1
|
Wei K, Hill BL, Miller ZA, Mueller A, Thompson JC, Lee RJ, Carey RM. Bitter Taste Receptor Agonists Induce Apoptosis in Papillary Thyroid Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.618693. [PMID: 39484580 PMCID: PMC11527002 DOI: 10.1101/2024.10.18.618693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background Papillary thyroid carcinoma (PTC) is the most common thyroid malignancy, with a 20% recurrence rate. Bitter taste receptors (T2Rs) and their genes ( TAS2Rs ) may regulate survival in solid tumors. This study examined T2R expression and function in PTC cells. Methods Three PTC cell lines (MDA-T32, MDA-T68, MDA-T85) were analyzed for expression using RT-qPCR and immunofluorescence. Live cell imaging measured calcium responses to six bitter agonists. Viability and apoptosis effects were assessed using crystal violet and caspase 3/7 activation assays. Genome analysis of survival was conducted. Results TAS2R14 was consistently highly expressed in all cell lines. Five bitter agonists produced significant calcium responses across all cell lines. All bitter agonists significantly decreased viability and induced apoptosis. Higher TAS2R14 expression correlated with better progression-free survival in patients (p<0.05). Conclusions T2R activation by bitter agonists induces apoptosis and higher TAS2R expression is associated with survival, suggesting potential therapeutic relevance in thyroid cancer management.
Collapse
|
2
|
Magdy N, Abdelkader NF, Zaki HF, Kamel AS. Potential exacerbation of polycystic ovary syndrome by saccharin sodium Via taste receptors in a letrozole rat model. Food Chem Toxicol 2024; 191:114874. [PMID: 39032681 DOI: 10.1016/j.fct.2024.114874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
The most common cause of anovulatory infertility is polycystic ovarian syndrome (PCOS), which is closely associated with obesity and metabolic syndrome. Artificial sweetener, notably saccharin sodium (SS), has been utilized in management of obesity in PCOS. However, accumulating evidence points towards SS deleterious effects on ovarian physiology, potentially through activation of ovarian sweet and bitter taste receptors, culminating in a phenotype reminiscent of PCOS. This research embarked on exploration of SS influence on ovarian functions within a PCOS paradigm. Rats were categorized into six groups: Control, Letrozole-model, two SS groups at 2 dose levels, and two groups receiving 2 doses of SS with Letrozole. The study underscored SS capability to potentiate PCOS-related anomalies. Elevated cystic profile with outer thin granulosa cells, were discernible. This owed to increased apoptotic markers as cleaved CASP-3, mirrored by high BAX and low BCL-2, with enhanced p38-MAPK/ERK1/2 pathway. This manifestation was accompanied by activation of taste receptors and disruption of steroidogenic factors; StAR, CYP11A1, and 17β-HSD. Thus, SS showed an escalation in testosterone, progesterone, estrogen, and LH/FSH ratio, insinuating a perturbation in endocrine regulation. It is found that there is an impact of taste receptor downstream signaling on ovarian steroidogenesis and apoptosis instigating pathophysiological milieu of PCOS.
Collapse
Affiliation(s)
- Nourhan Magdy
- Quality Assurance, National Food Safety Authority, Bab El-Louq, Cairo, Egypt
| | - Noha F Abdelkader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Egypt
| | - Hala F Zaki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Egypt
| | - Ahmed S Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Egypt.
| |
Collapse
|
3
|
Zhang M, Chen T, Lu X, Lan X, Chen Z, Lu S. G protein-coupled receptors (GPCRs): advances in structures, mechanisms, and drug discovery. Signal Transduct Target Ther 2024; 9:88. [PMID: 38594257 PMCID: PMC11004190 DOI: 10.1038/s41392-024-01803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of human membrane proteins and an important class of drug targets, play a role in maintaining numerous physiological processes. Agonist or antagonist, orthosteric effects or allosteric effects, and biased signaling or balanced signaling, characterize the complexity of GPCR dynamic features. In this study, we first review the structural advancements, activation mechanisms, and functional diversity of GPCRs. We then focus on GPCR drug discovery by revealing the detailed drug-target interactions and the underlying mechanisms of orthosteric drugs approved by the US Food and Drug Administration in the past five years. Particularly, an up-to-date analysis is performed on available GPCR structures complexed with synthetic small-molecule allosteric modulators to elucidate key receptor-ligand interactions and allosteric mechanisms. Finally, we highlight how the widespread GPCR-druggable allosteric sites can guide structure- or mechanism-based drug design and propose prospects of designing bitopic ligands for the future therapeutic potential of targeting this receptor family.
Collapse
Affiliation(s)
- Mingyang Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xun Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaobing Lan
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziqiang Chen
- Department of Orthopedics, Changhai Hospital, Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Shaoyong Lu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Miller ZA, Mueller A, Kim T, Jolivert JF, Ma RZ, Muthuswami S, Park A, McMahon DB, Nead KT, Carey RM, Lee RJ. Lidocaine induces apoptosis in head and neck squamous cell carcinoma through activation of bitter taste receptor T2R14. Cell Rep 2023; 42:113437. [PMID: 37995679 PMCID: PMC10842818 DOI: 10.1016/j.celrep.2023.113437] [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: 04/14/2023] [Revised: 09/22/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) have high mortality and significant treatment-related morbidity. It is vital to discover effective, minimally invasive therapies that improve survival and quality of life. Bitter taste receptors (T2Rs) are expressed in HNSCCs, and T2R activation can induce apoptosis. Lidocaine is a local anesthetic that also activates bitter taste receptor 14 (T2R14). Lidocaine has some anti-cancer effects, but the mechanisms are unclear. Here, we find that lidocaine causes intracellular Ca2+ mobilization through activation of T2R14 in HNSCC cells. T2R14 activation with lidocaine depolarizes mitochondria, inhibits proliferation, and induces apoptosis. Concomitant with mitochondrial Ca2+ influx, ROS production causes T2R14-dependent accumulation of poly-ubiquitinated proteins, suggesting that proteasome inhibition contributes to T2R14-induced apoptosis. Lidocaine may have therapeutic potential in HNSCCs as a topical gel or intratumor injection. In addition, we find that HPV-associated (HPV+) HNSCCs are associated with increased TAS2R14 expression. Lidocaine treatment may benefit these patients, warranting future clinical studies.
Collapse
Affiliation(s)
- Zoey A Miller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pharmacology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Arielle Mueller
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - TaeBeom Kim
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer F Jolivert
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ray Z Ma
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sahil Muthuswami
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - April Park
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Derek B McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin T Nead
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan M Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Robert J Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Schamarek I, Anders L, Chakaroun RM, Kovacs P, Rohde-Zimmermann K. The role of the oral microbiome in obesity and metabolic disease: potential systemic implications and effects on taste perception. Nutr J 2023; 22:28. [PMID: 37237407 DOI: 10.1186/s12937-023-00856-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity and its metabolic sequelae still comprise a challenge when it comes to understanding mechanisms, which drive these pandemic diseases. The human microbiome as a potential key player has attracted the attention of broader research for the past decade. Most of it focused on the gut microbiome while the oral microbiome has received less attention. As the second largest niche, the oral microbiome is associated with a multitude of mechanisms, which are potentially involved in the complex etiology of obesity and associated metabolic diseases. These mechanisms include local effects of oral bacteria on taste perception and subsequent food preference as well as systemic effects on adipose tissue function, the gut microbiome and systemic inflammation. This review summarizes a growing body of research, pointing towards a more prominent role of the oral microbiome in obesity and associated metabolic diseases than expected. Ultimately, our knowledge on the oral microbiome may support the development of new patient oriented therapeutic approaches inevitable to relieve the health burden of metabolic diseases and to reach long-term benefits in patients´ lives.
Collapse
Affiliation(s)
- Imke Schamarek
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University Leipzig and the University Clinic Leipzig, AöR, Liebigstraße 20, 04103, Leipzig, Germany.
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstraße 20, 04103, Leipzig, Germany.
| | - Lars Anders
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstraße 20, 04103, Leipzig, Germany
| | - Rima M Chakaroun
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstraße 20, 04103, Leipzig, Germany
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Peter Kovacs
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Liebigstraße 20, 04103, Leipzig, Germany
- Deutsches Zentrum Für Diabetesforschung, 85764, Neuherberg, Germany
| | - Kerstin Rohde-Zimmermann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University Leipzig and the University Clinic Leipzig, AöR, Liebigstraße 20, 04103, Leipzig, Germany
| |
Collapse
|
6
|
Costa AR, Duarte AC, Costa-Brito AR, Gonçalves I, Santos CRA. Bitter taste signaling in cancer. Life Sci 2023; 315:121363. [PMID: 36610638 DOI: 10.1016/j.lfs.2022.121363] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
Pharmacoresistance of cancer cells to many drugs used in chemotherapy remains a major challenge for the treatment of cancer. Multidrug resistance transporters, especially ATP-binding cassette (ABC) transporters, are a major cause of cancer drug resistance since they translocate a broad range of drug compounds across the cell membrane, extruding them out of the cells. The regulation of ABC transporters by bitter taste receptors (TAS2Rs), which might be activated by specific bitter tasting compounds, was described in several types of cells/organs, becoming a potential target for cancer therapy. TAS2Rs expression has been reported in many organs and several types of cancer, like breast, ovarian, prostate, and colorectal cancers, where their activation was shown to be involved in various biological actions (cell survival, apoptosis, molecular transport, among others). Moreover, many TAS2Rs' ligands, such as flavonoids and alkaloids, with well-recognized beneficial properties, including several anticancer effects, have been reported as potential adjuvants in cancer therapies. In this review, we discuss the potential therapeutic role of TAS2Rs and bitter tasting compounds in different types of cancer as a possible way to circumvent chemoresistance.
