1
|
Hao S, Guthrie B, Kim SK, Balanda S, Kubicek J, Murtaza B, Khan NA, Khakbaz P, Su J, Goddard WA. Steviol rebaudiosides bind to four different sites of the human sweet taste receptor (T1R2/T1R3) complex explaining confusing experiments. Commun Chem 2024; 7:236. [PMID: 39424933 PMCID: PMC11489721 DOI: 10.1038/s42004-024-01324-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Sucrose provides both sweetness and energy by binding to both Venus flytrap domains (VFD) of the heterodimeric sweet taste receptor (T1R2/T1R3). In contrast, non-caloric sweeteners such as sucralose and aspartame only bind to one specific domain (VFD2) of T1R2, resulting in high-intensity sweetness. In this study, we investigate the binding mechanism of various steviol glycosides, artificial sweeteners, and a negative allosteric modulator (lactisole) at four distinct binding sites: VFD2, VFD3, transmembrane domain 2 (TMD2), and TMD3 through binding experiments and computational docking studies. Our docking results reveal multiple binding sites for the tested ligands, including the radiolabeled ligands. Our experimental evidence demonstrates that the C20 carboxy terminus of the Gα protein can bind to the intracellular region of either TMD2 or TMD3, altering GPCR affinity to the high-affinity state for steviol glycosides. These findings provide a mechanistic understanding of the structure and function of this heterodimeric sweet taste receptor.
Collapse
Affiliation(s)
- Shuang Hao
- Wyant College of Optical Sciences and Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Brian Guthrie
- Global Core Research and Development Group, Cargill, Inc. 14800 28th Avenue N, Plymouth, MN, 55447, USA
| | - Soo-Kyung Kim
- Materials and Process Simulation Center (MSC), California Institute of Technology, Pasadena, CA, 91125, USA
| | - Sergej Balanda
- Cube Biotech, Creative Campus Monheim, Creative-Campus-Allee 12, 40789, Monheim, Germany
| | - Jan Kubicek
- Cube Biotech, Creative Campus Monheim, Creative-Campus-Allee 12, 40789, Monheim, Germany
| | - Babar Murtaza
- Physiologie de Nutrition & Toxicologie, UB 1231 Center for Translational & Molecular Medicine (CTM), Université de Bourgogne, 21000 Dijon, France
| | - Naim A Khan
- Physiologie de Nutrition & Toxicologie, UB 1231 Center for Translational & Molecular Medicine (CTM), Université de Bourgogne, 21000 Dijon, France
| | - Pouyan Khakbaz
- Global Core Research and Development Group, Cargill, Inc. 14800 28th Avenue N, Plymouth, MN, 55447, USA
| | - Judith Su
- Wyant College of Optical Sciences and Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
| | - William A Goddard
- Materials and Process Simulation Center (MSC), California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
2
|
Yoshida R, Ninomiya Y. Mechanisms and Functions of Sweet Reception in Oral and Extraoral Organs. Int J Mol Sci 2024; 25:7398. [PMID: 39000505 PMCID: PMC11242429 DOI: 10.3390/ijms25137398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The oral detection of sugars relies on two types of receptor systems. The first is the G-protein-coupled receptor TAS1R2/TAS1R3. When activated, this receptor triggers a downstream signaling cascade involving gustducin, phospholipase Cβ2 (PLCβ2), and transient receptor potential channel M5 (TRPM5). The second type of receptor is the glucose transporter. When glucose enters the cell via this transporter, it is metabolized to produce ATP. This ATP inhibits the opening of KATP channels, leading to cell depolarization. Beside these receptor systems, sweet-sensitive taste cells have mechanisms to regulate their sensitivity to sweet substances based on internal and external states of the body. Sweet taste receptors are not limited to the oral cavity; they are also present in extraoral organs such as the gastrointestinal tract, pancreas, and brain. These extraoral sweet receptors are involved in various functions, including glucose absorption, insulin release, sugar preference, and food intake, contributing to the maintenance of energy homeostasis. Additionally, sweet receptors may have unique roles in certain organs like the trachea and bone. This review summarizes past and recent studies on sweet receptor systems, exploring the molecular mechanisms and physiological functions of sweet (sugar) detection in both oral and extraoral organs.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Yuzo Ninomiya
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Li YH, Yang YS, Xue YB, Lei H, Zhang SS, Qian J, Yao Y, Zhou R, Huang L. G protein subunit G γ13-mediated signaling pathway is critical to the inflammation resolution and functional recovery of severely injured lungs. eLife 2024; 12:RP92956. [PMID: 38836551 DOI: 10.7554/elife.92956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.
Collapse
Affiliation(s)
- Yi-Hong Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Sen Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hao Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sai-Sai Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
- Monell Chemical Senses Center, Philadelphia, United States
| |
Collapse
|
4
|
Seong H, Song JW, Lee KH, Jang G, Shin DM, Shon WJ. Taste receptor type 1 member 3 regulates Western diet-induced male infertility. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159433. [PMID: 38007088 DOI: 10.1016/j.bbalip.2023.159433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Western diet (WD), characterized by a high intake of fats and sugary drinks, is a risk factor for male reproductive impairment. However, the molecular mechanisms underlying this remain unclear. Taste receptor type 1 member 3 (TAS1R3), activated by ligands of WD, is highly expressed in extra-oral tissues, particularly in the testes. Here, we investigated to determine the effects of WD intake on male reproduction and whether TAS1R3 mediates WD-induced impairment in male reproduction. Male C57BL/6 J wild-type (WT) and Tas1r3 knockout (KO) mice were fed either a normal diet and plain water (ND) or a 60 % high-fat-diet and 30 % (w/v) sucrose water (WD) for 18 weeks (n = 7-9/group). Long-term WD consumption significantly impaired sperm count, motility and testicular morphology in WT mice with marked Tas1r3 overexpression, whereas Tas1r3 KO mice were protected from WD-induced reproductive impairment. Testicular transcriptome analysis revealed downregulated AMP-activated protein kinase (AMPK) signaling and significantly elevated AMPK-targeted nuclear receptor 4A1 (Nr4a1) expression in WD-fed Tas1r3 KO mice. In vitro studies further validated that Tas1r3 knockdown in Leydig cells prevented the suppression of Nr4a1 and downstream steroidogenic genes (Star, Cyp11a1, Cyp17a1, and Hsd3b1) caused by high glucose, fructose, and palmitic acid levels, and maintained the levels of testosterone. Additionally, we analyzed the public human dataset to assess the clinical implications of our findings and confirmed a significant association between TAS1R3 and male-infertility-related diseases. Our findings suggest that TAS1R3 regulates WD-induced male reproductive impairment via the AMPK/NR4A1 signaling and can be a novel therapeutic target for male infertility.
Collapse
Affiliation(s)
- Hobin Seong
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Won Song
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea
| | - Keon-Hee Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea
| | - Goo Jang
- Department of Veterinary Clinical Science, College of Veterinary Medicine and the Research Institute of Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; Comparative Medicine Disease Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea.
| | - Woo-Jeong Shon
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
5
|
An JP, Wang Y, Munger SD, Tang X. A review on natural sweeteners, sweet taste modulators and bitter masking compounds: structure-activity strategies for the discovery of novel taste molecules. Crit Rev Food Sci Nutr 2024:1-24. [PMID: 38494695 DOI: 10.1080/10408398.2024.2326012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Growing demand for the tasty and healthy food has driven the development of low-calorie sweeteners, sweet taste modulators, and bitter masking compounds originated from natural sources. With the discovery of human taste receptors, increasing numbers of sweet taste modulators have been identified through human taste response and molecular docking techniques. However, the discovery of novel taste-active molecules in nature can be accelerated by using advanced spectrometry technologies based on structure-activity relationships (SARs). SARs explain why structurally similar compounds can elicit similar taste qualities. Given the characterization of structural information from reported data, strategies employing SAR techniques to find structurally similar compounds become an innovative approach to expand knowledge of sweeteners. This review aims to summarize the structural patterns of known natural non-nutritive sweeteners, sweet taste enhancers, and bitter masking compounds. Innovative SAR-based approaches to explore sweetener derivatives are also discussed. Most sweet-tasting flavonoids belong to either the flavanonols or the dihydrochalcones and known bitter masking molecules are flavanones. Based on SAR findings that structural similarities are related to the sensory properties, innovative methodologies described in this paper can be applied to screen and discover the derivatives of taste-active compounds or potential taste modulators.
Collapse
Affiliation(s)
- Jin-Pyo An
- Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Yu Wang
- Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Steven D Munger
- Center for Smell and Taste, Department of Pharmacology and Therapeutics, Department of Otolaryngology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Xixuan Tang
- Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| |
Collapse
|
6
|
Tu K, Zhou M, Tan JJ, Markos L, Cloud C, Zhou M, Hayashi N, Rawson NE, Margolskee RF, Wang H. Chronic social defeat stress broadly inhibits gene expression in the peripheral taste system and alters taste responses in mice. Physiol Behav 2024; 275:114446. [PMID: 38128683 PMCID: PMC10843841 DOI: 10.1016/j.physbeh.2023.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Human studies have linked stress exposure to unhealthy eating behavior. However, the mechanisms that drive stress-associated changes in eating behavior remain incompletely understood. The sense of taste plays important roles in food preference and intake. In this study, we use a chronic social defeat stress (CSDS) model in mice to address whether chronic stress impacts taste sensation and gene expression in taste buds and the gut. Our results showed that CSDS significantly elevated circulating levels of corticosterone and acylated ghrelin while lowering levels of leptin, suggesting a change in metabolic hormones that promotes food consumption. Stressed mice substantially increased their intake of food and water 3-5 days after the stress onset and gradually gained more body weight than that of controls. Moreover, CSDS significantly decreased the expression of multiple taste receptors and signaling molecules in taste buds and reduced mRNA levels of several taste progenitor/stem cell markers and regulators. Stressed mice showed significantly reduced sensitivity and response to umami and sweet taste compounds in behavioral tests. In the small intestine, the mRNA levels of Gnat3 and Tas1r2 were elevated in CSDS mice. The increased Gnat3 was mostly localized in a type of Gnat3+ and CD45+ immune cells, suggesting changes of immune cell distribution in the gut of stressed mice. Together, our study revealed broad effects of CSDS on the peripheral taste system and the gut, which may contribute to stress-associated changes in eating behavior.
