1
|
Feng X, Flüchter P, De Tenorio JC, Schneider C. Tuft cells in the intestine, immunity and beyond. Nat Rev Gastroenterol Hepatol 2024; 21:852-868. [PMID: 39327439 DOI: 10.1038/s41575-024-00978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/28/2024]
Abstract
Tuft cells have gained substantial attention over the past 10 years due to numerous reports linking them with type 2 immunity and microorganism-sensing capacity in many mucosal tissues. This heightened interest is fuelled by their unique ability to produce an array of biological effector molecules, including IL-25, allergy-related eicosanoids, and the neurotransmitter acetylcholine, enabling downstream responses in diverse cell types. Operating through G protein-coupled receptor-mediated signalling pathways reminiscent of type II taste cells in oral taste buds, tuft cells emerge as chemosensory sentinels that integrate luminal conditions, eliciting appropriate responses in immune, epithelial and neuronal populations. How tuft cells promote tissue alterations and adaptation to the variety of stimuli at mucosal surfaces has been explored in multiple studies in the past few years. Since the initial recognition of the role of tuft cells, the discovery of diverse tuft cell effector functions and associated feedback loops have also revealed the complexity of tuft cell biology. Although earlier work largely focused on extraintestinal tissues, novel genetic tools and recent mechanistic studies on intestinal tuft cells established fundamental concepts of tuft cell activation and functions. This Review is an overview of intestinal tuft cells, providing insights into their development, signalling and interaction modules in immunity and other states.
Collapse
Affiliation(s)
- Xiaogang Feng
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | - Pascal Flüchter
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
2
|
Montalbano C, Raia A, Caruso V, Migli L. Sodium valproate: cacosmia and dysgeusia as uncommon side effects. Arch Clin Cases 2023; 10:157-159. [PMID: 38026105 PMCID: PMC10660243 DOI: 10.22551/2023.41.1004.10265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
Smell and taste disturbances are potential adverse reactions of many drugs used in Psychiatry, such as antidepressants, anti-Parkinson agents, lithium, minor and major tranquilizers. To our knowledge, only one clinical case regarding valproate and cacosmia has been reported so far. However, several anticonvulsants are reported to cause taste and smell disturbances, although the underlying etiology is currently unclear. Our patient developed cacosmia and dysgeusia when taking valproic acid, both effects quickly disappeared upon drug discontinuation. In this article we not only report this uncommon side effect, but we discuss the plausible mechanisms behind such an adverse reaction. Our case is to date the second similar case in the literature. The aim of the present article is to make clinicians informed about this very uncommon and unpleasant side effect.
Collapse
Affiliation(s)
- Clara Montalbano
- Department of Psychiatry, Unit of Psychiatric Disorders in Medical Complex Pathologies, University of Pisa, Pisa, Italy
| | - Accursio Raia
- Department of Psychiatry, Unit of Psychiatric Disorders in Medical Complex Pathologies, University of Pisa, Pisa, Italy
| | - Valerio Caruso
- Department of Psychiatry, Unit of Psychiatric Disorders in Medical Complex Pathologies, University of Pisa, Pisa, Italy
| | - Lavinia Migli
- Department of Psychiatry, Unit of Psychiatric Disorders in Medical Complex Pathologies, University of Pisa, Pisa, Italy
| |
Collapse
|
3
|
Kawabata Y, Takai S, Sanematsu K, Yoshida R, Kawabata F, Shigemura N. The Antiarrhythmic Drug Flecainide Enhances Aversion to HCl in Mice. eNeuro 2023; 10:ENEURO.0048-23.2023. [PMID: 37696662 PMCID: PMC10515741 DOI: 10.1523/eneuro.0048-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Drug-induced taste disorders reduce quality of life, but little is known about the molecular mechanisms by which drugs induce taste disturbances. In this study, we investigated the short-term and long-term effects of the antiarrhythmic drug flecainide, which is known to cause taste dysfunction. Analyses of behavioral responses (licking tests) revealed that mice given a single intraperitoneal injection of flecainide exhibited a significant reduction in preference for a sour tastant (HCl) but not for other taste solutions (NaCl, quinine, sucrose, KCl and monopotassium glutamate) when compared with controls. Mice administered a single dose of flecainide also had significantly higher taste nerve responses to HCl but not to other taste solutions. Compared with controls, mice administered flecainide once-daily for 30 d showed a reduced preference for HCl without any changes in the behavioral responses to other taste solutions. The electrophysiological experiments using HEK293T cells transiently expressing otopetrin-1 (Otop1; the mouse sour taste receptor) showed that flecainide did not alter the responses to HCl. Taken together, our results suggest that flecainide specifically enhances the response to HCl in mice during short-term and long-term administration. Although further studies will be needed to elucidate the molecular mechanisms, these findings provide new insights into the pathophysiology of drug-induced taste disorders.
Collapse
Affiliation(s)
- Yuko Kawabata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
- Oral Health/Brain Health/Total Health Research Center, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Fuminori Kawabata
- Physiology of Domestic Animals, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
4
|
Ma Z, Paudel U, Foskett JK. Effects of temperature on action potentials and ion conductances in type II taste-bud cells. Am J Physiol Cell Physiol 2023; 325:C155-C171. [PMID: 37273235 PMCID: PMC10312327 DOI: 10.1152/ajpcell.00413.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/06/2023]
Abstract
Temperature strongly influences the intensity of taste, but it remains understudied despite its physiological, hedonic, and commercial implications. The relative roles of the peripheral gustatory and somatosensory systems innervating the oral cavity in mediating thermal effects on taste sensation and perception are poorly understood. Type II taste-bud cells, responsible for sensing sweet, bitter umami, and appetitive NaCl, release neurotransmitters to gustatory neurons by the generation of action potentials, but the effects of temperature on action potentials and the underlying voltage-gated conductances are unknown. Here, we used patch-clamp electrophysiology to explore the effects of temperature on acutely isolated type II taste-bud cell electrical excitability and whole cell conductances. Our data reveal that temperature strongly affects action potential generation, properties, and frequency and suggest that thermal sensitivities of underlying voltage-gated Na+ and K+ channel conductances provide a mechanism for how and whether voltage-gated Na+ and K+ channels in the peripheral gustatory system contribute to the influence of temperature on taste sensitivity and perception.NEW & NOTEWORTHY The temperature of food affects how it tastes. Nevertheless, the mechanisms involved are not well understood, particularly whether the physiology of taste-bud cells in the mouth is involved. Here we show that the electrical activity of type II taste-bud cells that sense sweet, bitter, and umami substances is strongly influenced by temperature. These results suggest a mechanism for the influence of temperature on the intensity of taste perception that resides in taste buds themselves.
Collapse
Affiliation(s)
- Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Usha Paudel
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
5
|
Wu X, Zhang H, Long H, Zhang D, Yang X, Liu D, E G. Genome-Wide Selection Signal Analysis to Investigate Wide Genomic Heredity Divergence between Eurasian Wild Boar and Domestic Pig. Animals (Basel) 2023; 13:2158. [PMID: 37443955 DOI: 10.3390/ani13132158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
As important livestock species, pigs provide essential meat resources for humans, so understanding the genetic evolution behind their domestic history could help with the genetic improvement of domestic pigs. This study aimed to investigate the evolution of convergence and divergence under selection in European and Asian domestic pigs by using public genome-wide data. A total of 164 and 108 candidate genes (CDGs) were obtained from the Asian group (wild boar vs. domestic pig) and the European group (wild boar vs. domestic pig), respectively, by taking the top 5% of intersected windows of a pairwise fixation index (FST) and a cross population extended haplotype homozygosity test (XPEHH). GO and KEGG annotated results indicated that most CDGs were related to reproduction and immunity in the Asian group. Conversely, rich CDGs were enriched in muscle development and digestion in the European group. Eight CDGs were subjected to parallel selection of Eurasian domestic pigs from local wild boars during domestication. These CDGs were mainly involved in olfactory transduction, metabolic pathways, and progesterone-mediated oocyte maturation. Moreover, 36 and 18 haplotypes of INPP5B and TRAK2 were identified in this study, respectively. In brief, this study did not only improve the understanding of the genetic evolution of domestication in pigs, but also provides valuable CDGs for future breeding and genetic improvement of pigs.
Collapse
Affiliation(s)
- Xinming Wu
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China
| | - Haoyuan Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China
| | - Haoyuan Long
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China
| | - Dongjie Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Guangxin E
- College of Animal Science and Technology, Southwest University, Chongqing 400716, China
| |
Collapse
|
6
|
Kouakou YI, Lee RJ. Interkingdom Detection of Bacterial Quorum-Sensing Molecules by Mammalian Taste Receptors. Microorganisms 2023; 11:1295. [PMID: 37317269 PMCID: PMC10221136 DOI: 10.3390/microorganisms11051295] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023] Open
Abstract
Bitter and sweet taste G protein-coupled receptors (known as T2Rs and T1Rs, respectively) were originally identified in type II taste cells on the tongue, where they signal perception of bitter and sweet tastes, respectively. Over the past ~15 years, taste receptors have been identified in cells all over the body, demonstrating a more general chemosensory role beyond taste. Bitter and sweet taste receptors regulate gut epithelial function, pancreatic β cell secretion, thyroid hormone secretion, adipocyte function, and many other processes. Emerging data from a variety of tissues suggest that taste receptors are also used by mammalian cells to "eavesdrop" on bacterial communications. These receptors are activated by several quorum-sensing molecules, including acyl-homoserine lactones and quinolones from Gram-negative bacteria such as Pseudomonas aeruginosa, competence stimulating peptides from Streptococcus mutans, and D-amino acids from Staphylococcus aureus. Taste receptors are an arm of immune surveillance similar to Toll-like receptors and other pattern recognition receptors. Because they are activated by quorum-sensing molecules, taste receptors report information about microbial population density based on the chemical composition of the extracellular environment. This review summarizes current knowledge of bacterial activation of taste receptors and identifies important questions remaining in this field.
