1
|
Iyer S, Montmayeur JP, Zolotukhin S, Dotson CD. Exogenous oral application of PYY and exendin-4 impacts upon taste-related behavior and taste perception in wild-type mice. Neuropharmacology 2025; 272:110408. [PMID: 40086622 PMCID: PMC12042652 DOI: 10.1016/j.neuropharm.2025.110408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Several gut peptides have been implicated in feeding and body mass accumulation. Glucagon-like peptide 1 (GLP-1) and peptide tyrosine-tyrosine (PYY) have been shown to mediate satiety and reduce food intake. While systemic administration of such peptides has been explored as a therapy for metabolic disease, the effects of these hormones on taste signaling should also be considered given the importance of taste to feeding decisions and considering the fact that components of these signaling systems are expressed in cells of the peripheral gustatory system. We previously demonstrated that genetic disruption of PYY signaling in mice can impact on taste responsiveness and feeding and that viral expression of PYY in the salivary glands of PYY knockout mice can rescue responsiveness. The present work uses adeno-associated virus-mediated salivary gland treatment with both GLP-1 receptor agonist exendin-4 and/or PYY encoding vectors to explore the effect of stimulating these orally present signaling systems on taste-related behavioral responsiveness in male wild-type mice with intact peptide signaling systems. Results showed a significant effect of salivary gland treatment on responsiveness to multiple taste qualities. Data gathered from taste bud cells in vitro suggest that these peptides directly influence the responsiveness of these primary sensory cells. Collectively, these findings show that taste perception can be modulated by the exogenous application of satiety peptides in wild-type mice and suggest that the taste bud is a promising substrate for food intake modulation.
Collapse
Affiliation(s)
- Satya Iyer
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA
| | | | - Sergei Zolotukhin
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Cedrick D Dotson
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
2
|
Abstract
Taste is one of our five primary senses, and taste impairment has been shown to increase with aging. The ability to taste allows us to enjoy the food we eat and to avoid foods that are potentially spoiled or poisonous. Recent advances in our understanding of the molecular mechanisms of taste receptor cells located within taste buds help us decipher how taste works. The discoveries of "classic" endocrine hormones in taste receptor cells point toward taste buds being actual endocrine organs. A better understanding of how taste works may help in reversing taste impairment associated with aging.
Collapse
Affiliation(s)
- Chee W Chia
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 3001 S. Hanover Street, 5th Floor, Room NM536, Baltimore, MD 21225, USA
| | - Shayna M Yeager
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 3001 S. Hanover Street, 5th Floor, Room NM547, Baltimore, MD 21225, USA
| | - Josephine M Egan
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 3001 S. Hanover Street, 5th Floor, Room NM527, Baltimore, MD 21225, USA.
| |
Collapse
|
3
|
Doyle ME, Premathilake HU, Yao Q, Mazucanti CH, Egan JM. Physiology of the tongue with emphasis on taste transduction. Physiol Rev 2023; 103:1193-1246. [PMID: 36422992 PMCID: PMC9942923 DOI: 10.1152/physrev.00012.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The tongue is a complex multifunctional organ that interacts and senses both interoceptively and exteroceptively. Although it is easily visible to almost all of us, it is relatively understudied and what is in the literature is often contradictory or is not comprehensively reported. The tongue is both a motor and a sensory organ: motor in that it is required for speech and mastication, and sensory in that it receives information to be relayed to the central nervous system pertaining to the safety and quality of the contents of the oral cavity. Additionally, the tongue and its taste apparatus form part of an innate immune surveillance system. For example, loss or alteration in taste perception can be an early indication of infection as became evident during the present global SARS-CoV-2 pandemic. Here, we particularly emphasize the latest updates in the mechanisms of taste perception, taste bud formation and adult taste bud renewal, and the presence and effects of hormones on taste perception, review the understudied lingual immune system with specific reference to SARS-CoV-2, discuss nascent work on tongue microbiome, as well as address the effect of systemic disease on tongue structure and function, especially in relation to taste.
Collapse
Affiliation(s)
- Máire E Doyle
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Hasitha U Premathilake
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Qin Yao
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Caio H Mazucanti
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M Egan
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
4
|
Andreozzi F, Di Fatta C, Spiga R, Mannino GC, Mancuso E, Averta C, De Caro C, Tallarico M, Leo A, Citraro R, Russo E, De Sarro G, Sesti G. Glucagon induces the hepatic expression of inflammatory markers in vitro and in vivo. Diabetes Obes Metab 2023; 25:556-569. [PMID: 36305474 DOI: 10.1111/dom.14902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/12/2022] [Accepted: 10/24/2022] [Indexed: 02/02/2023]
Abstract
Glucagon exerts multiple hepatic actions, including stimulation of glycogenolysis/gluconeogenesis. The liver plays a crucial role in chronic inflammation by synthesizing proinflammatory molecules, which are thought to contribute to insulin resistance and hyperglycaemia. Whether glucagon affects hepatic expression of proinflammatory cytokines and acute-phase reactants is unknown. Herein, we report a positive relationship between fasting glucagon levels and circulating interleukin (IL)-1β (r = 0.252, p = .042), IL-6 (r = 0.230, p = .026), fibrinogen (r = 0.193, p = .031), complement component 3 (r = 0.227, p = .024) and high sensitivity C-reactive protein (r = 0.230, p = .012) in individuals without diabetes. In CD1 mice, 4-week continuous treatment with glucagon induced a significant increase in circulating IL-1β (p = .02), and IL-6 (p = .001), which was countered by the contingent administration of the glucagon receptor antagonist, GRA-II. Consistent with these results, we detected a significant increase in the hepatic activation of inflammatory pathways, such as expression of NLRP3 (p < .02), and the phosphorylation of nuclear factor kappaB (NF-κB; p < .02) and STAT3 (p < .01). In HepG2 cells, we found that glucagon dose-dependently stimulated the expression of IL-1β (p < .002), IL-6 (p < .002), fibrinogen (p < .01), complement component 3 (p < .01) and C-reactive protein (p < .01), stimulated the activation of NLRP3 inflammasome (p < .01) and caspase-1 (p < .05), induced the phosphorylation of TRAF2 (p < .01), NF-κB (p < .01) and STAT3 (p < .01). Preincubating cells with GRA-II inhibited the ability of glucagon to induce an inflammatory response. Using HepaRG cells, we confirmed the dose-dependent ability of glucagon to stimulate the expression of NLRP3, the phosphorylation of NF-κB and STAT3, in the absence of GRA-II. These results suggest that glucagon has proinflammatory effects that may participate in the pathogenesis of hyperglycaemia and unfavourable cardiometabolic risk profile.