Collapse
Affiliation(s)
- Ana R Costa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; CPIRN-IPG - Centro de Potencial e Inovação de Recursos Naturais, Instituto Politécnico da Guarda, Guarda, Portugal
| | - Ana R Costa-Brito
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; Research Unit for Inland Development (UDI), Polytechnic of Guarda, Guarda, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal.
| |
Collapse
|
7
|
Savory Signaling: T1R Umami Receptor Modulates Endoplasmic Reticulum Calcium Store Content and Release Dynamics in Airway Epithelial Cells. Nutrients 2023; 15:nu15030493. [PMID: 36771200 PMCID: PMC9919336 DOI: 10.3390/nu15030493] [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: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
T1Rs are expressed in solitary chemosensory cells of the upper airway where they detect apical glucose levels and repress bitter taste receptor Ca2+ signaling pathways. Microbial growth leads to a decrease in apical glucose levels. T1Rs detect this change and liberate bitter taste receptor signaling, initiating an innate immune response to both kill and expel pathogens through releasing antimicrobial peptides and increasing nitric oxide production and ciliary beat frequency. However, chronic inflammation due to disease, smoking, or viral infections causes a remodeling of the epithelial airway. The resulting squamous metaplasia causes a loss of multi-ciliated cells and solitary chemosensory cells, replaced by basal epithelial cells. To understand how T1R function is altered during disease, we used basal epithelial cells as a model to study the function of T1R3 on Ca2+ signaling dynamics. We found that both T1R1 and T1R3 detect amino acids and signal via cAMP, increasing the responsiveness of the cells to Ca2+ signaling stimuli. Either knocking down T1R1/3 or treating wild-type cells with MEM amino acids caused a reduction in ER Ca2+ content through a non-cAMP signaled pathway. Treatment with amino acids led to a reduction in downstream denatonium-induced Ca2+-signaled caspase activity. Thus, amino acids may be used to reduce unwanted apoptosis signaling in treatments containing bitter compounds.
Collapse
|
8
|
Ponnusamy V, Subramanian G, Muthuswamy K, Shanmugamprema D, Krishnan V, Velusamy T, Subramaniam S. Genetic variation in sweet taste receptors and a mechanistic perspective on sweet and fat taste sensation in the context of obesity. Obes Rev 2022; 23:e13512. [PMID: 36282093 DOI: 10.1111/obr.13512] [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] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
Abstract
Taste sensation enables humans to make nutritionally important decisions such as food preference and consumption. It functions as deterministic factors for unpropitious eating behavior, leading to overweight and obesity. The hedonistic feeling on consumption of fat and sugar-rich meals, in particular, has a negative influence on health. In addition, impairment in the taste receptors alters the downstream signaling of taste transduction pathway. Hence, genetic polymorphism in typical taste receptors is a predictor of taste sensitivity variance across individuals. The present review summarizes the effect of a single nucleotide polymorphism (SNP) in sweet taste receptors (T1R2/T1R3) on taste perception among individuals of various body mass index (BMI). Furthermore, in the context of obesity, we discussed the possibility of crosstalk between fat and sweet receptors as well as taste dysfunction in diseased individuals. In overall, a greater understanding of the physiological relationship between taste receptors, altered taste sensitivity, and genetic polymorphisms should lead to more effective obesity prevention approaches.
Collapse
Affiliation(s)
- Vinithra Ponnusamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Gowtham Subramanian
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Karthi Muthuswamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Deepankumar Shanmugamprema
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Vasanth Krishnan
- Molecular Biology Laboratory, Department of Botany, School of Life Sciences, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Thirunavukkarasu Velusamy
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Selvakumar Subramaniam
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| |
Collapse
|
9
|
Reprogramming cultured human fungiform (HBO) taste cells into neuron-like cells through in vitro induction. In Vitro Cell Dev Biol Anim 2022; 58:817-829. [DOI: 10.1007/s11626-022-00724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/23/2022] [Indexed: 11/05/2022]
|
10
|
Carey RM, Hariri BM, Adappa ND, Palmer JN, Lee RJ. HSP90 Modulates T2R Bitter Taste Receptor Nitric Oxide Production and Innate Immune Responses in Human Airway Epithelial Cells and Macrophages. Cells 2022; 11:1478. [PMID: 35563784 PMCID: PMC9101439 DOI: 10.3390/cells11091478] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Bitter taste receptors (T2Rs) are G protein-coupled receptors (GPCRs) expressed in various cell types including ciliated airway epithelial cells and macrophages. T2Rs in these two innate immune cell types are activated by bitter products, including those secreted by Pseudomonas aeruginosa, leading to Ca2+-dependent activation of endothelial nitric oxide (NO) synthase (eNOS). NO enhances mucociliary clearance and has direct antibacterial effects in ciliated epithelial cells. NO also increases phagocytosis by macrophages. Using biochemistry and live-cell imaging, we explored the role of heat shock protein 90 (HSP90) in regulating T2R-dependent NO pathways in primary sinonasal epithelial cells, primary monocyte-derived macrophages, and a human bronchiolar cell line (H441). Immunofluorescence showed that H441 cells express eNOS and T2Rs and that the bitter agonist denatonium benzoate activates NO production in a Ca2+- and HSP90-dependent manner in cells grown either as submerged cultures or at the air-liquid interface. In primary sinonasal epithelial cells, we determined that HSP90 inhibition reduces T2R-stimulated NO production and ciliary beating, which likely limits pathogen clearance. In primary monocyte-derived macrophages, we found that HSP-90 is integral to T2R-stimulated NO production and phagocytosis of FITC-labeled Escherichia coli and pHrodo-Staphylococcus aureus. Our study demonstrates that HSP90 serves as an innate immune modulator by regulating NO production downstream of T2R signaling by augmenting eNOS activation without impairing upstream Ca2+ signaling. These findings suggest that HSP90 plays an important role in airway antibacterial innate immunity and may be an important target in airway diseases such as chronic rhinosinusitis, asthma, or cystic fibrosis.
Collapse
Affiliation(s)
- Ryan M. Carey
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - Benjamin M. Hariri
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - Nithin D. Adappa
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - James N. Palmer
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
| | - Robert J. Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.M.H.); (N.D.A.); (J.N.P.)
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Servant NB, Williams ME, Brust PF, Tang H, Wong MS, Chen Q, Lebl-Rinnova M, Adamski-Werner SL, Tachdjian C, Servant G. A Dynamic Mass Redistribution Assay for the Human Sweet Taste Receptor Uncovers G-Protein Dependent Biased Ligands. Front Pharmacol 2022; 13:832529. [PMID: 35250580 PMCID: PMC8893300 DOI: 10.3389/fphar.2022.832529] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/26/2022] Open
Abstract
The sweet taste receptor is rather unique, recognizing a diverse repertoire of natural or synthetic ligands, with a surprisingly large structural diversity, and with potencies stretching over more than six orders of magnitude. Yet, it is not clear if different cell-based assays can faithfully report the relative potencies and efficacies of these molecules. Indeed, up to now, sweet taste receptor agonists have been almost exclusively characterized using cell-based assays developed with overexpressed and promiscuous G proteins. This non-physiological coupling has allowed the quantification of receptor activity via phospholipase C activation and calcium mobilization measurements in heterologous cells on a FLIPR system, for example. Here, we developed a novel assay for the human sweet taste receptor where endogenous G proteins and signaling pathways are recruited by the activated receptor. The effects of several sweet taste receptor agonists and other types of modulators were recorded by measuring changes in dynamic mass redistribution (DMR) using an Epic® reader. Potency and efficacy values obtained in the DMR assay were compared to those results obtained with the classical FLIPR assay. Results demonstrate that for some ligands, the two assay systems provide similar information. However, a clear bias for the FLIPR assay was observed for one third of the agonists evaluated, suggesting that the use of non-physiological coupling may influence the potency and efficacy of sweet taste receptor ligands. Replacing the promiscuous G protein with a chimeric G protein containing the C-terminal tail 25 residues of the physiologically relevant G protein subunit Gαgustducin reduced or abrogated bias.
Collapse
|
12
|
Xie F, Shen J, Liu T, Zhou M, Johnston LJ, Zhao J, Zhang H, Ma X. Sensation of dietary nutrients by gut taste receptors and its mechanisms. Crit Rev Food Sci Nutr 2022; 63:5594-5607. [PMID: 34978220 DOI: 10.1080/10408398.2021.2021388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Nutrients sensing is crucial for fundamental metabolism and physiological functions, and it is also an essential component for maintaining body homeostasis. Traditionally, basic taste receptors exist in oral cavity to sense sour, sweet, bitter, umami, salty and et al. Recent studies indicate that gut can sense the composition of nutrients by activating relevant taste receptors, thereby exerting specific direct or indirect effects. Gut taste receptors, also named as intestinal nutrition receptors, including at least bitter, sweet and umami receptors, have been considered to be activated by certain nutrients and participate in important intestinal physiological activities such as eating behavior, intestinal motility, nutrient absorption and metabolism. Additionally, gut taste receptors can regulate appetite and body weight, as well as maintain homeostasis via targeting hormone secretion or regulating the gut microbiota. On the other hand, malfunction of gut taste receptors may lead to digestive disorders, and then result in obesity, type 2 diabetes and gastrointestinal diseases. At present, researchers have confirmed that the brain-gut axis may play indispensable roles in these diseases via the secretion of brain-gut peptides, but the mechanism is still not clear. In this review, we summarize the current observation of knowledge in gut taste systems in order to shed light on revealing their important nutritional functions and promoting clinical implications.