Collapse
Affiliation(s)
- Yobouet Ines Kouakou
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Robert J. Lee
- Department of Otorhinolaryngology and Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Dutta Banik D, Medler KF. Defining the role of TRPM4 in broadly responsive taste receptor cells. Front Cell Neurosci 2023; 17:1148995. [PMID: 37032837 PMCID: PMC10073513 DOI: 10.3389/fncel.2023.1148995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Peripheral taste receptor cells use multiple signaling pathways to transduce taste stimuli into output signals that are sent to the brain. We have previously identified a subpopulation of Type III taste cells that are broadly responsive (BR) and respond to multiple taste stimuli including bitter, sweet, umami, and sour. These BR cells use a PLCβ3/IP3R1 signaling pathway to detect bitter, sweet, and umami stimuli and use a separate pathway to detect sour. Currently, the downstream targets of the PLCβ3 signaling pathway are unknown. Here we identify TRPM4, a monovalent selective TRP channel, as an important downstream component in this signaling pathway. Using live cell imaging on isolated taste receptor cells from mice, we show that inhibition of TRPM4 abolished the taste-evoked sodium responses and significantly reduced the taste-evoked calcium responses in BR cells. Since BR cells are a subpopulation of Type III taste cells, they have conventional chemical synapses that require the activation of voltage-gated calcium channels (VGCCs) to cause neurotransmitter release. We found that TRPM4-dependent membrane depolarization selectively activates L-type VGCCs in these cells. The calcium influx through L-type VGCCs also generates a calcium-induced calcium release (CICR) via ryanodine receptors that enhances TRPM4 activity. Together these signaling events amplify the initial taste response to generate an appropriate output signal.
Collapse
|
8
|
Bigiani A, Tirindelli R, Bigiani L, Mapelli J. Changes of the biophysical properties of voltage-gated Na + currents during maturation of the sodium-taste cells in rat fungiform papillae. J Physiol 2022; 600:5119-5144. [PMID: 36250254 DOI: 10.1113/jp283636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/13/2022] [Indexed: 01/05/2023] Open
Abstract
Taste cells are a heterogeneous population of sensory receptors that undergo continuous turnover. Different chemo-sensitive cell lines rely on action potentials to release the neurotransmitter onto nerve endings. The electrical excitability is due to the presence of a tetrodotoxin-sensitive, voltage-gated sodium current (INa ) similar to that found in neurons. Since the biophysical properties of neuronal INa change during development, we wondered whether the same also occurred in taste cells. Here, we used the patch-clamp recording technique to study INa in salt-sensing cells (sodium cells) of rat fungiform papillae. We identified these cells by exploiting the known blocking effect of amiloride on ENaC, the sodium (salt) receptor. Based on the amplitude of INa , which is known to increase during development, we subdivided sodium cells into two groups: cells with small sodium current (SSC cells; INa < 1 nA) and cells with large sodium current (LSC cells; INa > 1 nA). We found that: the voltage dependence of activation and inactivation significantly differed between these subsets; a slowly inactivating sodium current was more prominent in LSC cells; membrane capacitance in SSC cells was larger than in LSC cells. mRNA expression analysis of the α-subunits of voltage-gated sodium channels in fungiform taste buds supported the functional data. Lucifer Yellow labelling of recorded cells revealed that our electrophysiological criterion for distinguishing two broad groups of taste cells was in good agreement with morphological observations for cell maturity. Thus, all these findings are consistent with developmental changes in the voltage-dependent properties of sodium-taste cells. KEY POINTS: Taste cells are sensory receptors that undergo continuous turnover while they detect food chemicals and communicate with afferent nerve fibres. The voltage-gated sodium current (INa ) is a key ion current for generating action potentials in fully differentiated and chemo-sensitive taste cells, which use electrical signalling to release neurotransmitters. Here we show that, during the maturation of rat taste cells involved in salt detection (sodium cells), the biophysical properties of INa , such as voltage dependence of activation and inactivation, change significantly. Our results help reveal how taste cells gain electrical excitability during turnover, a property critical to their operation as chemical detectors that relay sensory information to nerve fibres.
Collapse
Affiliation(s)
- Albertino Bigiani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Università di Modena e Reggio Emilia, Modena, Italy
| | - Roberto Tirindelli
- Dipartimento di Medicina e Chirurgia, SMart Laboratory, Università di Parma, Parma, Italy
| | | | - Jonathan Mapelli
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Università di Modena e Reggio Emilia, Modena, Italy
| |
Collapse
|
9
|
Ion rectification based on gel polymer electrolyte ionic diode. Nat Commun 2022; 13:6669. [PMID: 36335134 PMCID: PMC9637189 DOI: 10.1038/s41467-022-34429-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Biological ion channels rely on ions as charge carriers and unidirectional ion flow to produce and transmit signals. To realize artificial biological inspired circuitry and seamless human-machine communication, ion-transport-based rectification devices should be developed. In this research, poly(methyl methacrylate) (PMMA) and poly(vinylidene fluoride-co-hexafluoropropylene) (PVDF-HFP) gel polymer electrolytes (GPEs) are assembled to construct a novel ionic diode, enabling ion rectification through ion-diffusion/migration that emulates biological systems. This ion rectification results from the different diffusion/migration behaviors of mobile ions transporting in the GPE heterojunction. The electrical tests of the GPE heterojunction reveal outstanding rectifying ratio of 23.11. The GPE ionic diode operates in wide temperature window, from -20 °C (anti-freezing) to 125 °C (thermal tolerance). The absence of redox reactions is verified in the cyclic voltammogram. The GPE ionic diodes are used to construct ionic logic gates for signal communication. Furthermore, rectification of a triboelectric nanogenerator and potential for synaptic devices are demonstrated.
Collapse
|
10
|
Shi Y, Pu D, Zhou X, Zhang Y. Recent Progress in the Study of Taste Characteristics and the Nutrition and Health Properties of Organic Acids in Foods. Foods 2022; 11:3408. [PMID: 36360025 PMCID: PMC9654595 DOI: 10.3390/foods11213408] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 08/11/2023] Open
Abstract
Organic acids could improve the food flavor, maintain the nutritional value, and extend the shelf life of food. This review summarizes the detection methods and concentrations of organic acids in different foods, as well as their taste characteristics and nutritional properties. The composition of organic acids varies in different food. Fruits and vegetables often contain citric acid, creatine is a unique organic acid found in meat, fermented foods have a high content of acetic acid, and seasonings have a wide range of organic acids. Determination of the organic acid contents among different food matrices allows us to monitor the sensory properties, origin identification, and quality control of foods, and further provides a basis for food formulation design. The taste characteristics and the acid taste perception mechanisms of organic acids have made some progress, and binary taste interaction is the key method to decode multiple taste perception. Real food and solution models elucidated that the organic acid has an asymmetric interaction effect on the other four basic taste attributes. In addition, in terms of nutrition and health, organic acids can provide energy and metabolism regulation to protect the human immune and myocardial systems. Moreover, it also exhibited bacterial inhibition by disrupting the internal balance of bacteria and inhibiting enzyme activity. It is of great significance to clarify the synergistic dose-effect relationship between organic acids and other taste sensations and further promote the application of organic acids in food salt reduction.
Collapse
Affiliation(s)
- Yige Shi
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China Gengeral Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Dandan Pu
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China Gengeral Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Xuewei Zhou
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China Gengeral Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Yuyu Zhang
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China Gengeral Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
11
|
Sweet Taste Signaling: The Core Pathways and Regulatory Mechanisms. Int J Mol Sci 2022; 23:ijms23158225. [PMID: 35897802 PMCID: PMC9329783 DOI: 10.3390/ijms23158225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Sweet taste, a proxy for sugar-derived calories, is an important driver of food intake, and animals have evolved robust molecular and cellular machinery for sweet taste signaling. The overconsumption of sugar-derived calories is a major driver of obesity and other metabolic diseases. A fine-grained appreciation of the dynamic regulation of sweet taste signaling mechanisms will be required for designing novel noncaloric sweeteners with better hedonic and metabolic profiles and improved consumer acceptance. Sweet taste receptor cells express at least two signaling pathways, one mediated by a heterodimeric G-protein coupled receptor encoded by taste 1 receptor members 2 and 3 (TAS1R2 + TAS1R3) genes and another by glucose transporters and the ATP-gated potassium (KATP) channel. Despite these important discoveries, we do not fully understand the mechanisms regulating sweet taste signaling. We will introduce the core components of the above sweet taste signaling pathways and the rationale for having multiple pathways for detecting sweet tastants. We will then highlight the roles of key regulators of the sweet taste signaling pathways, including downstream signal transduction pathway components expressed in sweet taste receptor cells and hormones and other signaling molecules such as leptin and endocannabinoids.