Collapse
Affiliation(s)
- Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
- Research Center for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Concetta Di Fatta
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Rosangela Spiga
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Gaia Chiara Mannino
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Elettra Mancuso
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Carolina Averta
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Carmen De Caro
- Department of Science of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Martina Tallarico
- Department of Science of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonio Leo
- Department of Science of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Rita Citraro
- Department of Science of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Emilio Russo
- Department of Science of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, Rome, Italy
| |
Collapse
|
5
|
Karikkineth AC, Tang EY, Kuo PL, Ferrucci L, Egan JM, Chia CW. Longitudinal trajectories and determinants of human fungiform papillae density. Aging (Albany NY) 2021; 13:24989-25003. [PMID: 34857670 PMCID: PMC8714156 DOI: 10.18632/aging.203741] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/23/2021] [Indexed: 01/08/2023]
Abstract
Tongue fungiform papillae contain taste buds crucial for taste and hormone-producing taste receptor cells; therefore, they may be considered as endocrine organs and have important age-associated physiological implications. We examine the cross-sectional and longitudinal trajectories of fungiform papillae density in 1084 participants from the Baltimore Longitudinal Study of Aging using linear regression models and mixed effects models. At baseline, the mean age was 67.86 ± 14.20 years, with a mean follow-up time among those with repeat visits of 4.24 ± 1.70 years. Women (53%) were younger (66.85 ± 13.78 vs. 69.04 ± 14.61 years, p < 0.001) and had a higher fungiform papillae density than men (16.14 ± 9.54 vs. 13.77 ± 8.61 papillae/cm2, p < 0.001). Whites (67%) had a lower fungiform papillae density than non-Whites after adjusting for age and sex. Factors cross-sectionally associated with a lower fungiform papillae density included a higher waist-hip ratio (β = −8.525, p = 0.029), current smoking status (β = −5.133, p = 0.014), and alcohol use within the past 12 months (β = −1.571, p = 0.025). Longitudinally, fungiform papillae density decreased linearly with follow-up time (β = −0.646, p < 0.001). The rate of decline was not affected by sex, race, BMI, waist-hip ratio, smoking, or alcohol use. The longitudinal decline of fungiform papillae density over time needs to be explored further in order to identify other possible age-associated physiological determinants.
Collapse
Affiliation(s)
- Ajoy C Karikkineth
- Clinical Core Laboratory and Biorepository, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Eric Y Tang
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Pei-Lun Kuo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Chee W Chia
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
6
|
Jensterle M, DeVries JH, Battelino T, Battelino S, Yildiz B, Janez A. Glucagon-like peptide-1, a matter of taste? Rev Endocr Metab Disord 2021; 22:763-775. [PMID: 33123893 DOI: 10.1007/s11154-020-09609-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 01/22/2023]
Abstract
Understanding of gustatory coding helps to predict, and perhaps even modulate the ingestive decision circuitry, especially when eating behaviour becomes dysfunctional. Preclinical research demonstrated that glucagon like peptide 1 (GLP-1) is locally synthesized in taste bud cells in the tongue and that GLP-1 receptor exists on the gustatory nerves in close proximity to GLP-1 containing taste bud cells. In humans, the tongue has not yet been addressed as clinically relevant target for GLP-1 based therapies. The primary aim of the current review was to elaborate on the role of GLP- 1 in mammalian gustatory system, in particular in the perception of sweet. Secondly, we aimed to explore what modulates gustatory coding and whether the GLP-1 based therapies might be involved in regulation of taste perception. We performed a series of PubMed, Medline and Embase databases systemic searches. The Population-Intervention-Comparison-Outcome (PICO) framework was used to identify interventional studies. Based on the available data, GLP-1 is specifically involved in the perception of sweet. Aging, diabetes and obesity are characterized by diminished taste and sweet perception. Calorie restriction and bariatric surgery are associated with a diminished appreciation of sweet food. GLP-1 receptor agonists (RAs) modulate food preference, yet its modulatory potential in gustatory coding is currently unknown. Future studies should explore whether GLP-1 RAs modulate taste perception to the extent that changes of food preference and consumption ensue.
Collapse
Affiliation(s)
- Mojca Jensterle
- Division of Internal Medicine, Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška cesta, 7, 1000, Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - J Hans DeVries
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolism, University Children's Hospital, University Medical Centre Ljubljana, Bohoričeva 20, SI-1000, Ljubljana, Slovenia
- Department of Pediatrics, Faculty of Medicine, University of Ljubljana, Bohoričeva 20, SI-1000, Ljubljana, Slovenia
| | - Saba Battelino
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Bulent Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Hacettepe, 06100, Ankara, Turkey
| | - Andrej Janez
- Division of Internal Medicine, Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška cesta, 7, 1000, Ljubljana, Slovenia.
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia, Zaloška cesta 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
7
|
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
|
8
|
Jang JH, Kwon O, Moon SJ, Jeong YT. Recent Advances in Understanding Peripheral Taste Decoding I: 2010 to 2020. Endocrinol Metab (Seoul) 2021; 36:469-477. [PMID: 34139798 PMCID: PMC8258330 DOI: 10.3803/enm.2021.302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/20/2021] [Indexed: 01/03/2023] Open
Abstract
Taste sensation is the gatekeeper for direct decisions on feeding behavior and evaluating the quality of food. Nutritious and beneficial substances such as sugars and amino acids are represented by sweet and umami tastes, respectively, whereas noxious substances and toxins by bitter or sour tastes. Essential electrolytes including Na+ and other ions are recognized by the salty taste. Gustatory information is initially generated by taste buds in the oral cavity, projected into the central nervous system, and finally processed to provide input signals for food recognition, regulation of metabolism and physiology, and higher-order brain functions such as learning and memory, emotion, and reward. Therefore, understanding the peripheral taste system is fundamental for the development of technologies to regulate the endocrine system and improve whole-body metabolism. In this review article, we introduce previous widely-accepted views on the physiology and genetics of peripheral taste cells and primary gustatory neurons, and discuss key findings from the past decade that have raised novel questions or solved previously raised questions.
