Z, Lin R, Guerrerio AL, Tse CM, Donowitz M, Singh V. Type 1 diabetes human enteroid studies reveal major changes in the intestinal epithelial compartment. Sci Rep 2024; 14:11911. [PMID: 38789719 PMCID: PMC11126659 DOI: 10.1038/s41598-024-62282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Lack of understanding of the pathophysiology of gastrointestinal (GI) complications in type 1 diabetes (T1D), including altered intestinal transcriptomes and protein expression represents a major gap in the management of these patients. Human enteroids have emerged as a physiologically relevant model of the intestinal epithelium but establishing enteroids from individuals with long-standing T1D has proven difficult. We successfully established duodenal enteroids using endoscopic biopsies from pediatric T1D patients and compared them with aged-matched enteroids from healthy subjects (HS) using bulk RNA sequencing (RNA-seq), and functional analyses of ion transport processes. RNA-seq analysis showed significant differences in genes and pathways associated with cell differentiation and proliferation, cell fate commitment, and brush border membrane. Further validation of these results showed higher expression of enteroendocrine cells, and the proliferating cell marker Ki-67, significantly lower expression of NHE3, lower epithelial barrier integrity, and higher fluid secretion in response to cAMP and elevated calcium in T1D enteroids. Enteroids established from pediatric T1D duodenum identify characteristics of an abnormal intestinal epithelium and are distinct from HS. Our data supports the use of pediatric enteroids as an ex-vivo model to advance studies of GI complications and drug discovery in T1D patients.
Collapse
Affiliation(s)
- Vishwesh Bharadiya
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yan Rong
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zixin Zhang
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ruxian Lin
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | | | - C Ming Tse
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mark Donowitz
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Varsha Singh
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
30
|
Egan JM. Physiological Integration of Taste and Metabolism. N Engl J Med 2024; 390:1699-1710. [PMID: 38718360 DOI: 10.1056/nejmra2304578] [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: 05/18/2024]
Affiliation(s)
- Josephine M Egan
- From the Diabetes Section, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore
| |
Collapse
|
31
|
Konanur VR, Hurh SJ, Hsu TM, Roitman MF. Dopamine neuron activity evoked by sucrose and sucrose-predictive cues is augmented by peripheral and central manipulations of glucose availability. Eur J Neurosci 2024; 59:2419-2435. [PMID: 38057909 PMCID: PMC11108752 DOI: 10.1111/ejn.16214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/23/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023]
Abstract
Food deprivation drives eating through multiple signals and circuits. Decreased glucose availability (i.e., cytoglucopenia) drives eating and also increases the value of sucrose. Ventral tegmental area (VTA) dopamine neurons (DANs) contribute to the evaluation of taste stimuli, but their role in integrating glucoprivic signals remains unknown. We monitored VTA DAN activity via Cre-dependent expression of a calcium indicator with in vivo fibre photometry. In ad libitum fed rats, intraoral sucrose evoked a phasic increase in DAN activity. To manipulate glucose availability, we administered (intraperitoneal, lateral or fourth ventricular) the antiglycolytic agent 5-thio-D-glucose (5TG), which significantly augmented the phasic DAN activity to sucrose. 5TG failed to alter DAN activity to water or saccharin, suggesting the response was selective for caloric stimuli. 5TG enhancement of sucrose-evoked DAN activity was stronger after fourth ventricular administration, suggesting a critical node of action within the hindbrain. As 5TG also increases blood glucose, in a separate study, we used peripheral insulin, which stimulates eating, to decrease blood glucose-which was associated with increased DAN activity to intraoral sucrose. DAN activity developed to a cue predictive of intraoral sucrose. While 5TG augmented cue-evoked DAN activity, its action was most potent when delivered to the lateral ventricle. Together, the studies point to central glucose availability as a key modulator of phasic DAN activity to food and food-cues. As glucose sensing neurons are known to populate the hypothalamus and brainstem, results suggest differential modulation of cue-evoked and sucrose-evoked DAN activity.