Collapse
|
12
|
Abstract
Sour taste, the taste of acids, is one of the most enigmatic of the five basic taste qualities; its function is unclear and its receptor was until recently unknown. Sour tastes are transduced in taste buds on the tongue and palate epithelium by a subset of taste receptor cells, known as type III cells. Type III cells express a number of unique markers, including the PKD2L1 gene, which allow for their identification and manipulation. These cells respond to acid stimuli with action potentials and release neurotransmitters onto afferent nerve fibers, with cell bodies in geniculate and petrosal ganglia. Here, we review classical studies of sour taste leading up to the identification of the sour receptor as the proton channel, OTOP1. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Heather N Turner
- Department of Biological Sciences, Section of Neurobiology, University of Southern California, Los Angeles, California, USA; ,
| | - Emily R Liman
- Department of Biological Sciences, Section of Neurobiology, University of Southern California, Los Angeles, California, USA; ,
| |
Collapse
|
13
|
Kasahara Y, Narukawa M, Ishimaru Y, Kanda S, Umatani C, Takayama Y, Tominaga M, Oka Y, Kondo K, Kondo T, Takeuchi A, Misaka T, Abe K, Asakura T. TMC4 is a novel chloride channel involved in high-concentration salt taste sensation. J Physiol Sci 2021; 71:23. [PMID: 34429071 PMCID: PMC10717410 DOI: 10.1186/s12576-021-00807-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/16/2021] [Indexed: 12/27/2022]
Abstract
"Salty taste" sensation is evoked when sodium and chloride ions are present together in the oral cavity. The presence of an epithelial cation channel that receives Na+ has previously been reported. However, no molecular entity involving Cl- receptors has been elucidated. We report the strong expression of transmembrane channel-like 4 (TMC4) in the circumvallate and foliate papillae projected to the glossopharyngeal nerve, mediating a high-concentration of NaCl. Electrophysiological analysis using HEK293T cells revealed that TMC4 was a voltage-dependent Cl- channel and the consequent currents were completely inhibited by NPPB, an anion channel blocker. TMC4 allowed permeation of organic anions including gluconate, but their current amplitudes at positive potentials were less than that of Cl-. Tmc4-deficient mice showed significantly weaker glossopharyngeal nerve response to high-concentration of NaCl than the wild-type littermates. These results indicated that TMC4 is a novel chloride channel that responds to high-concentration of NaCl.
Collapse
Affiliation(s)
- Yoichi Kasahara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Masataka Narukawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshicho Imakumano Higashiyama, Kyoto, 605-8501, Japan
| | - Yoshiro Ishimaru
- Department of Agricultural Chemistry, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Shinji Kanda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Chie Umatani
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yasunori Takayama
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaijicho, Okazaki, Aichi, 444-8787, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaijicho, Okazaki, Aichi, 444-8787, Japan
- Thermal Biology Research Group, Exploratory Research Center On Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaijicho, Okazaki, Aichi, 444-8787, Japan
| | - Yoshitaka Oka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kaori Kondo
- Laboratory for Developmental Genetics, RIKEN-IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takashi Kondo
- Laboratory for Developmental Genetics, RIKEN-IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, and Life Science Innovation Center, University of Fukui, Fukui, 910-1193, Japan
| | - Takumi Misaka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Keiko Abe
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), LiSE 4F C-4, 3-25-13 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
| | - Tomiko Asakura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| |
Collapse
|
14
|
Koyama S, Kondo K, Ueha R, Kashiwadani H, Heinbockel T. Possible Use of Phytochemicals for Recovery from COVID-19-Induced Anosmia and Ageusia. Int J Mol Sci 2021; 22:8912. [PMID: 34445619 PMCID: PMC8396277 DOI: 10.3390/ijms22168912] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
The year 2020 became the year of the outbreak of coronavirus, SARS-CoV-2, which escalated into a worldwide pandemic and continued into 2021. One of the unique symptoms of the SARS-CoV-2 disease, COVID-19, is the loss of chemical senses, i.e., smell and taste. Smell training is one of the methods used in facilitating recovery of the olfactory sense, and it uses essential oils of lemon, rose, clove, and eucalyptus. These essential oils were not selected based on their chemical constituents. Although scientific studies have shown that they improve recovery, there may be better combinations for facilitating recovery. Many phytochemicals have bioactive properties with anti-inflammatory and anti-viral effects. In this review, we describe the chemical compounds with anti- inflammatory and anti-viral effects, and we list the plants that contain these chemical compounds. We expand the review from terpenes to the less volatile flavonoids in order to propose a combination of essential oils and diets that can be used to develop a new taste training method, as there has been no taste training so far. Finally, we discuss the possible use of these in clinical settings.
Collapse
Affiliation(s)
- Sachiko Koyama
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Kenji Kondo
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Tokyo 113-8655, Japan;
| | - Rumi Ueha
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Tokyo 113-8655, Japan;
- Swallowing Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Hideki Kashiwadani
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan;
| | - Thomas Heinbockel
- Department of Anatomy, College of Medicine, Howard University, Washington, DC 20059, USA
| |
Collapse
|
15
|
Ohtubo Y. Slow recovery from the inactivation of voltage-gated sodium channel Nav1.3 in mouse taste receptor cells. Pflugers Arch 2021; 473:953-968. [PMID: 33881614 DOI: 10.1007/s00424-021-02563-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 02/06/2023]
Abstract
Action potentials play an important role in neurotransmitter release in response to taste. Here, I have investigated voltage-gated Na+ channels, a primary component of action potentials, in respective cell types of mouse fungiform taste bud cells (TBCs) with in situ whole-cell clamping and single-cell RT-PCR techniques. The cell types of TBCs electrophysiologically examined were determined immunohistochemically using the type III inositol 1,4,5-triphoshate receptor as a type II cell marker and synaptosomal-associated protein 25 as a type III cell marker. I show that type II cells, type III cells, and TBCs not immunoreactive to these markers (likely type I cells) generate voltage-gated Na+ currents. The recovery following inactivation of these currents was well fitted with double exponential curves. The time constants in type III cells (~20 ms and ~ 1 s) were significantly slower than respective time constants in other cell types. RT-PCR analysis indicated the expression of Nav1.3, Nav1.5, Nav1.6, and β1 subunit mRNAs in TBCs. Pharmacological inhibition and single-cell RT-PCR studies demonstrated that type II and type III cells principally express tetrodotoxin (TTX)-sensitive Nav1.3 channels and that ~ 30% of type I cells express TTX-resistant Nav1.5 channels. The auxiliary β1 subunit that modulates gating kinetics was rarely detected in TBCs. As the β1 subunit co-expressed with an α subunit is known to accelerate the recovery from inactivation, it is likely that voltage-gated Na+ channels in TBCs may function without β subunits. Slow recovery from inactivation, especially in type III cells, may limit high-frequency firing in response to taste substances.
Collapse
Affiliation(s)
- Yoshitaka Ohtubo
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, Hibikino 2-4, Kitakyushu, 808-0196, Japan.
| |
Collapse
|
16
|
ANO7: Insights into topology, function, and potential applications as a biomarker and immunotherapy target. Tissue Cell 2021; 72:101546. [PMID: 33940566 DOI: 10.1016/j.tice.2021.101546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/21/2021] [Accepted: 04/11/2021] [Indexed: 01/01/2023]
Abstract
Anoctamin 7 (ANO7) is a member of the transmembrane protein TMEM16 family. It has a conservative topology similar to other members in this family, such as the typical eight-transmembrane domain, but it also has unique features. Although the ion channel role of ANO7 has been well accepted, evolutionary analyses and relevant studies suggest that ANO7 may be a multi-facet protein in function. Studies have shown that ANO7 may also function as a scramblase. ANO7 is highly expressed in prostate cancer as well as normal prostate tissues. A considerable amount of evidence has confirmed that ANO7 is associated with human physiology and pathology, particularly with the development of prostate cancer, which makes ANO7 a good candidate as a diagnostic and prognostic biomarker. In addition, ANO7 may be a potential target for prostate cancer immunotherapy. Antibody-based or T cell-mediated immunotherapies against prostate cancer by targeting ANO7 have been highly anticipated. ANO7 may also correlate with several other types of cancers or diseases, where further studies are warranted.
Collapse
|
17
|
Eaton M, Zhang J, Ma Z, Park AC, Lietzke E, Romero CM, Liu Y, Coleman ER, Chen X, Xiao T, Que Z, Lai S, Wu J, Lee JH, Palant S, Nguyen HP, Huang Z, Skarnes WC, Koss WA, Yang Y. Generation and basic characterization of a gene-trap knockout mouse model of Scn2a with a substantial reduction of voltage-gated sodium channel Na v 1.2 expression. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12725. [PMID: 33369088 DOI: 10.1111/gbb.12725] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Large-scale genetic studies revealed SCN2A as one of the most frequently mutated genes in patients with neurodevelopmental disorders. SCN2A encodes for the voltage-gated sodium channel isoform 1.2 (Nav 1.2) expressed in the neurons of the central nervous system. Homozygous knockout (null) of Scn2a in mice is perinatal lethal, whereas heterozygous knockout of Scn2a (Scn2a+/- ) results in mild behavior abnormalities. The Nav 1.2 expression level in Scn2a+/- mice is reported to be around 50-60% of the wild-type (WT) level, which indicates that a close to 50% reduction of Nav 1.2 expression may not be sufficient to lead to major behavioral phenotypes in mice. To overcome this barrier, we characterized a novel mouse model of severe Scn2a deficiency using a targeted gene-trap knockout (gtKO) strategy. This approach produces viable homozygous mice (Scn2agtKO/gtKO ) that can survive to adulthood, with about a quarter of Nav 1.2 expression compared to WT mice. Innate behaviors like nesting and mating were profoundly disrupted in Scn2agtKO/gtKO mice. Notably, Scn2agtKO/gtKO mice have a significantly decreased center duration compared to WT in the open field test, suggesting anxiety-like behaviors in a novel, open space. These mice also have decreased thermal and cold tolerance. Additionally, Scn2agtKO/gtKO mice have increased fix-pattern exploration in the novel object exploration test and a slight increase in grooming, indicating a detectable level of repetitive behaviors. They bury little to no marbles and have decreased interaction with novel objects. These Scn2a gene-trap knockout mice thus provide a unique model to study pathophysiology associated with severe Scn2a deficiency.