Collapse
Affiliation(s)
- Fei Xie
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiakun Shen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tianyi Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Min Zhou
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J Johnston
- West Central Research & Outreach Center, University of Minnesota, Morris, Minnesota, USA
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Abstract
Bitter taste-sensing type 2 receptors (TAS2Rs or T2Rs), belonging to the subgroup of family A G-protein coupled receptors (GPCRs), are of crucial importance in the perception of bitterness. Although in the first instance, TAS2Rs were considered to be exclusively distributed in the apical microvilli of taste bud cells, numerous studies have detected these sensory receptor proteins in several extra-oral tissues, such as in pancreatic or ovarian tissues, as well as in their corresponding malignancies. Critical points of extra-oral TAS2Rs biology, such as their structure, roles, signaling transduction pathways, extensive mutational polymorphism, and molecular evolution, have been currently broadly studied. The TAS2R cascade, for instance, has been recently considered to be a pivotal modulator of a number of (patho)physiological processes, including adipogenesis or carcinogenesis. The latest advances in taste receptor biology further raise the possibility of utilizing TAS2Rs as a therapeutic target or as an informative index to predict treatment responses in various disorders. Thus, the focus of this review is to provide an update on the expression and molecular basis of TAS2Rs functions in distinct extra-oral tissues in health and disease. We shall also discuss the therapeutic potential of novel TAS2Rs targets, which are appealing due to their ligand selectivity, expression pattern, or pharmacological profiles.
Collapse
Affiliation(s)
- Kamila Tuzim
- Department of Clinical Pathomorphology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Agnieszka Korolczuk
- Department of Clinical Pathomorphology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| |
Collapse
|
14
|
Tommonaro G, Morelli CF, Rabuffetti M, Nicolaus B, De Prisco R, Iodice C, Speranza G. Determination of flavor-potentiating compounds in different Italian tomato varieties. J Food Biochem 2021; 45:e13736. [PMID: 33870530 DOI: 10.1111/jfbc.13736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/05/2021] [Accepted: 04/01/2021] [Indexed: 01/07/2023]
Abstract
Umami taste, known as appetizing sensation, is mainly imparted by monosodium glutamate (MSG, the first identified umami factor) in synergistic combination with some 5' ribonucleotides such as inosine 5'-monophosphate, IMP, guanosine 5'-monophosphate, GMP, and adenosine 5'-monophoshate, AMP. The level of free glutamic acid in tomatoes is higher than in other vegetables or fruits and increases with ripening and industrial processing. In addition, due to the presence of bioactive metabolites, tomatoes and tomato-based products are among the most consumed healthy food items. The levels of the major umami compounds of tomato, that is, glutamate and 5'-ribonucleotides (GMP and AMP) were assessed in different parts (skin, outer flesh, and inner pulp) of known tomato varieties from southern Italy: San Marzano Originale, San Marzano 245, Black Tomato, Corbarino Corbara, Corbarino Nocera, and Superpomodoro (tomato hybrid). Such varieties were also investigated for their antioxidant properties through DMPD, DPPH, and ABTS assays, with San Marzano Originale showing the highest antioxidant power both in lipophilic and methanolic fractions. The concentration of umami compounds in tomato differs with the part of the fruit analyzed and is greatly dependent on the variety, being Corbarino Nocera the cultivar richest in glutamate and Superpomodoro in ribonucleotides. As for nutritional aspect, results confirm the great nutraceutical feature of San Marzano tomato, the most known variety used in industrial processes. PRACTICAL APPLICATIONS: This study was planned to develop a method to quantify the major umami compounds that strongly influence the organoleptic properties of many different tomato varieties. It is known that the sensory quality of fruits and vegetables is an important factor in consumer's choice. The analytical methods described here enabled the evaluation of the glutamate and 5'-ribonucleotides contents in six selected varieties of tomato from Campania region, and can be easily used to determine the sensory profile of commercial varieties, for example, those perceived as not very tasteful by consumers.
Collapse
Affiliation(s)
- Giuseppina Tommonaro
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | | | | | - Barbara Nicolaus
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Rocco De Prisco
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Carmine Iodice
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Giovanna Speranza
- Department of Chemistry, University of Milan, Milan, Italy.,Institute of Chemical Science and Technologies "G. Natta", CNR-SCITEC, Milan, Italy
| |
Collapse
|
15
|
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
|
16
|
Jiang J, Liu S, Qi L, Wei Q, Shi F. Activation of Ovarian Taste Receptors Inhibits Progesterone Production Potentially via NO/cGMP and Apoptotic Signaling. Endocrinology 2021; 162:6052298. [PMID: 33367902 DOI: 10.1210/endocr/bqaa240] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Indexed: 12/25/2022]
Abstract
Taste receptors are not only expressed in the taste buds, but also in other nongustatory tissues, including the reproductive system. Taste receptors can be activated by various tastants, thereby exerting relatively physiologic functions. The aim of this study was to investigate the effects and potential mechanisms underlying ovarian taste receptor activation on progesterone production using saccharin sodium as the receptor agonist in a pseudopregnant rat model. Taste 1 receptor member 2 (TAS1R2) and taste 2 receptor member 31 (TAS2R31) were demonstrated to be abundantly expressed in the corpora lutea of rats, and intraperitoneal injection of saccharin sodium can activate both of them and initiate their downstream signaling cascades. The activation of these ovarian taste receptors promoted nitric oxide (NO) production via endothelial nitric oxide synthase (eNOS). NO production then increased ovarian cyclic guanosine 3',5'-monophosphate (cGMP) levels, which, in turn, decreased ovarian cyclic adenosine 3',5'-monophosphate levels. In addition, the activation of ovarian taste receptors induced apoptosis, possibly through NO and mitogen-activated protein kinase signaling. As a result, the activation of ovarian taste receptors reduced the protein expression of steroidogenesis-related factors, causing the inhibition of ovarian progesterone production. In summary, our data suggest that the activation of ovarian taste receptors inhibits progesterone production in pseudopregnant rats, potentially via NO/cGMP and apoptotic signaling.
Collapse
Affiliation(s)
- Jingle Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Siyi Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lina Qi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
17
|
Behrens M. Pharmacology of TAS1R2/TAS1R3 Receptors and Sweet Taste. Handb Exp Pharmacol 2021; 275:155-175. [PMID: 33582884 DOI: 10.1007/164_2021_438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The detection of energy-rich sweet food items has been important for our survival during evolution, however, in light of the changing lifestyles in industrialized and developing countries our natural sweet preference is causing considerable problems. Hence, it is even more important to understand how our sense of sweetness works, and perhaps even, how we may deceive it for our own benefit. This chapter summarizes current knowledge about sweet tastants and sweet taste modulators on the compound side as well as insights into the structure and function of the sweet taste receptor and the transduction of sweet signals. Moreover, methods to assess the activity of sweet substances in vivo and in vitro are compared and discussed.
Collapse
Affiliation(s)
- Maik Behrens
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany.
| |
Collapse
|
18
|
Abstract
Umami, the fifth taste, has been recognized as a legitimate taste modality only recently relative to the other tastes. Dozens of compounds from vastly different chemical classes elicit a savory (also called umami) taste. The prototypical umami substance glutamic acid or its salt monosodium glutamate (MSG) is present in numerous savory food sources or ingredients such as kombu (edible kelp), beans, soy sauce, tomatoes, cheeses, mushrooms, and certain meats and fish. Derivatives of glutamate (Glu), other amino acids, nucleotides, and small peptides can also elicit or modulate umami taste. In addition, many potent umami tasting compounds structurally unrelated to amino acids, nucleotides, and MSG have been either synthesized or discovered as naturally occurring in plants and other substances. Over the last 20 years several receptors have been suggested to mediate umami taste, including members of the metabotropic and ionotropic Glu receptor families, and more recently, the heterodimeric G protein-coupled receptor, T1R1/T1R3. Careful assessment of representative umami tasting molecules from several different chemical classes shows activation of T1R1/T1R3 with the expected rank order of potency in cell-based assays. Moreover, 5'-ribonucleotides, molecules known to enhance the savory note of Glu, considerably enhance the effect of MSG on T1R1/T1R3 in vitro. Binding sites are found on at least 4 distinct locations on T1R1/T1R3, explaining the propensity of the receptor to being activated or modulated by many structurally distinct compounds and these binding sites allosterically interact to modulate receptor activity. Activation of T1R1/T1R3 by all known umami substances evaluated and the receptor's pharmacological properties are sufficient to explain the basic human sensory experience of savory taste and it is therefore unlikely that other receptors are involved.
