1
|
Wessels AG. Influence of the Gut Microbiome on Feed Intake of Farm Animals. Microorganisms 2022; 10:microorganisms10071305. [PMID: 35889024 PMCID: PMC9315566 DOI: 10.3390/microorganisms10071305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/04/2022] Open
Abstract
With the advancement of microbiome research, the requirement to consider the intestinal microbiome as the “last organ” of an animal emerged. Through the production of metabolites and/or the stimulation of the host’s hormone and neurotransmitter synthesis, the gut microbiota can potentially affect the host’s eating behavior both long and short-term. Based on current evidence, the major mediators appear to be short-chain fatty acids (SCFA), peptide hormones such as peptide YY (PYY) and glucagon-like peptide-1 (GLP-1), as well as the amino acid tryptophan with the associated neurotransmitter serotonin, dopamine and γ-Aminobutyrate (GABA). The influence appears to extend into central neuronal networks and the expression of taste receptors. An interconnection of metabolic processes with mechanisms of taste sensation suggests that the gut microbiota may even influence the sensations of their host. This review provides a summary of the current status of microbiome research in farm animals with respect to general appetite regulation and microbiota-related observations made on the influence on feed intake. This is briefly contrasted with the existing findings from research with rodent models in order to identify future research needs. Increasing our understanding of appetite regulation could improve the management of feed intake, feed frustration and anorexia related to unhealthy conditions in farm animals.
Collapse
Affiliation(s)
- Anna Grete Wessels
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
2
|
Treesukosol Y, Moran TH. Administration of Exendin-4 but not CCK alters lick responses and trial initiation to sucrose and intralipid during brief-access tests. Chem Senses 2022; 47:bjac004. [PMID: 35427413 PMCID: PMC9012268 DOI: 10.1093/chemse/bjac004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Administration of cholecystokinin (CCK) or the glucagon-like peptide 1 (GLP-1) receptor agonist Exendin-4 (Ex-4) reduces food intake. Findings in the literature suggest CCK reduces intake primarily as a satiety signal whereas GLP-1 may play a role in both satiety and reward-related feeding signals. Compounds that humans describe as âsweetâ and âfattyâ are palatable yet are signaled via separate transduction pathways. Here, unconditioned lick responses to sucrose and intralipid were measured in a brief-access lick procedure in food-restricted male rats in response to i.p. administration of Ex-4 (3 h before test), CCK (30 min before test), or a combination of both. The current experimental design measures lick responses to water and varying concentrations of both sucrose (0.03, 0.1, and 0.5 M) and intralipid (0.2%, 2%, and 20%) during 10-s trials across a 30-min single test session. This design minimized postingestive influences. Compared with saline-injected controls, CCK (1.0, 3.0, or 6.0 µg/kg) did not change lick responses to sucrose or intralipid. Number of trials initiated and lick responses to both sucrose and intralipid were reduced in rats injected with 3.0 µg/kg, but not 1.0 µg/kg Ex-4. The supplement of CCK did not alter lick responses or trials initiated compared with Ex-4 administration alone. These findings support a role for GLP-1 but not CCK in the oral responsiveness to palatable stimuli. Furthermore, Ex-4-induced reductions were observed for both sucrose and intralipid, compounds representing âsweetâ and âfat,â respectively.
Collapse
Affiliation(s)
- Yada Treesukosol
- Department of Psychology, California State University Long Beach, Long Beach, CA 90840, United States
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
3
|
Malone IG, Hunter BK, Rossow HL, Herzog H, Zolotukhin S, Munger SD, Dotson CD. Y1 receptors modulate taste-related behavioral responsiveness in male mice to prototypical gustatory stimuli. Horm Behav 2021; 136:105056. [PMID: 34509673 PMCID: PMC8640844 DOI: 10.1016/j.yhbeh.2021.105056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
Mammalian taste bud cells express receptors for numerous peptides implicated elsewhere in the body in the regulation of metabolism, nutrient assimilation, and satiety. The perturbation of several peptide signaling pathways in the gustatory periphery results in changes in behavioral and/or physiological responsiveness to subsets of taste stimuli. We previously showed that Peptide YY (PYY) - which is present in both saliva and in subsets of taste cells - can affect behavioral taste responsiveness and reduce food intake and body weight. Here, we investigated the contributions of taste bud-localized receptors for PYY and the related Neuropeptide Y (NPY) on behavioral taste responsiveness. Y1R, but not Y2R, null mice show reduced responsiveness to sweet, bitter, and salty taste stimuli in brief-access taste tests; similar results were seen when wildtype mice were exposed to Y receptor antagonists in the taste stimuli. Finally, mice in which the gene encoding the NPY propeptide was deleted also showed reduced taste responsiveness to sweet and bitter taste stimuli. Collectively, these results suggest that Y1R signaling, likely through its interactions with NPY, can modulate peripheral taste responsiveness in mice.
Collapse
Affiliation(s)
- Ian G Malone
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Brianna K Hunter
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Heidi L Rossow
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | - Sergei Zolotukhin
- Center for Smell and Taste, University of Florida, Gainesville, FL 32610, USA; Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Steven D Munger
- Center for Smell and Taste, University of Florida, Gainesville, FL 32610, USA; Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Cedrick D Dotson
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
4
|
Schalla MA, Taché Y, Stengel A. Neuroendocrine Peptides of the Gut and Their Role in the Regulation of Food Intake. Compr Physiol 2021; 11:1679-1730. [PMID: 33792904 DOI: 10.1002/cphy.c200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of food intake encompasses complex interplays between the gut and the brain. Among them, the gastrointestinal tract releases different peptides that communicate the metabolic state to specific nuclei in the hindbrain and the hypothalamus. The present overview gives emphasis on seven peptides that are produced by and secreted from specialized enteroendocrine cells along the gastrointestinal tract in relation with the nutritional status. These established modulators of feeding are ghrelin and nesfatin-1 secreted from gastric X/A-like cells, cholecystokinin (CCK) secreted from duodenal I-cells, glucagon-like peptide 1 (GLP-1), oxyntomodulin, and peptide YY (PYY) secreted from intestinal L-cells and uroguanylin (UGN) released from enterochromaffin (EC) cells. © 2021 American Physiological Society. Compr Physiol 11:1679-1730, 2021.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Roper SD. Chemical and electrical synaptic interactions among taste bud cells. CURRENT OPINION IN PHYSIOLOGY 2021; 20:118-125. [PMID: 33521414 DOI: 10.1016/j.cophys.2020.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chemical synapses between taste cells were first proposed based on electron microscopy of fish taste buds. Subsequently, researchers found considerable evidence for electrical coupling in fish, amphibian, and possibly mammalian taste buds. The development lingual slice and isolated cell preparations allowed detailed investigations of cell-cell interactions, both chemical and electrical, in taste buds. The identification of serotonin and ATP as taste neurotransmitters focused attention onto chemical synaptic interactions between taste cells and research on electrical coupling faded. Findings from Ca2+ imaging, electrophysiology, and molecular biology indicate that several neurotransmitters, including ATP, serotonin, GABA, acetylcholine, and norepinephrine, are secreted by taste cells and exert paracrine interactions in taste buds. Most work has been done on interactions between Type II and Type III taste cells. This brief review follows the trail of studies on cell-cell interactions in taste buds, from the initial ultrastructural observations to the most recent optogenetic manipulations.
Collapse
Affiliation(s)
- Stephen D Roper
- Department of Physiology & Biophysics and Department of Otolaryngology, Miller School of Medicine, University of Miami, FL 33136
| |
Collapse
|
6
|
An alternative pathway for sweet sensation: possible mechanisms and physiological relevance. Pflugers Arch 2020; 472:1667-1691. [PMID: 33030576 DOI: 10.1007/s00424-020-02467-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Sweet substances are detected by taste-bud cells upon binding to the sweet-taste receptor, a T1R2/T1R3 heterodimeric G protein-coupled receptor. In addition, experiments with mouse models lacking the sweet-taste receptor or its downstream signaling components led to the proposal of a parallel "alternative pathway" that may serve as metabolic sensor and energy regulator. Indeed, these mice showed residual nerve responses and behavioral attraction to sugars and oligosaccharides but not to artificial sweeteners. In analogy to pancreatic β cells, such alternative mechanism, to sense glucose in sweet-sensitive taste cells, might involve glucose transporters and KATP channels. Their activation may induce depolarization-dependent Ca2+ signals and release of GLP-1, which binds to its receptors on intragemmal nerve fibers. Via unknown neuronal and/or endocrine mechanisms, this pathway may contribute to both, behavioral attraction and/or induction of cephalic-phase insulin release upon oral sweet stimulation. Here, we critically review the evidence for a parallel sweet-sensitive pathway, involved signaling mechanisms, neural processing, interactions with endocrine hormonal mechanisms, and its sensitivity to different stimuli. Finally, we propose its physiological role in detecting the energy content of food and preparing for digestion.