Collapse
Affiliation(s)
- Muriel Eaton
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Jingliang Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Zhixiong Ma
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Anthony C Park
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Emma Lietzke
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Chloé M Romero
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Yushuang Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Emily R Coleman
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Xiaoling Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Tiange Xiao
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Zhefu Que
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Shirong Lai
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Jiaxiang Wu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Ji Hea Lee
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Sophia Palant
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Huynhvi P Nguyen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Zhuo Huang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - William C Skarnes
- Department of Cellular Engineering, The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Wendy A Koss
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
- Office of the Executive Vice President for Research and Partnerships, Purdue University, West Lafayette, Indiana, USA
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
18
|
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: 0.5] [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
|
19
|
Structure-Function Analyses of Human Bitter Taste Receptors-Where Do We Stand? Molecules 2020; 25:molecules25194423. [PMID: 32993119 PMCID: PMC7582848 DOI: 10.3390/molecules25194423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
The finding that bitter taste receptors are expressed in numerous tissues outside the oral cavity and fulfill important roles in metabolic regulation, innate immunity and respiratory control, have made these receptors important targets for drug discovery. Efficient drug discovery depends heavily on detailed knowledge on structure-function-relationships of the target receptors. Unfortunately, experimental structures of bitter taste receptors are still lacking, and hence, the field relies mostly on structures obtained by molecular modeling combined with functional experiments and point mutageneses. The present article summarizes the current knowledge on the structure–function relationships of human bitter taste receptors. Although these receptors are difficult to express in heterologous systems and their homology with other G protein-coupled receptors is very low, detailed information are available at least for some of these receptors.
Collapse
|
20
|
Gui Y, Thomas MH, Garcia P, Karout M, Halder R, Michelucci A, Kollmus H, Zhou C, Melmed S, Schughart K, Balling R, Mittelbronn M, Nadeau JH, Williams RW, Sauter T, Buttini M, Sinkkonen L. Pituitary Tumor Transforming Gene 1 Orchestrates Gene Regulatory Variation in Mouse Ventral Midbrain During Aging. Front Genet 2020; 11:566734. [PMID: 33173537 PMCID: PMC7538689 DOI: 10.3389/fgene.2020.566734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/27/2020] [Indexed: 01/07/2023] Open
Abstract
Dopaminergic neurons in the midbrain are of particular interest due to their role in diseases such as Parkinson’s disease and schizophrenia. Genetic variation between individuals can affect the integrity and function of dopaminergic neurons but the DNA variants and molecular cascades modulating dopaminergic neurons and other cells types of ventral midbrain remain poorly defined. Three genetically diverse inbred mouse strains – C57BL/6J, A/J, and DBA/2J – differ significantly in their genomes (∼7 million variants), motor and cognitive behavior, and susceptibility to neurotoxins. To further dissect the underlying molecular networks responsible for these variable phenotypes, we generated RNA-seq and ChIP-seq data from ventral midbrains of the 3 mouse strains. We defined 1000–1200 transcripts that are differentially expressed among them. These widespread differences may be due to altered activity or expression of upstream transcription factors. Interestingly, transcription factors were significantly underrepresented among the differentially expressed genes, and only one transcription factor, Pttg1, showed significant differences between all three strains. The changes in Pttg1 expression were accompanied by consistent alterations in histone H3 lysine 4 trimethylation at Pttg1 transcription start site. The ventral midbrain transcriptome of 3-month-old C57BL/6J congenic Pttg1–/– mutants was only modestly altered, but shifted toward that of A/J and DBA/2J in 9-month-old mice. Principle component analysis (PCA) identified the genes underlying the transcriptome shift and deconvolution of these bulk RNA-seq changes using midbrain single cell RNA-seq data suggested that the changes were occurring in several different cell types, including neurons, oligodendrocytes, and astrocytes. Taken together, our results show that Pttg1 contributes to gene regulatory variation between mouse strains and influences mouse midbrain transcriptome during aging.
Collapse
Affiliation(s)
- Yujuan Gui
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Mélanie H Thomas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg.,National Center of Pathology, Laboratoire National de Santé, Dudelange, Luxembourg.,Luxembourg Centre of Neuropathology, Dudelange, Luxembourg
| | - Mona Karout
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Alessandro Michelucci
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg.,Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Cuiqi Zhou
- Cedars Sinai Medical Centre, Los Angeles, CA, United States
| | - Shlomo Melmed
- Cedars Sinai Medical Centre, Los Angeles, CA, United States
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Infection Genetics, University of Veterinary Medicine Hannover, Hanover, Germany.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Michel Mittelbronn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg.,National Center of Pathology, Laboratoire National de Santé, Dudelange, Luxembourg.,Luxembourg Centre of Neuropathology, Dudelange, Luxembourg.,Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Joseph H Nadeau
- Pacific Northwest Research Institute, Seattle, WA, United States.,Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
21
|
Abstract
In the last few years, single-cell profiling of taste cells and ganglion cells has advanced our understanding of transduction, encoding, and transmission of information from taste buds as relayed to the central nervous system. This review focuses on new knowledge from these molecular approaches and attempts to place this in the context of previous questions and findings in the field. The individual taste cells within a taste bud are molecularly specialized for detection of one of the primary taste qualities: salt, sour, sweet, umami, and bitter. Transduction and transmitter release mechanisms differ substantially for taste cells transducing sour (Type III cells) compared with those transducing the qualities of sweet, umami, or bitter (Type II cells), although ultimately all transmission of taste relies on activation of purinergic P2X receptors on the afferent nerves. The ganglion cells providing innervation to the taste buds also appear divisible into functional and molecular subtypes, and each ganglion cell is primarily but not exclusively responsive to one taste quality.
Collapse
Affiliation(s)
- Sue C. Kinnamon
- Rocky Mountain Taste & Smell Center, Department of Otolaryngology and Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Thomas E. Finger
- Rocky Mountain Taste & Smell Center, Department of Otolaryngology and Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| |
Collapse
|
22
|
Cui Y, Wu H, Li Q, Liao J, Gao P, Sun F, Zhang H, Lu Z, Wei X, He C, Ma T, Wei X, Chen X, Zheng H, Yang G, Liu D, Zhu Z. Impairment of Bitter Taste Sensor Transient Receptor Potential Channel M5-Mediated Aversion Aggravates High-Salt Intake and Hypertension. Hypertension 2019; 74:1021-1032. [PMID: 31401881 DOI: 10.1161/hypertensionaha.119.13358] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Excessive salt consumption leads to cardiovascular diseases. Despite various measures designed to reduce salt intake, daily salt intake remains at a high level. Appropriate salt intake is balanced by salt taste preference triggered by epithelium sodium channel and salt taste aversion evoked by bitter taste sensor, transient receptor potential channel M5 (TRPM5). However, the behavioral mechanism of excessive salt intake remains largely elusive. In this study, wild type and TRPM5-/- mice were applied to study the influence of high-salt administration on epithelium sodium channel/TRPM5 and the associated behavior to salt consumption. We found that long-term high-salt intake impaired the aversive behavior to high-salt stimulation but did not alter the preference to low salt in mice. The mechanistic evidence demonstrated that high-salt intake blunted the TRPM5-mediated aversive behavior to noxious salt stimulation through inhibiting PKC (protein kinase C) activity and PKC-dependent threonine phosphorylation in the tongue epithelium but did not affect the epithelium sodium channel-dependent salt taste preference. Inhibition of TRPM5 also resulted in an impaired aversive response to high salt, with reduced taste perception in bitter cortical field of mice. TRPM5-/- mice showed a lowered aversion to high-salt diet and developed salt-induced hypertension. The impaired perception to bitter taste evoked by high-salt intake also existed in hypertensive patients with high-salt consumption. We demonstrate that long-term high-salt consumption impairs aversive response to concentrated salt by downregulating bitter taste sensor TRPM5. It suggests that enhancing TRPM5 function might antagonize excessive salt intake and high salt-induced hypertension.
Collapse
Affiliation(s)
- Yuanting Cui
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Hao Wu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Qiang Li
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Jianwen Liao
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Peng Gao
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Fang Sun
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Hexuan Zhang
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Zongshi Lu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Xiao Wei
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Chengkang He
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Tianyi Ma
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Xing Wei
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Xiaowei Chen
- Brain Research Center (X.C.), Third Military Medical University, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital (H.Z.), Third Military Medical University, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, China (G.Y.)
| | - Daoyan Liu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| | - Zhiming Zhu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (Y.C., H.W., Q.L., J.L., P.G., F.S., H.Z., Z.L., X.W., C.H., T.M., X.W., D.L., Z.Z.), Third Military Medical University, China
| |
Collapse
|
23
|
Zhou Y, Pan P, Tan ZY, Ji YH. Voltage-gated Sodium Channels in Sensory Information Processing. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:273-278. [DOI: 10.2174/1871527317666180627114849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/04/2018] [Accepted: 05/09/2018] [Indexed: 11/22/2022]
Abstract
Objective & Background:
Voltage-gated sodium channels (VGSCs) and potassium channels
are critical in the generation of action potentials in the nervous system. VGSCs and potassium
channels play important roles in the five fundamental senses of vision, audition, olfaction, taste and
touch. Dysfunctional VGSCs are associated with clinical sensory symptoms, such as hyperpselaphesia,
parosphresia, and so on.
Conclusion:
This short review highlights the recent advances in the study of VGSCs in sensory information
processing and discusses the potential role of VGSCs to serve as pharmacological targets for
the treatment of sensory system diseases.