Collapse
Affiliation(s)
- Katelyn Tu
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA; Haverford College, 370 Lancaster Ave., Haverford, PA 19041, USA
| | - Mary Zhou
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA; Haverford College, 370 Lancaster Ave., Haverford, PA 19041, USA
| | - Jidong J Tan
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA; Department of Chemistry, the University of Pennsylvania, 231 S. 34 St., Philadelphia, PA 19104, USA
| | - Loza Markos
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA
| | - Cameron Cloud
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA; Lafayette College, 730 High St., Easton, PA 18042, USA
| | - Minliang Zhou
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA
| | - Naoki Hayashi
- Department of Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Nancy E Rawson
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA
| | - Robert F Margolskee
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA
| | - Hong Wang
- The Monell Chemical Senses Center, 3500 Market St., Philadelphia, PA 19104, USA.
| |
Collapse
|
7
|
Ma D, Hu M, Yang X, Liu Q, Ye F, Cai W, Wang Y, Xu X, Chang S, Wang R, Yang W, Ye S, Su N, Fan M, Xu H, Guo J. Structural basis for sugar perception by Drosophila gustatory receptors. Science 2024; 383:eadj2609. [PMID: 38305684 DOI: 10.1126/science.adj2609] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
Insects rely on a family of seven transmembrane proteins called gustatory receptors (GRs) to encode different taste modalities, such as sweet and bitter. We report structures of Drosophila sweet taste receptors GR43a and GR64a in the apo and sugar-bound states. Both GRs form tetrameric sugar-gated cation channels composed of one central pore domain (PD) and four peripheral ligand-binding domains (LBDs). Whereas GR43a is specifically activated by the monosaccharide fructose that binds to a narrow pocket in LBDs, disaccharides sucrose and maltose selectively activate GR64a by binding to a larger and flatter pocket in LBDs. Sugar binding to LBDs induces local conformational changes, which are subsequently transferred to the PD to cause channel opening. Our studies reveal a structural basis for sugar recognition and activation of GRs.
Collapse
Affiliation(s)
- Demin Ma
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Meiqin Hu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310058, China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou 311121, China
| | - Xiaotong Yang
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310058, China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou 311121, China
| | - Qiang Liu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310058, China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou 311121, China
| | - Fan Ye
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Weijie Cai
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310058, China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou 311121, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ximing Xu
- Marine Biomedical Institute of Qingdao, School of Pharmacy and Medicine, Ocean University of China, Qingdao, Shandong 266100, China
| | - Shenghai Chang
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ruiying Wang
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Sheng Ye
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Nannan Su
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Minrui Fan
- CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Haoxing Xu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310058, China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou 311121, China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiangtao Guo
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
8
|
Lu P, Simas TAM, Delpapa E, ZhuGe R. Bitter taste receptors in the reproductive system: Function and therapeutic implications. J Cell Physiol 2024; 239:e31179. [PMID: 38219077 PMCID: PMC10922893 DOI: 10.1002/jcp.31179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 01/15/2024]
Abstract
Type 2 taste receptors (TAS2Rs), traditionally known for their role in bitter taste perception, are present in diverse reproductive tissues of both sexes. This review explores our current understanding of TAS2R functions with a particular focus on reproductive health. In males, TAS2Rs are believed to play potential roles in processes such as sperm chemotaxis and male fertility. Genetic insights from mouse models and human polymorphism studies provide some evidence for their contribution to male infertility. In female reproduction, it is speculated that TAS2Rs influence the ovarian milieu, shaping the functions of granulosa and cumulus cells and their interactions with oocytes. In the uterus, TAS2Rs contribute to uterine relaxation and hold potential as therapeutic targets for preventing preterm birth. In the placenta, they are proposed to function as vigilant sentinels, responding to infection and potentially modulating mechanisms of fetal protection. In the cervix and vagina, their analogous functions to those in other extraoral tissues suggest a potential role in infection defense. In addition, TAS2Rs exhibit altered expression patterns that profoundly affect cancer cell proliferation and apoptosis in reproductive cancers. Notably, TAS2R agonists show promise in inducing apoptosis and overcoming chemoresistance in these malignancies. Despite these advances, challenges remain, including a lack of genetic and functional studies. The application of techniques such as single-cell RNA sequencing and clustered regularly interspaced palindromic repeats (CRISPR)/CRISPR-associated endonuclease 9 gene editing could provide deeper insights into TAS2Rs in reproduction, paving the way for novel therapeutic strategies for reproductive disorders.
Collapse
Affiliation(s)
- Ping Lu
- Department of Microbiology and Physiological System, UMass Chan Medical School, 363 Plantation St., Worcester, MA, USA
| | - Tiffany A. Moore Simas
- Department of Obstetrics and Gynecology, UMass Chan Medical School/UMass Memorial Health, Memorial Campus 119 Belmont St., Worcester, MA, USA
| | - Ellen Delpapa
- Department of Obstetrics and Gynecology, UMass Chan Medical School/UMass Memorial Health, Memorial Campus 119 Belmont St., Worcester, MA, USA
| | - Ronghua ZhuGe
- Department of Microbiology and Physiological System, UMass Chan Medical School, 363 Plantation St., Worcester, MA, USA
| |
Collapse
|
9
|
Lemon CH. A non-singularity in sweet taste. Chem Senses 2024; 49:bjae006. [PMID: 38366583 DOI: 10.1093/chemse/bjae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 02/18/2024] Open
Affiliation(s)
- Christian H Lemon
- School of Biological Sciences, University of Oklahoma, Norman, OK 73019, United States
| |
Collapse
|
10
|
Yoshida A, Ito A, Yasui N, Yamashita A. Direct binding of calmodulin to the cytosolic C-terminal regions of sweet/umami taste receptors. J Biochem 2023; 174:451-459. [PMID: 37527916 PMCID: PMC11033526 DOI: 10.1093/jb/mvad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023] Open
Abstract
Sweet and umami taste receptors recognize chemicals such as sugars and amino acids on their extracellular side and transmit signals into the cytosol of the taste cell. In contrast to ligands that act on the extracellular side of these receptors, little is known regarding the molecules that regulate receptor functions within the cytosol. In this study, we analysed the interaction between sweet and umami taste receptors and calmodulin, a representative Ca2+-dependent cytosolic regulatory protein. High prediction scores for calmodulin binding were observed on the C-terminal cytosolic side of mouse taste receptor type 1 subunit 3 (T1r3), a subunit that is common to both sweet and umami taste receptors. Pull-down assay and surface plasmon resonance analyses showed different affinities of calmodulin to the C-terminal tails of distinct T1r subtypes. Furthermore, we found that T1r3 and T1r2 showed the highest and considerable binding to calmodulin, whereas T1r1 showed weaker binding affinity. Finally, the binding of calmodulin to T1rs was consistently higher in the presence of Ca2+ than in its absence. The results suggested a possibility of the Ca2+-dependent feedback regulation process of sweet and umami taste receptor signaling by calmodulin.
Collapse
Affiliation(s)
- Atsuki Yoshida
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ayumi Ito
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Norihisa Yasui
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Atsuko Yamashita
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
11
|
Kumar P, Redel U, Lang T, Korsching SI, Behrens M. Bitter taste receptors of the zebra finch ( Taeniopygia guttata). Front Physiol 2023; 14:1233711. [PMID: 37860623 PMCID: PMC10582322 DOI: 10.3389/fphys.2023.1233711] [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] [Received: 06/02/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Despite the important role of bitter taste for the rejection of potentially harmful food sources, birds have long been suspected to exhibit inferior bitter tasting abilities. Although more recent reports on the bitter recognition spectra of several bird species have cast doubt about the validity of this assumption, the bitter taste of avian species is still an understudied field. Previously, we reported the bitter activation profiles of three zebra finch receptors Tas2r5, -r6, and -r7, which represent orthologs of a single chicken bitter taste receptor, Tas2r1. In order to get a better understanding of the bitter tasting capabilities of zebra finches, we selected another Tas2r gene of this species that is similar to another chicken Tas2r. Using functional calcium mobilization experiments, we screened zebra finch Tas2r1 with 72 bitter compounds and observed responses for 7 substances. Interestingly, all but one of the newly identified bitter agonists were different from those previously identified for Tas2r5, -r6, and -r7 suggesting that the newly investigated receptor fills important gaps in the zebra finch bitter recognition profile. The most potent bitter agonist found in our study is cucurbitacin I, a highly toxic natural bitter substance. We conclude that zebra finch exhibits an exquisitely developed bitter taste with pronounced cucurbitacin I sensitivity suggesting a prominent ecological role of this compound for zebra finch.
Collapse
Affiliation(s)
- Praveen Kumar
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Ulrike Redel
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Tatjana Lang
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | | | - Maik Behrens
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| |
Collapse
|
12
|
Iwata S, Yoshida R, Takai S, Sanematsu K, Shigemura N, Ninomiya Y. Adrenomedullin Enhances Mouse Gustatory Nerve Responses to Sugars via T1R-Independent Sweet Taste Pathway. Nutrients 2023; 15:2941. [PMID: 37447268 DOI: 10.3390/nu15132941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
On the tongue, the T1R-independent pathway (comprising glucose transporters, including sodium-glucose cotransporter (SGLT1) and the KATP channel) detects only sugars, whereas the T1R-dependent (T1R2/T1R3) pathway can broadly sense various sweeteners. Cephalic-phase insulin release, a rapid release of insulin induced by sensory signals in the head after food-related stimuli, reportedly depends on the T1R-independent pathway, and the competitive sweet taste modulators leptin and endocannabinoids may function on these two different sweet taste pathways independently, suggesting independent roles of two oral sugar-detecting pathways in food intake. Here, we examined the effect of adrenomedullin (ADM), a multifunctional regulatory peptide, on sugar sensing in mice since it affects the expression of SGLT1 in rat enterocytes. We found that ADM receptor components were expressed in T1R3-positive taste cells. Analyses of chorda tympani (CT) nerve responses revealed that ADM enhanced responses to sugars but not to artificial sweeteners and other tastants. Moreover, ADM increased the apical uptake of a fluorescent D-glucose derivative into taste cells and SGLT1 mRNA expression in taste buds. These results suggest that the T1R-independent sweet taste pathway in mouse taste cells is a peripheral target of ADM, and the specific enhancement of gustatory nerve responses to sugars by ADM may contribute to caloric sensing and food intake.