Collapse
|
7
|
Song L, Chen K, Yan J, Zhang Y, Mao X, Lu B, Sun B. Maternal high-fat diet during gestation and lactation increases conditioned aversion threshold for sucrose and alters sweet taste receptors expression in taste buds in rat offspring. Physiol Behav 2019; 212:112709. [DOI: 10.1016/j.physbeh.2019.112709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 01/01/2023]
|
8
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [PMID: 31460832 DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 2692] [Impact Index Per Article: 448.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
9
|
Behrens M, Meyerhof W. A role for taste receptors in (neuro)endocrinology? J Neuroendocrinol 2019; 31:e12691. [PMID: 30712315 DOI: 10.1111/jne.12691] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/14/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022]
Abstract
The sense of taste is positioned at the forefront when it comes to the interaction of our body with foodborne chemicals. However, the role of our taste system, and in particular its associated taste receptors, is not limited to driving food preferences leading to ingestion or rejection before other organs take over responsibility for nutrient digestion, absorption and metabolic regulation. Taste sensory elements do much more. On the one hand, extra-oral taste receptors from the brain to the gut continue to sense nutrients and noxious substances after ingestion and, on the other hand, the nutritional state feeds back on the taste system. This intricate regulatory network is orchestrated by endocrine factors that are secreted in response to taste receptor signalling and, in turn regulate the taste receptor cells themselves. The present review summarises current knowledge on the endocrine regulation of the taste perceptual system and the release of hunger/satiety regulating factors by gastrointestinal taste receptors. Furthermore, the regulation of blood glucose levels via the activation of pancreatic sweet taste receptors and subsequent insulin secretion, as well as the influence of bitter compounds on thyroid hormone release, is addressed. Finally, the central effects of tastants are discussed briefly.
Collapse
Affiliation(s)
- Maik Behrens
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Wolfgang Meyerhof
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
10
|
Witt M. Anatomy and development of the human taste system. HANDBOOK OF CLINICAL NEUROLOGY 2019; 164:147-171. [DOI: 10.1016/b978-0-444-63855-7.00010-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
|
11
|
Yoshida R, Shin M, Yasumatsu K, Takai S, Inoue M, Shigemura N, Takiguchi S, Nakamura S, Ninomiya Y. The Role of Cholecystokinin in Peripheral Taste Signaling in Mice. Front Physiol 2017; 8:866. [PMID: 29163209 PMCID: PMC5671461 DOI: 10.3389/fphys.2017.00866] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/16/2017] [Indexed: 11/13/2022] Open
Abstract
Cholecystokinin (CCK) is a gut hormone released from enteroendocrine cells. CCK functions as an anorexigenic factor by acting on CCK receptors expressed on the vagal afferent nerve and hypothalamus with a synergistic interaction between leptin. In the gut, tastants such as amino acids and bitter compounds stimulate CCK release from enteroendocrine cells via activation of taste transduction pathways. CCK is also expressed in taste buds, suggesting potential roles of CCK in taste signaling in the peripheral taste organ. In the present study, we focused on the function of CCK in the initial responses to taste stimulation. CCK was coexpressed with type II taste cell markers such as Gα-gustducin, phospholipase Cβ2, and transient receptor potential channel M5. Furthermore, a small subset (~30%) of CCK-expressing taste cells expressed a sweet/umami taste receptor component, taste receptor type 1 member 3, in taste buds. Because type II taste cells are sweet, umami or bitter taste cells, the majority of CCK-expressing taste cells may be bitter taste cells. CCK-A and -B receptors were expressed in both taste cells and gustatory neurons. CCK receptor knockout mice showed reduced neural responses to bitter compounds compared with wild-type mice. Consistently, intravenous injection of CCK-Ar antagonist lorglumide selectively suppressed gustatory nerve responses to bitter compounds. Intravenous injection of CCK-8 transiently increased gustatory nerve activities in a dose-dependent manner whereas administration of CCK-8 did not affect activities of bitter-sensitive taste cells. Collectively, CCK may be a functionally important neurotransmitter or neuromodulator to activate bitter nerve fibers in peripheral taste tissues.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan.,OBT Research Center, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan
| | - Misa Shin
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan.,Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Keiko Yasumatsu
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan.,Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan
| | - Mayuko Inoue
- OBT Research Center, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan.,Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan
| | - Soichi Takiguchi
- National Kyushu Cancer Center, Institute for Clinical Research, Fukuoka, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuzo Ninomiya
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan.,Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan.,Monell Chemical Senses Center, Philadelphia, PA, United States
| |
Collapse
|
12
|
Oral Biosciences: The annual review 2016. J Oral Biosci 2017. [DOI: 10.1016/j.job.2016.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Gao S, Liu S, Yao J, Zhou T, Li N, Li Q, Dunham R, Liu Z. Taste receptors and gustatory associated G proteins in channel catfish, Ictalurus punctatus. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2016; 21:1-9. [PMID: 27806254 DOI: 10.1016/j.cbd.2016.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 11/18/2022]
Abstract
Taste sensation plays a pivotal role in nutrient identification and acquisition. This is particularly true for channel catfish (Ictalurus punctatus) that live in turbid waters with limited visibility. This biological process is mainly mediated by taste receptors expressed in taste buds that are distributed in several organs and tissues, including the barbels and skin. In the present study, we identified a complete repertoire of taste receptor and gustatory associated G protein genes in the channel catfish genome. A total of eight taste receptor genes were identified, including five type I and three type II taste receptor genes. Their genomic locations, phylogenetic relations, orthologies and expression were determined. Phylogenetic and collinear analyses provided understanding of the evolution dynamics of this gene family. Furthermore, the motif and dN/dS analyses indicated that selection pressures of different degrees were imposed on these receptors. Additionally, four genes of gustatory associated G proteins were also identified. It was indicated that expression patterns of catfish taste receptors and gustatory associated G proteins across organs mirror the distribution of taste buds across organs. Finally, the expression comparison between catfish and zebrafish organs provided evidence of potential roles of catfish skin and gill involved in taste sensation.
Collapse
Affiliation(s)
- Sen Gao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Shikai Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Jun Yao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Tao Zhou
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Ning Li
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Qi Li
- Key Laboratory of Mariculture of the Ministry of Education, Ocean University of China, Qingdao, China
| | - Rex Dunham
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Zhanjiang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
14
|
Meyer-Gerspach A, Wölnerhanssen B, Beglinger C. Functional roles of low calorie sweeteners on gut function. Physiol Behav 2016; 164:479-481. [DOI: 10.1016/j.physbeh.2016.01.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/29/2016] [Accepted: 01/30/2016] [Indexed: 01/07/2023]
|
15
|
Spector AC, le Roux CW, Munger SD, Travers SP, Sclafani A, Mennella JA. Proceedings of the 2015 ASPEN Research Workshop-Taste Signaling. JPEN J Parenter Enteral Nutr 2016; 41:113-124. [PMID: 26598504 DOI: 10.1177/0148607115617438] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article summarizes research findings from 6 experts in the field of taste and feeding that were presented at the 2015 American Society for Parenteral and Enteral Nutrition Research Workshop. The theme was focused on the interaction of taste signals with those of a postingestive origin and how this contributes to regulation of food intake through both physiological and learning processes. Gastric bypass results in exceptional loss of fat mass and increases in circulating levels of key gut peptides, some of which are also expressed along with their cognate receptors in taste buds. Changes in taste preference and food selection in both bariatric surgery patients and rodent models have been reported. Accordingly, the effects of this surgery on taste-related behavior were examined. The conservation of receptor and peptide signaling mechanisms in gustatory and extraoral tissues was discussed in the context of taste responsiveness and the regulation of metabolism. New findings detailing the features of neural circuits between the caudal nucleus of the solitary tract (NST), receiving visceral input from the vagus nerve, and the rostral NST, receiving taste input, were discussed, as was how early life experience with taste stimuli and learned associations between flavor and postoral consequences of nutrients can exert potent and long-lasting effects on feeding.
Collapse
Affiliation(s)
- Alan C Spector
- 1 Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Carel W le Roux
- 2 Diabetes Complications Research Centre, Conway Institute, University College, Dublin, Ireland
| | - Steven D Munger
- 3 Department of Pharmacology and Therapeutics; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism; Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Susan P Travers
- 4 Division of Biosciences, College of Dentistry, Ohio State University, Columbus, Ohio, USA
| | - Anthony Sclafani
- 5 Department of Psychology, Brooklyn College of the City University of New York, New York, New York, USA
| | - Julie A Mennella
- 6 Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Abstract
The taste system of animals is used to detect valuable nutrients and harmful compounds in foods. In humans and mice, sweet, bitter, salty, sour and umami tastes are considered the five basic taste qualities. Sweet and umami tastes are mediated by G-protein-coupled receptors, belonging to the T1R (taste receptor type 1) family. This family consists of three members (T1R1, T1R2 and T1R3). They function as sweet or umami taste receptors by forming heterodimeric complexes, T1R1+T1R3 (umami) or T1R2+T1R3 (sweet). Receptors for each of the basic tastes are thought to be expressed exclusively in taste bud cells. Sweet (T1R2+T1R3-expressing) taste cells were thought to be segregated from umami (T1R1+T1R3-expressing) taste cells in taste buds. However, recent studies have revealed that a significant portion of taste cells in mice expressed all T1R subunits and responded to both sweet and umami compounds. This suggests that sweet and umami taste cells may not be segregated. Mice are able to discriminate between sweet and umami tastes, and both tastes contribute to behavioural preferences for sweet or umami compounds. There is growing evidence that T1R3 is also involved in behavioural avoidance of calcium tastes in mice, which implies that there may be a further population of T1R-expressing taste cells that mediate aversion to calcium taste. Therefore the simple view of detection and segregation of sweet and umami tastes by T1R-expressing taste cells, in mice, is now open to re-examination.