Collapse
Affiliation(s)
- Vaibhav R. Konanur
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
- Current affiliation: Department of Biology, University of Illinois at Chicago, Chicago, IL
| | - Samantha J. Hurh
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
| | - Ted M. Hsu
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
| | | |
Collapse
|
32
|
Alsudayri A, Perelman S, Brewer M, Chura A, McDevitt M, Drerup C, Ye L. Gut microbiota regulate maturation and mitochondrial function of the nutrient-sensing enteroendocrine cell. Development 2024; 151:dev202544. [PMID: 38577841 PMCID: PMC11112165 DOI: 10.1242/dev.202544] [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: 11/14/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
Enteroendocrine cells (EECs) are crucial for sensing ingested nutrients and regulating feeding behavior. How gut microbiota regulate the nutrient-sensing EEC activity is unclear. Our transcriptomic analysis demonstrates that commensal microbiota colonization significantly increases the expression of many genes associated with mitochondrial function. Using new methods to image EEC cytoplasmic and mitochondrial Ca2+ activity in live zebrafish, our data revealed that it is dynamically regulated during the EEC development process. Mature EECs display an increased mitochondrial-to-cytoplasmic Ca2+ ratio. Mitochondria are evenly distributed in the cytoplasm of immature EECs. As EECs mature, their mitochondria are highly localized at the basal membrane where EEC vesicle secretion occurs. Conventionalized (CV) EECs, but not germ-free (GF) EECs, exhibit spontaneous low-amplitude Ca2+ fluctuation. The mitochondrial-to-cytoplasmic Ca2+ ratio is significantly higher in CV EECs. Nutrient stimulants, such as fatty acid, increase cytoplasmic Ca2+ in a subset of EECs and promote a sustained mitochondrial Ca2+ and ATP increase. However, the nutrient-induced EEC mitochondrial activation is nearly abolished in GF zebrafish. Together, our study reveals that commensal microbiota are crucial in supporting EEC mitochondrial function and maturation.
Collapse
Affiliation(s)
- Alfahdah Alsudayri
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Shane Perelman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Melissa Brewer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Annika Chura
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Madelyn McDevitt
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Catherine Drerup
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lihua Ye
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
33
|
Walmsley R, Chong L, Hii MW, Brown RM, Sumithran P. The effect of bariatric surgery on the expression of gastrointestinal taste receptors: A systematic review. Rev Endocr Metab Disord 2024; 25:421-446. [PMID: 38206483 PMCID: PMC10942945 DOI: 10.1007/s11154-023-09865-7] [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] [Accepted: 12/11/2023] [Indexed: 01/12/2024]
Abstract
Gastrointestinal nutrient sensing via taste receptors may contribute to weight loss, metabolic improvements, and a reduced preference for sweet and fatty foods following bariatric surgery. This review aimed to investigate the effect of bariatric surgery on the expression of oral and post-oral gastrointestinal taste receptors and associations between taste receptor alterations and clinical outcomes of bariatric surgery. A systematic review was conducted to capture data from both human and animal studies on changes in the expression of taste receptors in oral or post-oral gastrointestinal tissue following any type of bariatric surgery. Databases searched included Medline, Embase, Emcare, APA PsychInfo, Cochrane Library, and CINAHL. Two human and 21 animal studies were included. Bariatric surgery alters the quantity of many sweet, umami, and fatty acid taste receptors in the gastrointestinal tract. Changes to the expression of sweet and amino acid receptors occur most often in intestinal segments surgically repositioned more proximally, such as the alimentary limb after gastric bypass. Conversely, changes to fatty acid receptors were observed more frequently in the colon than in the small intestine. Significant heterogeneity in the methodology of included studies limited conclusions regarding the direction of change in taste receptor expression induced by bariatric surgeries. Few studies have investigated associations between taste receptor expression and clinical outcomes of bariatric surgery. As such, future studies should look to investigate the relationship between bariatric surgery-induced changes to gut taste receptor expression and function and the impact of surgery on taste preferences, food palatability, and eating behaviour.Registration code in PROSPERO: CRD42022313992.
Collapse
Affiliation(s)
- Rosalind Walmsley
- Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Surgery, St Vincent's Hospital Melbourne, University of Melbourne, Victoria, Australia
| | - Lynn Chong
- Department of Surgery, St Vincent's Hospital Melbourne, University of Melbourne, Victoria, Australia
| | - Michael W Hii
- Department of Surgery, St Vincent's Hospital Melbourne, University of Melbourne, Victoria, Australia
| | - Robyn M Brown
- Department of Pharmacology and Biochemistry, University of Melbourne, Victoria, Australia
| | - Priya Sumithran
- Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Parkville, VIC, 3052, Australia.
- Department of Surgery, Central Clinical School, Monash University, Victoria, Australia.