Collapse
Affiliation(s)
- You Zhou
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| | - Ping Pan
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| | - Zhi-Yong Tan
- Department of Pharmacology and Toxicology and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Yong-Hua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
24
|
CALHM1/CALHM3 channel is intrinsically sorted to the basolateral membrane of epithelial cells including taste cells. Sci Rep 2019; 9:2681. [PMID: 30804437 PMCID: PMC6390109 DOI: 10.1038/s41598-019-39593-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/12/2018] [Indexed: 12/20/2022] Open
Abstract
The CALHM1/CALHM3 channel in the basolateral membrane of polarized taste cells mediates neurotransmitter release. However, mechanisms regulating its localization remain unexplored. Here, we identified CALHM1/CALHM3 in the basolateral membrane of type II taste cells in discrete puncta localized close to afferent nerve fibers. As in taste cells, CALHM1/CALHM3 was present in the basolateral membrane of model epithelia, although it was distributed throughout the membrane and did not show accumulation in puncta. We identified canonical basolateral sorting signals in CALHM1 and CALHM3: tyrosine-based and dileucine motifs. However, basolateral sorting remained intact in mutated channels lacking those signals, suggesting that non-canonical signals reside elsewhere. Our study demonstrates intrinsic basolateral sorting of CALHM channels in polarized cells, and provides mechanistic insights.
Collapse
|
25
|
Xu J, Lewandowski BC, Miyazawa T, Shoji Y, Yee K, Bryant BP. Spilanthol Enhances Sensitivity to Sodium in Mouse Taste Bud Cells. Chem Senses 2019; 44:91-103. [PMID: 30364996 PMCID: PMC6350677 DOI: 10.1093/chemse/bjy069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Overconsumption of NaCl has been linked to increased hypertension-related morbidity. Compounds that can enhance NaCl responses in taste cells could help reduce human NaCl consumption without sacrificing perceived saltiness. Spilanthol is an unsaturated alkylamide isolated from the Jambu plant (Acmella oleracea) that can induce tingling, pungency, and numbing in the mouth. Structurally similar fatty acid amides, such as sanshool, elicit numbing and tingling sensations by inhibiting 2-pore-domain potassium leak channels on trigeminal sensory neurons. Even when insufficient to induce action potential firing, leak current inhibition causes depolarization and increased membrane resistance, which combine to make cells more sensitive to subsequent depolarizing stimuli, such as NaCl. Using calcium imaging, we tested whether spilanthol alters sensitivity to NaCl in isolated circumvallate taste bud cells and trigeminal sensory neurons of mice (Mus musculus). Micromolar spilanthol elicited little to no response in taste bud cells or trigeminal neurons. These same perithreshold concentrations of spilanthol significantly enhanced responses to NaCl (140 and 200 mM) in taste bud cells. Trigeminal neurons, however, exhibited response enhancement only at the highest concentrations of NaCl and spilanthol tested. Using a combination of potassium depolarization, immunohistochemistry, and Trpm5-GFP and Tas1r3-GFP mice to characterize taste bud cells by type, we found spilanthol enhancement of NaCl responses most prevalent in NaCl-responsive type III cells, and commonly observed in NaCl-responsive type II cells. Our results indicate that spilanthol enhances NaCl responses in taste bud cells and point to a family of compounds that may have utility as salty taste enhancers.
Collapse
Affiliation(s)
- Jiang Xu
- Monell Chemical Senses Center, Philadelphia, PA , USA
| | | | | | - Yasutaka Shoji
- Ogawa & Co. Ltd., Nihonbashi Honcho Chuo-ku, Tokyo, Japan
| | - Karen Yee
- Monell Chemical Senses Center, Philadelphia, PA , USA
| | | |
Collapse
|
26
|
Abstract
The study of taste has been guided throughout much of its history by the conceptual framework of psychophysics, where the focus was on quantification of the subjective experience of the taste sensations. By the mid-20th century, data from physiologic studies had accumulated sufficiently to assemble a model for the function of receptors that must mediate the initial stimulus of tastant molecules in contact with the tongue. But the study of taste as a receptor-mediated event did not gain momentum until decades later when the actual receptor proteins and attendant signaling mechanisms were identified and localized to the highly specialized taste-responsive cells of the tongue. With those discoveries a new opportunity to examine taste as a function of receptor activity has come into focus. Pharmacology is the science designed specifically for the experimental interrogation and quantitative characterization of receptor function at all levels of inquiry from molecules to behavior. This review covers the history of some of the major concepts that have shaped thinking and experimental approaches to taste, the seminal discoveries that have led to elucidation of receptors for taste, and how applying principles of receptor pharmacology can enhance understanding of the mechanisms of taste physiology and perception.
Collapse
Affiliation(s)
- R Kyle Palmer
- Opertech Bio, Inc., Pennovation Center, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Romanov RA, Lasher RS, High B, Savidge LE, Lawson A, Rogachevskaja OA, Zhao H, Rogachevsky VV, Bystrova MF, Churbanov GD, Adameyko I, Harkany T, Yang R, Kidd GJ, Marambaud P, Kinnamon JC, Kolesnikov SS, Finger TE. Chemical synapses without synaptic vesicles: Purinergic neurotransmission through a CALHM1 channel-mitochondrial signaling complex. Sci Signal 2018; 11:11/529/eaao1815. [PMID: 29739879 DOI: 10.1126/scisignal.aao1815] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Conventional chemical synapses in the nervous system involve a presynaptic accumulation of neurotransmitter-containing vesicles, which fuse with the plasma membrane to release neurotransmitters that activate postsynaptic receptors. In taste buds, type II receptor cells do not have conventional synaptic features but nonetheless show regulated release of their afferent neurotransmitter, ATP, through a large-pore, voltage-gated channel, CALHM1. Immunohistochemistry revealed that CALHM1 was localized to points of contact between the receptor cells and sensory nerve fibers. Ultrastructural and super-resolution light microscopy showed that the CALHM1 channels were consistently associated with distinctive, large (1- to 2-μm) mitochondria spaced 20 to 40 nm from the presynaptic membrane. Pharmacological disruption of the mitochondrial respiratory chain limited the ability of taste cells to release ATP, suggesting that the immediate source of released ATP was the mitochondrion rather than a cytoplasmic pool of ATP. These large mitochondria may serve as both a reservoir of releasable ATP and the site of synthesis. The juxtaposition of the large mitochondria to areas of membrane displaying CALHM1 also defines a restricted compartment that limits the influx of Ca2+ upon opening of the nonselective CALHM1 channels. These findings reveal a distinctive organelle signature and functional organization for regulated, focal release of purinergic signals in the absence of synaptic vesicles.
Collapse
Affiliation(s)
- Roman A Romanov
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia.,Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria.,Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia
| | - Robert S Lasher
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Brigit High
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Logan E Savidge
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Adam Lawson
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Olga A Rogachevskaja
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia
| | - Haitian Zhao
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Vadim V Rogachevsky
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia.,United Pushchino Center for Electron Microscopy, Pushchino, Moscow Region 142290, Russia
| | - Marina F Bystrova
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia
| | - Gleb D Churbanov
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia
| | - Igor Adameyko
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria.,Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria.,Department of Neuroscience, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Ruibiao Yang
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Grahame J Kidd
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, and 3D-Electron Microscopy, Renovo Neural Inc., Cleveland, OH 44195, USA
| | - Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - John C Kinnamon
- Rocky Mountain Taste and Smell Center, Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Stanislav S Kolesnikov
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia.
| | - Thomas E Finger
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
28
|
Influence of medications on taste and smell. World J Otorhinolaryngol Head Neck Surg 2018; 4:84-91. [PMID: 30035266 PMCID: PMC6051304 DOI: 10.1016/j.wjorl.2018.02.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/27/2018] [Indexed: 01/11/2023] Open
Abstract
Medications frequently have chemosensory side effects that can adversely affect compliance with medical treatment regimens. Hundreds of drugs have been reported to induce unpleasant tastes and/or odors as well as altered chemosensations when administered alone or in combination with other medications. Some chemosensory complaints are due to the sensory properties of the drug itself such as aversive bitter and metallic tastes. However, most chemosensory side effects of drugs are due to alterations in the transduction pathways, biochemical targets, enzymes, and transporters by the offending medications. Studies of chemosensory perception in medicated older individuals have found that taste and smell loss is greatest for those consuming the largest number of prescription drugs. There are no standard treatments for drug-induced chemosensory disorders because each drug has unique biological effects. However, there are a few treatment options to ameliorate chemosensory alterations including addition of simulated flavors to food to compensate for losses and to override offending tastes and smells.
Collapse
|
29
|
Sodium channel Na V1.3 is important for enterochromaffin cell excitability and serotonin release. Sci Rep 2017; 7:15650. [PMID: 29142310 PMCID: PMC5688111 DOI: 10.1038/s41598-017-15834-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/01/2017] [Indexed: 12/28/2022] Open
Abstract
In the gastrointestinal (GI) epithelium, enterochromaffin (EC) cells are enteroendocrine cells responsible for producing >90% of the body's serotonin (5-hydroxytryptamine, 5-HT). However, the molecular mechanisms of EC cell function are poorly understood. Here, we found that EC cells in mouse primary cultures fired spontaneous bursts of action potentials. We examined the repertoire of voltage-gated sodium channels (NaV) in fluorescence-sorted mouse EC cells and found that Scn3a was highly expressed. Scn3a-encoded NaV1.3 was specifically and densely expressed at the basal side of both human and mouse EC cells. Using electrophysiology, we found that EC cells expressed robust NaV1.3 currents, as determined by their biophysical and pharmacologic properties. NaV1.3 was not only critical for generating action potentials in EC cells, but it was also important for regulating 5-HT release by these cells. Therefore, EC cells use Scn3a-encoded voltage-gated sodium channel NaV1.3 for electrical excitability and 5-HT release. NaV1.3-dependent electrical excitability and its contribution to 5-HT release is a novel mechanism of EC cell function.