Collapse
|
19
|
Medapati MR, Bhagirath AY, Singh N, Chelikani P. Pharmacology of T2R Mediated Host-Microbe Interactions. Handb Exp Pharmacol 2021; 275:177-202. [PMID: 33580389 DOI: 10.1007/164_2021_435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bitter taste receptors (T2Rs) belong to the G protein-coupled receptor superfamily. Humans express 25 T2Rs that are known to detect several bitter compounds including bacterial quorum sensing molecules (QSM). Primarily found to be key receptors for bitter sensation T2Rs are known to play an important role in mediating innate immune responses in oral and extraoral tissues. Several studies have led to identification of Gram-negative and Gram-positive bacterial QSMs as agonists for T2Rs in airway epithelial cells and immune cells. However, the pharmacological characterization for many of the QSM-T2R interactions remains poorly defined. In this chapter, we discuss the extraoral roles including localization of T2Rs in extracellular vesicles, molecular pharmacology of QSM-T2R interactions, role of T2Rs in mediating innate immune responses, and some of the challenges in understanding T2R pharmacology.
Collapse
Affiliation(s)
- Manoj Reddy Medapati
- Manitoba Chemosensory Biology Research Group, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Anjali Y Bhagirath
- Manitoba Chemosensory Biology Research Group, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Nisha Singh
- Manitoba Chemosensory Biology Research Group, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Prashen Chelikani
- Manitoba Chemosensory Biology Research Group, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada.
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
20
|
Rohde K, Schamarek I, Blüher M. Consequences of Obesity on the Sense of Taste: Taste Buds as Treatment Targets? Diabetes Metab J 2020; 44:509-528. [PMID: 32431111 PMCID: PMC7453985 DOI: 10.4093/dmj.2020.0058] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/25/2020] [Indexed: 12/19/2022] Open
Abstract
Premature obesity-related mortality is caused by cardiovascular and pulmonary diseases, type 2 diabetes mellitus, physical disabilities, osteoarthritis, and certain types of cancer. Obesity is caused by a positive energy balance due to hyper-caloric nutrition, low physical activity, and energy expenditure. Overeating is partially driven by impaired homeostatic feedback of the peripheral energy status in obesity. However, food with its different qualities is a key driver for the reward driven hedonic feeding with tremendous consequences on calorie consumption. In addition to visual and olfactory cues, taste buds of the oral cavity process the earliest signals which affect the regulation of food intake, appetite and satiety. Therefore, taste buds may play a crucial role how food related signals are transmitted to the brain, particularly in priming the body for digestion during the cephalic phase. Indeed, obesity development is associated with a significant reduction in taste buds. Impaired taste bud sensitivity may play a causal role in the pathophysiology of obesity in children and adolescents. In addition, genetic variation in taste receptors has been linked to body weight regulation. This review discusses the importance of taste buds as contributing factors in the development of obesity and how obesity may affect the sense of taste, alterations in food preferences and eating behavior.
Collapse
Affiliation(s)
- Kerstin Rohde
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
| | - Imke Schamarek
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
| |
Collapse
|
21
|
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
|
22
|
Bloxham CJ, Foster SR, Thomas WG. A Bitter Taste in Your Heart. Front Physiol 2020; 11:431. [PMID: 32457649 PMCID: PMC7225360 DOI: 10.3389/fphys.2020.00431] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
The human genome contains ∼29 bitter taste receptors (T2Rs), which are responsible for detecting thousands of bitter ligands, including toxic and aversive compounds. This sentinel function varies between individuals and is underpinned by naturally occurring T2R polymorphisms, which have also been associated with disease. Recent studies have reported the expression of T2Rs and their downstream signaling components within non-gustatory tissues, including the heart. Though the precise role of T2Rs in the heart remains unclear, evidence points toward a role in cardiac contractility and overall vascular tone. In this review, we summarize the extra-oral expression of T2Rs, focusing on evidence for expression in heart; we speculate on the range of potential ligands that may activate them; we define the possible signaling pathways they activate; and we argue that their discovery in heart predicts an, as yet, unappreciated cardiac physiology.
Collapse
Affiliation(s)
- Conor J Bloxham
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Simon R Foster
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
23
|
Crowe MS, Wang H, Blakeney BA, Mahavadi S, Singh K, Murthy KS, Grider JR. Expression and function of umami receptors T1R1/T1R3 in gastric smooth muscle. Neurogastroenterol Motil 2020; 32:e13737. [PMID: 31721379 PMCID: PMC7008388 DOI: 10.1111/nmo.13737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/19/2019] [Accepted: 09/16/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND l-amino acids, such as monosodium glutamate (MSG), activate the umami receptor T1R1/T1R3. We previously showed increased peristalsis in response to activation of T1R1/T1R3 by MSG in mouse colon. However, the expression and function of these receptors in the different regions of the stomach are not clear. METHODS Mouse gastric smooth muscle cells (SMCs) were isolated and cultured in Dulbecco's Modified Eagle Medium. Expression of T1R1 and T1R3 was measured by RT-PCR and Western blot. The effect of MSG with and without inosine monophosphate (IMP, an allosteric activator of T1R1/T1R3) on acetylcholine (ACh)-induced contraction was measured in muscle strips and isolated SMCs by scanning micrometry. The effect of MSG with or without IMP on activation of G proteins and ACh-induced Ca2+ release was measured in SMCs. KEY RESULTS Monosodium glutamate inhibited ACh-induced contractions in muscle strips from both antrum and fundus and the effect of MSG was augmented by IMP; the effects were concentration-dependent and not affected by the nitric oxide synthase inhibitor, L-NNA, or tetrodotoxin suggesting a direct effect on SMCs. In isolated gastric SMCs, T1R1 and T1R3 transcripts and protein were identified. Addition of MSG with or without IMP inhibited ACh-induced Ca2+ release and muscle contraction; the effect on contraction was blocked by pertussis toxin suggesting activation of Gαi proteins. MSG in the presence of IMP selectively activated Gαi2 . CONCLUSIONS AND INFERENCES Umami receptors (T1R1/T1R3) are present on SMCs of the stomach, and activation of these receptors induces muscle relaxation by decreasing [Ca2+ ]i via Gαi2 .
Collapse
Affiliation(s)
- Molly S. Crowe
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| | - Hongxia Wang
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| | - Bryan A. Blakeney
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| | - Sunila Mahavadi
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| | - Kulpreet Singh
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| | - Karnam S. Murthy
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| | - John R. Grider
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences Virginia Commonwealth University Richmond VA USA
| |
Collapse
|
24
|
Roelse M, Wehrens R, Henquet MG, Witkamp RF, Hall RD, Jongsma MA. The Effect of Calcium Buffering and Calcium Sensor Type on the Sensitivity of an Array-Based Bitter Receptor Screening Assay. Chem Senses 2019; 44:497-505. [PMID: 31278864 PMCID: PMC7357244 DOI: 10.1093/chemse/bjz044] [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] [Indexed: 11/14/2022] Open
Abstract
The genetically encoded calcium sensor protein Cameleon YC3.6 has previously been applied for functional G protein-coupled receptor screening using receptor cell arrays. However, different types of sensors are available, with a wide range in [Ca2+] sensitivity, Hill coefficients, calcium binding domains, and fluorophores, which could potentially improve the performance of the assay. Here, we compared the responses of 3 structurally different calcium sensor proteins (Cameleon YC3.6, Nano140, and Twitch2B) simultaneously, on a single chip, at different cytosolic expression levels and in combination with 2 different bitter receptors, TAS2R8 and TAS2R14. Sensor concentrations were modified by varying the amount of calcium sensor DNA that was printed on the DNA arrays prior to reverse transfection. We found that ~2-fold lower concentrations of calcium sensor protein, by transfecting 4 times less sensor-coding DNA, resulted in more sensitive bitter responses. The best results were obtained with Twitch2B, where, relative to YC3.6 at the default DNA concentration, a 4-fold lower DNA concentration increased sensitivity 60-fold and signal strength 5- to 10-fold. Next, we compared the performance of YC3.6 and Twitch2B against an array with 11 different bitter taste receptors. We observed a 2- to 8-fold increase in sensitivity using Twitch2B compared with YC3.6. The bitter receptor arrays contained 300 spots and could be exposed to a series of 18 injections within 1 h resulting in 5400 measurements. These optimized sensor conditions provide a basis for enhancing receptomics calcium assays for receptors with poor Ca2+ signaling and will benefit future high-throughput receptomics experiments.
Collapse
Affiliation(s)
- Margriet Roelse
- BU Bioscience, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen, The Netherlands.,Laboratory of Plant Physiology, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen.,Nutritional Biology and Health, Wageningen University and Research, Stippeneng, WE Wageningen, The Netherlandsand
| | - Ron Wehrens
- BU Bioscience, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen, The Netherlands.,BU Biometris, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen
| | - Maurice Gl Henquet
- BU Bioscience, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen, The Netherlands
| | - Renger F Witkamp
- Nutritional Biology and Health, Wageningen University and Research, Stippeneng, WE Wageningen, The Netherlandsand
| | - Robert D Hall
- BU Bioscience, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen, The Netherlands.,Laboratory of Plant Physiology, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen
| | - Maarten A Jongsma
- BU Bioscience, Wageningen University and Research, Droevendaalsesteeg, PB Wageningen, The Netherlands
| |
Collapse
|
25
|
Role of Taste Receptors as Sentinels of Innate Immunity in the Upper Airway. J Pathog 2018; 2018:9541987. [PMID: 30363975 PMCID: PMC6188595 DOI: 10.1155/2018/9541987] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/01/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022] Open
Abstract
Evidence is emerging that shows taste receptors serve functions outside of taste sensation of the tongue. Taste receptors have been found in tissue across the human body, including the gastrointestinal tract, bladder, brain, and airway. These extraoral taste receptors appear to be important in modulating the innate immune response through detection of pathogens. This review discusses taste receptor signaling, focusing on the G-protein-coupled receptors that detect bitter and sweet compounds in the upper airway epithelium. Emphasis is given to recent studies which link the physiology of sinonasal taste receptors to clinical manifestation of upper airway disease.