Collapse
Affiliation(s)
- Jea Hwa Jang
- BK21 Graduate Program, Department of Biomedical Sciences, Yonsei University College of Dentistry, Seoul,
Korea
- Department of Pharmacology, Korea University College of Medicine, Yonsei University College of Dentistry, Seoul,
Korea
| | - Obin Kwon
- Departments of Biochemistry and Molecular Biology, Yonsei University College of Dentistry, Seoul,
Korea
- Biomedical Sciences, Seoul National University College of Medicine, Yonsei University College of Dentistry, Seoul,
Korea
| | - Seok Jun Moon
- Department of Oral Biology, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul,
Korea
| | - Yong Taek Jeong
- BK21 Graduate Program, Department of Biomedical Sciences, Yonsei University College of Dentistry, Seoul,
Korea
- Department of Pharmacology, Korea University College of Medicine, Yonsei University College of Dentistry, Seoul,
Korea
| |
Collapse
|
9
|
Jensterle M, Rizzo M, Janez A. Glucagon-Like Peptide 1 and Taste Perception: From Molecular Mechanisms to Potential Clinical Implications. Int J Mol Sci 2021; 22:ijms22020902. [PMID: 33477478 PMCID: PMC7830704 DOI: 10.3390/ijms22020902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/03/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
Preclinical studies provided some important insights into the action of glucagon-like peptide 1 (GLP-1) in taste perception. This review examines the literature to uncover some molecular mechanisms and connections between GLP-1 and the gustatory coding. Local GLP-1 production in the taste bud cells, the expression of GLP-1 receptor on the adjacent nerves, a functional continuum in the perception of sweet chemicals from the gut to the tongue and an identification of GLP-1 induced signaling pathways in peripheral and central gustatory coding all strongly suggest that GLP-1 is involved in the taste perception, especially sweet. However, the impact of GLP-1 based therapies on gustatory coding in humans remains largely unaddressed. Based on the molecular background we encourage further exploration of the tongue as a new treatment target for GLP-1 receptor agonists in clinical studies. Given that pharmacological manipulation of gustatory coding may represent a new potential strategy against obesity and diabetes, the topic is of utmost clinical relevance.
Collapse
Affiliation(s)
- Mojca Jensterle
- Diabetes and Metabolic Diseases, Division of Internal Medicine, Department of Endocrinology, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia;
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
| | - Manfredi Rizzo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of South Carolina, Columbia, SC 29208, USA;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90133 Palermo, Italy
| | - Andrej Janez
- Diabetes and Metabolic Diseases, Division of Internal Medicine, Department of Endocrinology, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia;
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-1-522-3114; Fax: +386-1-522-9359
| |
Collapse
|
10
|
Abduljabbar T, Alhamdan RS, Al Deeb M, AlAali KA, Vohra F. Association of Salivary Content Alteration and Early Ageusia Symptoms in COVID-19 Infections: A Systematic Review. Eur J Dent 2020; 14:S152-S158. [PMID: 33242917 PMCID: PMC7775251 DOI: 10.1055/s-0040-1716986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID 19) is a major threat to the health and prosperity of human life at present. It has resulted in loss of thousands of lives globally and has brought countries to the brink of economic, social, and health collapse. A major issue of this infection is the ease with which it transmits through salivary droplets and its survival for long durations outside the body. Therefore, its early detection is critical in prevention, diagnostic, and management efforts of COVID-19 patients. Loss of taste and smell is one of the early symptoms reported in these patients and the virus is abundantly found in the salivary secretion of the infected symptomatic and asymptomatic patients. Infection and inflammation of salivary glands are common among viral infections, particularly in the early stages, which lead to salivary composition changes. Chemosensory sensation of taste is critically dependent on the salivary flow rate and its inorganic constituents, protein levels, specific 3',5'-cyclic adenosine monophosphate and 3',5'-cyclic guanosine monophosphate levels, ghrelins, pH levels, and enzymes. Therefore, the question arises, "Does COVID-19 infection alter the salivary components and composition leading to early transient symptoms of Ageusia and hypogeusia?" This review shows association of the COVID-19 and Ageusia, in addition to the early viral infection of salivary glands and possible changes in salivary flow and content. Therefore, suggesting a potential association between early ageusia in COVID-19 infection and salivary compositional changes.
Collapse
Affiliation(s)
- Tariq Abduljabbar
- Department of Prosthetic Dental Science, Research Chair for Biological Research in Dental Health, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Rana S Alhamdan
- Department of Restorative Dental Science, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Modhi Al Deeb
- Department of Prosthetic Dental Science, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Khulud A AlAali
- Department of Clinical Dental Sciences, College of Dentistry, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Fahim Vohra
- Department of Prosthetic Dental Science, Research Chair for Biological Research in Dental Health, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
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
|
12
|
Crosson SM, Marques A, Dib P, Dotson CD, Munger SD, Zolotukhin S. Taste Receptor Cells in Mice Express Receptors for the Hormone Adiponectin. Chem Senses 2020; 44:409-422. [PMID: 31125082 DOI: 10.1093/chemse/bjz030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The metabolic hormone adiponectin is secreted into the circulation by adipocytes and mediates key biological functions, including insulin sensitivity, adipocyte development, and fatty acid oxidation. Adiponectin is also abundant in saliva, where its functions are poorly understood. Here we report that murine taste receptor cells (TRCs) express specific adiponectin receptors and may be a target for salivary adiponectin. This is supported by the presence of all three known adiponectin receptors in transcriptomic data obtained by RNA-seq analysis of purified circumvallate (CV) taste buds. As well, immunohistochemical analysis of murine CV papillae showed that two adiponectin receptors, ADIPOR1 and T-cadherin, are localized to subsets of TRCs. Immunofluorescence for T-cadherin was primarily co-localized with the Type 2 TRC marker phospholipase C β2, suggesting that adiponectin signaling could impact sweet, bitter, or umami taste signaling. However, adiponectin null mice showed no differences in behavioral lick responsiveness compared with wild-type controls in brief-access lick testing. AAV-mediated overexpression of adiponectin in the salivary glands of adiponectin null mice did result in a small but significant increase in behavioral lick responsiveness to the fat emulsion Intralipid. Together, these results suggest that salivary adiponectin can affect TRC function, although its impact on taste responsiveness and peripheral taste coding remains unclear.