- Department of Endocrinology and Diabetes, Alfred Health, Victoria, Australia.
| |
Collapse
|
34
|
Thomasi B, Valdetaro L, Gulbransen B, Tavares-Gomes AL. Neuroimmune Connectomes in the Gut and Their Implications in Parkinson's Disease. Mol Neurobiol 2024; 61:2081-2098. [PMID: 37840070 PMCID: PMC11151216 DOI: 10.1007/s12035-023-03679-z] [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: 08/26/2022] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
The gastrointestinal tract is the largest immune organ and it receives dense innervation from intrinsic (enteric) and extrinsic (sympathetic, parasympathetic, and somatosensory) neurons. The immune and neural systems of the gut communicate with each other and their interactions shape gut defensive mechanisms and neural-controlled gut functions such as motility and secretion. Changes in neuroimmune interactions play central roles in the pathogenesis of diseases such as Parkinson's disease (PD), which is a multicentric disorder that is heterogeneous in its manifestation and pathogenesis. Non-motor and premotor symptoms of PD are common in the gastrointestinal tract and the gut is considered a potential initiation site for PD in some cases. How the enteric nervous system and neuroimmune signaling contribute to PD disease progression is an emerging area of interest. This review focuses on intestinal neuroimmune loops such as the neuroepithelial unit, enteric glial cells and their immunomodulatory effects, anti-inflammatory cholinergic signaling and the relationship between myenteric neurons and muscularis macrophages, and the role of α-synuclein in gut immunity. Special consideration is given to the discussion of intestinal neuroimmune connectomes during PD and their possible implications for various aspects of the disease.
Collapse
Affiliation(s)
- Beatriz Thomasi
- Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building - Gulbransen lab, 567, Wilson Rd, Room 3199, East Lansing, MI, USA.
| | - Luisa Valdetaro
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, USA
| | - Brian Gulbransen
- Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building - Gulbransen lab, 567, Wilson Rd, Room 3199, East Lansing, MI, USA
| | - Ana Lúcia Tavares-Gomes
- Programa de Pós-Graduação Em Neurociências, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Ye J, Fan M, Zhang X, Liang Q, Zhang Y, Zhao X, Lin CT, Zhang D. A novel biomimetic electrochemical taste-biosensor based on conformational changes of the taste receptor. Biosens Bioelectron 2024; 249:116001. [PMID: 38199084 DOI: 10.1016/j.bios.2024.116001] [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: 07/20/2023] [Revised: 11/06/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Taste sensor, a useful tool which could detect and identify thousands of different chemical substances in liquid environments, has attracted continuous concern from beverage and foodstuff industry and its consumers. Although many taste sensing methods have been extensively developed, the assessment of tastant content remains challenging due to the limitations of sensor selectivity and sensitivity. Here we present a novel biomimetic electrochemical taste-biosensor based on bioactive sensing elements and immune amplification with nanomaterials carrier to address above concerns, while taking sweet taste perception as a model. The proposed biosensor based on ligand binding domain (T1R2 VFT) of human sweet taste receptor protein showed human mimicking character and initiated the application of immune recognition in gustation biosensor, which can precisely and sensitively distinguish sweet substances against other related gustation substances with detection limit of 5.1 pM, far less than that of taste sensors without immune amplification whose detection limit was 0.48 nM. The performance test demonstrated the biosensor has the capacity of monitoring the response of sweet substances in real food environments, which is crucial in practical. This biomimetic electrochemical taste-biosensor can work as a new screening platform for newly developed tastants and disclose sweet perception mechanism.
Collapse
Affiliation(s)
- Jing Ye
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China
| | - Minzhi Fan
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China
| | - Xiaoyu Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China
| | - Qi Liang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China; College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Yunshan Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China
| | - Xiaoyu Zhao
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China; College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Cheng-Te Lin
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Diming Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311121, China.
| |
Collapse
|
36
|
McMorrow HE, Lorch CM, Hayes NW, Fleps SW, Frydman JA, Xia JL, Samms RJ, Beutler LR. Incretin hormones and pharmacomimetics rapidly inhibit AgRP neuron activity to suppress appetite. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585583. [PMID: 38562891 PMCID: PMC10983981 DOI: 10.1101/2024.03.18.585583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Analogs of the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) have become mainstays of obesity and diabetes management. However, both the physiologic role of incretin hormones in the control of appetite and the pharmacologic mechanisms by which incretin-mimetic drugs suppress caloric intake remain incompletely understood. Hunger-promoting AgRP-expressing neurons are an important hypothalamic population that regulates food intake. Therefore, we set out to determine how incretins analogs affect their activity in vivo. Using fiber photometry, we observed that both GIP receptor (GIPR) and GLP-1 receptor (GLP-1R) agonism acutely inhibit AgRP neuron activity in fasted mice and reduce the response of AgRP neurons to food. Moreover, optogenetic stimulation of AgRP neurons partially attenuated incretin-induced feeding suppression, suggesting that AgRP neuron inhibition is necessary for the full appetite-suppressing effects of incretin-based therapeutics. Finally, we found that GIP but not GLP-1 is necessary for nutrient-mediated AgRP neuron inhibition, representing a novel physiologic role for GIP in maintaining energy balance. Taken together, these findings reveal neural mechanisms underlying the efficacy of incretin-mimetic obesity therapies. Understanding these drugs' mechanisms of action is crucial for the development of next-generation obesity pharmacotherapies with an improved therapeutic profile.