Collapse
|
26
|
Freund JR, Mansfield CJ, Doghramji LJ, Adappa ND, Palmer JN, Kennedy DW, Reed DR, Jiang P, Lee RJ. Activation of airway epithelial bitter taste receptors by Pseudomonas aeruginosa quinolones modulates calcium, cyclic-AMP, and nitric oxide signaling. J Biol Chem 2018; 293:9824-9840. [PMID: 29748385 DOI: 10.1074/jbc.ra117.001005] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
Bitter taste receptors (taste family 2 bitter receptor proteins; T2Rs), discovered in many tissues outside the tongue, have recently become potential therapeutic targets. We have shown previously that airway epithelial cells express several T2Rs that activate innate immune responses that may be important for treatment of airway diseases such as chronic rhinosinusitis. It is imperative to more clearly understand what compounds activate airway T2Rs as well as their full range of functions. T2R isoforms in airway motile cilia (T2R4, -14, -16, and -38) produce bactericidal levels of nitric oxide (NO) that also increase ciliary beating, promoting clearance of mucus and trapped pathogens. Bacterial quorum-sensing acyl-homoserine lactones activate T2Rs and stimulate these responses in primary airway cells. Quinolones are another type of quorum-sensing molecule used by Pseudomonas aeruginosa To elucidate whether bacterial quinolones activate airway T2Rs, we analyzed calcium, cAMP, and NO dynamics using a combination of fluorescent indicator dyes and FRET-based protein biosensors. T2R-transfected HEK293T cells, several lung epithelial cell lines, and primary sinonasal cells grown and differentiated at the air-liquid interface were tested with 2-heptyl-3-hydroxy-4-quinolone (known as Pseudomonas quinolone signal; PQS), 2,4-dihydroxyquinolone, and 4-hydroxy-2-heptylquinolone (HHQ). In HEK293T cells, PQS activated T2R4, -16, and -38, whereas HHQ activated T2R14. 2,4-Dihydroxyquinolone had no effect. PQS and HHQ increased calcium and decreased both baseline and stimulated cAMP levels in cultured and primary airway cells. In primary cells, PQS and HHQ activated levels of NO synthesis previously shown to be bactericidal. This study suggests that airway T2R-mediated immune responses are activated by bacterial quinolones as well as acyl-homoserine lactones.
Collapse
Affiliation(s)
- Jenna R Freund
- From the Departments of Otorhinolaryngology-Head and Neck Surgery and
| | | | | | - Nithin D Adappa
- From the Departments of Otorhinolaryngology-Head and Neck Surgery and
| | - James N Palmer
- From the Departments of Otorhinolaryngology-Head and Neck Surgery and
| | - David W Kennedy
- From the Departments of Otorhinolaryngology-Head and Neck Surgery and
| | - Danielle R Reed
- the Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Peihua Jiang
- the Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Robert J Lee
- From the Departments of Otorhinolaryngology-Head and Neck Surgery and .,Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104 and
| |
Collapse
|
27
|
Young RL, Lumsden AL, Martin AM, Schober G, Pezos N, Thazhath SS, Isaacs NJ, Cvijanovic N, Sun EWL, Wu T, Rayner CK, Nguyen NQ, Fontgalland DD, Rabbitt P, Hollington P, Sposato L, Due SL, Wattchow DA, Liou AP, Jackson VM, Keating DJ. Augmented capacity for peripheral serotonin release in human obesity. Int J Obes (Lond) 2018; 42:1880-1889. [PMID: 29568107 DOI: 10.1038/s41366-018-0047-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVES Evidence from animal studies highlights an important role for serotonin (5-HT), derived from gut enterochromaffin (EC) cells, in regulating hepatic glucose production, lipolysis and thermogenesis, and promoting obesity and dysglycemia. Evidence in humans is limited, although elevated plasma 5-HT concentrations are linked to obesity. SUBJECTS/METHODS We assessed (i) plasma 5-HT concentrations before and during intraduodenal glucose infusion (4 kcal/min for 30 min) in non-diabetic obese (BMI 44 ± 4 kg/m2, N = 14) and control (BMI 24 ± 1 kg/m2, N = 10) subjects, (ii) functional activation of duodenal EC cells (immunodetection of phospho-extracellular related-kinase, pERK) in response to glucose, and in separate subjects, (iii) expression of tryptophan hydroxylase-1 (TPH1) in duodenum and colon (N = 39), and (iv) 5-HT content in primary EC cells from these regions (N = 85). RESULTS Plasma 5-HT was twofold higher in obese than control responders prior to (P = 0.025), and during (iAUC, P = 0.009), intraduodenal glucose infusion, and related positively to BMI (R2 = 0.334, P = 0.003) and HbA1c (R2 = 0.508, P = 0.009). The density of EC cells in the duodenum was twofold higher at baseline in obese subjects than controls (P = 0.023), with twofold more EC cells activated by glucose infusion in the obese (EC cells co-expressing 5-HT and pERK, P = 0.001), while the 5-HT content of EC cells in duodenum and colon was similar; TPH1 expression was 1.4-fold higher in the duodenum of obese subjects (P = 0.044), and related positively to BMI (R2 = 0.310, P = 0.031). CONCLUSIONS Human obesity is characterized by an increased capacity to produce and release 5-HT from the proximal small intestine, which is strongly linked to higher body mass, and glycemic control. Gut-derived 5-HT is likely to be an important driver of pathogenesis in human obesity and dysglycemia.
Collapse
Affiliation(s)
- Richard L Young
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Amanda L Lumsden
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia
| | - Alyce M Martin
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia
| | - Gudrun Schober
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Nektaria Pezos
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Sony S Thazhath
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Nicole J Isaacs
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Nada Cvijanovic
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Emily W L Sun
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia
| | - Tongzhi Wu
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christopher K Rayner
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Nam Q Nguyen
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.,NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Dayan de Fontgalland
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Philippa Rabbitt
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Paul Hollington
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Luigi Sposato
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Steven L Due
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - David A Wattchow
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Alice P Liou
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, 02139, USA
| | - V Margaret Jackson
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, 02139, USA
| | - Damien J Keating
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia. .,Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
28
|
Jiang J, Qi L, Wei Q, Shi F. Effects of daily exposure to saccharin sodium and rebaudioside A on the ovarian cycle and steroidogenesis in rats. Reprod Toxicol 2018; 76:35-45. [DOI: 10.1016/j.reprotox.2017.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 10/18/2022]
|
29
|
Molecular mechanism of sweetness sensation. Physiol Behav 2016; 164:453-463. [DOI: 10.1016/j.physbeh.2016.03.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 11/17/2022]
|
30
|
Lossow K, Hübner S, Roudnitzky N, Slack JP, Pollastro F, Behrens M, Meyerhof W. Comprehensive Analysis of Mouse Bitter Taste Receptors Reveals Different Molecular Receptive Ranges for Orthologous Receptors in Mice and Humans. J Biol Chem 2016; 291:15358-77. [PMID: 27226572 DOI: 10.1074/jbc.m116.718544] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 11/06/2022] Open
Abstract
One key to animal survival is the detection and avoidance of potentially harmful compounds by their bitter taste. Variable numbers of taste 2 receptor genes expressed in the gustatory end organs enable bony vertebrates (Euteleostomi) to recognize numerous bitter chemicals. It is believed that the receptive ranges of bitter taste receptor repertoires match the profiles of bitter chemicals that the species encounter in their diets. Human and mouse genomes contain pairs of orthologous bitter receptor genes that have been conserved throughout evolution. Moreover, expansions in both lineages generated species-specific sets of bitter taste receptor genes. It is assumed that the orthologous bitter taste receptor genes mediate the recognition of bitter toxins relevant for both species, whereas the lineage-specific receptors enable the detection of substances differently encountered by mice and humans. By challenging 34 mouse bitter taste receptors with 128 prototypical bitter substances in a heterologous expression system, we identified cognate compounds for 21 receptors, 19 of which were previously orphan receptors. We have demonstrated that mouse taste 2 receptors, like their human counterparts, vary greatly in their breadth of tuning, ranging from very broadly to extremely narrowly tuned receptors. However, when compared with humans, mice possess fewer broadly tuned receptors and an elevated number of narrowly tuned receptors, supporting the idea that a large receptor repertoire is the basis for the evolution of specialized receptors. Moreover, we have demonstrated that sequence-orthologous bitter taste receptors have distinct agonist profiles. Species-specific gene expansions have enabled further diversification of bitter substance recognition spectra.