Collapse
Affiliation(s)
- Shusuke Iwata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Department of Oral Physiology, Asahi University School of Dentistry, Gifu 501-0296, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Dent-Craniofacial Development and Regeneration Center, Graduate School of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
- OBT Research Center, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
| | - Yuzo Ninomiya
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Oral Science Research Center, Tokyo Dental College, Tokyo 101-0061, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Ziegler F, Steuer A, Di Pizio A, Behrens M. Physiological activation of human and mouse bitter taste receptors by bile acids. Commun Biol 2023; 6:612. [PMID: 37286811 DOI: 10.1038/s42003-023-04971-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/23/2023] [Indexed: 06/09/2023] Open
Abstract
Beside the oral cavity, bitter taste receptors are expressed in several non-gustatory tissues. Whether extra-oral bitter taste receptors function as sensors for endogenous agonists is unknown. To address this question, we devised functional experiments combined with molecular modeling approaches to investigate human and mouse receptors using a variety of bile acids as candidate agonists. We show that five human and six mouse receptors are responsive to an array of bile acids. Moreover, their activation threshold concentrations match published data of bile acid concentrations in human body fluids, suggesting a putative physiological activation of non-gustatory bitter receptors. We conclude that these receptors could serve as sensors for endogenous bile acid levels. These results also indicate that bitter receptor evolution may not be driven solely by foodstuff or xenobiotic stimuli, but also depend on endogenous ligands. The determined bitter receptor activation profiles of bile acids now enable detailed physiological model studies.
Collapse
Affiliation(s)
- Florian Ziegler
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Alexandra Steuer
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Maik Behrens
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany.
| |
Collapse
|
14
|
Chometton S, Tsan L, Hayes AMR, Kanoski SE, Schier LA. Early-life influences of low-calorie sweetener consumption on sugar taste. Physiol Behav 2023; 264:114133. [PMID: 36801464 PMCID: PMC11062773 DOI: 10.1016/j.physbeh.2023.114133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
Children and adolescents are the highest consumers of added sugars, particularly from sugar-sweetened beverages (SSB). Regular consumption of SSB early in life induces a variety of negative consequences on health that can last into adulthood. Low-calorie sweeteners (LCS) are increasingly used as an alternative to added sugars because they provide a sweet sensation without adding calories to the diet. However, the long-term effects of early-life consumption of LCS are not well understood. Considering LCS engage at least one of the same taste receptors as sugars and potentially modulate cellular mechanisms of glucose transport and metabolism, it is especially important to understand how early-life LCS consumption impacts intake of and regulatory responses to caloric sugars. In our recent study, we found that habitual intake of LCS during the juvenile-adolescence period significantly changed how rats responded to sugar later in life. Here, we review evidence that LCS and sugars are sensed via common and distinct gustatory pathways, and then discuss the implications this has for shaping sugar-associated appetitive, consummatory, and physiological responses. Ultimately, the review highlights the diverse gaps in knowledge that will be necessary to fill to understand the consequences of regular LCS consumption during important phases of development.
Collapse
Affiliation(s)
- Sandrine Chometton
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Linda Tsan
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Anna M R Hayes
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Lindsey A Schier
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Shon WJ, Song JW, Oh SH, Lee KH, Seong H, You HJ, Seong JK, Shin DM. Gut taste receptor type 1 member 3 is an intrinsic regulator of Western diet-induced intestinal inflammation. BMC Med 2023; 21:165. [PMID: 37118698 PMCID: PMC10148556 DOI: 10.1186/s12916-023-02848-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 03/24/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Long-term intake of a Western diet (WD), characterized by a high-fat content and sugary drinks, is hypothesized to contribute to the development of inflammatory bowel disease (IBD). Despite the identified clinical association, the molecular mechanisms by which dietary changes contribute to IBD development remain unknown. Therefore, we examined the influence of long-term intake of a WD on intestinal inflammation and the mechanisms by which WD intake affects IBD development. METHODS Mice fed normal diet or WD for 10 weeks, and bowel inflammation was evaluated through pathohistological and infiltrated inflammatory cell assessments. To understand the role of intestinal taste receptor type 1 member 3 (TAS1R3) in WD-induced intestinal inflammation, cultured enteroendocrine cells harboring TAS1R3, subjected to RNA interference or antagonist treatment, and Tas1r3-deficient mice were used. RNA-sequencing, flow cytometry, 16S metagenomic sequencing, and bioinformatics analyses were performed to examine the involved mechanisms. To demonstrate their clinical relevance, intestinal biopsies from patients with IBD and mice with dextran sulfate sodium-induced colitis were analyzed. RESULTS Our study revealed for the first time that intestinal TAS1R3 is a critical mediator of WD-induced intestinal inflammation. WD-fed mice showed marked TAS1R3 overexpression with hallmarks of serious bowel inflammation. Conversely, mice lacking TAS1R3 failed to exhibit inflammatory responses to WD. Mechanistically, intestinal transcriptome analysis revealed that Tas1r3 deficiency suppressed mTOR signaling, significantly increasing the expression of PPARγ (a major mucosal defense enhancer) and upregulating the expression of PPARγ target-gene (tight junction protein and antimicrobial peptide). The gut microbiota of Tas1r3-deficient mice showed expansion of butyrate-producing Clostridia. Moreover, an increased expression of host PPARγ-signaling pathway proteins was positively correlated with butyrate-producing microbes, suggesting that intestinal TAS1R3 regulates the relationship between host metabolism and gut microflora in response to dietary factors. In cultured intestinal cells, regulation of the TAS1R3-mTOR-PPARγ axis was critical for triggering an inflammatory response via proinflammatory cytokine production and secretion. Abnormal regulation of the axis was observed in patients with IBD. CONCLUSIONS Our findings suggest that the TAS1R3-mTOR-PPARγ axis in the gut links Western diet consumption with intestinal inflammation and is a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Woo-Jeong Shon
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea.
| | - Jae Won Song
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Seung Hoon Oh
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Keon-Hee Lee
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Hobin Seong
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Hyun Ju You
- Bio-MAX/N-Bio, Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Ascencio Gutierrez V, Simental Ramos A, Khayoyan S, Schier LA. Dietary experience with glucose and fructose fosters heightened avidity for glucose-containing sugars independent of TRPM5 taste transduction in mice. Nutr Neurosci 2023; 26:345-356. [PMID: 35311614 PMCID: PMC9810270 DOI: 10.1080/1028415x.2022.2050092] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Experience with metabolically distinct sugars, glucose and fructose, enhances attraction to the orosensory properties of glucose over fructose. To gain insight into which sensory signals are affected, we investigated how this nutritive learning reshapes behavioral responding to various sugars in brief access taste tests in C57BL6/J (B6) mice and assessed whether sugar-exposed mice lacking the TRPM5 channel involved in G-protein coupled taste transduction could acquire these types of preferences for glucose-containing sugars. METHODS B6, TRPM5 knockout (KO), and TRPM5 heterozygous (Het) mice were given extensive access to water (sugar naïve) or 0.316, 0.56, and 1.1 M glucose and fructose (sugar-exposed) and then tested, whilst food deprived, for their relative avidities for glucose, fructose, sucrose, maltose, and/or a non-metabolizable glucose analog in a series of taste tests. RESULTS Sugar-exposed B6 mice licked relatively more for glucose than fructose, driven by an increased avidity for glucose, not an avoidance of fructose, and licked more for maltose, compared to their sugar-naïve counterparts. Sugar-exposed B6 mice did not lick with such avidity for a non-metabolizable glucose analog. TRPM5 KO mice took longer to acquire the sugar discrimination than the Het controls, but both groups ultimately licked significantly more for glucose than fructose. Het mice displayed clear preferential licking for sucrose over fructose, while licking comparably high for glucose, sucrose, and maltose. KO mice licked significantly more for maltose than sucrose. CONCLUSIONS Collectively, the findings suggest that ingestive experience with glucose and fructose primarily reprograms behavioral responding to a TRPM5-independent orosensory signal generated by glucose-containing sugars.
Collapse
Affiliation(s)
| | | | - Shushanna Khayoyan
- Department of Biological Sciences, University of Southern California, Los Angeles, CA
| | - Lindsey A. Schier
- Department of Biological Sciences, University of Southern California, Los Angeles, CA
| |
Collapse
|
17
|
Stavrou MR, So SS, Finch AM, Ballouz S, Smith NJ. Gene expression analyses of TAS1R taste receptors relevant to the treatment of cardiometabolic disease. Chem Senses 2023; 48:bjad027. [PMID: 37539767 DOI: 10.1093/chemse/bjad027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Indexed: 08/05/2023] Open
Abstract
The sweet taste receptor (STR) is a G protein-coupled receptor (GPCR) responsible for mediating cellular responses to sweet stimuli. Early evidence suggests that elements of the STR signaling system are present beyond the tongue in metabolically active tissues, where it may act as an extraoral glucose sensor. This study aimed to delineate expression of the STR in extraoral tissues using publicly available RNA-sequencing repositories. Gene expression data was mined for all genes implicated in the structure and function of the STR, and control genes including highly expressed metabolic genes in relevant tissues, other GPCRs and effector G proteins with physiological roles in metabolism, and other GPCRs with expression exclusively outside the metabolic tissues. Since the physiological role of the STR in extraoral tissues is likely related to glucose sensing, expression was then examined in diseases related to glucose-sensing impairment such as type 2 diabetes. An aggregate co-expression network was then generated to precisely determine co-expression patterns among the STR genes in these tissues. We found that STR gene expression was negligible in human pancreatic and adipose tissues, and low in intestinal tissue. Genes encoding the STR did not show significant co-expression or connectivity with other functional genes in these tissues. In addition, STR expression was higher in mouse pancreatic and adipose tissues, and equivalent to human in intestinal tissue. Our results suggest that STR expression in mice is not representative of expression in humans, and the receptor is unlikely to be a promising extraoral target in human cardiometabolic disease.