Collapse
Affiliation(s)
- Hayley E McMorrow
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
- Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL, USA
| | - Carolyn M Lorch
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Nikolas W Hayes
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
- Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL, USA
| | - Stefan W Fleps
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Northwestern University, Chicago, IL, USA
| | - Joshua A Frydman
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jessica L Xia
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ricardo J Samms
- Diabetes, Obesity and Complications Therapeutic Area, Eli Lilly, Indianapolis, IN, USA
| | - Lisa R Beutler
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Chicago, IL 60611, USA
- Lead contact
| |
Collapse
|
37
|
Cao Y, Li R, Bai L. Vagal sensory pathway for the gut-brain communication. Semin Cell Dev Biol 2024; 156:228-243. [PMID: 37558522 DOI: 10.1016/j.semcdb.2023.07.009] [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: 11/21/2022] [Revised: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
The communication between the gut and brain is crucial for regulating various essential physiological functions, such as energy balance, fluid homeostasis, immune response, and emotion. The vagal sensory pathway plays an indispensable role in connecting the gut to the brain. Recently, our knowledge of the vagal gut-brain axis has significantly advanced through molecular genetic studies, revealing a diverse range of vagal sensory cell types with distinct peripheral innervations, response profiles, and physiological functions. Here, we review the current understanding of how vagal sensory neurons contribute to gut-brain communication. First, we highlight recent transcriptomic and genetic approaches that have characterized different vagal sensory cell types. Then, we focus on discussing how different subtypes encode numerous gut-derived signals and how their activities are translated into physiological and behavioral regulations. The emerging insights into the diverse cell types and functional properties of vagal sensory neurons have paved the way for exciting future directions, which may provide valuable insights into potential therapeutic targets for disorders involving gut-brain communication.
Collapse
Affiliation(s)
- Yiyun Cao
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Rui Li
- Chinese Institute for Brain Research, Beijing 102206, China; State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Ling Bai
- Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
38
|
Borgmann D, Fenselau H. Vagal pathways for systemic regulation of glucose metabolism. Semin Cell Dev Biol 2024; 156:244-252. [PMID: 37500301 DOI: 10.1016/j.semcdb.2023.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Maintaining blood glucose at an appropriate physiological level requires precise coordination of multiple organs and tissues. The vagus nerve bidirectionally connects the central nervous system with peripheral organs crucial to glucose mobilization, nutrient storage, and food absorption, thereby presenting a key pathway for the central control of blood glucose levels. However, the precise mechanisms by which vagal populations that target discrete tissues participate in glucoregulation are much less clear. Here we review recent advances unraveling the cellular identity, neuroanatomical organization, and functional contributions of both vagal efferents and vagal afferents in the control of systemic glucose metabolism. We focus on their involvement in relaying glucoregulatory cues from the brain to peripheral tissues, particularly the pancreatic islet, and by sensing and transmitting incoming signals from ingested food to the brain. These recent findings - largely driven by advances in viral approaches, RNA sequencing, and cell-type selective manipulations and tracings - have begun to clarify the precise vagal neuron populations involved in the central coordination of glucose levels, and raise interesting new possibilities for the treatment of glucose metabolism disorders such as diabetes.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Physical Activity Research (CFAS), Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
39
|
Bhatia A, Hanna J, Stuart T, Kasper KA, Clausen DM, Gutruf P. Wireless Battery-free and Fully Implantable Organ Interfaces. Chem Rev 2024; 124:2205-2280. [PMID: 38382030 DOI: 10.1021/acs.chemrev.3c00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Advances in soft materials, miniaturized electronics, sensors, stimulators, radios, and battery-free power supplies are resulting in a new generation of fully implantable organ interfaces that leverage volumetric reduction and soft mechanics by eliminating electrochemical power storage. This device class offers the ability to provide high-fidelity readouts of physiological processes, enables stimulation, and allows control over organs to realize new therapeutic and diagnostic paradigms. Driven by seamless integration with connected infrastructure, these devices enable personalized digital medicine. Key to advances are carefully designed material, electrophysical, electrochemical, and electromagnetic systems that form implantables with mechanical properties closely matched to the target organ to deliver functionality that supports high-fidelity sensors and stimulators. The elimination of electrochemical power supplies enables control over device operation, anywhere from acute, to lifetimes matching the target subject with physical dimensions that supports imperceptible operation. This review provides a comprehensive overview of the basic building blocks of battery-free organ interfaces and related topics such as implantation, delivery, sterilization, and user acceptance. State of the art examples categorized by organ system and an outlook of interconnection and advanced strategies for computation leveraging the consistent power influx to elevate functionality of this device class over current battery-powered strategies is highlighted.