Collapse
|
30
|
Ramos-Lopez O, Arpón A, Riezu-Boj JI, Milagro FI, Mansego ML, Martinez JA. DNA methylation patterns at sweet taste transducing genes are associated with BMI and carbohydrate intake in an adult population. Appetite 2017; 120:230-239. [PMID: 28888730 DOI: 10.1016/j.appet.2017.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/09/2017] [Accepted: 09/05/2017] [Indexed: 02/07/2023]
Abstract
Individual differences in taste perception may influence appetite, dietary intakes, and subsequently, disease risk. Correlations of DNA methylation patterns at taste transducing genes with BMI and dietary intakes were studied. A nutriepigenomic analysis within the Methyl Epigenome Network Association (MENA) project was conducted in 474 adults. DNA methylation in peripheral white blood cells was analyzed by a microarray approach. KEGG pathway analyses were performed concerning the characterization and discrimination of genes involved in the taste transduction pathway. Adjusted FDR values (p < 0.0001) were used to select those CpGs that showed best correlation with BMI. A total of 29 CpGs at taste transducing genes met the FDR criteria. However, only 12 CpGs remained statistically significant after linear regression analyses adjusted for age and sex. These included cg15743657 (TAS1R2), cg02743674 (TRPM5), cg01790523 (SCN9A), cg15947487 (CALHM1), cg11658986 (ADCY6), cg04149773 (ADCY6), cg02841941 (P2RY1), cg02315111 (P2RX2), cg08273233 (HTR1E), cg14523238 (GABBR2), cg12315353 (GABBR1) and cg05579652 (CACNA1C). Interestingly, most of them were implicated in the sweet taste signaling pathway, except CACNA1C (sour taste). In addition, TAS1R2 methylation at cg15743657 was strongly correlated with total energy (p < 0.0001) and carbohydrate intakes (p < 0.0001). This study suggests that methylation in genes related to sweet taste could be an epigenetic mechanism associated with obesity.
Collapse
Affiliation(s)
- O Ramos-Lopez
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain
| | - A Arpón
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain
| | - J I Riezu-Boj
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - F I Milagro
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Carlos III Institute, Madrid, Spain
| | - M L Mansego
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain
| | - J A Martinez
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Carlos III Institute, Madrid, Spain; Madrid Institute of Advanced Studies (IMDEA Food), Madrid, Spain.
| | | |
Collapse
|
31
|
Yamaci RF, Fraser SP, Battaloglu E, Kaya H, Erguler K, Foster CS, Djamgoz MB. Neonatal Nav1.5 protein expression in normal adult human tissues and breast cancer. Pathol Res Pract 2017; 213:900-907. [DOI: 10.1016/j.prp.2017.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 05/03/2017] [Accepted: 06/04/2017] [Indexed: 01/15/2023]
|
32
|
Bolz F, Kasper S, Bufe B, Zufall F, Pyrski M. Organization and Plasticity of Sodium Channel Expression in the Mouse Olfactory and Vomeronasal Epithelia. Front Neuroanat 2017; 11:28. [PMID: 28420967 PMCID: PMC5376585 DOI: 10.3389/fnana.2017.00028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
To understand the molecular basis of neuronal excitation in the mammalian olfactory system, we conducted a systematic analysis of the organization of voltage-gated sodium (Nav) channel subunits in the main olfactory epithelium (MOE) and vomeronasal organ (VNO) of adult mice. We also analyzed changes in Nav channel expression during development in these two systems and during regeneration of the MOE. Quantitative PCR shows that Nav1.7 is the predominant isoform in both adult MOE and VNO. We detected pronounced immunoreactivity for Nav1.7 and Nav1.3 in axons of olfactory and vomeronasal sensory neurons (VSNs). Analysis of Nav1.2 and Nav1.6 revealed an unexpected subsystem-specific distribution. In the MOE, these Nav channels are absent from olfactory sensory neurons (OSNs) but present in non-neuronal olfactory cell types. In the VNO, Nav1.2 and Nav1.6 are confined to VSNs, with Nav1.2-immunoreactive somata solely present in the basal layer of the VNO. The subcellular localization of Nav1.3 and Nav1.7 in OSNs can change dramatically during periods of heightened plasticity in the MOE. During the first weeks of development and during regeneration of the olfactory epithelium following chemical lesion, expression of Nav1.3 and Nav1.7 is transiently enhanced in the somata of mature OSNs. Our results demonstrate a highly complex organization of Nav channel expression in the mouse olfactory system, with specific commonalities but also differences between the MOE and the VNO. On the basis of their subcellular localization, Nav1.3 and Nav1.7 should play major roles in action potential propagation in both MOE and VNO, whereas Nav1.2 and Nav1.6 are specific to the function of VSNs. The plasticity of Nav channel expression in OSNs during early development and recovery from injury could reflect important physiological requirements in a variety of activity-dependent mechanisms.
Collapse
Affiliation(s)
- Florian Bolz
- Center for Integrative Physiology and Molecular Medicine, Saarland UniversityHomburg, Germany
| | - Stephanie Kasper
- Center for Integrative Physiology and Molecular Medicine, Saarland UniversityHomburg, Germany
| | - Bernd Bufe
- Center for Integrative Physiology and Molecular Medicine, Saarland UniversityHomburg, Germany
| | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland UniversityHomburg, Germany
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland UniversityHomburg, Germany
| |
Collapse
|
33
|
Ma Z, Saung WT, Foskett JK. Action potentials and ion conductances in wild-type and CALHM1-knockout type II taste cells. J Neurophysiol 2017; 117:1865-1876. [PMID: 28202574 DOI: 10.1152/jn.00835.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 11/22/2022] Open
Abstract
Taste bud type II cells fire action potentials in response to tastants, triggering nonvesicular ATP release to gustatory neurons via voltage-gated CALHM1-associated ion channels. Whereas CALHM1 regulates mouse cortical neuron excitability, its roles in regulating type II cell excitability are unknown. In this study, we compared membrane conductances and action potentials in single identified TRPM5-GFP-expressing circumvallate papillae type II cells acutely isolated from wild-type (WT) and Calhm1 knockout (KO) mice. The activation kinetics of large voltage-gated outward currents were accelerated in cells from Calhm1 KO mice, and their associated nonselective tail currents, previously shown to be highly correlated with ATP release, were completely absent in Calhm1 KO cells, suggesting that CALHM1 contributes to all of these currents. Calhm1 deletion did not significantly alter resting membrane potential or input resistance, the amplitudes and kinetics of Na+ currents either estimated from action potentials or recorded from steady-state voltage pulses, or action potential threshold, overshoot peak, afterhyperpolarization, and firing frequency. However, Calhm1 deletion reduced the half-widths of action potentials and accelerated the deactivation kinetics of transient outward currents, suggesting that the CALHM1-associated conductance becomes activated during the repolarization phase of action potentials.NEW & NOTEWORTHY CALHM1 is an essential ion channel component of the ATP neurotransmitter release mechanism in type II taste bud cells. Its contribution to type II cell resting membrane properties and excitability is unknown. Nonselective voltage-gated currents, previously associated with ATP release, were absent in cells lacking CALHM1. Calhm1 deletion was without effects on resting membrane properties or voltage-gated Na+ and K+ channels but contributed modestly to the kinetics of action potentials.
Collapse
Affiliation(s)
- Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Wint Thu Saung
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Thennavan A, Sharma M, Chandrashekar C, Hunter K, Radhakrishnan R. Exploring the potential of laser capture microdissection technology in integrated oral biosciences. Oral Dis 2016; 23:737-748. [DOI: 10.1111/odi.12578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/10/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022]
Affiliation(s)
- A Thennavan
- Department of Oral Pathology; Manipal College of Dental Sciences; Manipal Karnataka India
| | - M Sharma
- Pacific Academy of Higher Education and Research (PAHER) University; Udaipur Rajasthan India
- Department of Oral Pathology; ITS Dental College; Hospital and Research Center; Greater Noida India
| | - C Chandrashekar
- Department of Oral Pathology; Manipal College of Dental Sciences; Manipal Karnataka India
| | - K Hunter
- School of Clinical Dentistry; The University of Sheffield; Sheffield UK
| | - R Radhakrishnan
- Department of Oral Pathology; Manipal College of Dental Sciences; Manipal Karnataka India
- School of Clinical Dentistry; The University of Sheffield; Sheffield UK
| |
Collapse
|
35
|
Iamshanova O, Mariot P, Lehen'kyi V, Prevarskaya N. Comparison of fluorescence probes for intracellular sodium imaging in prostate cancer cell lines. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:765-777. [PMID: 27660079 PMCID: PMC5045488 DOI: 10.1007/s00249-016-1173-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/23/2016] [Accepted: 09/02/2016] [Indexed: 10/25/2022]
Abstract
Sodium (Na+) ions are known to regulate many signaling pathways involved in both physiological and pathological conditions. In particular, alterations in intracellular concentrations of Na+ and corresponding changes in membrane potential are known to be major actors of cancer progression to metastatic phenotype. Though the functionality of Na+ channels and the corresponding Na+ currents can be investigated using the patch-clamp technique, the latter is rather invasive and a technically difficult method to study intracellular Na+ transients compared to Na+ fluorescence imaging. Despite the fact that Na+ signaling is considered an important controller of cancer progression, only few data using Na+ imaging approaches are available so far, suggesting the persisting challenge within the scientific community. In this study, we describe in detail the approach for application of Na+ imaging technique to measure intracellular Na+ variations in human prostate cancer cells. Accordingly, we used three Na+-specific fluorescent dyes-Na+-binding benzofuran isophthalate (SBFI), CoroNa™ Green (Corona) and Asante NaTRIUM Green-2 (ANG-2). These dyes have been assessed for optimal loading conditions, dissociation constant and working range after different calibration methods, and intracellular Na+ sensitivity, in order to determine which probe can be considered as the most reliable to visualize Na+ fluctuations in vitro.