Collapse
Affiliation(s)
- Mariah R Stavrou
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Sean Souchiart So
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Angela M Finch
- Department of Pharmacology, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Sara Ballouz
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Computer Science and Engineering, Faculty of Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - Nicola J Smith
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Melis M, Mastinu M, Naciri LC, Muroni P, Tomassini Barbarossa I. Associations between Sweet Taste Sensitivity and Polymorphisms (SNPs) in the TAS1R2 and TAS1R3 Genes, Gender, PROP Taster Status, and Density of Fungiform Papillae in a Genetically Homogeneous Sardinian Cohort. Nutrients 2022; 14:4903. [PMID: 36432589 PMCID: PMC9696868 DOI: 10.3390/nu14224903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Individual differences in sweet taste sensitivity can affect dietary preferences as well as nutritional status. Despite the lack of consensus, it is believed that sweet taste is impacted by genetic and environmental variables. Here we determined the effect of well-established factors influencing the general taste variability, such as gender and fungiform papillae density, specific genetic variants (SNPs of TAS1R2 and TAS1R3 receptors genes), and non-specific genetic factors (PROP phenotype and genotype), on the threshold and suprathreshold sweet taste sensitivity. Suprathreshold measurements showed that the sweet taste response increased in a dose-dependent manner, and this was related to PROP phenotype, gender, rs35874116 SNP in the TAS1R2 gene, and rs307355 SNP in the TAS1R3 gene. The threshold values and density of fungiform papillae exhibited a strong correlation, and both varied according to PROP phenotype. Our data confirm the role of PROP taste status in the sweet perception related to fungiform papilla density, show a higher sweet sensitivity in females who had lower BMI than males, and demonstrate for the first time the involvement of the rs35874116 SNP of TAS1R2 in the sweet taste sensitivity of normal weight subjects with body mass index (BMI) ranging from 20.2 to 24.8 kg/m2. These results may have an important impact on nutrition and health mostly in subjects with low taste ability for sweets and thus with high vulnerability to developing obesity or metabolic disease.
Collapse
Affiliation(s)
- Melania Melis
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, CA, Italy
| | | | | | | | | |
Collapse
|
19
|
Abstract
When it comes to food, one tempting substance is sugar. Although sweetness is detected by the tongue, the desire to consume sugar arises from the gut. Even when sweet taste is impaired, animals can distinguish sugars from non-nutritive sweeteners guided by sensory cues arising from the gut epithelium. Here, we review the molecular receptors, cells, circuits and behavioural consequences associated with sugar sensing in the gut. Recent work demonstrates that some duodenal cells, termed neuropod cells, can detect glucose using sodium-glucose co-transporter 1 and release glutamate onto vagal afferent neurons. Based on these and other data, we propose a model in which specific populations of vagal neurons relay these sensory cues to distinct sets of neurons in the brain, including neurons in the caudal nucleus of the solitary tract, dopaminergic reward circuits in the basal ganglia and homeostatic feeding circuits in the hypothalamus, that alter current and future sugar consumption. This emerging model highlights the critical role of the gut in sensing the chemical properties of ingested nutrients to guide appetitive decisions.
Collapse
Affiliation(s)
- Winston W Liu
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
20
|
Xu W, Wu L, Liu S, Liu X, Cao X, Zhou C, Zhang J, Fu Y, Guo Y, Wu Y, Tan Q, Wang L, Liu J, Jiang L, Fan Z, Pei Y, Yu J, Cheng J, Zhao S, Hao X, Liu ZJ, Hua T. Structural basis for strychnine activation of human bitter taste receptor TAS2R46. Science 2022; 377:1298-1304. [DOI: 10.1126/science.abo1633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Taste sensing is a sophisticated chemosensory process, and bitter taste perception is mediated by type 2 taste receptors (TAS2Rs), or class T G protein–coupled receptors. Understanding the detailed molecular mechanisms behind taste sensation is hindered by a lack of experimental receptor structures. Here, we report the cryo–electron microscopy structures of human TAS2R46 complexed with chimeric mini–G protein gustducin, in both strychnine-bound and apo forms. Several features of TAS2R46 are disclosed, including distinct receptor structures that compare with known GPCRs, a new “toggle switch,” activation-related motifs, and precoupling with mini–G protein gustducin. Furthermore, the dynamic extracellular and more-static intracellular parts of TAS2R46 suggest possible diverse ligand-recognition and activation processes. This study provides a basis for further exploration of other bitter taste receptors and their therapeutic applications.
Collapse
Affiliation(s)
- Weixiu Xu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Shenhui Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiao Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoling Cao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Cui Zhou
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinyi Zhang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - You Fu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yu Guo
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Qiwen Tan
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Ling Wang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Longquan Jiang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhongbo Fan
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Pei
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Jingyi Yu
- School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Plant Resource in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650210, China
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
21
|
Ki SY, Jeong YT. Taste Receptors beyond Taste Buds. Int J Mol Sci 2022; 23:ijms23179677. [PMID: 36077074 PMCID: PMC9455917 DOI: 10.3390/ijms23179677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Taste receptors are responsible for detecting their ligands not only in taste receptor cells (TRCs) but also in non-gustatory organs. For several decades, many research groups have accumulated evidence for such “ectopic” expression of taste receptors. More recently, some of the physiologic functions (apart from taste) of these ectopic taste receptors have been identified. Here, we summarize our current understanding of these ectopic taste receptors across multiple organs. With a particular focus on the specialized epithelial cells called tuft cells, which are now considered siblings of type II TRCs, we divide the ectopic expression of taste receptors into two categories: taste receptors in TRC-like cells outside taste buds and taste receptors with surprising ectopic expression in completely different cell types.
Collapse
Affiliation(s)
- Su Young Ki
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Yong Taek Jeong
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
- Correspondence: ; Tel.: +82-2-2286-1295
| |
Collapse
|
22
|
Lee J, Kim SJ, Choi GE, Yi E, Park HJ, Choi WS, Jang YJ, Kim HS. Sweet taste receptor agonists attenuate macrophage IL-1β expression and eosinophilic inflammation linked to autophagy deficiency in myeloid cells. Clin Transl Med 2022; 12:e1021. [PMID: 35988262 PMCID: PMC9393075 DOI: 10.1002/ctm2.1021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/28/2022] [Accepted: 08/04/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Eosinophilic inflammation is a hallmark of refractory chronic rhinosinusitis (CRS) and considered a major therapeutic target. Autophagy deficiency in myeloid cells plays a causal role in eosinophilic CRS (ECRS) via macrophage IL-1β overproduction, thereby suggesting autophagy regulation as a potential therapeutic modality. Trehalose is a disaccharide sugar with known pro-autophagy activity and effective in alleviating diverse inflammatory diseases. We sought to investigate the therapeutic potential of autophagy-enhancing agent, trehalose, or related sugar compounds, and the underlying mechanism focusing on macrophage IL-1β production in ECRS pathogenesis. METHODS We investigated the therapeutic effects of trehalose and saccharin on macrophage IL-1β production and eosinophilia in the mouse model of ECRS with myeloid cell-specific autophagy-related gene 7 (Atg7) deletion. The mechanisms underlying their anti-inflammatory effects were assessed using specific inhibitor, genetic knockdown or knockout, and overexpression of cognate receptors. RESULTS Unexpectedly, trehalose significantly attenuated eosinophilia and disease pathogenesis in ECRS mice caused by autophagy deficiency in myeloid cells. This autophagy-independent effect was associated with reduced macrophage IL-1β expression. Various sugars recapitulated the anti-inflammatory effect of trehalose, and saccharin was particularly effective amongst other sugars. The mechanistic study revealed an involvement of sweet taste receptor (STR), especially T1R3, in alleviating macrophage IL-1β production and eosinophilia in CRS, which was supported by genetic depletion of T1R3 or overexpression of T1R2/T1R3 in macrophages and treatment with the T1R3 antagonist gurmarin. CONCLUSION Our results revealed a previously unappreciated anti-inflammatory effect of STR agonists, particularly trehalose and saccharin, and may provide an alternative strategy to autophagy modulation in the ECRS treatment.
Collapse
Affiliation(s)
- Jinju Lee
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - So Jeong Kim
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Go Eun Choi
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Department of Clinical Laboratory ScienceCatholic University of PusanBusanKorea
| | - Eunbi Yi
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Hyo Jin Park
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Woo Seon Choi
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Yong Ju Jang
- Department of OtolaryngologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| | - Hun Sik Kim
- Department of Biomedical SciencesAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Department of MicrobiologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Stem Cell Immunomodulation Research Center (SCIRC)Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
| |
Collapse
|
23
|
Zhang C, Miao Y, Feng Y, Wang J, Tian Z, Dong J, Gao B, Zhang L. Umami polypeptide detection system targeting the human T1R1 receptor and its taste-presenting mechanism. Biomaterials 2022; 287:121660. [PMID: 35792387 DOI: 10.1016/j.biomaterials.2022.121660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/24/2022] [Indexed: 01/30/2023]
Abstract
Umami is one of five basic tastes, the elucidation of its mechanism by the study of the interaction between umami polypeptides and hT1R1 umami receptors is of great significance. However, research on umami peptides targeting human T1R1 receptors is lacking, and the molecular mechanism remains elusive. Here, we successfully established a system to detect umami peptides targeting human T1R1 receptors by fluorescence spectroscopy, Surface Plasmon Resonance (SPR) and computational simulation. The sensory evaluation, calculated Kd value, and experimental affinity results between the four selected umami peptides (GRVSNCAA, KGDEESLA, KGGGGP, and TGDPEK) and glutamate were tested using this system, and all matched well. The maximum Ka value of GRVSNCAA was 479.55 M-1, and the minimum affinity of TGDPEK was 2.67 M-1. Computational simulations showed that the different peptide binding sites in the hT1R1 binding pocket occupied due to conformational changes are important factors for different taste thresholds, and that peptide hydrophobicity plays an important role in regulating affinity. Thus, our study enables rapid screening of high-intensity umami peptides and the development of T1R1 receptor-based umami detection sensors.