Collapse
|
17
|
Cao X, Zhou X, Liu XM, Zhou LH. Liraglutide alters DPP4 in the circumvallate papillae of type 2 diabetic rats. J Mol Endocrinol 2016; 57:13-21. [PMID: 27151572 DOI: 10.1530/jme-16-0001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/04/2016] [Indexed: 11/08/2022]
Abstract
Liraglutide, a human glucagon-like peptide (GLP1) analog that partially inhibits dipeptidyl-peptidase 4 (DPP4), can decrease glucose levels and suppress appetite in patients with type 2 diabetes (T2DM). GLP1 and its receptor (GLP1R) also exist in the taste buds of rodents and regulate taste sensitivity. DPP4, a protease, functions in homeostasis of blood glucose, lipids, and body weight. Interactions among GLP1, GLP1R, and DPP4 likely affect taste and food-intake behavior. The aim of the present study was to investigate DPP4 expression in the taste buds of the circumvallate papillae (CV) in T2DM rats, and determine the effects of liraglutide treatment. Rats were divided into diabetic control (T2DM-C), normal control (NC), and liraglutide-treated diabetic (T2DM+LIR) groups. DPP4 localization and gene expression levels were evaluated by immunohistochemistry and quantitative reverse transcription-polymerase chain reaction (RT-qPCR), respectively. DPP4 immunoreactive cells were localized in the taste buds of the rat CV. RT-qPCR showed significantly higher expression of Dpp4 mRNA in both the taste buds and hypothalamus of T2DM-C rats compared with NC rats. However, in the T2DM+LIR group, Dpp4 expression differed between the taste buds and hypothalamus, with significantly higher and lower levels compared with the T2DM-C group, respectively. Dpp4 mRNA expression is increased in the taste buds of the CV of T2DM rats. Liraglutide simultaneously upregulated (taste buds) and downregulated (hypothalamus) Dpp4 expression in T2DM rats. Therefore, DPP4 may be closely associated with the anorexigenic signaling and weight loss induced by the treatment of liraglutide in type 2 diabetic patients.
Collapse
Affiliation(s)
- Xun Cao
- Department of EndocrinologyHarbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiao Zhou
- Department of Human Anatomy and EmbryologyHarbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiao-Min Liu
- Department of EndocrinologyHarbin Medical University, Harbin, Heilongjiang Province, China
| | - Li-Hong Zhou
- Department of EndocrinologyHarbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
18
|
Abstract
From the age of 2 years, an American child is more likely to consume a sugar-sweetened product than a fruit or vegetable on any given day-a troubling statistic, given that food preferences are established early in childhood, as well as the strong association between this dietary pattern and increased risk of developing a number of chronic diseases. Here, we review the ontogeny and biopsychology of sweet taste, highlighting how a biological drive to prefer sweetness at high concentrations during childhood, which would have conferred an advantage in environments of scarcity, now predisposes children to overconsume all that is sweet in a modern food system replete with added sugars. We review the power of sweet taste to blunt expressions of pain and mask bad tastes in foods as well as factors that predispose some to consume high-sugar diets, including experiential learning and taste preferences driven in part by genetics. Understanding children's unique vulnerability to our current food environment, rich in both nutritive and nonnutritive sweeteners, is highlighted as a priority for future research to develop evidence-based strategies to help establish healthy dietary behaviors early in life.
Collapse
Affiliation(s)
- Julie A Mennella
- Monell Chemical Senses Center, Philadelphia, PA, 19104-3308, USA.
| | - Nuala K Bobowski
- Monell Chemical Senses Center, Philadelphia, PA, 19104-3308, USA
| | - Danielle R Reed
- Monell Chemical Senses Center, Philadelphia, PA, 19104-3308, USA
| |
Collapse
|
19
|
|
20
|
Loper HB, La Sala M, Dotson C, Steinle N. Taste perception, associated hormonal modulation, and nutrient intake. Nutr Rev 2016; 73:83-91. [PMID: 26024495 DOI: 10.1093/nutrit/nuu009] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
It is well known that taste perception influences food intake. After ingestion, gustatory receptors relay sensory signals to the brain, which segregates, evaluates, and distinguishes the stimuli, leading to the experience known as "flavor." It is well accepted that five taste qualities – sweet, salty, bitter, sour, and umami – can be perceived by animals. In this review, the anatomy and physiology of human taste buds, the hormonal modulation of taste function, the importance of genetic chemosensory variation, and the influence of gustatory functioning on macronutrient selection and eating behavior are discussed. Individual genotypic variation results in specific phenotypes of food preference and nutrient intake. Understanding the role of taste in food selection and ingestive behavior is important for expanding our understanding of the factors involved in body weight maintenance and the risk of chronic diseases including obesity, atherosclerosis, cancer, diabetes, liver disease, and hypertension.
Collapse
Affiliation(s)
- Hillary B Loper
- H.B. Loper is with the Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA. M. La Sala and C. Dotson are with the Division of Addiction Medicine, Center for Smell and Taste, Department of Neuroscience and Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA. N Steinle is with the Baltimore Veterans Administration Medical Center and University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael La Sala
- H.B. Loper is with the Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA. M. La Sala and C. Dotson are with the Division of Addiction Medicine, Center for Smell and Taste, Department of Neuroscience and Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA. N Steinle is with the Baltimore Veterans Administration Medical Center and University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cedrick Dotson
- H.B. Loper is with the Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA. M. La Sala and C. Dotson are with the Division of Addiction Medicine, Center for Smell and Taste, Department of Neuroscience and Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA. N Steinle is with the Baltimore Veterans Administration Medical Center and University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nanette Steinle
- H.B. Loper is with the Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA. M. La Sala and C. Dotson are with the Division of Addiction Medicine, Center for Smell and Taste, Department of Neuroscience and Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA. N Steinle is with the Baltimore Veterans Administration Medical Center and University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Abstract
Nesfatin-1, an anorexigenic peptide derived from nucleobindin 2 (NUCB2), is closely involved in feeding behavior, glycometabolism, and satiety regulation. Some studies show that NUCB2/nesfatin-1 is highly expressed and interacts with many appetite-regulating peptides that are co-expressed in the gastrointestinal tract. However, it remains unclear whether nesfatin-1 is expressed and interacts similarly in taste buds. Glucagon-like peptide-1 (GLP-1), a well-known appetite down-regulating peptide, is associated with changes in the expression of nesfatin-1. Therefore, we measured the expression of the NUCB2 gene and the distribution of nesfatin-1-immunoreactive cells and investigated whether these variables change in taste buds of circumvallate papillae (CV) from rats with type 2 diabetes (T2DM) after treatment with liraglutide, a GLP-1 receptor agonist. The results showed that nesfatin-1 immunoreactive cells were localized in the taste buds of rat CV. Quantitative RT-PCR showed a significantly lower expression of NUCB2 mRNA in the taste buds of diabetic control rats (T2DM-C) than in those of the normal control group (NC) and a higher level of NUCB2 in the liraglutide treated group (T2DM + LIR) than either the T2DM-C or the NC groups. Changes in the expression of NUCB2 in the rat hypothalamus were opposite to those in CV taste buds. In summary, we found that rat CV taste buds express NUCB2/nesfatin-1, and that this expression decreases significantly in T2DM and increases after treatment with liraglutide in rat CV. This indicates that nesfatin-1 could be an important factor in the regulation of gustatory function, feeding and perhaps energy homeostasis.
Collapse
Affiliation(s)
- Xun Cao
- Department of Endocrinology, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | | | | | | | | |
Collapse
|
22
|
Pepino MY. Metabolic effects of non-nutritive sweeteners. Physiol Behav 2015; 152:450-5. [PMID: 26095119 DOI: 10.1016/j.physbeh.2015.06.024] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/20/2015] [Accepted: 06/17/2015] [Indexed: 12/22/2022]
Abstract
Until recently, the general belief was that non-nutritive sweeteners (NNSs) were healthy sugar substitutes because they provide sweet taste without calories or glycemic effects. However, data from several epidemiological studies have found that consumption of NNSs, mainly in diet sodas, is associated with increased risk to develop obesity, metabolic syndrome, and type 2 diabetes. The main purpose of this article is to review recent scientific evidence supporting potential mechanisms that explain how "metabolically inactive" NNSs, which have few, if any, calories, might promote metabolic dysregulation. Three potential mechanisms, which are not mutually exclusive, are presented: 1) NNSs interfere with learned responses that contribute to control glucose and energy homeostasis, 2) NNSs interfere with gut microbiota and induce glucose intolerance, and 3) NNSs interact with sweet-taste receptors expressed throughout the digestive system that play a role in glucose absorption and trigger insulin secretion. In addition, recent findings from our laboratory showing an association between individual taste sensitivity to detect sucralose and sucralose's acute effects on metabolic response to an oral glucose load are reported. Taken as a whole, data support the notion that NNSs have metabolic effects. More research is needed to elucidate the mechanisms by which NNSs may drive metabolic dysregulation and better understand potential effects of these commonly used food additives.