Collapse
Affiliation(s)
- Oksana Iamshanova
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, Université des Sciences et Technologies de Lille, 59656, Villeneuve d'Ascq, France
| | - Pascal Mariot
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, Université des Sciences et Technologies de Lille, 59656, Villeneuve d'Ascq, France
| | - V'yacheslav Lehen'kyi
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, Université des Sciences et Technologies de Lille, 59656, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, Université des Sciences et Technologies de Lille, 59656, Villeneuve d'Ascq, France.
| |
Collapse
|
36
|
Carey RM, Adappa ND, Palmer JN, Lee RJ, Cohen NA. Taste Receptors: Regulators of Sinonasal Innate Immunity. Laryngoscope Investig Otolaryngol 2016; 1:88-95. [PMID: 27819057 PMCID: PMC5089074 DOI: 10.1002/lio2.26] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Taste receptors in the oral cavity guide our preferences for foods, preventing toxic ingestions and encouraging proper nutrient consumption. More recently, expression of taste receptors has been demonstrated in other locations throughout the body, including the airway, gastrointestinal tract, pancreas, and brain. The extent and specific roles of extraoral taste receptors are largely unknown, but a growing body of evidence suggests that taste receptors in the airway serve a critical role in sensing bacteria and regulating innate immunity. This review will focus on the function of bitter and sweet taste receptors in the human airway, with particular emphasis on T2R38, a bitter taste receptor found in sinonasal ciliated cells, and the bitter and sweet receptors found on specialized sinonasal solitary chemosensory cells. The importance of these novel taste receptor‐immune circuits in the human airway and their clinical relevance in airway disease will also be reviewed.
Collapse
Affiliation(s)
- Ryan M Carey
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5 floor, Philadelphia, PA 19104
| | - Nithin D Adappa
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5 floor, Philadelphia, PA 19104
| | - James N Palmer
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5 floor, Philadelphia, PA 19104
| | - Robert J Lee
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5 floor, Philadelphia, PA 19104
| | - Noam A Cohen
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Ravdin Building, 5 floor, Philadelphia, PA 19104
| |
Collapse
|
37
|
Kumarhia D, He L, McCluskey LP. Inflammatory stimuli acutely modulate peripheral taste function. J Neurophysiol 2016; 115:2964-75. [PMID: 27009163 DOI: 10.1152/jn.01104.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/23/2016] [Indexed: 12/30/2022] Open
Abstract
Inflammation-mediated changes in taste perception can affect health outcomes in patients, but little is known about the underlying mechanisms. In the present work, we hypothesized that proinflammatory cytokines directly modulate Na(+) transport in taste buds. To test this, we measured acute changes in Na(+) flux in polarized fungiform taste buds loaded with a Na(+) indicator dye. IL-1β elicited an amiloride-sensitive increase in Na(+) transport in taste buds. In contrast, TNF-α dramatically and reversibly decreased Na(+) flux in polarized taste buds via amiloride-sensitive and amiloride-insensitive Na(+) transport systems. The speed and partial amiloride sensitivity of these changes in Na(+) flux indicate that IL-1β and TNF-α modulate epithelial Na(+) channel (ENaC) function. A portion of the TNF-mediated decrease in Na(+) flux is also blocked by the TRPV1 antagonist capsazepine, although TNF-α further reduced Na(+) transport independently of both amiloride and capsazepine. We also assessed taste function in vivo in a model of infection and inflammation that elevates these and additional cytokines. In rats administered systemic lipopolysaccharide (LPS), CT responses to Na(+) were significantly elevated between 1 and 2 h after LPS treatment. Low, normally preferred concentrations of NaCl and sodium acetate elicited high response magnitudes. Consistent with this outcome, codelivery of IL-1β and TNF-α enhanced Na(+) flux in polarized taste buds. These results demonstrate that inflammation elicits swift changes in Na(+) taste function, which may limit salt consumption during illness.
Collapse
Affiliation(s)
- Devaki Kumarhia
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia; and Graduate Program in Molecular Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Lianying He
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| | - Lynnette Phillips McCluskey
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| |
Collapse
|
38
|
Boggs K, Venkatesan N, Mederacke I, Komatsu Y, Stice S, Schwabe RF, Mistretta CM, Mishina Y, Liu HX. Contribution of Underlying Connective Tissue Cells to Taste Buds in Mouse Tongue and Soft Palate. PLoS One 2016; 11:e0146475. [PMID: 26741369 PMCID: PMC4704779 DOI: 10.1371/journal.pone.0146475] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023] Open
Abstract
Taste buds, the sensory organs for taste, have been described as arising solely from the surrounding epithelium, which is in distinction from other sensory receptors that are known to originate from neural precursors, i.e., neural ectoderm that includes neural crest (NC). Our previous study suggested a potential contribution of NC derived cells to early immature fungiform taste buds in late embryonic (E18.5) and young postnatal (P1-10) mice. In the present study we demonstrated the contribution of the underlying connective tissue (CT) to mature taste buds in mouse tongue and soft palate. Three independent mouse models were used for fate mapping of NC and NC derived connective tissue cells: (1) P0-Cre/R26-tdTomato (RFP) to label NC, NC derived Schwann cells and derivatives; (2) Dermo1-Cre/RFP to label mesenchymal cells and derivatives; and (3) Vimentin-CreER/mGFP to label Vimentin-expressing CT cells and derivatives upon tamoxifen treatment. Both P0-Cre/RFP and Dermo1-Cre/RFP labeled cells were abundant in mature taste buds in lingual taste papillae and soft palate, but not in the surrounding epithelial cells. Concurrently, labeled cells were extensively distributed in the underlying CT. RFP signals were seen in the majority of taste buds and all three types (I, II, III) of differentiated taste bud cells, with the neuronal-like type III cells labeled at a greater proportion. Further, Vimentin-CreER labeled cells were found in the taste buds of 3-month-old mice whereas Vimentin immunoreactivity was only seen in the CT. Taken together, our data demonstrate a previously unrecognized origin of taste bud cells from the underlying CT, a conceptually new finding in our knowledge of taste bud cell derivation, i.e., from both the surrounding epithelium and the underlying CT that is primarily derived from NC.
Collapse
Affiliation(s)
- Kristin Boggs
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| | - Nandakumar Venkatesan
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| | - Ingmar Mederacke
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Yoshihiro Komatsu
- Department of Pediatrics, Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Steve Stice
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| | - Robert F. Schwabe
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Charlotte M. Mistretta
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, United States of America
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, United States of America
| | - Hong-Xiang Liu
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States of America
| |
Collapse
|
39
|
Kim JM, Ren D, Reverter A, Roura E. A regulatory gene network related to the porcine umami taste receptor (TAS1R1/TAS1R3). Anim Genet 2015; 47:114-9. [PMID: 26554867 DOI: 10.1111/age.12374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2015] [Indexed: 11/28/2022]
Abstract
Taste perception plays an important role in the mediation of food choices in mammals. The first porcine taste receptor genes identified, sequenced and characterized, TAS1R1 and TAS1R3, were related to the dimeric receptor for umami taste. However, little is known about their regulatory network. The objective of this study was to unfold the genetic network involved in porcine umami taste perception. We performed a meta-analysis of 20 gene expression studies spanning 480 porcine microarray chips and screened 328 taste-related genes by selective mining steps among the available 12,320 genes. A porcine umami taste-specific regulatory network was constructed based on the normalized coexpression data of the 328 genes across 27 tissues. From the network, we revealed the 'taste module' and identified a coexpression cluster for the umami taste according to the first connector with the TAS1R1/TAS1R3 genes. Our findings identify several taste-related regulatory genes and extend previous genetic background of porcine umami taste.
Collapse
Affiliation(s)
- J M Kim
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation Hartley Teakle 83, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - D Ren
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation Hartley Teakle 83, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - A Reverter
- CSIRO Agriculture Flagship, Queensland Bioscience Precinct, 306 Carmody Road, St Lucia, Queensland, 4067, Australia
| | - E Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation Hartley Teakle 83, The University of Queensland, St Lucia, Queensland, 4072, Australia
| |
Collapse
|
40
|
Abstract
Levels of obesity have reached epidemic proportions on a global scale, which has led to considerable increases in health problems and increased risk of several diseases, including cardiovascular and pulmonary diseases, cancer and diabetes mellitus. People with obesity consume more food than is needed to maintain an ideal body weight, despite the discrimination that accompanies being overweight and the wealth of available information that overconsumption is detrimental to health. The relationship between energy expenditure and energy intake throughout an individual's lifetime is far more complicated than previously thought. An improved comprehension of the relationships between taste, palatability, taste receptors and hedonic responses to food might lead to increased understanding of the biological underpinnings of energy acquisition, as well as why humans sometimes eat more than is needed and more than we know is healthy. This Review discusses the role of taste receptors in the tongue, gut, pancreas and brain and their hormonal involvement in taste perception, as well as the relationship between taste perception, overeating and the development of obesity.