Collapse
Affiliation(s)
- Chuanxi Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China; School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China; Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai , 200240, China
| | - Yulu Miao
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Yinghui Feng
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Jiawei Wang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China; School of Health Science and Engineering, University of Shanghai for Science and Tecchnology, Shanghai, 200093, China
| | - Zhuoli Tian
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Juan Dong
- School of Food Science and Technology, Shihezi University, Shihezi, 832000, China
| | - Bei Gao
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.
| | - Lujia Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China; NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, 200062, China.
| |
Collapse
|
24
|
Sweet Taste Signaling: The Core Pathways and Regulatory Mechanisms. Int J Mol Sci 2022; 23:ijms23158225. [PMID: 35897802 PMCID: PMC9329783 DOI: 10.3390/ijms23158225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Sweet taste, a proxy for sugar-derived calories, is an important driver of food intake, and animals have evolved robust molecular and cellular machinery for sweet taste signaling. The overconsumption of sugar-derived calories is a major driver of obesity and other metabolic diseases. A fine-grained appreciation of the dynamic regulation of sweet taste signaling mechanisms will be required for designing novel noncaloric sweeteners with better hedonic and metabolic profiles and improved consumer acceptance. Sweet taste receptor cells express at least two signaling pathways, one mediated by a heterodimeric G-protein coupled receptor encoded by taste 1 receptor members 2 and 3 (TAS1R2 + TAS1R3) genes and another by glucose transporters and the ATP-gated potassium (KATP) channel. Despite these important discoveries, we do not fully understand the mechanisms regulating sweet taste signaling. We will introduce the core components of the above sweet taste signaling pathways and the rationale for having multiple pathways for detecting sweet tastants. We will then highlight the roles of key regulators of the sweet taste signaling pathways, including downstream signal transduction pathway components expressed in sweet taste receptor cells and hormones and other signaling molecules such as leptin and endocannabinoids.
Collapse
|
25
|
Kondo T, Uebanso T, Arao N, Shimohata T, Mawatari K, Takahashi A. Effect of T1R3 Taste Receptor Gene Deletion on Dextran Sulfate Sodium-Induced Colitis in Mice. J Nutr Sci Vitaminol (Tokyo) 2022; 68:204-212. [PMID: 35768251 DOI: 10.3177/jnsv.68.204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Taste receptor type 1 member 3 (T1R3) recognize umami or sweet tastes and also contributes type 2 immunity and autophagy in small intestine and muscle cells, respectively. Since imbalance of type 1 and type 2 immunity and autophagy affect intestinal bowel disease (IBD), we hypothesized that T1R3 have a potential role in the incidence and progression of colitis. In the present study, we investigated whether genetic deletion of T1R3 impacted aggravation of DSS-induced colitis in mice. We found that T1R3-KO mice showed reduction in colon damage, including reduced inflammation and colon shrinking relative to those of WT mice following DSS treatment. mRNA expression of tight junction components, particularly claudin1 was significantly lower in T1R3-KO mice with trend to lower inflammation related gene mRNA expression in colon. Other parameters, such as response to microbial stimuli in splenic lymphocytes and peritoneal macrophages, gut microbiota composition, and expression of autophagy-related proteins, were similar between WT and KO mice. Together, these results indicated that deletion of T1R3 has a minor role in intestinal inflammation induced by DSS-induced acute colitis in mice.
Collapse
Affiliation(s)
- Tsubasa Kondo
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Takashi Uebanso
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Natsuki Arao
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Takaaki Shimohata
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School.,Faculty of Marine Biosciences, Fukui Prefectural University
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Akira Takahashi
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School
| |
Collapse
|
26
|
Behrens M, Lang T. Extra-Oral Taste Receptors-Function, Disease, and Perspectives. Front Nutr 2022; 9:881177. [PMID: 35445064 PMCID: PMC9014832 DOI: 10.3389/fnut.2022.881177] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Taste perception is crucial for the critical evaluation of food constituents in human and other vertebrates. The five basic taste qualities salty, sour, sweet, umami (in humans mainly the taste of L-glutamic acid) and bitter provide important information on the energy content, the concentration of electrolytes and the presence of potentially harmful components in food items. Detection of the various taste stimuli is facilitated by specialized receptor proteins that are expressed in taste buds distributed on the tongue and the oral cavity. Whereas, salty and sour receptors represent ion channels, the receptors for sweet, umami and bitter belong to the G protein-coupled receptor superfamily. In particular, the G protein-coupled taste receptors have been located in a growing number of tissues outside the oral cavity, where they mediate important processes. This article will provide a brief introduction into the human taste perception, the corresponding receptive molecules and their signal transduction. Then, we will focus on taste receptors in the gastrointestinal tract, which participate in a variety of processes including the regulation of metabolic functions, hunger/satiety regulation as well as in digestion and pathogen defense reactions. These important non-gustatory functions suggest that complex selective forces have contributed to shape taste receptors during evolution.
Collapse
Affiliation(s)
- Maik Behrens
- Leibniz Institute of Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Tatjana Lang
- Leibniz Institute of Food Systems Biology at the Technical University of Munich, Freising, Germany
| |
Collapse
|
27
|
Drogalis-Kim D, Cheifetz I, Robbins N. Early nutritional influences of cardiovascular health. Expert Rev Cardiovasc Ther 2021; 19:1063-1073. [PMID: 34927523 DOI: 10.1080/14779072.2021.2021070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Increasing evidence shows that nutritional choices during children's formative years, including prenatally, impacts the development of adult onset cardiovascular diseases (CVDs), such as hypertension, myocardial infarction, or stroke. AREAS COVERED This literature review aims to synthesize the current body of evidence on nutritional factors, from conception through adolescence, which may influence a person's risk factors for future development of CVD. EXPERT OPINION Given the escalating healthcare costs associated with CVD, it is imperative that medical professionals and scientists remain steadfast in prioritizing and promoting early CVD prevention, even within the first few years of life. Though not the only contributing risk factor, diet is a modifiable risk factor and has been shown to have a profound impact on the reduction of cardiovascular morbidity and mortality in adult literature. Nutritional choices should be targeted on multiple levels: prenatally with the mother, individually with the patient, in conjunction with their family unit, and also within the broader community wherein they reside. Healthcare providers can play a key advocacy role for local and national food environment policy changes.
Collapse
Affiliation(s)
- Diana Drogalis-Kim
- Division of Pediatric Cardiology, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Ira Cheifetz
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Divisions of Pediatric Cardiac Critical Care and Cardiology, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Nathaniel Robbins
- Division of Pediatric Cardiology, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| |
Collapse
|
28
|
von Molitor E, Riedel K, Krohn M, Hafner M, Rudolf R, Cesetti T. Sweet Taste Is Complex: Signaling Cascades and Circuits Involved in Sweet Sensation. Front Hum Neurosci 2021; 15:667709. [PMID: 34239428 PMCID: PMC8258107 DOI: 10.3389/fnhum.2021.667709] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Sweetness is the preferred taste of humans and many animals, likely because sugars are a primary source of energy. In many mammals, sweet compounds are sensed in the tongue by the gustatory organ, the taste buds. Here, a group of taste bud cells expresses a canonical sweet taste receptor, whose activation induces Ca2+ rise, cell depolarization and ATP release to communicate with afferent gustatory nerves. The discovery of the sweet taste receptor, 20 years ago, was a milestone in the understanding of sweet signal transduction and is described here from a historical perspective. Our review briefly summarizes the major findings of the canonical sweet taste pathway, and then focuses on molecular details, about the related downstream signaling, that are still elusive or have been neglected. In this context, we discuss evidence supporting the existence of an alternative pathway, independent of the sweet taste receptor, to sense sugars and its proposed role in glucose homeostasis. Further, given that sweet taste receptor expression has been reported in many other organs, the physiological role of these extraoral receptors is addressed. Finally, and along these lines, we expand on the multiple direct and indirect effects of sugars on the brain. In summary, the review tries to stimulate a comprehensive understanding of how sweet compounds signal to the brain upon taste bud cells activation, and how this gustatory process is integrated with gastro-intestinal sugar sensing to create a hedonic and metabolic representation of sugars, which finally drives our behavior. Understanding of this is indeed a crucial step in developing new strategies to prevent obesity and associated diseases.
Collapse
Affiliation(s)
- Elena von Molitor
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| | | | | | - Mathias Hafner
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Tiziana Cesetti
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| |
Collapse
|
29
|
Lin C, Tordoff MG, Li X, Bosak NP, Inoue M, Ishiwatari Y, Chen L, Beauchamp GK, Bachmanov AA, Reed DR. Genetic controls of Tas1r3-independent sucrose consumption in mice. Mamm Genome 2021; 32:70-93. [PMID: 33710367 DOI: 10.1007/s00335-021-09860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/11/2021] [Indexed: 10/21/2022]
Abstract
We have previously used crosses between C57BL/6ByJ (B6) and 129P3/J (129) inbred strains to map a quantitative trait locus (QTL) on mouse chromosome (Chr) 4 that affects behavioral and neural responses to sucrose. We have named it the sucrose consumption QTL 2 (Scon2), and shown that it corresponds to the Tas1r3 gene, which encodes a sweet taste receptor subunit TAS1R3. To discover other sucrose consumption QTLs, we have intercrossed B6 inbred and 129.B6-Tas1r3 congenic mice to produce F2 hybrids, in which Scon2 (Tas1r3) does not segregate, and hence does not contribute to phenotypical variation. Chromosome mapping using this F2 intercross identified two main-effect QTLs, Scon3 (Chr9) and Scon10 (Chr14), and an epistatically interacting QTL pair Scon3 (Chr9)-Scon4 (Chr1). Using serial backcrosses, congenic and consomic strains, we conducted high-resolution mapping of Scon3 and Scon4 and analyzed their epistatic interactions. We used mice with different Scon3 or Scon4 genotypes to understand whether these two QTLs influence sucrose intake via gustatory or postoral mechanisms. These studies found no evidence for involvement of the taste mechanisms, but suggested involvement of energy metabolism. Mice with the B6 Scon4 genotype drank less sucrose in two-bottle tests, and also had a higher respiratory exchange ratio and lower energy expenditure under basal conditions (when they had only chow and water available). Our results provide evidence that Scon3 and Scon4 influence mouse-to-mouse variation in sucrose intake and that both likely act through a common postoral mechanism.