Collapse
Affiliation(s)
- M Yanina Pepino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
23
|
Abstract
Levels of obesity have reached epidemic proportions on a global scale, which has led to considerable increases in health problems and increased risk of several diseases, including cardiovascular and pulmonary diseases, cancer and diabetes mellitus. People with obesity consume more food than is needed to maintain an ideal body weight, despite the discrimination that accompanies being overweight and the wealth of available information that overconsumption is detrimental to health. The relationship between energy expenditure and energy intake throughout an individual's lifetime is far more complicated than previously thought. An improved comprehension of the relationships between taste, palatability, taste receptors and hedonic responses to food might lead to increased understanding of the biological underpinnings of energy acquisition, as well as why humans sometimes eat more than is needed and more than we know is healthy. This Review discusses the role of taste receptors in the tongue, gut, pancreas and brain and their hormonal involvement in taste perception, as well as the relationship between taste perception, overeating and the development of obesity.
Collapse
Affiliation(s)
- Sara Santa-Cruz Calvo
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Biomedical Research Center, Room 09B133, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Biomedical Research Center, Room 09B133, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, USA
| |
Collapse
|
24
|
Takai S, Yasumatsu K, Inoue M, Iwata S, Yoshida R, Shigemura N, Yanagawa Y, Drucker DJ, Margolskee RF, Ninomiya Y. Glucagon-like peptide-1 is specifically involved in sweet taste transmission. FASEB J 2015; 29:2268-80. [PMID: 25678625 DOI: 10.1096/fj.14-265355] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/22/2015] [Indexed: 11/11/2022]
Abstract
Five fundamental taste qualities (sweet, bitter, salty, sour, umami) are sensed by dedicated taste cells (TCs) that relay quality information to gustatory nerve fibers. In peripheral taste signaling pathways, ATP has been identified as a functional neurotransmitter, but it remains to be determined how specificity of different taste qualities is maintained across synapses. Recent studies demonstrated that some gut peptides are released from taste buds by prolonged application of particular taste stimuli, suggesting their potential involvement in taste information coding. In this study, we focused on the function of glucagon-like peptide-1 (GLP-1) in initial responses to taste stimulation. GLP-1 receptor (GLP-1R) null mice had reduced neural and behavioral responses specifically to sweet compounds compared to wild-type (WT) mice. Some sweet responsive TCs expressed GLP-1 and its receptors were expressed in gustatory neurons. GLP-1 was released immediately from taste bud cells in response to sweet compounds but not to other taste stimuli. Intravenous administration of GLP-1 elicited transient responses in a subset of sweet-sensitive gustatory nerve fibers but did not affect other types of fibers, and this response was suppressed by pre-administration of the GLP-1R antagonist Exendin-4(3-39). Thus GLP-1 may be involved in normal sweet taste signal transmission in mice.
Collapse
Affiliation(s)
- Shingo Takai
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Keiko Yasumatsu
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Mayuko Inoue
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Shusuke Iwata
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Ryusuke Yoshida
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Noriatsu Shigemura
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Yuchio Yanagawa
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Daniel J Drucker
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Robert F Margolskee
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Yuzo Ninomiya
- *Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, and JST, CREST, Maebashi, Japan; The Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Abstract
In taste cells, taste receptors, their coupled G proteins and downstream signalling elements mediate the detection and transduction of sweet, bitter and umami compounds. In some intestinal endocrine cells, taste receptors and gustducin contribute to the release of glucagon-like peptide 1 (GLP-1) and other gut hormones in response to glucose and non-energetic sweeteners. Conversely, taste cells have been found to express multiple hormones typically found in intestinal endocrine cells, e.g. GLP-1, glucagon, somatostatin and ghrelin. In the present study, by immunohistochemistry, multiple subsets of taste cells were found to express GLP-1. The release of GLP-1 from 'endocrine taste cells' into the bloodstream was examined. In wild-type mice, even after oesophagectomy and vagotomy, oral stimulation with glucose induced an elevation of GLP-1 levels in the bloodstream within 10 min. Stimulation of taste cell explants from wild-type mice with glucose led to the release of GLP-1 into the medium. Knocking out of the Tas1r3 gene did not eliminate glucose-stimulated GLP-1 release from taste cells in vivo. The present results indicate that a portion of the cephalic-phase rise in circulating GLP-1 levels is mediated by the direct release of GLP-1 from taste cells into the bloodstream.
Collapse
|
26
|
Passilly-Degrace P, Chevrot M, Bernard A, Ancel D, Martin C, Besnard P. Is the taste of fat regulated? Biochimie 2014; 96:3-7. [DOI: 10.1016/j.biochi.2013.07.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/25/2013] [Indexed: 12/25/2022]
|
27
|
Abstract
Over the past 30 years, it has been established that hormones produced by the gut, pancreas, and adipose tissue are key players in the control of body weight. These hormones act through a complex neuroendocrine system, including the hypothalamus, to regulate metabolism and energy homeostasis. In obesity, this homeostatic balance is disrupted, either through alterations in the levels of these hormones or through resistance to their actions. Alterations in gut hormone secretion following gastric bypass surgery are likely to underlie the dramatic and persistent loss of weight following this procedure, as well as the observed amelioration in type 2 diabetes mellitus. Medications based on the gut hormone GLP-1 are currently in clinical use to treat type 2 diabetes mellitus and have been shown to produce weight loss. Further therapies for obesity based on other gut hormones are currently in development.
Collapse
Affiliation(s)
- Rebecca Scott
- Division of Diabetes, Endocrinology, Metabolism, Hammersmith Hospital, Imperial College London, London, United Kingdom.
| | | | | |
Collapse
|
28
|
Zhu X, He L, McCluskey LP. Ingestion of bacterial lipopolysaccharide inhibits peripheral taste responses to sucrose in mice. Neuroscience 2013; 258:47-61. [PMID: 24215981 DOI: 10.1016/j.neuroscience.2013.10.072] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/17/2013] [Accepted: 10/22/2013] [Indexed: 12/22/2022]
Abstract
A fundamental role of the taste system is to discriminate between nutritive and toxic foods. However, it is unknown whether bacterial pathogens that might contaminate food and water modulate the transmission of taste input to the brain. We hypothesized that exogenous, bacterially-derived lipopolysaccharide (LPS), modulates neural responses to taste stimuli. Neurophysiological responses from the chorda tympani nerve, which innervates taste cells on the anterior tongue, were unchanged by acute exposure to LPS. Instead, neural responses to sucrose were selectively inhibited in mice that drank LPS during a single overnight period. Decreased sucrose sensitivity appeared 7days after LPS ingestion, in parallel with decreased lingual expression of Tas1r2 and Tas1r3 transcripts, which are translated to T1R2+T1R3 subunits forming the sweet taste receptor. Tas1r2 and Tas1r3 mRNA expression levels and neural responses to sucrose were restored by 14 days after LPS consumption. Ingestion of LPS, rather than contact with taste receptor cells, appears to be necessary to suppress sucrose responses. Furthermore, mice lacking the Toll-like receptor (TLR) 4 for LPS were resistant to neurophysiological changes following LPS consumption. These findings demonstrate that ingestion of LPS during a single period specifically and transiently inhibits neural responses to sucrose. We suggest that LPS drinking initiates TLR4-dependent hormonal signals that downregulate sweet taste receptor genes in taste buds. Delayed inhibition of sweet taste signaling may influence food selection and the complex interplay between gastrointestinal bacteria and obesity.