Collapse
Affiliation(s)
- Sean M Crosson
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida, Gainesville, FL, USA.,Graduate Program in Biomedical Sciences, University of Florida, Gainesville, FL, USA
| | - Andrew Marques
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL, USA
| | - Peter Dib
- Graduate Program in Biomedical Sciences, University of Florida, Gainesville, FL, USA.,Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, USA
| | - Cedrick D Dotson
- Center for Smell and Taste, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Steven D Munger
- Center for Smell and Taste, University of Florida, Gainesville, FL, USA.,Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism; University of Florida, Gainesville, FL, USA
| | - Sergei Zolotukhin
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida, Gainesville, FL, USA
| |
Collapse
|
13
|
Kittah E, Camilleri M, Jensen MD, Vella A. A Pilot Study Examining the Effects of GLP-1 Receptor Blockade Using Exendin-(9,39) on Gastric Emptying and Caloric Intake in Subjects With and Without Bariatric Surgery. Metab Syndr Relat Disord 2020; 18:406-412. [PMID: 32833560 DOI: 10.1089/met.2020.0049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Obesity causes significant morbidity and mortality and continues to be a significant public health concern. Unfortunately, lifestyle modification and pharmacotherapy do not produce durable results. This has led to bariatric surgical procedures playing an increasingly prominent role in the management of medically complicated obesity. Roux-en-Y gastric bypass and sleeve gastrectomy are the most commonly performed bariatric surgeries in North America and produce mechanical restriction with accelerated gastrointestinal transit accompanied by increased postprandial secretion of glucagon-like peptide-1 (GLP-1). GLP-1 is a gastrointestinal hormone that delays gastric emptying and causes satiety and weight loss. This raises the possibility that the postprandial rise in GLP-1 might affect feeding behavior over and above the mechanical restriction produced by bariatric surgery. Methods: We, therefore, sought to determine the effects of GLP-1 receptor blockade using exendin-9,39-a competitive antagonist of the actions of GLP-1 at its receptor-on caloric intake and gastrointestinal transit in subjects after sleeve gastrectomy and after Roux-en-Y gastric bypass compared with weight-matched controls. Results: GLP-1 receptor blockade did not alter caloric intake in people after bariatric surgery. However, caloric intake was decreased in age-, weight- and sex-matched control subjects, and the mechanisms require further study. Conclusions: Given the known effects of GLP-1 on gastric accommodation, future studies should ascertain effects of GLP-1 receptor blockade on gastric accommodation, which might be a useful and novel strategy to decrease caloric intake in humans with an intact upper gastrointestinal tract. The Clinical Trial Resigtration number is NCT02779075.
Collapse
Affiliation(s)
- Esi Kittah
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael D Jensen
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
14
|
Rohde K, Schamarek I, Blüher M. Consequences of Obesity on the Sense of Taste: Taste Buds as Treatment Targets? Diabetes Metab J 2020; 44:509-528. [PMID: 32431111 PMCID: PMC7453985 DOI: 10.4093/dmj.2020.0058] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/25/2020] [Indexed: 12/19/2022] Open
Abstract
Premature obesity-related mortality is caused by cardiovascular and pulmonary diseases, type 2 diabetes mellitus, physical disabilities, osteoarthritis, and certain types of cancer. Obesity is caused by a positive energy balance due to hyper-caloric nutrition, low physical activity, and energy expenditure. Overeating is partially driven by impaired homeostatic feedback of the peripheral energy status in obesity. However, food with its different qualities is a key driver for the reward driven hedonic feeding with tremendous consequences on calorie consumption. In addition to visual and olfactory cues, taste buds of the oral cavity process the earliest signals which affect the regulation of food intake, appetite and satiety. Therefore, taste buds may play a crucial role how food related signals are transmitted to the brain, particularly in priming the body for digestion during the cephalic phase. Indeed, obesity development is associated with a significant reduction in taste buds. Impaired taste bud sensitivity may play a causal role in the pathophysiology of obesity in children and adolescents. In addition, genetic variation in taste receptors has been linked to body weight regulation. This review discusses the importance of taste buds as contributing factors in the development of obesity and how obesity may affect the sense of taste, alterations in food preferences and eating behavior.
Collapse
Affiliation(s)
- Kerstin Rohde
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
| | - Imke Schamarek
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
| |
Collapse
|
15
|
Aji GK, Warren FJ, Roura E. Salivary α-Amylase Activity and Starch-Related Sweet Taste Perception in Humans. Chem Senses 2020; 44:249-256. [PMID: 30753419 DOI: 10.1093/chemse/bjz010] [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] [Indexed: 12/15/2022] Open
Abstract
Starch-related sweet taste perception plays an important role as a part of the dietary nutrient sensing mechanisms in the oral cavity. However, the release of sugars from starchy foods eliciting sweetness has been less studied in humans than in laboratory rodents. Thus, 28 respondents were recruited and evaluated for their starch-related sweet taste perception, salivary alpha-amylase (sAA) activity, oral release of reducing sugars, and salivary leptin. The results demonstrated that a 2-min oral mastication of starchy chewing gum produced an oral concentration of maltose above the sweet taste threshold and revealed that the total amount of maltose equivalent reducing sugars produced was positively correlated with the sAA activity. In addition, respondents who consistently identified the starch-related sweet taste in two sessions (test and retest) generated a higher maltose equivalent reducing sugar concentration compared to respondents who could not detect starch-related sweet taste at all (51.52 ± 2.85 and 29.96 ± 15.58 mM, respectively). In our study, salivary leptin levels were not correlated with starch-related sweet taste perception. The data contribute to the overall understanding of oral nutrient sensing and potentially to the control of food intake in humans. The results provide insight on how starchy foods without added glucose can elicit variable sweet taste perception in humans after mastication as a result of the maltose generated. The data contribute to the overall understanding of oral sensing of simple and complex carbohydrates in humans.
Collapse
Affiliation(s)
- Galih Kusuma Aji
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, St. Lucia, Australia.,Centre of Technology for Agro-Industry, The Agency for Assessment and Application of Technology, Kompleks Perkantoran Puspiptek, Tangerang Selatan, Indonesia
| | | | - Eugeni Roura
- Centre for Nutrition and Food Sciences (CNAFS), Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, St. Lucia, Australia
| |
Collapse
|
16
|
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
|
17
|
Al-Najim W, Docherty NG, le Roux CW. Food Intake and Eating Behavior After Bariatric Surgery. Physiol Rev 2018; 98:1113-1141. [PMID: 29717927 DOI: 10.1152/physrev.00021.2017] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Obesity is an escalating global chronic disease. Bariatric surgery is a very efficacious treatment for obesity and its comorbidities. Alterations to gastrointestinal anatomy during bariatric surgery result in neurological and physiological changes affecting hypothalamic signaling, gut hormones, bile acids, and gut microbiota, which coalesce to exert a profound influence on eating behavior. A thorough understanding of the mechanisms underlying eating behavior is essential in the management of patients after bariatric surgery. Studies investigating candidate mechanisms have expanded dramatically in the last decade. Herein we review the proposed mechanisms governing changes in eating behavior, food intake, and body weight after bariatric surgery. Additive or synergistic effects of both conditioned and unconditioned factors likely account for the complete picture of changes in eating behavior. Considered application of strategies designed to support the underlying principles governing changes in eating behavior holds promise as a means of optimizing responses to surgery and long-term outcomes.