Collapse
Affiliation(s)
- Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | | - Xia Li
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Sonora Quest Laboratories, Phoenix, AZ, USA
| | | | - Masashi Inoue
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Laboratory of Cellular Neurobiology, School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan
| | - Yutaka Ishiwatari
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Ajinomoto Co. Inc, Tokyo, Japan
| | - Longhui Chen
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Tannbach Capital, Hong Kong, China
| | | | - Alexander A Bachmanov
- Monell Chemical Senses Center, Philadelphia, PA, USA.,GlaxoSmithKline, Collegeville, PA, USA
| | | |
Collapse
|
30
|
Smith NJ, Grant JN, Moon JI, So SS, Finch AM. Critically evaluating sweet taste receptor expression and signaling through a molecular pharmacology lens. FEBS J 2021; 288:2660-2672. [DOI: 10.1111/febs.15768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/04/2021] [Accepted: 02/15/2021] [Indexed: 12/26/2022]
Affiliation(s)
- Nicola J. Smith
- Orphan Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
| | - Jennifer N. Grant
- Orphan Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
- G Protein‐Coupled Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
| | - Justin I. Moon
- Orphan Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
- G Protein‐Coupled Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
| | - Sean S. So
- Orphan Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
| | - Angela M. Finch
- G Protein‐Coupled Receptor Laboratory School of Medical Sciences UNSW Sydney Kensington Australia
| |
Collapse
|
31
|
Immune Regulatory Roles of Cells Expressing Taste Signaling Elements in Nongustatory Tissues. Handb Exp Pharmacol 2021; 275:271-293. [PMID: 33945029 DOI: 10.1007/164_2021_468] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled taste receptors and their downstream signaling elements, including Gnat3 (also known as α-gustducin) and TrpM5, were first identified in taste bud cells. Subsequent studies, however, revealed that some cells in nongustatory tissues also express taste receptors and/or their signaling elements. These nongustatory-tissue-expressed taste receptors and signaling elements play important roles in a number of physiological processes, including metabolism and immune responses. Special populations of cells expressing taste signaling elements in nongustatory tissues have been described as solitary chemosensory cells (SCCs) and tuft cells, mainly based on their morphological features and their expression of taste signaling elements as a critical molecular signature. These cells are typically scattered in barrier epithelial tissues, and their functions were largely unknown until recently. Emerging evidence shows that SCCs and tuft cells play important roles in immune responses to microbes and parasites. Additionally, certain immune cells also express taste receptors or taste signaling elements, suggesting a direct link between chemosensation and immune function. In this chapter, we highlight our current understanding of the functional roles of these "taste-like" cells and taste signaling pathways in different tissues, focusing on their activities in immune regulation.
Collapse
|
32
|
Abstract
All organisms have the ability to detect chemicals in the environment, which likely evolved out of organisms' needs to detect food sources and avoid potentially harmful compounds. The taste system detects chemicals and is used to determine whether potential food items will be ingested or rejected. The sense of taste detects five known taste qualities: bitter, sweet, salty, sour, and umami, which is the detection of amino acids, specifically glutamate. These different taste qualities encompass a wide variety of chemicals that differ in their structure and as a result, the peripheral taste utilizes numerous and diverse mechanisms to detect these stimuli. In this chapter, we will summarize what is currently known about the signaling mechanisms used by taste cells to transduce stimulus signals.
Collapse
Affiliation(s)
- Debarghya Dutta Banik
- Department of Biological Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathryn F Medler
- Department of Biological Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
33
|
Wolstenholme JT, Younis RM, Toma W, Damaj MI. Adolescent low-dose ethanol drinking in the dark increases ethanol intake later in life in C57BL/6J, but not DBA/2J mice. Alcohol 2020; 89:85-91. [PMID: 32860857 PMCID: PMC7721983 DOI: 10.1016/j.alcohol.2020.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023]
Abstract
Alcohol is the most widely used and abused drug among youth in the United States. Youths aged 12-20 years old drink almost 11% of all alcohol consumed in the United States, and typically these young people are consuming alcohol in the form of binge drinking. Particularly concerning is that the risk of developing an alcohol use disorder over their lifetime increases the younger one begins to drink. Here we investigated the impact of ethanol drinking in early adolescence on adult ethanol intake using C57BL/6J and DBA/2J mice. We modeled low-dose drinking in adolescent mice using a modified Drinking in the Dark (DID) model where the total ethanol intake during adolescence was similar between the strains to specifically ask whether low-dose ethanol exposure in the high-alcohol preferring C57BL/6J strain will also lead to increased ethanol intake in adulthood. Our results show that low-dose ethanol drinking in early adolescence dramatically increases adult intake, but only in the alcohol-preferring C57BL/6J strain. Early adolescent ethanol exposure had no effect on ethanol intake in the alcohol-nonpreferring DBA/2J mice. These data add to the growing evidence that low-dose ethanol exposures, below the pharmacologically relevant dose, can also contribute to increased drinking in adulthood, but the effect may be influenced by genetic background.
Collapse
Affiliation(s)
- Jennifer T Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; Virginia Commonwealth University, Alcohol Research Center, Richmond, VA, United States.
| | - Rabha M Younis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
34
|
Ahmad R, Dalziel JE. G Protein-Coupled Receptors in Taste Physiology and Pharmacology. Front Pharmacol 2020; 11:587664. [PMID: 33390961 PMCID: PMC7774309 DOI: 10.3389/fphar.2020.587664] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein-coupled receptors (GPCRs) comprise the largest receptor family in mammals and are responsible for the regulation of most physiological functions. Besides mediating the sensory modalities of olfaction and vision, GPCRs also transduce signals for three basic taste qualities of sweet, umami (savory taste), and bitter, as well as the flavor sensation kokumi. Taste GPCRs reside in specialised taste receptor cells (TRCs) within taste buds. Type I taste GPCRs (TAS1R) form heterodimeric complexes that function as sweet (TAS1R2/TAS1R3) or umami (TAS1R1/TAS1R3) taste receptors, whereas Type II are monomeric bitter taste receptors or kokumi/calcium-sensing receptors. Sweet, umami and kokumi receptors share structural similarities in containing multiple agonist binding sites with pronounced selectivity while most bitter receptors contain a single binding site that is broadly tuned to a diverse array of bitter ligands in a non-selective manner. Tastant binding to the receptor activates downstream secondary messenger pathways leading to depolarization and increased intracellular calcium in TRCs, that in turn innervate the gustatory cortex in the brain. Despite recent advances in our understanding of the relationship between agonist binding and the conformational changes required for receptor activation, several major challenges and questions remain in taste GPCR biology that are discussed in the present review. In recent years, intensive integrative approaches combining heterologous expression, mutagenesis and homology modeling have together provided insight regarding agonist binding site locations and molecular mechanisms of orthosteric and allosteric modulation. In addition, studies based on transgenic mice, utilizing either global or conditional knock out strategies have provided insights to taste receptor signal transduction mechanisms and their roles in physiology. However, the need for more functional studies in a physiological context is apparent and would be enhanced by a crystallized structure of taste receptors for a more complete picture of their pharmacological mechanisms.
Collapse
Affiliation(s)
- Raise Ahmad
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - Julie E Dalziel
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| |
Collapse
|
35
|
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: 4] [Impact Index Per Article: 1.0] [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
|
36
|
Structure-Function Analyses of Human Bitter Taste Receptors-Where Do We Stand? Molecules 2020; 25:molecules25194423. [PMID: 32993119 PMCID: PMC7582848 DOI: 10.3390/molecules25194423] [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: 08/31/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
The finding that bitter taste receptors are expressed in numerous tissues outside the oral cavity and fulfill important roles in metabolic regulation, innate immunity and respiratory control, have made these receptors important targets for drug discovery. Efficient drug discovery depends heavily on detailed knowledge on structure-function-relationships of the target receptors. Unfortunately, experimental structures of bitter taste receptors are still lacking, and hence, the field relies mostly on structures obtained by molecular modeling combined with functional experiments and point mutageneses. The present article summarizes the current knowledge on the structure–function relationships of human bitter taste receptors. Although these receptors are difficult to express in heterologous systems and their homology with other G protein-coupled receptors is very low, detailed information are available at least for some of these receptors.
Collapse
|
37
|
Gutierrez R, Fonseca E, Simon SA. The neuroscience of sugars in taste, gut-reward, feeding circuits, and obesity. Cell Mol Life Sci 2020; 77:3469-3502. [PMID: 32006052 PMCID: PMC11105013 DOI: 10.1007/s00018-020-03458-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 12/19/2022]
Abstract
Throughout the animal kingdom sucrose is one of the most palatable and preferred tastants. From an evolutionary perspective, this is not surprising as it is a primary source of energy. However, its overconsumption can result in obesity and an associated cornucopia of maladies, including type 2 diabetes and cardiovascular disease. Here we describe three physiological levels of processing sucrose that are involved in the decision to ingest it: the tongue, gut, and brain. The first section describes the peripheral cellular and molecular mechanisms of sweet taste identification that project to higher brain centers. We argue that stimulation of the tongue with sucrose triggers the formation of three distinct pathways that convey sensory attributes about its quality, palatability, and intensity that results in a perception of sweet taste. We also discuss the coding of sucrose throughout the gustatory pathway. The second section reviews how sucrose, and other palatable foods, interact with the gut-brain axis either through the hepatoportal system and/or vagal pathways in a manner that encodes both the rewarding and of nutritional value of foods. The third section reviews the homeostatic, hedonic, and aversive brain circuits involved in the control of food intake. Finally, we discuss evidence that overconsumption of sugars (or high fat diets) blunts taste perception, the post-ingestive nutritional reward value, and the circuits that control feeding in a manner that can lead to the development of obesity.