Collapse
Affiliation(s)
- X Zhu
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, United States; Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - L He
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, United States
| | - L P McCluskey
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, United States.
| |
Collapse
|
29
|
Cai H, Cong WN, Daimon CM, Wang R, Tschöp MH, Sévigny J, Martin B, Maudsley S. Altered lipid and salt taste responsivity in ghrelin and GOAT null mice. PLoS One 2013; 8:e76553. [PMID: 24124572 PMCID: PMC3790684 DOI: 10.1371/journal.pone.0076553] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 08/26/2013] [Indexed: 02/06/2023] Open
Abstract
Taste perception plays an important role in regulating food preference, eating behavior and energy homeostasis. Taste perception is modulated by a variety of factors, including gastric hormones such as ghrelin. Ghrelin can regulate growth hormone release, food intake, adiposity, and energy metabolism. Octanoylation of ghrelin by ghrelin O-acyltransferase (GOAT) is a specific post-translational modification which is essential for many biological activities of ghrelin. Ghrelin and GOAT are both widely expressed in many organs including the gustatory system. In the current study, overall metabolic profiles were assessed in wild-type (WT), ghrelin knockout (ghrelin−/−), and GOAT knockout (GOAT−/−) mice. Ghrelin−/− mice exhibited decreased food intake, increased plasma triglycerides and increased ketone bodies compared to WT mice while demonstrating WT-like body weight, fat composition and glucose control. In contrast GOAT−/− mice exhibited reduced body weight, adiposity, resting glucose and insulin levels compared to WT mice. Brief access taste behavioral tests were performed to determine taste responsivity in WT, ghrelin−/− and GOAT−/− mice. Ghrelin and GOAT null mice possessed reduced lipid taste responsivity. Furthermore, we found that salty taste responsivity was attenuated in ghrelin−/− mice, yet potentiated in GOAT−/− mice compared to WT mice. Expression of the potential lipid taste regulators Cd36 and Gpr120 were reduced in the taste buds of ghrelin and GOAT null mice, while the salt-sensitive ENaC subunit was increased in GOAT−/− mice compared with WT mice. The altered expression of Cd36, Gpr120 and ENaC may be responsible for the altered lipid and salt taste perception in ghrelin−/− and GOAT−/− mice. The data presented in the current study potentially implicates ghrelin signaling activity in the modulation of both lipid and salt taste modalities.
Collapse
Affiliation(s)
- Huan Cai
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Wei-na Cong
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Caitlin M. Daimon
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Rui Wang
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Matthias H. Tschöp
- Institute for Diabetes and Obesity, Helmholtz Centre Munich, Munich, Germany
| | - Jean Sévigny
- Centre de recherche en Rhumatologie et Immunologie, Centre de recherche du CHU de Québec, QC and Département de microbiologie-infectiologie et d′immunologie, Faculté de médecine, Université Laval, Québec, Quebec City, Canada
| | - Bronwen Martin
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Stuart Maudsley
- Receptor Pharmacology Unit, Laboratory of Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| |
Collapse
|
30
|
Gustatory stimuli representing different perceptual qualities elicit distinct patterns of neuropeptide secretion from taste buds. J Neurosci 2013; 33:7559-64. [PMID: 23616560 DOI: 10.1523/jneurosci.0372-13.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Taste stimuli that evoke different perceptual qualities (e.g., sweet, umami, bitter, sour, salty) are detected by dedicated subpopulations of taste bud cells that use distinct combinations of sensory receptors and transduction molecules. Here, we report that taste stimuli also elicit unique patterns of neuropeptide secretion from taste buds that are correlated with those perceptual qualities. We measured tastant-dependent secretion of glucagon-like peptide-1 (GLP-1), glucagon, and neuropeptide Y (NPY) from circumvallate papillae of Tas1r3(+/+), Tas1r3(+/-) and Tas1r3 (-/-) mice. Isolated tongue epithelia were mounted in modified Ussing chambers, permitting apical stimulation of taste buds; secreted peptides were collected from the basal side and measured by specific ELISAs. Appetitive stimuli (sweet: glucose, sucralose; umami: monosodium glutamate; polysaccharide: Polycose) elicited GLP-1 and NPY secretion and inhibited basal glucagon secretion. Sweet and umami stimuli were ineffective in Tas1r3(-/-) mice, indicating an obligatory role for the T1R3 subunit common to the sweet and umami taste receptors. Polycose responses were unaffected by T1R3 deletion, consistent with the presence of a distinct polysaccharide taste receptor. The effects of sweet stimuli on peptide secretion also required the closing of ATP-sensitive K(+) (KATP) channels, as the KATP channel activator diazoxide inhibited the effects of glucose and sucralose on both GLP-1 and glucagon release. Both sour citric acid and salty NaCl increased NPY secretion but had no effects on GLP-1 or glucagon. Bitter denatonium showed no effects on these peptides. Together, these results suggest that taste stimuli of different perceptual qualities elicit unique patterns of neuropeptide secretion from taste buds.
Collapse
|
31
|
Zolotukhin S. Metabolic hormones in saliva: origins and functions. Oral Dis 2013; 19:219-29. [PMID: 22994880 PMCID: PMC3530011 DOI: 10.1111/odi.12015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 08/26/2012] [Accepted: 08/27/2012] [Indexed: 12/13/2022]
Abstract
The salivary proteome consists of thousands of proteins, which include, among others, hormonal modulators of energy intake and output. Although the functions of this prominent category of hormones in whole body energy metabolism are well characterized, their functions in the oral cavity, whether as a salivary component, or when expressed in taste cells, are less studied and poorly understood. The respective receptors for the majority of salivary metabolic hormones have been also shown to be expressed in salivary glands (SGs), taste cells, or other cells in the oral mucosa. This review provides a comprehensive account of the gastrointestinal hormones, adipokines, and neuropeptides identified in saliva, SGs, or lingual epithelium, as well as their respective cognate receptors expressed in the oral cavity. Surprisingly, few functions are assigned to salivary metabolic hormones, and these functions are mostly associated with the modulation of taste perception. Because of the well-characterized correlation between impaired oral nutrient sensing and increased energy intake and body mass index, a conceptually provocative point of view is introduced, whereupon it is argued that targeted changes in the composition of saliva could affect whole body metabolism in response to the activation of cognate receptors expressed locally in the oral mucosa.
Collapse
Affiliation(s)
- S Zolotukhin
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
32
|
Hevér H, Altdorfer K, Zelles T, Batbayar B, Fehér E. [Changes in the innervation of the taste buds in diabetic rats]. Orv Hetil 2013; 154:443-8. [PMID: 23506800 DOI: 10.1556/oh.2013.29570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Abnormal sensations such as pain and impairment of taste are symptoms of approximately 10% of patients having diabetes mellitus. AIM The aim of the study was to investigate and quantify the different neuropeptide containing nerve fibres in the vallate papilla of the diabetic rat. METHODS Immunohistochemical methods were used to study the changes of the number of different neuropeptide containing nerve terminals located in the vallate papillae in diabetic rats. Diabetes was induced in the rats with streptozotocin. RESULTS Two weeks after streptozotocin treatment the number of the substance P, galanin, vasoactive intestinal polypeptide and neuropeptide Y immunoreactive nerve terminals was significantly increased (p<0.05) in the tunica mucosa of the tongue. The number of the lymphocytes and mast cells was also increased significantly. Some of the immunoreactive nerve terminals were located in the lingual epithelium both intragemmally and extragemmally and were seen to comprise dense bundles in the lamina propria just beneath the epithelium. No taste cells were immunoreactive for any of the investigated peptides. Vasoactive intestinal polypeptide and neuropeptide Y immunoreactive nerve fibres were not detected in the taste buds. For weeks after streptozotocin administration the number of the substance P, calcitonin gene related peptide and galanin immunoreactive nerve terminals was decreased both intragemmally and intergemmally. In case of immediate insulin treatment, the number of the immunoreactive nerve terminals was similar to that of the controls, however, insulin treatment given 1 week later to diabetic rats produced a decreased number of nerve fibers. Morphometry revealed no significant difference in papilla size between the control and diabetic groups, but there were fewer taste buds (per papilla). CONCLUSIONS Increased number of immunoreactive nerve terminals and mast cells 2 weeks after the development of diabetes was the consequence of neurogenic inflammation which might cause vasoconstriction and lesions of the oral mucosa. Taste impairment, which developed 4 weeks after streptozotocin treatment could be caused by neuropathic defects and degeneration or morphological changes in the taste buds and nerve fibres.
Collapse
Affiliation(s)
- Helén Hevér
- Semmelweis Egyetem, Fogorvos-tudományi Kar Budapest
| | | | | | | | | |
Collapse
|
33
|
Dotson CD, Geraedts MCP, Munger SD. Peptide regulators of peripheral taste function. Semin Cell Dev Biol 2013; 24:232-9. [PMID: 23348523 DOI: 10.1016/j.semcdb.2013.01.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/10/2013] [Indexed: 12/25/2022]
Abstract
The peripheral sensory organ of the gustatory system, the taste bud, contains a heterogeneous collection of sensory cells. These taste cells can differ in the stimuli to which they respond and the receptors and other signaling molecules they employ to transduce and encode those stimuli. This molecular diversity extends to the expression of a varied repertoire of bioactive peptides that appear to play important functional roles in signaling taste information between the taste cells and afferent sensory nerves and/or in processing sensory signals within the taste bud itself. Here, we review studies that examine the expression of bioactive peptides in the taste bud and the impact of those peptides on taste functions. Many of these peptides produced in taste buds are known to affect appetite, satiety or metabolism through their actions in the brain, pancreas and other organs, suggesting a functional link between the gustatory system and the neural and endocrine systems that regulate feeding and nutrient utilization.