Collapse
Affiliation(s)
- Kristina Lossow
- From the Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Sandra Hübner
- From the Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Natacha Roudnitzky
- From the Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Jay P Slack
- the Givaudan Flavors Corporation, Cincinnati, Ohio 45216, and
| | - Federica Pollastro
- the Department of Drug Sciences, University of Eastern Piemonte, 28100 Novara, Italy
| | - Maik Behrens
- From the Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany,
| | - Wolfgang Meyerhof
- From the Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| |
Collapse
|
31
|
Comparing Class A GPCRs to bitter taste receptors: Structural motifs, ligand interactions and agonist-to-antagonist ratios. Methods Cell Biol 2015; 132:401-27. [PMID: 26928553 DOI: 10.1016/bs.mcb.2015.10.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) are seven transmembrane (TM) proteins that play a key role in human physiology. The GPCR superfamily comprises about 800 members, classified into several classes, with rhodopsin-like Class A being the largest and most studied thus far. A huge component of the human repertoire consists of the chemosensory GPCRs, including ∼400 odorant receptors, 25 bitter taste receptors (TAS2Rs), which are thought to guard the organism from consuming poisons, and sweet and umami TAS1R heteromers, which indicate the nutritive value of food. The location of the binding site of TAS2Rs is similar to that of Class A GPCRs. However, most of the known bitter ligands are agonists, with only a few antagonists documented thus far. The agonist-to-antagonist ratios of Class A GPCRs vary, but in general are much lower than for TAS2Rs. For a set of well-studied GPCRs, a gradual change in agonists-to-antagonists ratios is observed when comparing low (10 μM)- and high (10 nM)-affinity ligand sets from ChEMBL and the DrugBank set of drugs. This shift reflects pharmaceutical bias toward the therapeutically desirable pharmacology for each of these GPCRs, while the 10 μM sets possibly represent the native tendency of the receptors toward either agonists or antagonists. Analyzing ligand-GPCR interactions in 56 X-ray structures representative of currently available structural data, we find that the N-terminus, TM1 and TM2 are more involved in binding of antagonists than of agonists. On the other hand, ECL2 tends to be more involved in binding of agonists. This is of interest, since TAS2Rs harbor variations on the typical Class A sequence motifs, including the absence of the ECL2-TM3 disulfide bridge. This suggests an alternative mode of regulation of conformational states for TAS2Rs, with potentially less stabilized inactive state. The comparison of TAS2Rs and Class A GPCRs structural features and the pharmacology of the their ligands highlights the intricacies of GPCR architecture and provides a framework for rational design of new ligands.
Collapse
|
32
|
Avau B, Rotondo A, Thijs T, Andrews CN, Janssen P, Tack J, Depoortere I. Targeting extra-oral bitter taste receptors modulates gastrointestinal motility with effects on satiation. Sci Rep 2015; 5:15985. [PMID: 26541810 PMCID: PMC4635351 DOI: 10.1038/srep15985] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/06/2015] [Indexed: 01/01/2023] Open
Abstract
Bitter taste receptors (TAS2Rs) are present in extra-oral tissues, including gut endocrine cells. This study explored the presence and mechanism of action of TAS2R agonists on gut smooth muscle in vitro and investigated functional effects of intra-gastric administration of TAS2R agonists on gastric motility and satiation. TAS2Rs and taste signalling elements were expressed in smooth muscle tissue along the mouse gut and in human gastric smooth muscle cells (hGSMC). Bitter tastants induced concentration and region-dependent contractility changes in mouse intestinal muscle strips. Contractions induced by denatonium benzoate (DB) in gastric fundus were mediated via increases in intracellular Ca2+ release and extracellular Ca2+-influx, partially masked by a hyperpolarizing K+-efflux. Intra-gastric administration of DB in mice induced a TAS2R-dependent delay in gastric emptying. In hGSMC, bitter compounds evoked Ca2+-rises and increased ERK-phosphorylation. Healthy volunteers showed an impaired fundic relaxation in response to nutrient infusion and a decreased nutrient volume tolerance and increased satiation during an oral nutrient challenge test after intra-gastric DB administration. These findings suggest a potential role for intestinal TAS2Rs as therapeutic targets to alter gastrointestinal motility and hence to interfere with hunger signalling.
Collapse
Affiliation(s)
- Bert Avau
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Alessandra Rotondo
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Theo Thijs
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Christopher N Andrews
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Pieter Janssen
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Lee RJ, Cohen NA. Sinonasal solitary chemosensory cells "taste" the upper respiratory environment to regulate innate immunity. Am J Rhinol Allergy 2015; 28:366-73. [PMID: 25198020 DOI: 10.2500/ajra.2014.28.4077] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND It is not fully understood how sinonasal epithelial cells detect the presence of pathogens and activate innate defense responses necessary for protecting the upper airway from infection. One mechanism is through bitter taste receptors (T2Rs), which are expressed in the sinonasal cavity. One T2R isoform, T2R38, is expressed in ciliated cells and detects quorum-sensing molecules from gram-negative bacteria, activating antimicrobial nitric oxide production. More recent studies have examined the role of T2Rs expressed in a sinonasal cell type that has only recently been identified in humans, the solitary chemosensory cell (SCC). We sought to provide an overview of SCCs and taste receptor function in human sinonasal defense as well as implications for chronic rhinosinusitis (CRS). METHODS A literature review of the current knowledge of SCCs and taste receptors in sinonasal physiology and CRS was conducted. RESULTS Human sinonasal SCCs express both bitter T2R and sweet T1R2/3 receptors. Activation of SCC T2Rs activates a calcium signal that propagates to the surrounding epithelial cells and causes secretion of antimicrobial peptides. T1R2/3 sweet receptor activation by physiological airway surface liquid (ASL) glucose concentrations attenuates the T2R response, likely as a mechanism to prevent full activation of the T2R pathway except during times of infection, when pathogens may consume ASL glucose and reduce its concentration. CONCLUSION SCCs appear to be important mediators of upper airway innate immunity, as the SCC T2Rs regulate antimicrobial peptide secretion, but further study is needed to determine the specific T2R isoforms involved as well as whether polymorphisms in these isoforms affect susceptibility to infection or patient outcomes in CRS. The inhibitory role of T1R2/3 sweet receptor suggests that T1R2/3 blockers may have therapeutic potential in some CRS patients, particularly those with diabetes mellitus. However, further clinical study of the relationship between infection and T1R2/3 genotype is required.
Collapse
Affiliation(s)
- Robert J Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
34
|
Lee RJ, Cohen NA. Taste receptors in innate immunity. Cell Mol Life Sci 2015; 72:217-36. [PMID: 25323130 PMCID: PMC4286424 DOI: 10.1007/s00018-014-1736-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/11/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023]
Abstract
Taste receptors were first identified on the tongue, where they initiate a signaling pathway that communicates information to the brain about the nutrient content or potential toxicity of ingested foods. However, recent research has shown that taste receptors are also expressed in a myriad of other tissues, from the airway and gastrointestinal epithelia to the pancreas and brain. The functions of many of these extraoral taste receptors remain unknown, but emerging evidence suggests that bitter and sweet taste receptors in the airway are important sentinels of innate immunity. This review discusses taste receptor signaling, focusing on the G-protein-coupled receptors that detect bitter, sweet, and savory tastes, followed by an overview of extraoral taste receptors and in-depth discussion of studies demonstrating the roles of taste receptors in airway innate immunity. Future research on extraoral taste receptors has significant potential for identification of novel immune mechanisms and insights into host-pathogen interactions.
Collapse
Affiliation(s)
- Robert J. Lee
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5th floor, Philadelphia, PA 19104 USA
| | - Noam A. Cohen
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5th floor, Philadelphia, PA 19104 USA
- Philadelphia Veterans Affairs Medical Center Surgical Services, 3900 Woodland Ave, Philadelphia, PA 19104 USA
| |
Collapse
|
35
|
Foster SR, Blank K, Hoe LES, Behrens M, Meyerhof W, Peart JN, Thomas WG. Bitter taste receptor agonists elicit G‐protein‐dependent negative inotropy in the murine heart. FASEB J 2014; 28:4497-508. [DOI: 10.1096/fj.14-256305] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Simon R. Foster
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Kristina Blank
- Department of Molecular GeneticsGerman Institute of Human Nutrition (DIfE) Potsdam‐RehbrückeNuthetalGermany
| | - Louise E. See Hoe
- Griffith Health InstituteGriffith UniversityGold CoastQueenslandAustralia
| | - Maik Behrens
- Department of Molecular GeneticsGerman Institute of Human Nutrition (DIfE) Potsdam‐RehbrückeNuthetalGermany
| | - Wolfgang Meyerhof
- Department of Molecular GeneticsGerman Institute of Human Nutrition (DIfE) Potsdam‐RehbrückeNuthetalGermany
| | - Jason N. Peart
- Griffith Health InstituteGriffith UniversityGold CoastQueenslandAustralia
| | - Walter G. Thomas
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
36
|
Sawano S, Seto E, Mori T, Hayashi Y. G-Protein-Dependent and -Independent Pathways in Denatonium Signal Transduction. Biosci Biotechnol Biochem 2014; 69:1643-51. [PMID: 16195580 DOI: 10.1271/bbb.69.1643] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To clarify the involvement of G protein in denatonium signal transduction, we carried out a whole-cell patch-clamp analysis with isolated taste cells in mice. Two different responses were observed by applying GDP-beta-S, a G-protein inhibitor. One response to denatonium was reduced by GDP-beta-S (G-protein-dependent), whereas the other was not affected (G-protein-independent). These different patterns were also observed by concurrently inhibiting the phospholipase C beta2 and phosphodiesterase pathways via G protein. These data suggest dual, G-protein-dependent and -independent mechanisms for denatonium. Moreover, the denatonium responses were not attenuated by singly inhibiting the phospholipase C beta2 or phosphodiesterase pathway, implying that both pathways were involved in G-protein-dependent transduction. In the G-protein-independent cells, the response was abolished by the depletion of calcium ions within the intracellular store. These results suggest that Ca2+ release from the intracellular store is an important factor. Our data demonstrate multiple transduction pathways for denatonium in mammalian taste cells.