Collapse
Affiliation(s)
- Ranier Gutierrez
- Laboratory of Neurobiology of Appetite, Department of Pharmacology, CINVESTAV, 07360, Mexico City, Mexico.
| | - Esmeralda Fonseca
- Laboratory of Neurobiology of Appetite, Department of Pharmacology, CINVESTAV, 07360, Mexico City, Mexico
| | - Sidney A Simon
- Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA
| |
Collapse
|
38
|
Research on sensing characteristics of three human umami receptors via receptor‐based biosensor. FLAVOUR FRAG J 2020. [DOI: 10.1002/ffj.3608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
39
|
Allelic variation of the Tas1r3 taste receptor gene affects sweet taste responsiveness and metabolism of glucose in F1 mouse hybrids. PLoS One 2020; 15:e0235913. [PMID: 32673349 PMCID: PMC7365461 DOI: 10.1371/journal.pone.0235913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/25/2020] [Indexed: 11/25/2022] Open
Abstract
In mammals, inter- and intraspecies differences in consumption of sweeteners largely depend on allelic variation of the Tas1r3 gene (locus Sac) encoding the T1R3 protein, a sweet taste receptor subunit. To assess the influence of Tas1r3 polymorphisms on feeding behavior and metabolism, we examined the phenotype of F1 male hybrids obtained from crosses between the following inbred mouse strains: females from 129SvPasCrl (129S2) bearing the recessive Tas1r3 allele and males from either C57BL/6J (B6), carrying the dominant allele, or the Tas1r3-gene knockout strain C57BL/6J-Tas1r3tm1Rfm (B6-Tas1r3-/-). The hybrids 129S2B6F1 and 129S2B6-Tas1r3-/-F1 had identical background genotypes and different sets of Tas1r3 alleles. The effect of Tas1r3 hemizygosity was analyzed by comparing the parental strain B6 (Tas1r3 homozygote) and hemizygous F1 hybrids B6 × B6-Tas1r3-/-. Data showed that, in 129S2B6-Tas1r3-/-F1 hybrids, the reduction of glucose tolerance, along with lower consumption of and lower preference for sweeteners during the initial licking responses, is due to expression of the recessive Tas1r3 allele. Hemizygosity of Tas1r3 did not influence these behavioral and metabolic traits. However, the loss of the functional Tas1r3 allele was associated with a small decline in the long-term intake and preference for sweeteners and reduction of plasma insulin and body, liver, and fat mass.
Collapse
|
40
|
Current Progress in Understanding the Structure and Function of Sweet Taste Receptor. J Mol Neurosci 2020; 71:234-244. [PMID: 32607758 DOI: 10.1007/s12031-020-01642-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 06/19/2020] [Indexed: 10/24/2022]
Abstract
The sweet taste receptor, which was identified approximately 20 years ago, mediates sweet taste recognition in humans and other vertebrates. With the development of genomics, metabonomics, structural biology, evolutionary biology, physiology, and neuroscience, as well as technical advances in these areas, our understanding of this important protein has resulted in substantial progress. This article reviews the structure, function, genetics, and evolution of the sweet taste receptor and offers meaningful insights into this G protein-coupled receptor, which may be helpful guidances for personalized feeding, diet, and medicine. Prospective directions for research on sweet taste receptors have also been proposed.
Collapse
|
41
|
Sensing Senses: Optical Biosensors to Study Gustation. SENSORS 2020; 20:s20071811. [PMID: 32218129 PMCID: PMC7180777 DOI: 10.3390/s20071811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 12/11/2022]
Abstract
The five basic taste modalities, sweet, bitter, umami, salty and sour induce changes of Ca2+ levels, pH and/or membrane potential in taste cells of the tongue and/or in neurons that convey and decode gustatory signals to the brain. Optical biosensors, which can be either synthetic dyes or genetically encoded proteins whose fluorescence spectra depend on levels of Ca2+, pH or membrane potential, have been used in primary cells/tissues or in recombinant systems to study taste-related intra- and intercellular signaling mechanisms or to discover new ligands. Taste-evoked responses were measured by microscopy achieving high spatial and temporal resolution, while plate readers were employed for higher throughput screening. Here, these approaches making use of fluorescent optical biosensors to investigate specific taste-related questions or to screen new agonists/antagonists for the different taste modalities were reviewed systematically. Furthermore, in the context of recent developments in genetically encoded sensors, 3D cultures and imaging technologies, we propose new feasible approaches for studying taste physiology and for compound screening.
Collapse
|
42
|
Myers KP, Summers MY, Geyer-Roberts E, Schier LA. The Role of Post-Ingestive Feedback in the Development of an Enhanced Appetite for the Orosensory Properties of Glucose over Fructose in Rats. Nutrients 2020; 12:nu12030807. [PMID: 32197514 PMCID: PMC7146512 DOI: 10.3390/nu12030807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 01/30/2023] Open
Abstract
The simple sugars glucose and fructose share a common “sweet” taste quality mediated by the T1R2+T1R3 taste receptor. However, when given the opportunity to consume each sugar, rats learn to affectively discriminate between glucose and fructose on the basis of cephalic chemosensory cues. It has been proposed that glucose has a unique sensory property that becomes more hedonically positive through learning about the relatively more rewarding post-ingestive effects that are associated with glucose as compared to fructose. We tested this theory using intragastric (IG) infusions to manipulate the post-ingestive consequences of glucose and fructose consumption. Food-deprived rats with IG catheters repeatedly consumed multiple concentrations of glucose and fructose in separate sessions. For rats in the “Matched” group, each sugar was accompanied by IG infusion of the same sugar. For the “Mismatched” group, glucose consumption was accompanied by IG fructose, and vice versa. This condition gave rats orosensory experience with each sugar but precluded the differential post-ingestive consequences. Following training, avidity for each sugar was assessed in brief access and licking microstructure tests. The Matched group displayed more positive evaluation of glucose relative to fructose than the Mismatched group. A second experiment used a different concentration range and compared responses of the Matched and Mismatched groups to a control group kept naïve to the orosensory properties of sugar. Consistent with results from the first experiment, the Matched group, but not the Mismatched or Control group, displayed elevated licking responses to glucose. These experiments yield additional evidence that glucose and fructose have discriminable sensory properties and directly demonstrate that their different post-ingestive effects are responsible for the experience-dependent changes in the motivation for glucose versus fructose.
Collapse
Affiliation(s)
- Kevin P. Myers
- Department of Psychology, Bucknell University, Lewisburg, PA 17837, USA;
- Neuroscience Program, Bucknell University, Lewisburg, PA 17837, USA;
| | - Megan Y. Summers
- Neuroscience Program, Bucknell University, Lewisburg, PA 17837, USA;
| | | | - Lindsey A. Schier
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: ; Tel.: +1-213-740-6633
| |
Collapse
|
43
|
Spaggiari G, Di Pizio A, Cozzini P. Sweet, umami and bitter taste receptors: State of the art of in silico molecular modeling approaches. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2019.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Howitt MR, Cao YG, Gologorsky MB, Li JA, Haber AL, Biton M, Lang J, Michaud M, Regev A, Garrett WS. The Taste Receptor TAS1R3 Regulates Small Intestinal Tuft Cell Homeostasis. Immunohorizons 2020; 4:23-32. [PMID: 31980480 PMCID: PMC7197368 DOI: 10.4049/immunohorizons.1900099] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/08/2020] [Indexed: 01/06/2023] Open
Abstract
Tuft cells are an epithelial cell type critical for initiating type 2 immune responses to parasites and protozoa in the small intestine. To respond to these stimuli, intestinal tuft cells use taste chemosensory signaling pathways, but the role of taste receptors in type 2 immunity is poorly understood. In this study, we show that the taste receptor TAS1R3, which detects sweet and umami in the tongue, also regulates tuft cell responses in the distal small intestine. BALB/c mice, which have an inactive form of TAS1R3, as well as Tas1r3-deficient C57BL6/J mice both have severely impaired responses to tuft cell–inducing signals in the ileum, including the protozoa Tritrichomonas muris and succinate. In contrast, TAS1R3 is not required to mount an immune response to the helminth Heligmosomoides polygyrus, which infects the proximal small intestine. Examination of uninfected Tas1r3−/− mice revealed a modest reduction in the number of tuft cells in the proximal small intestine but a severe decrease in the distal small intestine at homeostasis. Together, these results suggest that TAS1R3 influences intestinal immunity by shaping the epithelial cell landscape at steady-state. ImmunoHorizons, 2020, 4: 23–32.