Collapse
Affiliation(s)
- Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin , Dublin , Ireland ; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; and Investigative Science, Imperial College London , London , United Kingdom
| |
Collapse
|
18
|
Ma J, Vella A. What Has Bariatric Surgery Taught Us About the Role of the Upper Gastrointestinal Tract in the Regulation of Postprandial Glucose Metabolism? Front Endocrinol (Lausanne) 2018; 9:324. [PMID: 29997575 PMCID: PMC6028568 DOI: 10.3389/fendo.2018.00324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/31/2018] [Indexed: 02/06/2023] Open
Abstract
The interaction between the upper gastrointestinal tract and the endocrine system is important in the regulation of metabolism and of weight. The gastrointestinal tract has a heterogeneous cellular content and comprises a variety of cells that elaborate paracrine and endocrine mediators that collectively form the entero-endocrine system. The advent of therapy that utilizes these pathways as well as the association of bariatric surgery with diabetes remission has (re-)kindled interest in the role of the gastrointestinal tract in glucose homeostasis. In this review, we will use the changes wrought by bariatric surgery to provide insights into the various gut-pancreas interactions that maintain weight, regulate satiety, and limit glucose excursions after meal ingestion.
Collapse
Affiliation(s)
- Jing Ma
- Division of Endocrinology and Metabolism, Shanghai Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic College of Medicine, Rochester, NY, United States
| | - Adrian Vella
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic College of Medicine, Rochester, NY, United States
| |
Collapse
|
19
|
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
|
20
|
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]
|
21
|
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
|
22
|
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
|
23
|
Recent Advances in Molecular Mechanisms of Taste Signaling and Modifying. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:71-106. [PMID: 26944619 DOI: 10.1016/bs.ircmb.2015.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sense of taste conveys crucial information about the quality and nutritional value of foods before it is ingested. Taste signaling begins with taste cells via taste receptors in oral cavity. Activation of these receptors drives the transduction systems in taste receptor cells. Then particular transmitters are released from the taste cells and activate corresponding afferent gustatory nerve fibers. Recent studies have revealed that taste sensitivities are defined by distinct taste receptors and modulated by endogenous humoral factors in a specific group of taste cells. Such peripheral taste generations and modifications would directly influence intake of nutritive substances. This review will highlight current understanding of molecular mechanisms for taste reception, signal transduction in taste bud cells, transmission between taste cells and nerves, regeneration from taste stem cells, and modification by humoral factors at peripheral taste organs.
Collapse
|
24
|
|
25
|
Ciullo DL, Dotson CD. Using Animal Models to Determine the Role of Gustatory Neural Input in the Control of Ingestive Behavior and the Maintenance of Body Weight. CHEMOSENS PERCEPT 2015; 8:61-77. [PMID: 26557212 PMCID: PMC4636125 DOI: 10.1007/s12078-015-9190-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Decades of research have suggested that nutritional intake contributes to the development of human disease, mainly by influencing the development of obesity and obesity-related conditions. A relatively large body of research indicates that functional variation in human taste perception can influence nutritional intake as well as body mass accumulation. However, there are a considerable number of studies that suggest that no link between these variables actually exists. These discrepancies in the literature likely result from the confounding influence of a variety of other, uncontrolled, factors that can influence ingestive behavior. STRATEGY In this review, the use of controlled animal experimentation to alleviate at least some of these issues related to the lack of control of experimental variables is discussed. Specific examples of the use of some of these techniques are examined. DISCUSSION AND CONCLUSIONS The review will close with some specific suggestions aimed at strengthening the link between gustatory neural input and its putative influence on ingestive behaviors and the maintenance of body weight.
Collapse
Affiliation(s)
- Dana L Ciullo
- Departments of Neuroscience and Psychiatry, Division of Addiction Medicine, University of Florida College of Medicine, and Center for Smell and Taste, University of Florida, Gainesville, FL 32611, USA,
| | - Cedrick D Dotson
- Departments of Neuroscience and Psychiatry, Division of Addiction Medicine, University of Florida College of Medicine, and Center for Smell and Taste, University of Florida, Gainesville, FL 32611, USA,
| |
Collapse
|
26
|
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
|
27
|
Glendinning JI, Elson AET, Kalik S, Sosa Y, Patterson CM, Myers MG, Munger SD. Taste responsiveness to sweeteners is resistant to elevations in plasma leptin. Chem Senses 2015; 40:223-31. [PMID: 25740302 DOI: 10.1093/chemse/bju075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
There is uncertainty about the relationship between plasma leptin and sweet taste in mice. Whereas 2 studies have reported that elevations in plasma leptin diminish responsiveness to sweeteners, another found that they enhanced responsiveness to sucrose. We evaluated the impact of plasma leptin on sweet taste in C57BL/6J (B6) and leptin-deficient ob/ob mice. Although mice expressed the long-form leptin receptor (LepRb) selectively in Type 2 taste cells, leptin failed to activate a critical leptin-signaling protein, STAT3, in taste cells. Similarly, we did not observe any impact of intraperitoneal (i.p.) leptin treatment on chorda tympani nerve responses to sweeteners in B6 or ob/ob mice. Finally, there was no effect of leptin treatment on initial licking responses to several sucrose concentrations in B6 mice. We confirmed that basal plasma leptin levels did not exceed 10ng/mL, regardless of time of day, physiological state, or body weight, suggesting that taste cell LepRb were not desensitized to leptin in our studies. Furthermore, i.p. leptin injections produced plasma leptin levels that exceeded those previously reported to exert taste effects. We conclude that any effect of plasma leptin on taste responsiveness to sweeteners is subtle and manifests itself only under specific experimental conditions.