Collapse
Affiliation(s)
- Cedrick D Dotson
- Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32611, USA
| | | | | |
Collapse
|
34
|
Page AJ, Symonds E, Peiris M, Blackshaw LA, Young RL. Peripheral neural targets in obesity. Br J Pharmacol 2012; 166:1537-58. [PMID: 22432806 PMCID: PMC3419899 DOI: 10.1111/j.1476-5381.2012.01951.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 12/15/2022] Open
Abstract
Interest in pharmacological treatments for obesity that act in the brain to reduce appetite has increased exponentially over recent years, but failures of clinical trials and withdrawals due to adverse effects have so far precluded any success. Treatments that do not act within the brain are, in contrast, a neglected area of research and development. This is despite the fact that a vast wealth of molecular mechanisms exists within the gut epithelium and vagal afferent system that could be manipulated to increase satiety. Here we discuss mechano- and chemosensory pathways from the gut involved in appetite suppression, and distinguish between gastric and intestinal vagal afferent pathways in terms of their basic physiology and activation by enteroendocrine factors. Gastric bypass surgery makes use of this system by exposing areas of the intestine to greater nutrient loads resulting in greater satiety hormone release and reduced food intake. A non-surgical approach to this system is preferable for many reasons. This review details where the opportunities may lie for such approaches by describing nutrient-sensing mechanisms throughout the gastrointestinal tract.
Collapse
Affiliation(s)
- Amanda J Page
- Nerve-Gut Research Laboratory, Discipline of Medicine, South Australia, Australia
| | | | | | | | | |
Collapse
|
35
|
Sclafani A, Ackroff K. Role of gut nutrient sensing in stimulating appetite and conditioning food preferences. Am J Physiol Regul Integr Comp Physiol 2012; 302:R1119-33. [PMID: 22442194 PMCID: PMC3362145 DOI: 10.1152/ajpregu.00038.2012] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 03/14/2012] [Indexed: 12/17/2022]
Abstract
The discovery of taste and nutrient receptors (chemosensors) in the gut has led to intensive research on their functions. Whereas oral sugar, fat, and umami taste receptors stimulate nutrient appetite, these and other chemosensors in the gut have been linked to digestive, metabolic, and satiating effects that influence nutrient utilization and inhibit appetite. Gut chemosensors may have an additional function as well: to provide positive feedback signals that condition food preferences and stimulate appetite. The postoral stimulatory actions of nutrients are documented by flavor preference conditioning and appetite stimulation produced by gastric and intestinal infusions of carbohydrate, fat, and protein. Recent findings suggest an upper intestinal site of action, although postabsorptive nutrient actions may contribute to flavor preference learning. The gut chemosensors that generate nutrient conditioning signals remain to be identified; some have been excluded, including sweet (T1R3) and fatty acid (CD36) sensors. The gut-brain signaling pathways (neural, hormonal) are incompletely understood, although vagal afferents are implicated in glutamate conditioning but not carbohydrate or fat conditioning. Brain dopamine reward systems are involved in postoral carbohydrate and fat conditioning but less is known about the reward systems mediating protein/glutamate conditioning. Continued research on the postoral stimulatory actions of nutrients may enhance our understanding of human food preference learning.
Collapse
Affiliation(s)
- Anthony Sclafani
- Department of Psychology, Brooklyn College, City University of New York, Brooklyn, NY 11210, USA.
| | | |
Collapse
|
36
|
|
37
|
|
38
|
Abstract
In response to gustatory stimulation, taste bud cells release a transmitter, ATP, that activates P2X2 and P2X3 receptors on gustatory afferent fibers. Taste behavior and gustatory neural responses are largely abolished in mice lacking P2X2 and P2X3 receptors [P2X2 and P2X3 double knock-out (DKO) mice]. The assumption has been that eliminating P2X2 and P2X3 receptors only removes postsynaptic targets but that transmitter secretion in mice is normal. Using functional imaging, ATP biosensor cells, and a cell-free assay for ATP, we tested this assumption. Surprisingly, although gustatory stimulation mobilizes Ca(2+) in taste Receptor (Type II) cells from DKO mice, as from wild-type (WT) mice, taste cells from DKO mice fail to release ATP when stimulated with tastants. ATP release could be elicited by depolarizing DKO Receptor cells with KCl, suggesting that ATP-release machinery remains functional in DKO taste buds. To explore the difference in ATP release across genotypes, we used reverse transcriptase (RT)-PCR, immunostaining, and histochemistry for key proteins underlying ATP secretion and degradation: Pannexin1, TRPM5, and NTPDase2 (ecto-ATPase) are indistinguishable between WT and DKO mice. The ultrastructure of contacts between taste cells and nerve fibers is also normal in the DKO mice. Finally, quantitative RT-PCR show that P2X4 and P2X7, potential modulators of ATP secretion, are similarly expressed in taste buds in WT and DKO taste buds. Importantly, we find that P2X2 is expressed in WT taste buds and appears to function as an autocrine, positive feedback signal to amplify taste-evoked ATP secretion.
Collapse
|
39
|
Abstract
Peptide YY3-36 is a satiation hormone released postprandially into the bloodstream from L-endocrine cells in the gut epithelia. In the current report, we demonstrate PYY3-36 is also present in murine as well as in human saliva. In mice, salivary PYY3-36 derives from plasma and is also synthesized in the taste cells in taste buds of the tongue. Moreover, the cognate receptor Y2R is abundantly expressed in the basal layer of the progenitor cells of the tongue epithelia and von Ebner's gland. The acute augmentation of salivary PYY3-36 induced stronger satiation as demonstrated in feeding behavioral studies. The effect is mediated through the activation of the specific Y2 receptor expressed in the lingual epithelial cells. In a long-term study involving diet-induced obese (DIO) mice, a sustained increase in PYY3-36 was achieved using viral vector-mediated gene delivery targeting salivary glands. The chronic increase in salivary PYY3-36 resulted in a significant long-term reduction in food intake (FI) and body weight (BW). Thus this study provides evidence for new functions of the previously characterized gut peptide PYY3-36 suggesting a potential simple and efficient alternative therapeutic approach for the treatment of obesity.
Collapse
|
40
|
Zhang XJ, Zhou LH, Ban X, Liu DX, Jiang W, Liu XM. Decreased expression of CD36 in circumvallate taste buds of high-fat diet induced obese rats. Acta Histochem 2011; 113:663-7. [PMID: 20926122 DOI: 10.1016/j.acthis.2010.09.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 08/21/2010] [Accepted: 09/07/2010] [Indexed: 11/24/2022]
Abstract
Mammals spontaneously prefer lipid rich foods. Overconsumption of high-fat diet leads to obesity and related diseases. Recent findings indicate that taste may participate in the orosensory perception of dietary lipids and the fatty taste may contribute to a preference for and excessive consumption of dietary fat. CD36, a trans-membrane glycoprotein, which is located in the taste buds of circumvallate papillae of rodents, appears to be a plausible receptor for this fatty taste. Obese subjects present a stronger preference for fatty foods, though the mechanisms involved are complex and are not fully investigated. Our data from immunofluorescence and real-time RT-PCR showed that the expression levels of CD36 in circumvallate taste buds were significantly lower in high-fat diet induced obese rats as compared with that of control rats fed a normal diet. These results suggest that decreased expression of CD36 in circumvallate taste buds of high-fat diet induced obese rats may be associated with diminished fatty taste sensitivity and in order to compensate the preference for dietary fat, rats consume more fatty foods. Therapeutic strategies designed to alter or manipulate CD36 expression or function in taste buds may have important implications in treating obesity and related diseases.
Collapse
|
41
|
Up-regulation of intestinal type 1 taste receptor 3 and sodium glucose luminal transporter-1 expression and increased sucrose intake in mice lacking gut microbiota. Br J Nutr 2011; 107:621-30. [PMID: 21781379 DOI: 10.1017/s0007114511003412] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The chemosensory components shared by both lingual and intestinal epithelium play a critical role in food consumption and the regulation of intestinal functions. In addition to nutrient signals, other luminal contents, including micro-organisms, are important in signalling across the gastrointestinal mucosa and initiating changes in digestive functions. A potential role of gut microbiota in influencing food intake, energy homeostasis and weight gain has been suggested. However, whether gut microbiota modulates the expression of nutrient-responsive receptors and transporters, leading to altered food consumption, is unknown. Thus, we examined the preference for nutritive (sucrose) and non-nutritive (saccharin) sweet solutions in germ-free (GF, C57BL/6J) mice compared with conventional (CV, C57BL/6J) control mice using a two-bottle preference test. Then, we quantified mRNA and protein expression of the sweet signalling protein type 1 taste receptor 3 (T1R3) and α-gustducin and Na glucose luminal transporter-1 (SGLT-1) of the intestinal epithelium of both CV and GF mice. Additionally, we measured gene expression of T1R2, T1R3 and α-gustducin in the lingual epithelium. We found that, while the preference for sucrose was similar between the groups, GF mice consumed more of the high concentration (8 %) of sucrose solution than CV mice. There was no difference in either the intake of or the preference for saccharin. GF mice expressed significantly more T1R3 and SGLT-1 mRNA and protein in the intestinal epithelium compared with CV mice; however, lingual taste receptor mRNA expression was similar between the groups. We conclude that the absence of intestinal microbiota alters the expression of sweet taste receptors and GLUT in the proximal small intestine, which is associated with increased consumption of nutritive sweet solutions.