Collapse
Affiliation(s)
- Aman Bhatia
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jessica Hanna
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Tucker Stuart
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Kevin Albert Kasper
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - David Marshall Clausen
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Philipp Gutruf
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
- Department of Electrical and Computer Engineering, The University of Arizona, Tucson, Arizona 85721, United States
- Bio5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
- Neuroscience Graduate Interdisciplinary Program (GIDP), The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
40
|
Sun Y, Yu B, Yu Y, Wang B, Tan X, Lu Y, Wang Y, Zhang K, Wang N. Sweetened Beverages, Genetic Susceptibility, and Incident Atrial Fibrillation: A Prospective Cohort Study. Circ Arrhythm Electrophysiol 2024; 17:e012145. [PMID: 38440895 DOI: 10.1161/circep.123.012145] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 12/15/2023] [Indexed: 03/06/2024]
Abstract
BACKGROUND An association between sweetened beverages and several cardiometabolic diseases has been reported, but their association with atrial fibrillation (AF) is unclear. We aimed to investigate the associations between consumption of sugar-sweetened beverages (SSB), artificially sweetened beverages (ASB), and pure fruit juice (PJ) and risk of consumption with AF risk and further evaluate whether genetic susceptibility modifies these associations. METHODS A total of 201 856 participants who were free of baseline AF, had genetic data available, and completed a 24-hour diet questionnaire were included. Cox proportional hazard models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS During a median follow-up of 9.9 years, 9362 incident AF cases were documented. Compared with nonconsumers, individuals who consumed >2 L/wk of SSB or ASB had an increased risk of AF (HR, 1.10 [95% CI, 1.01-1.20] and HR, 1.20 [95% CI, 1.10-1.31]) in the multivariable-adjusted model. A negative association was observed between the consumption of ≤1 L/wk of PJ and the risk of AF (HR, 0.92 [95% CI, 0.87-0.97]). The highest HRs (95% CIs) of AF were observed for participants at high genetic risk who consumed >2 L/wk of ASB (HR, 3.51 [95% CI, 2.94-4.19]), and the lowest HR were observed for those at low genetic risk who consumed ≤1 L/wk of PJ (HR, 0.77 [95% CI, 0.65-0.92]). No significant interactions were observed between the consumption of SSB, ASB, or PJ and genetic predisposition to AF. CONCLUSIONS Consumption of SSB and ASB at >2 L/wk was associated with an increased risk for AF. PJ consumption ≤1 L/wk was associated with a modestly lower risk for AF. The association between sweetened beverages and AF risk persisted after adjustment for genetic susceptibility to AF. This study does not demonstrate that consumption of SSB and ASB alters AF risk but rather that the consumption of SSB and ASB may predict AF risk beyond traditional risk factors.
Collapse
Affiliation(s)
- Ying Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| | - Bowei Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| | - Yuefeng Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| | - Bin Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| | - Xiao Tan
- School of Public Health, Zhejiang University, Hangzhou, China (X.T.)
- Department of Medical Sciences, Uppsala University, Sweden (X.T.)
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| | - Yu Wang
- Department of Cardiology, Shidong Hospital, University of Shanghai for Science and Technology, China (Y.W.)
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China (Y.S., B.Y., Y.Y., B.W., Y.L., K.Z., N.W.)
| |
Collapse
|
41
|
Aguayo-Guerrero JA, Méndez-García LA, Solleiro-Villavicencio H, Viurcos-Sanabria R, Escobedo G. Sucralose: From Sweet Success to Metabolic Controversies-Unraveling the Global Health Implications of a Pervasive Non-Caloric Artificial Sweetener. Life (Basel) 2024; 14:323. [PMID: 38541649 PMCID: PMC10971371 DOI: 10.3390/life14030323] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 01/04/2025] Open
Abstract
Sucralose is a food additive initially used to mitigate glycemic peaks and calorie intake in patients with diabetes and obesity. Although sucralose has been considered safe for human consumption, the World Health Organization (WHO) issued a global alert in 2023 concerning the potential health implications of this artificial sweetener. This review aims to comprehensively explore the effects of sucralose intake on human health by understanding sucralose absorption, metabolism, and excretion. We also outline the role of the sweet taste 1 receptor 3 (T1R3) in mediating sucralose-dependent signaling pathways that regulate satiety, incretin release, and insulin response. Finally, we discuss the impact of sucralose on microbiome dysbiosis, inflammatory response origin, liver damage, and toxicity. Gaining a deeper understanding of the manifold effects of sucralose on human physiology will help promote further studies to ensure its consumption is deemed safe for a broader population, including children, adolescents, and pregnant women.
