Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters










Publication year range
1.
Physiol Behav ; 275: 114446, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38128683

ABSTRACT

Human studies have linked stress exposure to unhealthy eating behavior. However, the mechanisms that drive stress-associated changes in eating behavior remain incompletely understood. The sense of taste plays important roles in food preference and intake. In this study, we use a chronic social defeat stress (CSDS) model in mice to address whether chronic stress impacts taste sensation and gene expression in taste buds and the gut. Our results showed that CSDS significantly elevated circulating levels of corticosterone and acylated ghrelin while lowering levels of leptin, suggesting a change in metabolic hormones that promotes food consumption. Stressed mice substantially increased their intake of food and water 3-5 days after the stress onset and gradually gained more body weight than that of controls. Moreover, CSDS significantly decreased the expression of multiple taste receptors and signaling molecules in taste buds and reduced mRNA levels of several taste progenitor/stem cell markers and regulators. Stressed mice showed significantly reduced sensitivity and response to umami and sweet taste compounds in behavioral tests. In the small intestine, the mRNA levels of Gnat3 and Tas1r2 were elevated in CSDS mice. The increased Gnat3 was mostly localized in a type of Gnat3+ and CD45+ immune cells, suggesting changes of immune cell distribution in the gut of stressed mice. Together, our study revealed broad effects of CSDS on the peripheral taste system and the gut, which may contribute to stress-associated changes in eating behavior.


Subject(s)
Taste Buds , Taste , Mice , Humans , Animals , Taste/physiology , Social Defeat , Body Weight/physiology , Taste Buds/physiology , RNA, Messenger , Gene Expression , Stress, Psychological/genetics , Mice, Inbred C57BL
2.
Biochem Biophys Res Commun ; 682: 138-140, 2023 11 19.
Article in English | MEDLINE | ID: mdl-37806252

ABSTRACT

Coronavirus disease 19 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has remained a public health threat since late 2019. Among the strategies rapidly developed to prevent and treat COVID-19, the antiviral medication Paxlovid (nirmatrelvir/ritonavir combination) has shown remarkable efficacy in reducing viral load and relieving clinical symptoms. Unexpectedly, a persistent bitter/bad taste, referred to as "Paxlovid mouth", has been frequently noted. Consistent with this, dysgeusia (altered taste) is listed as a main adverse effect of Paxlovid based on clinical trial data. Nirmatrelvir inhibits Mpro, a SARS-CoV-2 main protease, whereas ritonavir prolongs the activity of nirmatrelvir by slowing its metabolism. Prior usage of ritonavir in other conditions has not been linked to a persistent bad taste, despite the fact that ritonavir tastes bitter. Therefore, we hypothesized that nirmatrelvir may account for Paxlovid mouth by activating one or more of the 25 human TAS2R bitter taste receptors. Here, we show that TAS2R1 is the primary bitter receptor activated by nirmatrelvir, at concentrations as low as 15 µM, which overlaps with plasma concentrations of nirmatrelvir in a subset of patients. We also show that saccharin, a non-nutritive sweetener that may block the activity of TAS2R1, has little or no effect on nirmatrelvir-stimulated TAS2R1 activity. Such findings may help identify novel strategies to alleviate Paxlovid mouth and increase treatment compliance.


Subject(s)
COVID-19 , Dysgeusia , Humans , Dysgeusia/chemically induced , Taste , Ritonavir , Mouth , Antiviral Agents/pharmacology
3.
iScience ; 26(6): 106920, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37283808

ABSTRACT

T2R bitter receptors, encoded by Tas2r genes, are not only critical for bitter taste signal transduction but also important for defense against bacteria and parasites. However, little is known about whether and how Tas2r gene expression are regulated. Here, we show that in an inflammation model mimicking bacterial infection using lipopolysaccharide, the expression of many Tas2rs was significantly upregulated and mice displayed markedly increased neural and behavioral responses to bitter compounds. Using single-cell assays for transposase-accessible chromatin with sequencing (scATAC-seq), we found that the chromatin accessibility of Tas2rs was highly celltype specific and lipopolysaccharide increased the accessibility of many Tas2rs. scATAC-seq also revealed substantial chromatin remodeling in immune response genes in taste tissue stem cells, suggesting potential long-lasting effects. Together, our results suggest an epigenetic mechanism connecting inflammation, Tas2r gene regulation, and altered bitter taste, which may explain heightened bitter taste that can occur with infections and cancer treatments.