Collapse
|
42
|
Stengel A, Taché Y. Interaction between gastric and upper small intestinal hormones in the regulation of hunger and satiety: ghrelin and cholecystokinin take the central stage. Curr Protein Pept Sci 2011; 12:293-304. [PMID: 21428875 PMCID: PMC3670092 DOI: 10.2174/138920311795906673] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 03/23/2011] [Indexed: 12/13/2022]
Abstract
Several peptides are produced and released from endocrine cells scattered within the gastric oxyntic and the small intestinal mucosa. These peptide hormones are crucially involved in the regulation of gastrointestinal functions and food intake by conveying their information to central regulatory sites located in the brainstem as well as in the forebrain, such as hypothalamic nuclei. So far, ghrelin is the only known hormone that is peripherally produced in gastric X/A-like cells and centrally acting to stimulate food intake, whereas the suppression of feeding seems to be much more redundantly controlled by a number of gut peptides. Cholecystokinin produced in the duodenum is a well established anorexigenic hormone that interacts with ghrelin to modulate food intake indicating a regulatory network located at the first site of contact with nutrients in the stomach and upper small intestine. In addition, a number of peptides including leptin, urocortin 2, amylin and glucagon-like peptide 1 interact synergistically with CCK to potentiate its satiety signaling effect. New developments have led to the identification of additional peptides in X/A-like cells either derived from the pro-ghrelin gene by alternative splicing and posttranslational processing (obestatin) or a distinct gene (nucleobindin2/nesfatin-1) which have been investigated for their influence on food intake.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yvette Taché
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
43
|
Merigo F, Benati D, Cristofoletti M, Osculati F, Sbarbati A. Glucose transporters are expressed in taste receptor cells. J Anat 2011. [PMID: 21592100 DOI: 10.1111/j.1469‐7580.2011.01385.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In the intestine, changes of sugar concentration generated in the lumen during digestion induce adaptive responses of glucose transporters in the epithelium. A close matching between the intestinal expression of glucose transporters and the composition and amount of the diet has been provided by several experiments. Functional evidence has demonstrated that the regulation of glucose transporters into enterocytes is induced by the sensing of sugar of the enteroendocrine cells through activation of sweet taste receptors (T1R2 and T1R3) and their associated elements of G-protein-linked signaling pathways (e.g. α-gustducin, phospholipase C β type 2 and transient receptor potential channel M5), which are signaling molecules also involved in the perception of sweet substances in the taste receptor cells (TRCs) of the tongue. Considering this phenotypical similarity between the intestinal cells and TRCs, we evaluated whether the TRCs themselves possess proteins of the glucose transport mechanism. Therefore, we investigated the expression of the typical intestinal glucose transporters (i.e. GLUT2, GLUT5 and SGLT1) in rat circumvallate papillae, using immunohistochemistry, double-labeling immunofluorescence, immunoelectron microscopy and reverse transcriptase-polymerase chain reaction analysis. The results showed that GLUT2, GLUT5 and SGLT1 are expressed in TRCs; their immunoreactivity was also observed in cells that displayed staining for α-gustducin and T1R3 receptor. The immunoelectron microscopic results confirmed that GLUT2, GLUT5 and SGLT1 were predominantly expressed in cells with ultrastructural characteristics of chemoreceptor cells. The presence of glucose transporters in TRCs adds a further link between chemosensory information and cellular responses to sweet stimuli that may have important roles in glucose homeostasis, contributing to a better understanding of the pathways implicated in glucose metabolism.
Collapse
Affiliation(s)
- Flavia Merigo
- Department of Neurological, Neuropsychological, Morphological and Movement Sciences, Human Anatomy and Histology Section, School of Medicine, University of Verona, Verona, Italy.
| | | | | | | | | |
Collapse
|
44
|
Merigo F, Benati D, Cristofoletti M, Osculati F, Sbarbati A. Glucose transporters are expressed in taste receptor cells. J Anat 2011; 219:243-52. [PMID: 21592100 DOI: 10.1111/j.1469-7580.2011.01385.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In the intestine, changes of sugar concentration generated in the lumen during digestion induce adaptive responses of glucose transporters in the epithelium. A close matching between the intestinal expression of glucose transporters and the composition and amount of the diet has been provided by several experiments. Functional evidence has demonstrated that the regulation of glucose transporters into enterocytes is induced by the sensing of sugar of the enteroendocrine cells through activation of sweet taste receptors (T1R2 and T1R3) and their associated elements of G-protein-linked signaling pathways (e.g. α-gustducin, phospholipase C β type 2 and transient receptor potential channel M5), which are signaling molecules also involved in the perception of sweet substances in the taste receptor cells (TRCs) of the tongue. Considering this phenotypical similarity between the intestinal cells and TRCs, we evaluated whether the TRCs themselves possess proteins of the glucose transport mechanism. Therefore, we investigated the expression of the typical intestinal glucose transporters (i.e. GLUT2, GLUT5 and SGLT1) in rat circumvallate papillae, using immunohistochemistry, double-labeling immunofluorescence, immunoelectron microscopy and reverse transcriptase-polymerase chain reaction analysis. The results showed that GLUT2, GLUT5 and SGLT1 are expressed in TRCs; their immunoreactivity was also observed in cells that displayed staining for α-gustducin and T1R3 receptor. The immunoelectron microscopic results confirmed that GLUT2, GLUT5 and SGLT1 were predominantly expressed in cells with ultrastructural characteristics of chemoreceptor cells. The presence of glucose transporters in TRCs adds a further link between chemosensory information and cellular responses to sweet stimuli that may have important roles in glucose homeostasis, contributing to a better understanding of the pathways implicated in glucose metabolism.
Collapse
Affiliation(s)
- Flavia Merigo
- Department of Neurological, Neuropsychological, Morphological and Movement Sciences, Human Anatomy and Histology Section, School of Medicine, University of Verona, Verona, Italy.
| | | | | | | | | |
Collapse
|
45
|
Young RL. Sensing via intestinal sweet taste pathways. Front Neurosci 2011; 5:23. [PMID: 21519398 PMCID: PMC3080736 DOI: 10.3389/fnins.2011.00023] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Accepted: 02/10/2011] [Indexed: 12/15/2022] Open
Abstract
The detection of nutrients in the gastrointestinal (GI) tract is of fundamental significance to the control of motility, glycemia and energy intake, and yet we barely know the most fundamental aspects of this process. This is in stark contrast to the mechanisms underlying the detection of lingual taste, which have been increasingly well characterized in recent years, and which provide an excellent starting point for characterizing nutrient detection in the intestine. This review focuses on the form and function of sweet taste transduction mechanisms identified in the intestinal tract; it does not focus on sensors for fatty acids or proteins. It examines the intestinal cell types equipped with sweet taste transduction molecules in animals and humans, their location, and potential signals that transduce the presence of nutrients to neural pathways involved in reflex control of GI motility.
Collapse
Affiliation(s)
- Richard L Young
- Discipline of Medicine, School of Medicine, University of Adelaide Adelaide, SA, Australia
| |
Collapse
|
46
|
Vandenbeuch A, Zorec R, Kinnamon SC. Capacitance measurements of regulated exocytosis in mouse taste cells. J Neurosci 2010; 30:14695-701. [PMID: 21048127 PMCID: PMC3064517 DOI: 10.1523/jneurosci.1570-10.2010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 08/30/2010] [Accepted: 09/07/2010] [Indexed: 11/21/2022] Open
Abstract
Exocytosis, consisting of the merger of vesicle and plasma membrane, is a common mechanism used by different types of nucleated cells to release their vesicular contents. Taste cells possess vesicles containing various neurotransmitters to communicate with adjacent taste cells and afferent nerve fibers. However, whether these vesicles engage in exocytosis on a stimulus is not known. Since vesicle membrane merger with the plasma membrane is reflected in plasma membrane area fluctuations, we measured membrane capacitance (C(m)), a parameter linearly related to membrane surface area. To investigate whether taste cells undergo regulated exocytosis, we used the compensated tight-seal whole-cell recording technique to monitor depolarization-induced changes in C(m) in the different types of taste cells. To identify taste cell types, mice expressing green fluorescent protein from the TRPM5 promoter or from the GAD67 promoter were used to discriminate type II and type III taste cells, respectively. Moreover, the cell types were also identified by monitoring their voltage-current properties. The results demonstrate that only type III taste cells show significant depolarization-induced increases in C(m), which were correlated to the voltage-activated calcium currents. The results suggest that type III, but neither type II nor type I cells exhibit depolarization-induced regulated exocytosis to release transmitter and activate gustatory afferent nerve fibers.