Collapse
Affiliation(s)
- Michael R Howitt
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115; .,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115.,Department of Pathology, Stanford University, Stanford, CA 94305
| | - Y Grace Cao
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115.,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | | | - Jessica A Li
- Department of Pathology, Stanford University, Stanford, CA 94305
| | - Adam L Haber
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Moshe Biton
- Broad Institute of MIT and Harvard, Cambridge, MA 02142.,Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Jessica Lang
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115.,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Monia Michaud
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115.,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142; and
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115; .,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115.,Broad Institute of MIT and Harvard, Cambridge, MA 02142.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215
| |
Collapse
|
45
|
Governini L, Semplici B, Pavone V, Crifasi L, Marrocco C, De Leo V, Arlt E, Gudermann T, Boekhoff I, Luddi A, Piomboni P. Expression of Taste Receptor 2 Subtypes in Human Testis and Sperm. J Clin Med 2020; 9:E264. [PMID: 31963712 PMCID: PMC7019805 DOI: 10.3390/jcm9010264] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/31/2022] Open
Abstract
Taste receptors (TASRs) are expressed not only in the oral cavity but also throughout the body, thus suggesting that they may play different roles in organ systems beyond the tongue. Recent studies showed the expression of several TASRs in mammalian testis and sperm, indicating an involvement of these receptors in male gametogenesis and fertility. This notion is supported by an impaired reproductive phenotype of mouse carrying targeted deletion of taste receptor genes, as well as by a significant correlation between human semen parameters and specific polymorphisms of taste receptor genes. To better understand the biological and thus clinical significance of these receptors for human reproduction, we analyzed the expression of several members of the TAS2Rs family of bitter receptors in human testis and in ejaculated sperm before and after in vitro selection and capacitation. Our results provide evidence for the expression of TAS2R genes, with TAS2R14 being the most expressed bitter receptor subtype in both testis tissue and sperm cells, respectively. In addition, it was observed that in vitro capacitation significantly affects both the expression and the subcellular localization of these receptors in isolated spermatozoa. Interestingly, α-gustducin and α-transducin, two Gα subunits expressed in taste buds on the tongue, are also expressed in human spermatozoa; moreover, a subcellular redistribution of both G protein α-subunits to different sub-compartments of sperm was registered upon in vitro capacitation. Finally, we shed light on the possible downstream transduction pathway initiated upon taste receptor activation in the male reproductive system. Performing ultrasensitive droplets digital PCR assays to quantify RNA copy numbers of a distinct gene, we found a significant correlation between the expression of TAS2Rs and TRPM5 (r = 0.87), the cation channel involved in bitter but also sweet and umami taste transduction in taste buds on the tongue. Even if further studies are needed to clarify the precise functional role of taste receptors for successful reproduction, the presented findings significantly extend our knowledge of the biological role of TAS2Rs for human male fertility.
Collapse
Affiliation(s)
- Laura Governini
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Bianca Semplici
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Valentina Pavone
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Laura Crifasi
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Camilla Marrocco
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Vincenzo De Leo
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Muenchen, Germany; (E.A.); (T.G.); (I.B.)
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Muenchen, Germany; (E.A.); (T.G.); (I.B.)
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80336 Muenchen, Germany; (E.A.); (T.G.); (I.B.)
| | - Alice Luddi
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| | - Paola Piomboni
- Department of Molecular and Developmental Medicine, Siena University, 53100 Siena, Italy; (L.G.); (B.S.); (V.P.); (L.C.); (C.M.); (V.D.L.); (P.P.)
| |
Collapse
|
46
|
The function and allosteric control of the human sweet taste receptor. FROM STRUCTURE TO CLINICAL DEVELOPMENT: ALLOSTERIC MODULATION OF G PROTEIN-COUPLED RECEPTORS 2020; 88:59-82. [DOI: 10.1016/bs.apha.2020.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
47
|
Malik B, Elkaddi N, Turkistani J, Spielman AI, Ozdener MH. Mammalian Taste Cells Express Functional Olfactory Receptors. Chem Senses 2019; 44:289-301. [PMID: 31140574 PMCID: PMC6538964 DOI: 10.1093/chemse/bjz019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The peripheral taste and olfactory systems in mammals are separate and independent sensory systems. In the current model of chemosensation, gustatory, and olfactory receptors are genetically divergent families expressed in anatomically distinct locations that project to disparate downstream targets. Although information from the 2 sensory systems merges to form the perception of flavor, the first cross talk is thought to occur centrally, in the insular cortex. Recent studies have shown that gustatory and olfactory receptors are expressed throughout the body and serve as chemical sensors in multiple tissues. Olfactory receptor cDNA has been detected in the tongue, yet the presence of physiologically functional olfactory receptors in taste cells has not yet been demonstrated. Here we report that olfactory receptors are functionally expressed in taste papillae. We found expression of olfactory receptors in the taste papillae of green fluorescent protein-expressing transgenic mice and, using immunocytochemistry and real-time quantitative polymerase chain reaction experiments, the presence of olfactory signal transduction molecules and olfactory receptors in cultured human fungiform taste papilla (HBO) cells. Both HBO cells and mouse taste papilla cells responded to odorants. Knockdown of adenylyl cyclase mRNA by specific small inhibitory RNA and pharmacological block of adenylyl cyclase eliminated these responses, leading us to hypothesize that the gustatory system may receive olfactory information in the periphery. These results provide the first direct evidence of the presence of functional olfactory receptors in mammalian taste cells. Our results also demonstrate that the initial integration of gustatory and olfactory information may occur as early as the taste receptor cells.
Collapse
Affiliation(s)
- Bilal Malik
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Nadia Elkaddi
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
48
|
Schier LA, Inui-Yamamoto C, Blonde GD, Spector AC. T1R2+T1R3-independent chemosensory inputs contributing to behavioral discrimination of sugars in mice. Am J Physiol Regul Integr Comp Physiol 2019; 316:R448-R462. [PMID: 30624973 PMCID: PMC6589602 DOI: 10.1152/ajpregu.00255.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/06/2018] [Accepted: 01/08/2019] [Indexed: 11/22/2022]
Abstract
Simple sugars are thought to elicit a unitary sensation, principally via the "sweet" taste receptor type 1 taste receptor (T1R)2+T1R3, yet we previously found that rats with experience consuming two metabolically distinct sugars, glucose and fructose, subsequently licked more for glucose than fructose, even when postingestive influences were abated. The results pointed to the existence of an orosensory receptor that binds one sugar but not the other and whose signal is channeled into neural circuits that motivate ingestion. Here we sought to determine the chemosensory nature of this signal. First, we assessed whether T1R2 and/or T1R3 are necessary to acquire this behavioral discrimination, replicating our rat study in T1R2+T1R3 double-knockout (KO) mice and their wild-type counterparts as well as in two common mouse strains that vary in their sensitivity to sweeteners [C57BL/6 (B6) and 129X1/SvJ (129)]. These studies showed that extensive exposure to multiple concentrations of glucose and fructose in daily one-bottle 30-min sessions enhanced lick responses for glucose over fructose in brief-access tests. This was true even for KO mice that lacked the canonical "sweet" taste receptor. Surgical disconnection of olfactory inputs to the forebrain (bulbotomy) in B6 mice severely disrupted the ability to express this experience-dependent sugar discrimination. Importantly, these bulbotomized B6 mice exhibited severely blunted responsiveness to both sugars relative to water in brief-access lick tests, despite the fact that they have intact T1R2+T1R3 receptors. The results highlight the importance of other sources of chemosensory and postingestive inputs in shaping and maintaining "hardwired" responses to sugar.
Collapse
Affiliation(s)
- Lindsey A Schier
- Department of Biological Sciences, University of Southern California , Los Angeles, California
| | - Chizuko Inui-Yamamoto
- Department of Oral Anatomy and Development, Osaka University Graduate School of Dentistry , Osaka , Japan
- Department of Psychology, Program in Neuroscience, Florida State University , Tallahassee, Florida
| | - Ginger D Blonde
- Department of Psychology, Program in Neuroscience, Florida State University , Tallahassee, Florida
| | - Alan C Spector
- Department of Psychology, Program in Neuroscience, Florida State University , Tallahassee, Florida
| |
Collapse
|
49
|
Lim J, Pullicin AJ. Oral carbohydrate sensing: Beyond sweet taste. Physiol Behav 2019; 202:14-25. [DOI: 10.1016/j.physbeh.2019.01.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/15/2019] [Accepted: 01/23/2019] [Indexed: 01/28/2023]
|
50
|
Nakagita T, Ishida A, Matsuya T, Kobayashi T, Narukawa M, Hirokawa T, Hashimoto M, Misaka T. Structural insights into the differences among lactisole derivatives in inhibitory mechanisms against the human sweet taste receptor. PLoS One 2019; 14:e0213552. [PMID: 30883570 PMCID: PMC6422327 DOI: 10.1371/journal.pone.0213552] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/23/2019] [Indexed: 11/22/2022] Open
Abstract
Lactisole, an inhibitor of the human sweet taste receptor, has a 2-phenoxypropionic acid skeleton and has been shown to interact with the transmembrane domain of the T1R3 subunit (T1R3-TMD) of the receptor. Another inhibitor, 2,4-DP, which shares the same molecular skeleton as lactisole, was confirmed to be approximately 10-fold more potent in its inhibitory activity than lactisole; however the structural basis of their inhibitory mechanisms against the receptor remains to be elucidated. Crystal structures of the TMD of metabotropic glutamate receptors, which along with T1Rs are categorized as class C G-protein coupled receptors, have recently been reported and made it possible to create an accurate structural model for T1R3-TMD. In this study, the detailed structural mechanism underlying sweet taste inhibition was characterized by comparing the action of lactisole on T1R3-TMD with that of 2,4-DP. We first performed a series of experiments using cultured cells expressing the sweet taste receptor with mutations and examined the interactions with these inhibitors. Based on the results, we next performed docking simulations and then applied molecular dynamics-based energy minimization. Our analyses clearly revealed that the (S)-isomers of both lactisole and 2,4-DP, interacted with the same seven residues in T1R3-TMD and that the inhibitory potencies of those inhibitors were mainly due to stabilizing interactions mediated via their carboxyl groups in the vertical dimension of the ligand pocket of T1R3-TMD. In addition, 2,4-DP engaged in a hydrophobic interaction mediated by its o-Cl group, and this interaction may be chiefly responsible for the higher inhibitory potency of 2,4-DP.
Collapse
Affiliation(s)
- Tomoya Nakagita
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiko Ishida
- Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Takumi Matsuya
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takuya Kobayashi
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masataka Narukawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takatsugu Hirokawa
- Molecular Profiling Research Center for Drug Discovery, National Institutes of Advanced Industrial Science and Technology, Tokyo, Japan
- Department of Chemical Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Makoto Hashimoto
- Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Takumi Misaka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|