Collapse
Affiliation(s)
- José Alfredo Aguayo-Guerrero
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06720, Mexico; (J.A.A.-G.)
| | - Lucía Angélica Méndez-García
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06720, Mexico; (J.A.A.-G.)
| | | | - Rebeca Viurcos-Sanabria
- Department of Plastic and Hand Surgery, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Galileo Escobedo
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06720, Mexico; (J.A.A.-G.)
| |
Collapse
|
42
|
Sclafani A, Ackroff K. Glucose appetition in C57BL/6J mice: Influence of nonnutritive sweetener experience, food deprivation state and sex differences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582331. [PMID: 38464099 PMCID: PMC10925266 DOI: 10.1101/2024.02.27.582331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
In addition to its sweet taste, glucose has potent and rapid postoral actions (appetition) that enhance its reward value. This has been demonstrated by the experience-induced preference for glucose over initially preferred nonnutritive sweetener solutions in 24-h choice tests. However, some sweetener solutions (e.g., 0.8% sucralose) have inhibitory postoral actions that may exaggerate glucose appetition whereas others (e.g., 0.1% sucralose + 0.1% saccharin, S+S) do not. Experiment 1 revealed that food-restricted (FR) male C57BL/6J mice displayed similar rapid glucose appetition effects (stimulation of glucose licking within minutes) and conditioned flavor preferences following 1-h experience with flavored 0.8% sucralose or 0.1% S+S and 8% glucose solutions. Thus, the inhibitory effects of 0.8% sucralose observed in 24-h tests were not apparent in 1-h tests. Experiment 2 evaluated the effects of food deprivation state on 1-h glucose appetition. Unlike FR female mice, ad libitum (AL) fed mice displayed no or delayed stimulation of glucose licking depending upon the training solutions used (0.1% S+S vs. 8% glucose, or 0.2% S+S vs. 16% glucose). Both AL groups, like the FR group, developed a preference for the glucose-paired flavor over the S+S paired flavor. Thus, food restriction promotes glucose appetition but is not required for a conditioned preference. Overall, male and female mice showed similar glucose appetition responses although females displayed a more rapid initial glucose response.
Collapse
|
43
|
Wang RL, Chang RB. The Coding Logic of Interoception. Annu Rev Physiol 2024; 86:301-327. [PMID: 38061018 PMCID: PMC11103614 DOI: 10.1146/annurev-physiol-042222-023455] [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] [Indexed: 02/13/2024]
Abstract
Interoception, the ability to precisely and timely sense internal body signals, is critical for life. The interoceptive system monitors a large variety of mechanical, chemical, hormonal, and pathological cues using specialized organ cells, organ innervating neurons, and brain sensory neurons. It is important for maintaining body homeostasis, providing motivational drives, and regulating autonomic, cognitive, and behavioral functions. However, compared to external sensory systems, our knowledge about how diverse body signals are coded at a system level is quite limited. In this review, we focus on the unique features of interoceptive signals and the organization of the interoceptive system, with the goal of better understanding the coding logic of interoception.
Collapse
Affiliation(s)
- Ruiqi L Wang
- Department of Neuroscience and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Rui B Chang
- Department of Neuroscience and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
44
|
Kraimi N, Ross T, Pujo J, De Palma G. The gut microbiome in disorders of gut-brain interaction. Gut Microbes 2024; 16:2360233. [PMID: 38949979 PMCID: PMC11218806 DOI: 10.1080/19490976.2024.2360233] [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: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 07/03/2024] Open
Abstract
Functional gastrointestinal disorders (FGIDs), chronic disorders characterized by either abdominal pain, altered intestinal motility, or their combination, have a worldwide prevalence of more than 40% and impose a high socioeconomic burden with a significant decline in quality of life. Recently, FGIDs have been reclassified as disorders of gut-brain interaction (DGBI), reflecting the key role of the gut-brain bidirectional communication in these disorders and their impact on psychological comorbidities. Although, during the past decades, the field of DGBIs has advanced significantly, the molecular mechanisms underlying DGBIs pathogenesis and pathophysiology, and the role of the gut microbiome in these processes are not fully understood. This review aims to discuss the latest body of literature on the complex microbiota-gut-brain interactions and their implications in the pathogenesis of DGBIs. A better understanding of the existing communication pathways between the gut microbiome and the brain holds promise in developing effective therapeutic interventions for DGBIs.