Collapse
Affiliation(s)
- Aurelie Vandenbeuch
- Department of Otolaryngology and
- Rocky Mountain Taste and Smell Center, University of Colorado, Denver, Aurora, Colorado 80045, and
| | - Robert Zorec
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, and
- Laboratory of Cell Engineering, Celica, Biomedical Center, 1000 Ljubljana, Slovenia
| | - Sue C. Kinnamon
- Department of Otolaryngology and
- Rocky Mountain Taste and Smell Center, University of Colorado, Denver, Aurora, Colorado 80045, and
| |
Collapse
|
47
|
Expression of GABAergic receptors in mouse taste receptor cells. PLoS One 2010; 5:e13639. [PMID: 21049022 PMCID: PMC2964312 DOI: 10.1371/journal.pone.0013639] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 10/04/2010] [Indexed: 12/05/2022] Open
Abstract
Background Multiple excitatory neurotransmitters have been identified in the mammalian taste transduction, with few studies focused on inhibitory neurotransmitters. Since the synthetic enzyme glutamate decarboxylase (GAD) for gamma-aminobutyric acid (GABA) is expressed in a subset of mouse taste cells, we hypothesized that other components of the GABA signaling pathway are likely expressed in this system. GABA signaling is initiated by the activation of either ionotropic receptors (GABAA and GABAC) or metabotropic receptors (GABAB) while it is terminated by the re-uptake of GABA through transporters (GATs). Methodology/Principal Findings Using reverse transcriptase-PCR (RT-PCR) analysis, we investigated the expression of different GABA signaling molecules in the mouse taste system. Taste receptor cells (TRCs) in the circumvallate papillae express multiple subunits of the GABAA and GABAB receptors as well as multiple GATs. Immunocytochemical analyses examined the distribution of the GABA machinery in the circumvallate papillae. Both GABAA-and GABAB- immunoreactivity were detected in the peripheral taste receptor cells. We also used transgenic mice that express green fluorescent protein (GFP) in either the Type II taste cells, which can respond to bitter, sweet or umami taste stimuli, or in the Type III GAD67 expressing taste cells. Thus, we were able to identify that GABAergic receptors are expressed in some Type II and Type III taste cells. Mouse GAT4 labeling was concentrated in the cells surrounding the taste buds with a few positively labeled TRCs at the margins of the taste buds. Conclusions/Significance The presence of GABAergic receptors localized on Type II and Type III taste cells suggests that GABA is likely modulating evoked taste responses in the mouse taste bud.
Collapse
|
48
|
Elson AE, Dotson CD, Egan JM, Munger SD. Glucagon signaling modulates sweet taste responsiveness. FASEB J 2010; 24:3960-9. [PMID: 20547661 PMCID: PMC2996909 DOI: 10.1096/fj.10-158105] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 05/27/2010] [Indexed: 11/11/2022]
Abstract
The gustatory system provides critical information about the quality and nutritional value of food before it is ingested. Thus, physiological mechanisms that modulate taste function in the context of nutritional needs or metabolic status could optimize ingestive decisions. We report that glucagon, which plays important roles in the maintenance of glucose homeostasis, enhances sweet taste responsiveness through local actions in the mouse gustatory epithelium. Using immunohistochemistry and confocal microscopy, we found that glucagon and its receptor (GlucR) are coexpressed in a subset of mouse taste receptor cells. Most of these cells also express the T1R3 taste receptor implicated in sweet and/or umami taste. Genetic or pharmacological disruption of glucagon signaling in behaving mice indicated a critical role for glucagon in the modulation of taste responsiveness. Scg5(-/-) mice, which lack mature glucagon, had significantly reduced responsiveness to sucrose as compared to wild-type littermates in brief-access taste tests. No significant differences were seen in responses to prototypical salty, sour, or bitter stimuli. Taste responsiveness to sucrose was similarly reduced upon acute and local disruption of glucagon signaling by the GlucR antagonist L-168,049. Together, these data indicate a role for local glucagon signaling in the peripheral modulation of sweet taste responsiveness.
Collapse
Affiliation(s)
- Amanda E.T. Elson
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; and
| | - Cedrick D. Dotson
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; and
| | - Josephine M. Egan
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, USA
| | - Steven D. Munger
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; and
| |
Collapse
|
49
|
Shin YK, Martin B, Kim W, White CM, Ji S, Sun Y, Smith RG, Sévigny J, Tschöp MH, Maudsley S, Egan JM. Ghrelin is produced in taste cells and ghrelin receptor null mice show reduced taste responsivity to salty (NaCl) and sour (citric acid) tastants. PLoS One 2010; 5:e12729. [PMID: 20856820 PMCID: PMC2939079 DOI: 10.1371/journal.pone.0012729] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 08/11/2010] [Indexed: 12/14/2022] Open
Abstract
Background The gustatory system plays a critical role in determining food preferences, food intake and energy balance. The exact mechanisms that fine tune taste sensitivity are currently poorly defined, but it is clear that numerous factors such as efferent input and specific signal transduction cascades are involved. Methodology/Principal Findings Using immunohistochemical analyses, we show that ghrelin, a hormone classically considered to be an appetite-regulating hormone, is present within the taste buds of the tongue. Prepro-ghrelin, prohormone convertase 1/3 (PC 1/3), ghrelin, its cognate receptor (GHSR), and ghrelin-O-acyltransferase (GOAT , the enzyme that activates ghrelin) are expressed in Type I, II, III and IV taste cells of mouse taste buds. In addition, ghrelin and GHSR co-localize in the same taste cells, suggesting that ghrelin works in an autocrine manner in taste cells. To determine a role for ghrelin in modifying taste perception, we performed taste behavioral tests using GHSR null mice. GHSR null mice exhibited significantly reduced taste responsivity to sour (citric acid) and salty (sodium chloride) tastants. Conclusions/Significance These findings suggest that ghrelin plays a local modulatory role in determining taste bud signaling and function and could be a novel mechanism for the modulation of salty and sour taste responsivity.
Collapse
Affiliation(s)
- Yu-Kyong Shin
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Bronwen Martin
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Wook Kim
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Caitlin M. White
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Sunggoan Ji
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yuxiang Sun
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, United States of America
| | - Roy G. Smith
- Department of Metabolism and Aging, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Jean Sévigny
- Centre de Recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec City, Québec, Canada
| | - Matthias H. Tschöp
- Division of Endocrinology, Departments of Medicine and Psychiatry, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Stuart Maudsley
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Josephine M. Egan
- National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
50
|
Martin B, Shin YK, White CM, Ji S, Kim W, Carlson OD, Napora JK, Chadwick W, Chapter M, Waschek JA, Mattson MP, Maudsley S, Egan JM. Vasoactive intestinal peptide-null mice demonstrate enhanced sweet taste preference, dysglycemia, and reduced taste bud leptin receptor expression. Diabetes 2010; 59:1143-52. [PMID: 20150284 PMCID: PMC2857894 DOI: 10.2337/db09-0807] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE It is becoming apparent that there is a strong link between taste perception and energy homeostasis. Recent evidence implicates gut-related hormones in taste perception, including glucagon-like peptide 1 and vasoactive intestinal peptide (VIP). We used VIP knockout mice to investigate VIP's specific role in taste perception and connection to energy regulation. RESEARCH DESIGN AND METHODS Body weight, food intake, and plasma levels of multiple energy-regulating hormones were measured and pancreatic morphology was determined. In addition, the immunocytochemical profile of taste cells and gustatory behavior were examined in wild-type and VIP knockout mice. RESULTS VIP knockout mice demonstrate elevated plasma glucose, insulin, and leptin levels, with no islet beta-cell number/topography alteration. VIP and its receptors (VPAC1, VPAC2) were identified in type II taste cells of the taste bud, and VIP knockout mice exhibit enhanced taste preference to sweet tastants. VIP knockout mouse taste cells show a significant decrease in leptin receptor expression and elevated expression of glucagon-like peptide 1, which may explain sweet taste preference of VIP knockout mice. CONCLUSIONS This study suggests that the tongue can play a direct role in modulating energy intake to correct peripheral glycemic imbalances. In this way, we could view the tongue as a sensory mechanism that is bidirectionally regulated and thus forms a bridge between available foodstuffs and the intricate hormonal balance in the animal itself.
Collapse
Affiliation(s)
- Bronwen Martin
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Yu-Kyong Shin
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Caitlin M. White
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Sunggoan Ji
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Wook Kim
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Olga D. Carlson
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Joshua K. Napora
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Wayne Chadwick
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Megan Chapter
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - James A. Waschek
- Department of Psychiatry and Behavioral Sciences, Mental Retardation Research Center, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California
| | - Mark P. Mattson
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Stuart Maudsley
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
| | - Josephine M. Egan
- National Institutes of Health, National Institute on Aging, Baltimore, Maryland
- Corresponding author: Josephine Egan,
| |
Collapse
|