Collapse
Affiliation(s)
- Sara Santa-Cruz Calvo
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Biomedical Research Center, Room 09B133, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Biomedical Research Center, Room 09B133, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, USA
| |
Collapse
|
41
|
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: 9.6] [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
|
42
|
Feng P, Chai J, Zhou M, Simon N, Huang L, Wang H. Interleukin-10 is produced by a specific subset of taste receptor cells and critical for maintaining structural integrity of mouse taste buds. J Neurosci 2014; 34:2689-701. [PMID: 24523558 PMCID: PMC3921433 DOI: 10.1523/jneurosci.3074-13.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/17/2013] [Accepted: 01/11/2014] [Indexed: 12/21/2022] Open
Abstract
Although inflammatory responses are a critical component in defense against pathogens, too much inflammation is harmful. Mechanisms have evolved to regulate inflammation, including modulation by the anti-inflammatory cytokine interleukin-10 (IL-10). Previously we have shown that taste buds express various molecules involved in innate immune responses, including the proinflammatory cytokine tumor necrosis factor (TNF). Here, using a reporter mouse strain, we show that taste cells also express the anti-inflammatory cytokine IL-10. Remarkably, IL-10 is produced by only a specific subset of taste cells, which are different from the TNF-producing cells in mouse circumvallate and foliate taste buds: IL-10 expression was found exclusively in the G-protein gustducin-expressing bitter receptor cells, while TNF was found in sweet and umami receptor cells as reported previously. In contrast, IL-10R1, the ligand-binding subunit of the IL-10 receptor, is predominantly expressed by TNF-producing cells, suggesting a novel cellular hierarchy for regulating TNF production and effects in taste buds. In response to inflammatory challenges, taste cells can increase IL-10 expression both in vivo and in vitro. These findings suggest that taste buds use separate populations of taste receptor cells that coincide with sweet/umami and bitter taste reception to modulate local inflammatory responses, a phenomenon that has not been previously reported. Furthermore, IL-10 deficiency in mice leads to significant reductions in the number and size of taste buds, as well as in the number of taste receptor cells per taste bud, suggesting that IL-10 plays critical roles in maintaining structural integrity of the peripheral gustatory system.
Collapse
Affiliation(s)
- Pu Feng
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Jinghua Chai
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Minliang Zhou
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Nirvine Simon
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Liquan Huang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| | - Hong Wang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania 19104
| |
Collapse
|
43
|
Kimura K, Ohtubo Y, Tateno K, Takeuchi K, Kumazawa T, Yoshii K. Cell-type-dependent action potentials and voltage-gated currents in mouse fungiform taste buds. Eur J Neurosci 2013; 39:24-34. [PMID: 24152110 DOI: 10.1111/ejn.12388] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/11/2013] [Indexed: 01/24/2023]
Abstract
Taste receptor cells fire action potentials in response to taste substances to trigger non-exocytotic neurotransmitter release in type II cells and exocytotic release in type III cells. We investigated possible differences between these action potentials fired by mouse taste receptor cells using in situ whole-cell recordings, and subsequently we identified their cell types immunologically with cell-type markers, an IP3 receptor (IP3 R3) for type II cells and a SNARE protein (SNAP-25) for type III cells. Cells not immunoreactive to these antibodies were examined as non-IRCs. Here, we show that type II cells and type III cells fire action potentials using different ionic mechanisms, and that non-IRCs also fire action potentials with either of the ionic mechanisms. The width of action potentials was significantly narrower and their afterhyperpolarization was deeper in type III cells than in type II cells. Na(+) current density was similar in type II cells and type III cells, but it was significantly smaller in non-IRCs than in the others. Although outwardly rectifying current density was similar between type II cells and type III cells, tetraethylammonium (TEA) preferentially suppressed the density in type III cells and the majority of non-IRCs. Our mathematical model revealed that the shape of action potentials depended on the ratio of TEA-sensitive current density and TEA-insensitive current one. The action potentials of type II cells and type III cells under physiological conditions are discussed.
Collapse
Affiliation(s)
- Kenji Kimura
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Hibikino 2-4, Kitakyushu-shi, 808-0196, Japan
| | | | | | | | | | | |
Collapse
|
44
|
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: 5.8] [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
|
45
|
Taruno A, Matsumoto I, Ma Z, Marambaud P, Foskett JK. How do taste cells lacking synapses mediate neurotransmission? CALHM1, a voltage-gated ATP channel. Bioessays 2013; 35:1111-8. [PMID: 24105910 DOI: 10.1002/bies.201300077] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CALHM1 was recently demonstrated to be a voltage-gated ATP-permeable ion channel and to serve as a bona fide conduit for ATP release from sweet-, umami-, and bitter-sensing type II taste cells. Calhm1 is expressed in taste buds exclusively in type II cells and its product has structural and functional similarities with connexins and pannexins, two families of channel protein candidates for ATP release by type II cells. Calhm1 knockout in mice leads to loss of perception of sweet, umami, and bitter compounds and to impaired gustatory nerve responses to these tastants. These new studies validate the concept of ATP as the primary neurotransmitter from type II cells to gustatory neurons. Furthermore, they identify voltage-gated ATP release through CALHM1 as an essential molecular mechanism of ATP release in taste buds. We discuss these new findings, as well as unresolved issues in peripheral taste signaling that we hope will stimulate future research.
Collapse
Affiliation(s)
- Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | |
Collapse
|
46
|
Heimann D, Lötsch J, Hummel T, Doehring A, Oertel BG. Linkage between increased nociception and olfaction via a SCN9A haplotype. PLoS One 2013; 8:e68654. [PMID: 23874707 PMCID: PMC3707874 DOI: 10.1371/journal.pone.0068654] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/30/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND AIMS Mutations reducing the function of Nav1.7 sodium channels entail diminished pain perception and olfactory acuity, suggesting a link between nociception and olfaction at ion channel level. We hypothesized that if such link exists, it should work in both directions and gain-of-function Nav1.7 mutations known to be associated with increased pain perception should also increase olfactory acuity. METHODS SCN9A variants were assessed known to enhance pain perception and found more frequently in the average population. Specifically, carriers of SCN9A variants rs41268673C>A (P610T; n = 14) or rs6746030C>T (R1150W; n = 21) were compared with non-carriers (n = 40). Olfactory function was quantified by assessing odor threshold, odor discrimination and odor identification using an established olfactory test. Nociception was assessed by measuring pain thresholds to experimental nociceptive stimuli (punctate and blunt mechanical pressure, heat and electrical stimuli). RESULTS The number of carried alleles of the non-mutated SCN9A haplotype rs41268673C/rs6746030C was significantly associated with the comparatively highest olfactory threshold (0 alleles: threshold at phenylethylethanol dilution step 12 of 16 (n = 1), 1 allele: 10.6±2.6 (n = 34), 2 alleles: 9.5±2.1 (n = 40)). The same SCN9A haplotype determined the pain threshold to blunt pressure stimuli (0 alleles: 21.1 N/m(2), 1 allele: 29.8±10.4 N/m(2), 2 alleles: 33.5±10.2 N/m(2)). CONCLUSIONS The findings established a working link between nociception and olfaction via Nav1.7 in the gain-of-function direction. Hence, together with the known reduced olfaction and pain in loss-of-function mutations, a bidirectional genetic functional association between nociception and olfaction exists at Nav1.7 level.
Collapse
Affiliation(s)
- Dirk Heimann
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt am Main, Germany
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt am Main, Germany
- Fraunhofer Institute of Molecular Biology and Applied Ecology - Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt am Main, Germany
| | - Thomas Hummel
- Smell and Taste Clinic, Department of Otorhinolaryngology, University of Dresden Medical School, Dresden, Germany
| | - Alexandra Doehring
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt am Main, Germany
| | - Bruno G. Oertel
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt am Main, Germany
- Fraunhofer Institute of Molecular Biology and Applied Ecology - Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt am Main, Germany
| |
Collapse
|
47
|
Romanov RA, Rogachevskaja OA, Bystrova MF, Kolesnikov SS. Electrical excitability of taste cells. Mechanisms and possible physiological significance. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2012. [DOI: 10.1134/s1990747812010126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
48
|
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: 0.8] [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
|
49
|
Soe AK, Nahavandi S, Khoshmanesh K. Neuroscience goes on a chip. Biosens Bioelectron 2012; 35:1-13. [DOI: 10.1016/j.bios.2012.02.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 02/02/2012] [Accepted: 02/06/2012] [Indexed: 01/09/2023]
|
50
|
Ohtubo Y, Iwamoto M, Yoshii K. Subtype-dependent postnatal development of taste receptor cells in mouse fungiform taste buds. Eur J Neurosci 2012; 35:1661-71. [PMID: 22462540 DOI: 10.1111/j.1460-9568.2012.08068.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Taste buds contain two types of taste receptor cells, inositol 1,4,5-triphosphate receptor type 3-immunoreactive cells (type II cells) and synaptosomal-associating protein-25-immunoreactive cells (type III cells). We investigated their postnatal development in mouse fungiform taste buds immunohistochemically and electrophysiologically. The cell density, i.e. the number of cells per taste bud divided by the maximal area of the horizontal cross-section of the taste bud, of type II cells increased by postnatal day (PD)49, where as that of type III cells was unchanged throughout the postnatal observation period and was equal to that of the adult cells at PD1. The immunoreactivity of taste bud cell subtypes was the same as that of their respective subtypes in adult mice throughout the postnatal observation period. Almost all type II cells were immunoreactive to gustducin at PD1, and then the ratio of gustducin-immunoreactive type II cells to all type II cells decreased to a saturation level, ∼60% of all type II cells, by PD15. Type II and III cells generated voltage-gated currents similar to their respective adult cells even at PD3. These results show that infant taste receptor cells are as excitable as those of adults and propagate in a subtype-dependent manner. The relationship between the ratio of each taste receptor cell subtype to all cells and taste nerve responses are discussed.
Collapse
Affiliation(s)
- Yoshitaka Ohtubo
- Kyushu Institute of Technology, Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu, Japan
| | | | | |
Collapse
|