Collapse
Affiliation(s)
- Narjis Kraimi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Taylor Ross
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Julien Pujo
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| |
Collapse
|
45
|
Schwarz A, Hernandez L, Arefin S, Sartirana E, Witasp A, Wernerson A, Stenvinkel P, Kublickiene K. Sweet, bloody consumption - what we eat and how it affects vascular ageing, the BBB and kidney health in CKD. Gut Microbes 2024; 16:2341449. [PMID: 38686499 PMCID: PMC11062370 DOI: 10.1080/19490976.2024.2341449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
In today's industrialized society food consumption has changed immensely toward heightened red meat intake and use of artificial sweeteners instead of grains and vegetables or sugar, respectively. These dietary changes affect public health in general through an increased incidence of metabolic diseases like diabetes and obesity, with a further elevated risk for cardiorenal complications. Research shows that high red meat intake and artificial sweeteners ingestion can alter the microbial composition and further intestinal wall barrier permeability allowing increased transmission of uremic toxins like p-cresyl sulfate, indoxyl sulfate, trimethylamine n-oxide and phenylacetylglutamine into the blood stream causing an array of pathophysiological effects especially as a strain on the kidneys, since they are responsible for clearing out the toxins. In this review, we address how the burden of the Western diet affects the gut microbiome in altering the microbial composition and increasing the gut permeability for uremic toxins and the detrimental effects thereof on early vascular aging, the kidney per se and the blood-brain barrier, in addition to the potential implications for dietary changes/interventions to preserve the health issues related to chronic diseases in future.
Collapse
Affiliation(s)
- Angelina Schwarz
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Leah Hernandez
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Samsul Arefin
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Sartirana
- Department of Translational Medicine, Nephrology and Kidney Transplantation Unit, University of Piemonte Orientale, Novara, Italy
| | - Anna Witasp
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika Wernerson
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Karolina Kublickiene
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Glendinning JI, Archambeau A, Conlin C, Drimmer Z, Griffith G, Isber R, Koffler K, Ortiz G, Prakash A, Sollitto C, Srinivasan H. Mice learn to identify and discriminate sugar solutions based on odor cues. Chem Senses 2024; 49:bjae024. [PMID: 38824409 DOI: 10.1093/chemse/bjae024] [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: 07/15/2023] [Indexed: 06/03/2024] Open
Abstract
This study examined how olfaction impacts ingestive responses of mice to sugar solutions. Experiment 1 asked whether naïve C57BL/6 (B6) mice could identify 1 M glucose, fructose, or sucrose solutions based on odor cues, during a 30-min 2-bottle acceptability test. We tested mice both before and after they were rendered anosmic with ZnSO4 treatment. We used 2 indirect measures of odor-mediated response: number of trials initiated and latency to initiate licking. Before ZnSO4 treatment, the mice learned how to identify 1 M glucose and fructose (but not sucrose) solutions based on odor cues. ZnSO4 treatment eliminated their ability to identify the glucose and fructose solutions. Experiment 2 asked whether 2 d of exposure to a 1 M glucose, fructose, or sucrose solution improved the identification of the same sugar solution. Following exposure, the B6 mice identified all 3 sugar solutions based on odor cues. Experiment 3 asked whether T1R3 knockout mice (i.e. mice lacking the T1R3 subunit of the T1R2 + R3 sweet taste receptor) could learn to discriminate 0.44 M glucose and fructose solutions based on odor cues. All mice were subjected to a 1-h preference test, both before and after exposure to the 0.44 M glucose and fructose solutions. During exposure, the experimental mice received ZnSO4 treatment, whereas the control mice received saline treatment. Before exposure, neither type of mouse preferred the glucose solution. After exposure, the control mice preferred the glucose solution, whereas the experimental mice did not. Our results reveal that mice can learn to use odor cues to identify and discriminate between sugar solutions.