Collapse
Affiliation(s)
- Shoko Sawano
- Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | | | | | | |
Collapse
|
37
|
Lee RJ, Chen B, Redding KM, Margolskee RF, Cohen NA. Mouse nasal epithelial innate immune responses to Pseudomonas aeruginosa quorum-sensing molecules require taste signaling components. Innate Immun 2013; 20:606-17. [PMID: 24045336 DOI: 10.1177/1753425913503386] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 08/02/2013] [Indexed: 11/17/2022] Open
Abstract
We previously observed that the human bitter taste receptor T2R38 is an important component of upper respiratory innate defense because it detects acyl homoserine lactone (AHL) quorum-sensing molecules secreted by Gram-negative bacteria. T2R38 activation in human sinonasal epithelial cells stimulates calcium and NO signals that increase mucociliary clearance, the major physical respiratory defense against inhaled pathogens. While mice do not have a clear T2R38 ortholog, they do have bitter taste receptors capable of responding to T2R38 agonists, suggesting that T2R-mediated innate immune mechanisms may be conserved in mice. We examined whether AHLs activate calcium and NO signaling in mouse nasal epithelial cells, and utilized pharmacology, as well as cells from knockout mice lacking important components of canonical taste signal transduction pathways, to determine if AHL-stimulated responses require taste signaling molecules. We found that AHLs stimulate calcium-dependent NO production that increases mucociliary clearance and thus likely serves an innate immune role against Gram-negative bacteria. These responses require PLCβ2 and TRPM5 taste signaling components, but not α-gustducin. These data suggest the mouse may be a useful model for further studies of T2R-mediated innate immunity.
Collapse
Affiliation(s)
- Robert J Lee
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Bei Chen
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin M Redding
- Monell Chemical Senses Center and Philadelphia VA Medical Center Surgical Services, Philadelphia, PA, USA
| | - Robert F Margolskee
- Monell Chemical Senses Center and Philadelphia VA Medical Center Surgical Services, Philadelphia, PA, USA
| | - Noam A Cohen
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA Surgical Services, Philadelphia Veterans Administration Medical Center, Philadelphia, PA, USA
| |
Collapse
|
38
|
Liu Z, Fenech C, Cadiou H, Grall S, Tili E, Laugerette F, Wiencis A, Grosmaitre X, Montmayeur JP. Identification of new binding partners of the chemosensory signaling protein Gγ13 expressed in taste and olfactory sensory cells. Front Cell Neurosci 2012; 6:26. [PMID: 22737109 PMCID: PMC3380295 DOI: 10.3389/fncel.2012.00026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 05/31/2012] [Indexed: 01/19/2023] Open
Abstract
Tastant detection in the oral cavity involves selective receptors localized at the apical extremity of a subset of specialized taste bud cells called taste receptor cells (TRCs). The identification of the genes coding for the taste receptors involved in this process have greatly improved our understanding of the molecular mechanisms underlying detection. However, how these receptors signal in TRCs, and whether the components of the signaling cascades interact with each other or are organized in complexes is mostly unexplored. Here we report on the identification of three new binding partners for the mouse G protein gamma 13 subunit (Gγ13), a component of the bitter taste receptors signaling cascade. For two of these Gγ13 associated proteins, namely GOPC and MPDZ, we describe the expression in taste bud cells for the first time. Furthermore, we demonstrate by means of a yeast two-hybrid interaction assay that the C terminal PDZ binding motif of Gγ13 interacts with selected PDZ domains in these proteins. In the case of the PDZ domain-containing protein zona occludens-1 (ZO-1), a major component of the tight junction defining the boundary between the apical and baso-lateral region of TRCs, we identified the first PDZ domain as the site of strong interaction with Gγ13. This association was further confirmed by co-immunoprecipitation experiments in HEK 293 cells. In addition, we present immunohistological data supporting partial co-localization of GOPC, MPDZ, or ZO-1, and Gγ13 in taste buds cells. Finally, we extend this observation to olfactory sensory neurons (OSNs), another type of chemosensory cells known to express both ZO-1 and Gγ13. Taken together our results implicate these new interaction partners in the sub-cellular distribution of Gγ13 in olfactory and gustatory primary sensory cells.
Collapse
Affiliation(s)
- Zhenhui Liu
- Chemosensory Perception, Centre des Sciences du Goût et de l'Alimentation, UMR-6265 CNRS, UMR-1324 INRA Dijon, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bruen CM, O'Halloran F, Cashman KD, Giblin L. The effects of food components on hormonal signalling in gastrointestinal enteroendocrine cells. Food Funct 2012; 3:1131-43. [DOI: 10.1039/c2fo30086a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Young RL. Sensing via intestinal sweet taste pathways. Front Neurosci 2011; 5:23. [PMID: 21519398 PMCID: PMC3080736 DOI: 10.3389/fnins.2011.00023] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Accepted: 02/10/2011] [Indexed: 12/15/2022] Open
Abstract
The detection of nutrients in the gastrointestinal (GI) tract is of fundamental significance to the control of motility, glycemia and energy intake, and yet we barely know the most fundamental aspects of this process. This is in stark contrast to the mechanisms underlying the detection of lingual taste, which have been increasingly well characterized in recent years, and which provide an excellent starting point for characterizing nutrient detection in the intestine. This review focuses on the form and function of sweet taste transduction mechanisms identified in the intestinal tract; it does not focus on sensors for fatty acids or proteins. It examines the intestinal cell types equipped with sweet taste transduction molecules in animals and humans, their location, and potential signals that transduce the presence of nutrients to neural pathways involved in reflex control of GI motility.
Collapse
Affiliation(s)
- Richard L Young
- Discipline of Medicine, School of Medicine, University of Adelaide Adelaide, SA, Australia
| |
Collapse
|
41
|
Urwyler S. Allosteric modulation of family C G-protein-coupled receptors: from molecular insights to therapeutic perspectives. Pharmacol Rev 2011; 63:59-126. [PMID: 21228259 DOI: 10.1124/pr.109.002501] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Allosteric receptor modulation is an attractive concept in drug targeting because it offers important potential advantages over conventional orthosteric agonism or antagonism. Allosteric ligands modulate receptor function by binding to a site distinct from the recognition site for the endogenous agonist. They often have no effect on their own and therefore act only in conjunction with physiological receptor activation. This article reviews the current status of allosteric modulation at family C G-protein coupled receptors in the light of their specific structural features on the one hand and current concepts in receptor theory on the other hand. Family C G-protein-coupled receptors are characterized by a large extracellular domain containing the orthosteric agonist binding site known as the "venus flytrap module" because of its bilobal structure and the dynamics of its activation mechanism. Mutational analysis and chimeric constructs have revealed that allosteric modulators of the calcium-sensing, metabotropic glutamate and GABA(B) receptors bind to the seven transmembrane domain, through which they modify signal transduction after receptor activation. This is in contrast to taste-enhancing molecules, which bind to different parts of sweet and umami receptors. The complexity of interactions between orthosteric and allosteric ligands is revealed by a number of adequate biochemical and electrophysiological assay systems. Many allosteric family C GPCR modulators show in vivo efficacy in behavioral models for a variety of clinical indications. The positive allosteric calcium sensing receptor modulator cinacalcet is the first drug of this type to enter the market and therefore provides proof of principle in humans.
Collapse
Affiliation(s)
- Stephan Urwyler
- Department of Chemistry and Biochemistry, University of Berne, P/A Weissensteinweg 3, CH-3303 Jegenstorf, Berne, Switzerland.
| |
Collapse
|
42
|
Lioe HN, Selamat J, Yasuda M. Soy sauce and its umami taste: a link from the past to current situation. J Food Sci 2010; 75:R71-6. [PMID: 20492309 DOI: 10.1111/j.1750-3841.2010.01529.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Soy sauce taste has become a focus of umami taste research. Umami taste is a 5th basic taste, which is associated to a palatable and pleasurable taste of food. Soy sauce has been used as an umami seasoning since the ancient time in Asia. The complex fermentation process occurred to soy beans, as the raw material in the soy sauce production, gives a distinct delicious taste. The recent investigation on Japanese and Indonesian soy sauces revealed that this taste is primarily due to umami components which have molecular weights lower than 500 Da. Free amino acids are the low molecular compounds that have an important role to the taste, in the presence of sodium salt. The intense umami taste found in the soy sauces may also be a result from the interaction between umami components and other tastants. Small peptides are also present, but have very low, almost undetected umami taste intensities investigated in their fractions.