Collapse
Affiliation(s)
- John I Glendinning
- Department of Biology, Barnard College, Columbia University, 3009 Broadway New York, NY 10027, USA,
| | - Amanda E T Elson
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 S. Penn St., Baltimore, MD 21201, USA
| | - Salina Kalik
- Department of Biology, Barnard College, Columbia University, 3009 Broadway New York, NY 10027, USA
| | - Yvett Sosa
- Department of Biology, Barnard College, Columbia University, 3009 Broadway New York, NY 10027, USA
| | - Christa M Patterson
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA and
| | - Martin G Myers
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA and
| | - Steven D Munger
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 S. Penn St., Baltimore, MD 21201, USA, Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, HH-495, Baltimore, MD 21201, USA
| |
Collapse
|
28
|
Abstract
Glucagon action is transduced by a G protein-coupled receptor located in liver, kidney, intestinal smooth muscle, brain, adipose tissue, heart, pancreatic β-cells, and placenta. Genetically modified animal models have provided important clues about the role of glucagon and its receptor (Gcgr) beyond glucose control. The PubMed database was searched for articles published between 1995 and 2014 using the key terms glucagon, glucagon receptor, signaling, and animal models. Lack of Gcgr signaling has been associated with: i) hypoglycemic pregnancies, altered placentation, poor fetal growth, and increased fetal-neonatal death; ii) pancreatic glucagon cell hyperplasia and hyperglucagonemia; iii) altered body composition, energy state, and protection from diet-induced obesity; iv) impaired hepatocyte survival; v) altered glucose, lipid, and hormonal milieu; vi) altered metabolic response to prolonged fasting and exercise; vii) reduced gastric emptying and increased intestinal length; viii) altered retinal function; and ix) prevention of the development of diabetes in insulin-deficient mice. Similar phenotypic findings were observed in the hepatocyte-specific deletion of Gcgr. Glucagon action has been involved in the modulation of sweet taste responsiveness, inotropic and chronotropic effects in the heart, satiety, glomerular filtration rate, secretion of insulin, cortisol, ghrelin, GH, glucagon, and somatostatin, and hypothalamic signaling to suppress hepatic glucose production. Glucagon (α) cells under certain conditions can transdifferentiate into insulin (β) cells. These findings suggest that glucagon signaling plays an important role in multiple organs. Thus, treatment options designed to block Gcgr activation in diabetics may have implications beyond glucose homeostasis.
Collapse
Affiliation(s)
- Maureen J Charron
- Departments of BiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA Departments of BiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA Departments of BiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Patricia M Vuguin
- Departments of BiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| |
Collapse
|
29
|
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: 60] [Impact Index Per Article: 6.0] [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
|
30
|
Yang H, Cong WN, Yoon JS, Egan JM. Vismodegib, an antagonist of hedgehog signaling, directly alters taste molecular signaling in taste buds. Cancer Med 2014; 4:245-52. [PMID: 25354792 PMCID: PMC4329008 DOI: 10.1002/cam4.350] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/25/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
Vismodegib, a highly selective inhibitor of hedgehog (Hh) pathway, is an approved treatment for basal-cell carcinoma. Patients on treatment with vismodegib often report profound alterations in taste sensation. The cellular mechanisms underlying the alterations have not been studied. Sonic Hh (Shh) signaling is required for cell growth and differentiation. In taste buds, Shh is exclusively expressed in type IV taste cells, which are undifferentiated basal cells and the precursors of the three types of taste sensing cells. Thus, we investigated if vismodegib has an inhibitory effect on taste cell turnover because of its known effects on Hh signaling. We gavaged C57BL/6J male mice daily with either vehicle or 30 mg/kg vismodegib for 15 weeks. The gustatory behavior and immunohistochemical profile of taste cells were examined. Vismodegib-treated mice showed decreased growth rate and behavioral responsivity to sweet and bitter stimuli, compared to vehicle-treated mice. We found that vismodegib-treated mice had significant reductions in taste bud size and numbers of taste cells per taste bud. Additionally, vismodegib treatment resulted in decreased numbers of Ki67- and Shh-expressing cells in taste buds. The numbers of phospholipase Cβ2- and α-gustducin-expressing cells, which contain biochemical machinery for sweet and bitter sensing, were reduced in vismodegib-treated mice. Furthermore, vismodegib treatment resulted in reduction in numbers of T1R3, glucagon-like peptide-1, and glucagon-expressing cells, which are known to modulate sweet taste sensitivity. These results suggest that inhibition of Shh signaling by vismodegib treatment directly results in alteration of taste due to local effects in taste buds.
Collapse
Affiliation(s)
- Hyekyung Yang
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | | | | | | |
Collapse
|
31
|
Independent, reciprocal neuromodulatory control of sweet and bitter taste sensitivity during starvation in Drosophila. Neuron 2014; 84:806-20. [PMID: 25451195 DOI: 10.1016/j.neuron.2014.09.032] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2014] [Indexed: 11/22/2022]
Abstract
An organism's behavioral decisions often depend upon the relative strength of appetitive and aversive sensory stimuli, the relative sensitivity to which can be modified by internal states like hunger. However, whether sensitivity to such opposing influences is modulated in a unidirectional or bidirectional manner is not clear. Starved flies exhibit increased sugar and decreased bitter sensitivity. It is widely believed that only sugar sensitivity changes, and that this masks bitter sensitivity. Here we use gene- and circuit-level manipulations to show that sweet and bitter sensitivity are independently and reciprocally regulated by starvation in Drosophila. We identify orthogonal neuromodulatory cascades that oppositely control peripheral taste sensitivity for each modality. Moreover, these pathways are recruited at increasing hunger levels, such that low-risk changes (higher sugar sensitivity) precede high-risk changes (lower sensitivity to potentially toxic resources). In this way, state-intensity-dependent, reciprocal regulation of appetitive and aversive peripheral gustatory sensitivity permits flexible, adaptive feeding decisions.