4.
Elife ; 122023 02 28.
Article in English | MEDLINE | ID: mdl-36852482

ABSTRACT

Salt taste sensation is multifaceted: NaCl at low or high concentrations is preferably or aversively perceived through distinct pathways. Cl- is thought to participate in taste sensation through an unknown mechanism. Here, we describe Cl- ion binding and the response of taste receptor type 1 (T1r), a receptor family composing sweet/umami receptors. The T1r2a/T1r3 heterodimer from the medaka fish, currently the sole T1r amenable to structural analyses, exhibited a specific Cl- binding in the vicinity of the amino-acid-binding site in the ligand-binding domain (LBD) of T1r3, which is likely conserved across species, including human T1r3. The Cl- binding induced a conformational change in T1r2a/T1r3LBD at sub- to low-mM concentrations, similar to canonical taste substances. Furthermore, oral Cl- application to mice increased impulse frequencies of taste nerves connected to T1r-expressing taste cells and promoted their behavioral preferences attenuated by a T1r-specific blocker or T1r3 knock-out. These results suggest that the Cl- evokes taste sensations by binding to T1r, thereby serving as another preferred salt taste pathway at a low concentration.


Humans perceive taste when proteins called taste receptors on the surface of the tongue are activated by molecules of food. These receptors turn on nerve cells that send signals the brain can read as sweet, sour, salty, bitter, or umami, depending on which receptor was activated. Most animals with backbones share the same five types of taste receptors. In food, salty flavors are usually the result of adding table salt, which has two components: a sodium ion and chloride ion. The main taste receptors that signal to the brain that a food is salty become activated when they bind to the sodium ion. However, some studies have shown that salt is also perceived as sweet when eaten in minuscule amounts. It is poorly understood why this happens, but it is possible that the chloride half of salt drives the sweet taste. In 2017, scientists worked out the structure of a taste receptor from a fish, that is equivalent to the sweet receptor in humans. Curiously, one part of this receptor, known as T1r2a/T1r3LBD, was bound to a chloride ion. This prompted Atsumi, Yasumatsu et al. to think about the 'sweet' taste of salt, leading them to take a closer look at T1r2a/T1r3LBD and whether chloride could indeed activate it. Atsumi, Yasumatsu et al. used structural biology techniques to examine T1r2a/T1r3LBD and found evidence that the receptor might be binding chloride. Further biophysical experiments confirmed that chloride does indeed bind to the receptor, and that it also causes it to change shape. Usually, changes in shape are hallmarks of receptor activation, suggesting that chloride may activate T1r2a/T1r3LBD. Next, Atsumi, Yasumatsu et al. checked whether chloride could stimulate the neurons that signal when food tastes sweet, by using an approach known as electrophysiology to measure the activity of these neurons in mice. The results showed that the neurons became active when a solution containing small amounts of chloride was placed on the mouse's tongue. This activity went away when a compound that can block the receptor's activity was delivered alongside the chloride. Additionally, when mice were given a choice of plain water or water containing chloride, they seemed to prefer the latter. This confirmed that mice recognized the sweetness of chloride via the activation of sweet taste receptors and neurons. Based on these findings, Atsumi, Yasumatsu et al. propose that small amounts of salt may taste sweet because the chloride ions in the salt activate sweet taste receptors and their linked neurons. Their results also suggest that animals sense salt in many ways, likely because balanced salt levels are essential for the body to work properly. Future experiments on human taste receptors may reveal how these pathways help assess salt levels in humans.


Subject(s)
Taste Buds , Taste , Animals , Humans , Mice , Chlorides , Ligands , Sodium Chloride , Sodium Chloride, Dietary , Extracellular Space/metabolism
5.
bioRxiv ; 2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36798225

ABSTRACT

T2R bitter receptors, encoded by Tas2r genes, are not only critical for bitter taste signal transduction but also important for defense against bacteria and parasites. However, little is known about whether and how Tas2r gene expression are regulated. Here we show that, in an inflammation model mimicking bacterial infection, the expression of many Tas2rs are significantly up-regulated and mice displayed markedly increased neural and behavioral responses to bitter compounds. Using single-cell assays for transposase-accessible chromatin with sequencing (scATAC-seq), we found that the chromatin accessibility of Tas2rs was highly cell type specific and inflammation increased the accessibility of many Tas2rs . scATAC-seq also revealed substantial chromatin remodeling in immune response genes in taste tissue stem cells, suggesting potential long-term effects. Together, our results suggest an epigenetic mechanism connecting inflammation, Tas2r gene regulation, and altered bitter taste, which may explain heightened bitter taste that can occur with infections and cancer treatments.