Collapse
Affiliation(s)
- Hanifah Nuryani Lioe
- Centre of Excellence for Food Safety Research, Faculty of Food Science and Technology, Univ. Putra Malaysia, UPM Serdang, Selangor 43400, Malaysia.
| | | | | |
Collapse
|
43
|
Meyerhof W, Batram C, Kuhn C, Brockhoff A, Chudoba E, Bufe B, Appendino G, Behrens M. The Molecular Receptive Ranges of Human TAS2R Bitter Taste Receptors. Chem Senses 2009; 35:157-70. [DOI: 10.1093/chemse/bjp092] [Citation(s) in RCA: 757] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
44
|
Fenech C, Patrikainen L, Kerr DS, Grall S, Liu Z, Laugerette F, Malnic B, Montmayeur JP. Ric-8A, a Galpha protein guanine nucleotide exchange factor potentiates taste receptor signaling. Front Cell Neurosci 2009; 3:11. [PMID: 19847316 PMCID: PMC2763893 DOI: 10.3389/neuro.03.011.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 09/22/2009] [Indexed: 11/18/2022] Open
Abstract
Taste receptors for sweet, bitter and umami tastants are G-protein-coupled receptors (GPCRs). While much effort has been devoted to understanding G-protein-receptor interactions and identifying the components of the signalling cascade downstream of these receptors, at the level of the G-protein the modulation of receptor signal transduction remains relatively unexplored. In this regard a taste-specific regulator of G-protein signaling (RGS), RGS21, has recently been identified. To study whether guanine nucleotide exchange factors (GEFs) are involved in the transduction of the signal downstream of the taste GPCRs we investigated the expression of Ric-8A and Ric-8B in mouse taste cells and their interaction with G-protein subunits found in taste buds. Mammalian Ric-8 proteins were initially identified as potent GEFs for a range of Gα subunits and Ric-8B has recently been shown to amplify olfactory signal transduction. We find that both Ric-8A and Ric-8B are expressed in a large portion of taste bud cells and that most of these cells contain IP3R-3 a marker for sweet, umami and bitter taste receptor cells. Ric-8A interacts with Gα-gustducin and Gαi2 through which it amplifies the signal transduction of hTas2R16, a receptor for bitter compounds. Overall, these findings are consistent with a role for Ric-8 in mammalian taste signal transduction.
Collapse
Affiliation(s)
- Claire Fenech
- UMR 5170 CNRS, Centre des Sciences du Goût Dijon, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Ide N, Sato E, Ohta K, Masuda T, Kitabatake N. Interactions of the sweet-tasting proteins thaumatin and lysozyme with the human sweet-taste receptor. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2009; 57:5884-5890. [PMID: 19489607 DOI: 10.1021/jf803956f] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
This study investigated the sweetness of the sweet-tasting protein thaumatin and lysozyme by both an in vitro cell-based assay and an in vivo sensory analysis to elucidate the differences between in vitro and in vivo response profiles. Hek293 cells were constructed that stably expressed the human T1R2+T1R3 sweet-taste receptor, and their responses to thaumatin and lysozyme were analyzed by monitoring the levels of intracellular cAMP. The results indicated that thaumatin and lysozyme as well as aspartame induced a decrease in the intracellular cAMP accumulation of the T1R2+T1R3-transfected cells and that EC(50) values of thaumatin and lysozyme determined by cell-based assay are well-consistent with the results of the sweetness threshold value determined by sensory analysis in the presence of 140 mM NaCl. The results of both in vitro and in vivo experiments confirmed that the sweetness inhibitor lactisole significantly suppressed the sweetness of thaumatin and lysozyme.
Collapse
Affiliation(s)
- Nobuyuki Ide
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto 611-0011, Japan
| | | | | | | | | |
Collapse
|
46
|
Little TJ, Gupta N, Case RM, Thompson DG, McLaughlin JT. Sweetness and bitterness taste of meals per se does not mediate gastric emptying in humans. Am J Physiol Regul Integr Comp Physiol 2009; 297:R632-9. [PMID: 19535679 DOI: 10.1152/ajpregu.00090.2009] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In cell line and animal models, sweet and bitter tastants induce secretion of signaling peptides (e.g., glucagon-like peptide-1 and cholecystokinin) and slow gastric emptying (GE). Whether human GE and appetite responses are regulated by the sweetness or bitterness per se of ingested food is, however, unknown. We aimed to determine whether intragastric infusion of "equisweet" (Study A) or "equibitter" (Study B) solutions slow GE to the same extent, and whether a glucose solution made sweeter by the addition of saccharin will slow GE more potently than glucose alone. Healthy nonobese subjects were studied in a single-blind, randomized fashion. Subjects received 500-ml intragastric infusions of predetermined equisweet solutions of glucose (560 mosmol/kgH(2)O), fructose (290 mosmol/kgH(2)O), aspartame (200 mg), and saccharin (50 mg); twice as sweet glucose + saccharin, water (volumetric control) (Study A); or equibitter solutions of quinine (0.198 mM), naringin (1 mM), or water (Study B). GE was evaluated using a [(13)C]acetate breath test, and hunger and fullness were scored using visual analog scales. In Study A, equisweet solutions did not empty similarly. Fructose, aspartame, and saccharin did not slow GE compared with water, but glucose did (P < 0.05). There was no additional effect of the sweeter glucose + saccharin solution (P > 0.05, compared with glucose alone). In Study B, neither bitter tastant slowed GE compared with water. None of the solutions modulated perceptions of hunger or fullness. We conclude that, in humans, the presence of sweetness and bitterness taste per se in ingested solutions does not appear to signal to influence GE or appetite perceptions.
Collapse
Affiliation(s)
- Tanya J Little
- Univ. of Manchester, Section of Gastrointestinal Sciences, Clinical Sciences Bldg., Salford Royal NHS Foundation Trust, Stott Lane, Salford, United Kingdom, M6 8HD.
| | | | | | | | | |
Collapse
|
47
|
Abstract
A greater understanding of the molecular mechanisms of sweet taste has profound significance for the food industry as well as for consumers. Understanding the mechanism by which sweet taste is elicited by saccharides, peptides, and proteins will assist science and industry in their search for sweet substances with fewer negative health effects. The original AH-B theories have been supplanted by detailed structural models. Recent identification of the human sweet receptor as a dimeric G-protein coupled receptor comprising T1R2 and T1R3 subunits has greatly increased the understanding of the mechanisms involved in sweet molecule binding and sweet taste transduction. This review discusses early theories of the sweet receptor, recent research of sweetener chemoreception of nonprotein and protein ligands, homology modeling, the transduction pathway, the possibility of the sweet receptor functioning allosterically, as well as the implications of allelic variation.
Collapse
Affiliation(s)
- B Meyers
- NutraSweet Co., Chicago, IL 60654, USA
| | | |
Collapse
|
48
|
Abstract
Bitter taste in mammals is achieved by a family of approximately 30 bitter taste receptor genes. The main function of bitter taste is to protect the organism against the ingestion of, frequently bitter, toxic food metabolites. The field of taste research has advanced rapidly during the last several years. This is especially true for the G-protein-coupled-receptor-mediated taste qualities, sweet, umami, and bitter. This review summarizes current knowledge of bitter taste receptor gene expression, signal transduction, the structure-activity relationship of bitter taste receptor proteins, as well as their variability leading to a high degree of individualization of this taste quality in mammals.
Collapse
Affiliation(s)
- M Behrens
- German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.
| | | |
Collapse
|
49
|
Abstract
Astringency plays an important role in the sensory experience of many foods and beverages, ranging from wine to nuts. Given the recent trend toward fortifying consumables with astringent compounds and the evidence regarding the health benefits of some astringents, the mechanisms and perceptual characteristics of astringency warrant further discussion and investigation. This paper reviews the current state of the literature, including consideration of new methods for describing and measuring astringency, and provides an overview of research concerned with elucidating the physical, physiological, and psychological factors that underlie and mediate perception of this sensation.
Collapse
Affiliation(s)
- Martha R Bajec
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | | |
Collapse
|
50
|
Tizzano M, Dvoryanchikov G, Barrows JK, Kim S, Chaudhari N, Finger TE. Expression of Galpha14 in sweet-transducing taste cells of the posterior tongue. BMC Neurosci 2008; 9:110. [PMID: 19014514 PMCID: PMC2596171 DOI: 10.1186/1471-2202-9-110] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2008] [Accepted: 11/13/2008] [Indexed: 12/03/2022] Open
Abstract
Background "Type II"/Receptor cells express G protein-coupled receptors (GPCRs) for sweet, umami (T1Rs and mGluRs) or bitter (T2Rs), as well as the proteins for downstream signalling cascades. Transduction downstream of T1Rs and T2Rs relies on G-protein and PLCβ2-mediated release of stored Ca2+. Whereas Gαgus (gustducin) couples to the T2R (bitter) receptors, which Gα-subunit couples to the sweet (T1R2 + T1R3) receptor is presently not known. We utilized RT-PCR, immunocytochemistry and single-cell gene expression profiling to examine the expression of the Gαq family (q, 11, 14) in mouse taste buds. Results By RT-PCR, Gα14 is expressed strongly and in a taste selective manner in posterior (vallate and foliate), but not anterior (fungiform and palate) taste fields. Gαq and Gα11, although detectable, are not expressed in a taste-selective fashion. Further, expression of Gα14 mRNA is limited to Type II/Receptor cells in taste buds. Immunocytochemistry on vallate papillae using a broad Gαq family antiserum reveals specific staining only in Type II taste cells (i.e. those expressing TrpM5 and PLCβ2). This staining persists in Gαq knockout mice and immunostaining with a Gα11-specific antiserum shows no immunoreactivity in taste buds. Taken together, these data show that Gα14 is the dominant Gαq family member detected. Immunoreactivity for Gα14 strongly correlates with expression of T1R3, the taste receptor subunit present in taste cells responsive to either umami or sweet. Single cell gene expression profiling confirms a tight correlation between the expression of Gα14 and both T1R2 and T1R3, the receptor combination that forms sweet taste receptors. Conclusion Gα14 is co-expressed with the sweet taste receptor in posterior tongue, although not in anterior tongue. Thus, sweet taste transduction may rely on different downstream transduction elements in posterior and anterior taste fields.
Collapse
Affiliation(s)
- Marco Tizzano
- Rocky Mountain Taste & Smell Center, University of Colorado Denver School of Medicine, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|