Collapse
|
32
|
|
33
|
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
|
34
|
Wang R, van Keeken NMA, Siddiqui S, Dijksman LM, Maudsley S, Derval D, van Dam PS, Martin B. Higher TNF-α, IGF-1, and Leptin Levels are Found in Tasters than Non-Tasters. Front Endocrinol (Lausanne) 2014; 5:125. [PMID: 25120532 PMCID: PMC4114300 DOI: 10.3389/fendo.2014.00125] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/11/2014] [Indexed: 12/03/2022] Open
Abstract
Taste perception is controlled by taste cells that are present in the tongue that produce and secrete various metabolic hormones. Recent studies have demonstrated that taste receptors in tongue, gut, and pancreas are associated with local hormone secretion. The aim of this study was to determine whether there is a link between taste sensitivity and levels of circulating metabolic hormones in humans and whether taste sensitivity is potentially related to peripheral metabolic regulation. Thirty-one subjects were recruited and separated into tasters and non-tasters based on their phenol thiocarbamide (PTC) bitter taste test results. Fasting plasma and saliva were collected and levels of hormones and cytokines were assayed. We observed significant differences in both hormone levels and hormone-body mass index (BMI) correlation between tasters and non-tasters. Tasters had higher plasma levels of leptin (p = 0.05), tumor necrosis factor-α (TNF-α) (p = 0.04), and insulin-like growth factor 1 (IGF-1) (p = 0.03). There was also a trend toward increased IGF-1 levels in the saliva of tasters (p = 0.06). We found a positive correlation between plasma levels of glucose and BMI (R = 0.4999, p = 0.04) exclusively in non-tasters. In contrast, plasma C-peptide levels were found to be positively correlated to BMI (R = 0.5563, p = 0.03) in tasters. Saliva TNF-α levels were negatively correlated with BMI in tasters (R = -0.5908, p = 0.03). Our findings demonstrate that there are differences in circulating levels of leptin, TNF-α, and IGF-1 between tasters and non-tasters. These findings indicate that in addition to the regulation of food consumption, taste perception also appears to be tightly linked to circulating metabolic hormone levels. People with different taste sensitivity may respond differently to the nutrient stimulation. Further work investigating the link between taste perception and peripheral metabolic control could potentially lead to the development of novel therapies for obesity or Type 2 diabetes.
Collapse
Affiliation(s)
- Rui Wang
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - Sana Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Lea M. Dijksman
- Teaching Hospital, Onze Lieve Vrouwe Gasthuis, Amsterdam, Netherlands
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- VIB Department of Molecular Genetics, University of Antwerp, Antwerp, Belgium
| | - Diana Derval
- Better Immune System Foundation, Amsterdam, Netherlands
| | - P. Sytze van Dam
- Department of Internal Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, Netherlands
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- *Correspondence: Bronwen Martin, Metabolism Unit, National Institute on Aging, 251 Bayview Blvd., Suite 100 Room 08C009, Baltimore, MD 21224, USA e-mail:
| |
Collapse
|
35
|
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: 18] [Impact Index Per Article: 1.5] [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
|
36
|
La Sala MS, Hurtado MD, Brown AR, Bohórquez DV, Liddle RA, Herzog H, Zolotukhin S, Dotson CD. Modulation of taste responsiveness by the satiation hormone peptide YY. FASEB J 2013; 27:5022-33. [PMID: 24043261 DOI: 10.1096/fj.13-228064] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It has been hypothesized that the peripheral taste system may be modulated in the context of an animal's metabolic state. One purported mechanism for this phenomenon is that circulating gastrointestinal peptides modulate the functioning of the peripheral gustatory system. Recent evidence suggests endocrine signaling in the oral cavity can influence food intake (FI) and satiety. We hypothesized that these hormones may be affecting FI by influencing taste perception. We used immunohistochemistry along with genetic knockout models and the specific reconstitution of peptide YY (PYY) in saliva using gene therapy protocols to identify a role for PYY signaling in taste. We show that PYY is expressed in subsets of taste cells in murine taste buds. We also show, using brief-access testing with PYY knockouts, that PYY signaling modulates responsiveness to bitter-tasting stimuli, as well as to lipid emulsions. We show that salivary PYY augmentation, via viral vector therapy, rescues behavioral responsiveness to a lipid emulsion but not to bitter stimuli and that this response is likely mediated via activation of Y2 receptors localized apically in taste cells. Our findings suggest distinct functions for PYY produced locally in taste cells vs. that circulating systemically.
Collapse
Affiliation(s)
- Michael S La Sala
- 1Department of Neuroscience, University of Florida McKnight Brain Institute, 1149 Newell Dr., Box 100244, Gainesville, FL 32611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Sugita M, Yamamoto K, Hirono C, Shiba Y. Functional dissection of sweet and bitter taste pathways. J Oral Biosci 2013. [DOI: 10.1016/j.job.2013.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
39
|
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: 44] [Impact Index Per Article: 3.7] [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
|
40
|
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
|
41
|
Knaapila A, Hwang LD, Lysenko A, Duke FF, Fesi B, Khoshnevisan A, James RS, Wysocki CJ, Rhyu M, Tordoff MG, Bachmanov AA, Mura E, Nagai H, Reed DR. Genetic analysis of chemosensory traits in human twins. Chem Senses 2012; 37:869-81. [PMID: 22977065 DOI: 10.1093/chemse/bjs070] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We explored genetic influences on the perception of taste and smell stimuli. Adult twins rated the chemosensory aspects of water, sucrose, sodium chloride, citric acid, ethanol, quinine hydrochloride, phenylthiocarbamide (PTC), potassium chloride, calcium chloride, cinnamon, androstenone, Galaxolide™, cilantro, and basil. For most traits, individual differences were stable over time and some traits were heritable (h(2) from 0.41 to 0.71). Subjects were genotyped for 44 single nucleotide polymorphisms within and near genes related to taste and smell. The results of these association analyses confirmed previous genotype-phenotype results for PTC, quinine, and androstenone. New associations were detected for ratings of basil and a bitter taste receptor gene, TAS2R60, and between cilantro and variants in three genes (TRPA1, GNAT3, and TAS2R50). The flavor of ethanol was related to variation within an olfactory receptor gene (OR7D4) and a gene encoding a subunit of the epithelial sodium channel (SCNN1D). Our study demonstrates that person-to-person differences in the taste and smell perception of simple foods and drinks are partially accounted for by genetic variation within chemosensory pathways.