Collapse
Affiliation(s)
- John I Glendinning
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
- Department of Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Alix Archambeau
- Department of Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Catherine Conlin
- Department of Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Zoee Drimmer
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Gabriel Griffith
- Department of Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Rayna Isber
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Kayla Koffler
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Gabriella Ortiz
- Department of Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Anisha Prakash
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Catherine Sollitto
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Harini Srinivasan
- Department of Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, United States
| |
Collapse
|
47
|
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
|
48
|
Chandra R, Sokratian A, Chavez KR, King S, Swain SM, Snyder JC, West AB, Liddle RA. Gut mucosal cells transfer α-synuclein to the vagus nerve. JCI Insight 2023; 8:e172192. [PMID: 38063197 PMCID: PMC10795834 DOI: 10.1172/jci.insight.172192] [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: 05/11/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023] Open
Abstract
Epidemiological and histopathological findings have raised the possibility that misfolded α-synuclein protein might spread from the gut to the brain and increase the risk of Parkinson's disease. Although past experimental studies in mouse models have relied on gut injections of exogenous recombinant α-synuclein fibrils to study gut-to-brain α-synuclein transfer, the possible origins of misfolded α-synuclein within the gut have remained elusive. We recently demonstrated that sensory cells of intestinal mucosa express α-synuclein. Here, we employed mouse intestinal organoids expressing human α-synuclein to observe the transfer of α-synuclein protein from epithelial cells in organoids to cocultured nodose neurons devoid of α-synuclein. In mice expressing human α-synuclein, but no mouse α-synuclein, α-synuclein fibril-templating activity emerged in α-synuclein-seeded fibril aggregation assays in intestine, vagus nerve, and dorsal motor nucleus. In newly engineered transgenic mice that restrict pathological human α-synuclein expression to intestinal epithelial cells, α-synuclein fibril-templating activity transfered to the vagus nerve and dorsal motor nucleus. Subdiaphragmatic vagotomy prior to induction of α-synuclein expression in intestinal epithelial cells effectively protected the hindbrain from emergence of α-synuclein fibril-templating activity. Overall, these findings highlight a potential non-neuronal source of fibrillar α-synuclein protein that might arise in gut mucosal cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Joshua C. Snyder
- Department of Surgery, and
- Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Andrew B. West
- Department of Pharmacology and Cancer Biology
- Duke Institute for Brain Sciences, Durham, North Carolina, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Rodger A. Liddle
- Department of Medicine
- Duke Institute for Brain Sciences, Durham, North Carolina, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Veterans Affairs, Chevy Chase, Maryland, USA
| |
Collapse
|
49
|
Glendinning JI, Williams N. Chronic sugar exposure increases daily intake of sugars but decreases avidity for sweeteners in mice. Appetite 2023; 191:107077. [PMID: 37813162 DOI: 10.1016/j.appet.2023.107077] [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: 07/20/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/11/2023]
Abstract
Little is known about how chronic sugar consumption impacts avidity for and daily intake of sugars. This issue is topical because modern humans exhibit high daily intakes of sugar. Here, we exposed sugar-naïve C57BL/6 mice (across two 28-day exposure periods, EP1 and EP2) to a control (chow and water) or experimental (chow, water and a 11 or 34% sugar solution) diet. The sugar solutions contained sucrose, glucose syrups, or high-fructose syrups. We used brief-access tests to measure appetitive responses to sucralose and sucrose solutions at three time points: baseline (before EP1), after EP1, and after EP2. We used lick rates to infer palatability, and number of trials initiated/test to infer motivation. Exposure to the control diet had no impact on lick rates or number of trials initiated for sucralose and sucrose. In contrast, exposure to the experimental diets reduced licking for the sweeteners to varying degrees. Lick rates were reduced by exposure to sugar solutions containing the 11% glucose syrups, 34% sucrose, 34% glucose syrups and 34% high-fructose syrups. The number of trials initiated was reduced by exposure to all of the sugar solutions. Despite the exposure-induced reductions in avidity for the sweetener solutions, daily intakes of virtually all of the sugar solutions increased across the exposure periods. We conclude that (i) chronic consumption of sugar solutions reduced avidity for the sweetened solutions, (ii) the extent of this effect depended on the concentration and type of sugar, and (iii) avidity for sweet-tasting solutions could not explain the persistently high daily intake of sugar solutions in mice.
Collapse
Affiliation(s)
- John I Glendinning
- Departments of Biology and Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA.
| | - Niki Williams
- Departments of Biology and Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| |
Collapse
|
50
|
Lai TT, Liou CW, Tsai YH, Lin YY, Wu WL. Butterflies in the gut: the interplay between intestinal microbiota and stress. J Biomed Sci 2023; 30:92. [PMID: 38012609 PMCID: PMC10683179 DOI: 10.1186/s12929-023-00984-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
Psychological stress is a global issue that affects at least one-third of the population worldwide and increases the risk of numerous psychiatric disorders. Accumulating evidence suggests that the gut and its inhabiting microbes may regulate stress and stress-associated behavioral abnormalities. Hence, the objective of this review is to explore the causal relationships between the gut microbiota, stress, and behavior. Dysbiosis of the microbiome after stress exposure indicated microbial adaption to stressors. Strikingly, the hyperactivated stress signaling found in microbiota-deficient rodents can be normalized by microbiota-based treatments, suggesting that gut microbiota can actively modify the stress response. Microbiota can regulate stress response via intestinal glucocorticoids or autonomic nervous system. Several studies suggest that gut bacteria are involved in the direct modulation of steroid synthesis and metabolism. This review provides recent discoveries on the pathways by which gut microbes affect stress signaling and brain circuits and ultimately impact the host's complex behavior.
Collapse
Affiliation(s)
- Tzu-Ting Lai
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Chia-Wei Liou
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Hsuan Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yuan-Yuan Lin
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Wei-Li Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|