Collapse
Affiliation(s)
- Antti Knaapila
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Geraedts MCP, Takahashi T, Vigues S, Markwardt ML, Nkobena A, Cockerham RE, Hajnal A, Dotson CD, Rizzo MA, Munger SD. Transformation of postingestive glucose responses after deletion of sweet taste receptor subunits or gastric bypass surgery. Am J Physiol Endocrinol Metab 2012; 303:E464-74. [PMID: 22669246 PMCID: PMC3423100 DOI: 10.1152/ajpendo.00163.2012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/05/2012] [Indexed: 01/06/2023]
Abstract
The glucose-dependent secretion of the insulinotropic hormone glucagon-like peptide-1 (GLP-1) is a critical step in the regulation of glucose homeostasis. Two molecular mechanisms have separately been suggested as the primary mediator of intestinal glucose-stimulated GLP-1 secretion (GSGS): one is a metabotropic mechanism requiring the sweet taste receptor type 2 (T1R2) + type 3 (T1R3) while the second is a metabolic mechanism requiring ATP-sensitive K(+) (K(ATP)) channels. By quantifying sugar-stimulated hormone secretion in receptor knockout mice and in rats receiving Roux-en-Y gastric bypass (RYGB), we found that both of these mechanisms contribute to GSGS; however, the mechanisms exhibit different selectivity, regulation, and localization. T1R3(-/-) mice showed impaired glucose and insulin homeostasis during an oral glucose challenge as well as slowed insulin granule exocytosis from isolated pancreatic islets. Glucose, fructose, and sucralose evoked GLP-1 secretion from T1R3(+/+), but not T1R3(-/-), ileum explants; this secretion was not mimicked by the K(ATP) channel blocker glibenclamide. T1R2(-/-) mice showed normal glycemic control and partial small intestine GSGS, suggesting that T1R3 can mediate GSGS without T1R2. Robust GSGS that was K(ATP) channel-dependent and glucose-specific emerged in the large intestine of T1R3(-/-) mice and RYGB rats in association with elevated fecal carbohydrate throughout the distal gut. Our results demonstrate that the small and large intestines utilize distinct mechanisms for GSGS and suggest novel large intestine targets that could mimic the improved glycemic control seen after RYGB.
Collapse
Affiliation(s)
- Maartje C P Geraedts
- Division of Endocrinology, Diabetes, and Nutrition, Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Fernstrom JD, Munger SD, Sclafani A, de Araujo IE, Roberts A, Molinary S. Mechanisms for sweetness. J Nutr 2012; 142:1134S-41S. [PMID: 22573784 PMCID: PMC3738222 DOI: 10.3945/jn.111.149567] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A remarkable amount of information has emerged in the past decade regarding sweet taste physiology. This article reviews these data, with a particular focus on the elucidation of the sweet taste receptor, its location and actions in taste transduction in the mouth, its nontaste functions in the gastrointestinal tract (e.g., in enteroendocrine cells), and the brain circuitry involved in the sensory processing of sweet taste. Complications in the use of rodents to model human sweet taste perception and responses are also considered. In addition, information relating to low-calorie sweeteners (LCS) is discussed in the context of these issues. Particular consideration is given to the known effects of LCS on enteroendocrine cell function.
Collapse
Affiliation(s)
- John D. Fernstrom
- University of Pittsburgh School of Medicine, Pittsburgh, PA,To whom correspondence should be addressed. E-mail:
| | | | | | - Ivan E. de Araujo
- John B. Pierce Laboratory and Yale University School of Medicine, New Haven, CT
| | | | | |
Collapse
|
44
|
|
45
|
|
46
|
Shin YK, Cong WN, Cai H, Kim W, Maudsley S, Egan JM, Martin B. Age-related changes in mouse taste bud morphology, hormone expression, and taste responsivity. J Gerontol A Biol Sci Med Sci 2011; 67:336-44. [PMID: 22056740 DOI: 10.1093/gerona/glr192] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Normal aging is a complex process that affects every organ system in the body, including the taste system. Thus, we investigated the effects of the normal aging process on taste bud morphology, function, and taste responsivity in male mice at 2, 10, and 18 months of age. The 18-month-old animals demonstrated a significant reduction in taste bud size and number of taste cells per bud compared with the 2- and 10-month-old animals. The 18-month-old animals exhibited a significant reduction of protein gene product 9.5 and sonic hedgehog immunoreactivity (taste cell markers). The number of taste cells expressing the sweet taste receptor subunit, T1R3, and the sweet taste modulating hormone, glucagon-like peptide-1, were reduced in the 18-month-old mice. Concordant with taste cell alterations, the 18-month-old animals demonstrated reduced sweet taste responsivity compared with the younger animals and the other major taste modalities (salty, sour, and bitter) remained intact.
Collapse
Affiliation(s)
- Yu-Kyong Shin
- Laboratory of Clinical Investigation, Diabetes Section, National Institute on Aging, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Fadool DA, Tucker K, Pedarzani P. Mitral cells of the olfactory bulb perform metabolic sensing and are disrupted by obesity at the level of the Kv1.3 ion channel. PLoS One 2011; 6:e24921. [PMID: 21966386 PMCID: PMC3178571 DOI: 10.1371/journal.pone.0024921] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 08/19/2011] [Indexed: 12/12/2022] Open
Abstract
Sixty-five percent of Americans are over-weight. While the neuroendocrine controls of energy homeostasis are well known, how sensory systems respond to and are impacted by obesity is scantily understood. The main accepted function of the olfactory system is to provide an internal depiction of our external chemical environment, starting from the detection of chemosensory cues. We hypothesized that the system additionally functions to encode internal chemistry via the detection of chemicals that are important indicators of metabolic state. We here uncovered that the olfactory bulb (OB) subserves as an internal sensor of metabolism via insulin-induced modulation of the potassium channel Kv1.3. Using an adult slice preparation of the olfactory bulb, we found that evoked neural activity in Kv1.3-expressing mitral cells is enhanced following acute insulin application. Insulin mediated changes in mitral cell excitability are predominantly due to the modulation of Kv1.3 channels as evidenced by the lack of effect in slices from Kv1.3-null mice. Moreover, a selective Kv1.3 peptide blocker (ShK186) inhibits more than 80% of the outward current in parallel voltage-clamp studies, whereby insulin significantly decreases the peak current magnitude without altering the kinetics of inactivation or deactivation. Mice that were chronically administered insulin using intranasal delivery approaches exhibited either an elevation in basal firing frequency or fired a single cluster of action potentials. Following chronic administration of the hormone, mitral cells were inhibited by application of acute insulin rather than excited. Mice made obese through a diet of ∼32% fat exhibited prominent changes in mitral cell action potential shape and clustering behavior, whereby the subsequent response to acute insulin stimulation was either attenuated or completely absent. Our results implicate an inappropriate neural function of olfactory sensors following exposure to chronic levels of the hormone insulin (diabetes) or increased body weight (obesity).
Collapse
Affiliation(s)
- Debra Ann Fadool
- Program in Neuroscience, The Florida State University, Tallahassee, Florida, United States of America
- Department of Biological Sciences, The Florida State University, Tallahassee, Florida, United States of America
- Institute of Molecular Biophysics, The Florida State University, Tallahassee, Florida, United States of America
- * E-mail:
| | - Kristal Tucker
- Program in Neuroscience, The Florida State University, Tallahassee, Florida, United States of America
| | - Paola Pedarzani
- Research Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
49
|
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
|
50
|
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
|