6.
PLoS Biol ; 21(1): e3001647, 2023 01.
Article in English | MEDLINE | ID: mdl-36634039

ABSTRACT

The oral microbiome is second only to its intestinal counterpart in diversity and abundance, but its effects on taste cells remains largely unexplored. Using single-cell RNASeq, we found that mouse taste cells, in particular, sweet and umami receptor cells that express taste 1 receptor member 3 (Tas1r3), have a gene expression signature reminiscent of Microfold (M) cells, a central player in immune surveillance in the mucosa-associated lymphoid tissue (MALT) such as those in the Peyer's patch and tonsils. Administration of tumor necrosis factor ligand superfamily member 11 (TNFSF11; also known as RANKL), a growth factor required for differentiation of M cells, dramatically increased M cell proliferation and marker gene expression in the taste papillae and in cultured taste organoids from wild-type (WT) mice. Taste papillae and organoids from knockout mice lacking Spib (SpibKO), a RANKL-regulated transcription factor required for M cell development and regeneration on the other hand, failed to respond to RANKL. Taste papillae from SpibKO mice also showed reduced expression of NF-κB signaling pathway components and proinflammatory cytokines and attracted fewer immune cells. However, lipopolysaccharide-induced expression of cytokines was strongly up-regulated in SpibKO mice compared to their WT counterparts. Like M cells, taste cells from WT but not SpibKO mice readily took up fluorescently labeled microbeads, a proxy for microbial transcytosis. The proportion of taste cell subtypes are unaltered in SpibKO mice; however, they displayed increased attraction to sweet and umami taste stimuli. We propose that taste cells are involved in immune surveillance and may tune their taste responses to microbial signaling and infection.


Subject(s)
Taste Buds , Taste , Animals , Mice , Intestines , Mucous Membrane , Cytokines/metabolism , Mice, Inbred C57BL , Mice, Knockout
7.
J Neurovirol ; 29(1): 35-44, 2023 02.
Article in English | MEDLINE | ID: mdl-36719595

ABSTRACT

Clinical manifestations of human coronavirus (HCoV)-related diseases are mostly related to the respiratory system, although secondary complications such as headache, anosmia, ageusia, and myalgia have been reported. HCoV infection and replication in chemosensory cells associated with ageusia and anosmia is poorly understood. Here, we characterized HCoV-OC43 and SARS-CoV-2 infection in two types of chemosensory cells, olfactory and taste cells, with their unique molecular and histological characteristics. We first assessed HCoV-OC43 infection in in vitro cultured human olfactory epithelial cells (hOECs) and fungiform taste papilla (HBO) cells. Interestingly, while both cell types were susceptible to HCoV-OC43 infection, viral replication rates were significantly reduced in HBO cells compared to hOECs. More interestingly, while culture media from hOECs was able to produce secondary infection in Vero cells, there was very limited secondary infection from HBO cells, suggesting that HBO cells may not be able to release infectious virus. On the other hand, unlike HCoV-OC43, SARS-CoV-2 showed comparable levels of viral infection rates in both hOECs and HBO cells. Furthermore, our RT-qPCR-based gene array studies revealed that several key genes involved in taste and olfactory functions were significantly altered by SARS-CoV-2 infection. These results may suggest a possible mechanism associated with chemosensory symptoms, such as anosmia and ageusia in patients infected with SARS-CoV-2.


Subject(s)
Ageusia , COVID-19 , Coinfection , Coronavirus OC43, Human , Animals , Chlorocebus aethiops , Humans , Vero Cells , Anosmia , SARS-CoV-2 , Coronavirus OC43, Human/genetics
8.
Chem Senses ; 472022 01 01.
Article in English | MEDLINE | ID: mdl-36484118

ABSTRACT

Taste perception, initiated by activation of taste receptors in taste bud cells, is crucial for regulating nutrient intake. Genetic polymorphisms in taste receptor genes cannot fully explain the wide individual variations of taste sensitivity. Alternative splicing (AS) is a ubiquitous posttranscriptional mode of gene regulation that enriches the functional diversity of proteins. Here, we report the identification of a novel splicing variant of sweet taste receptor gene Tas1r2 (Tas1r2_∆e4) in mouse taste buds and the mechanism by which it diminishes sweet taste responses in vitro and in vivo. Skipping of Tas1r2 exon 4 in Tas1r2_∆e4 led to loss of amino acids in the extracellular Venus flytrap domain, and the truncated isoform reduced the response of sweet taste receptors (STRs) to all sweet compounds tested by generating nonfunctional T1R2/T1R3 STR heterodimers. The splicing factor PTBP1 (polypyrimidine tract-binding protein 1) promoted Tas1r2_∆e4 generation through binding to a polypyrimidine-rich splicing silencer in Tas1r2 exon 4, thus decreasing STR function and sweet taste perception in mice. Taken together, these data reveal the existence of a regulated AS event in Tas1r2 expression and its effect on sweet taste perception, providing a novel mechanism for modulating taste sensitivity at the posttranscriptional level.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins , Taste Perception , Mice , Animals , Polypyrimidine Tract-Binding Protein/genetics
9.
Brain Behav Immun ; 103: 145-153, 2022 07.
Article in English | MEDLINE | ID: mdl-35447301

ABSTRACT

Inducible nitric oxide synthase (iNOS) is expressed when cells are induced or stimulated by proinflammatory cytokines and/or bacterial lipopolysaccharide (LPS). iNOS is a downstream gene of the NF-κB pathway. Our previous studies demonstrated that five Nfkb genes are expressed in mouse taste epithelium and taste organoids. However, it is unclear whether activation of the NF-κB pathway could induce iNOS gene expression and increase nitric oxide (NO) production in taste buds. In this study, we investigated the expression of iNOS mRNA and protein after LPS stimulation. Our results showed that a subset of taste bud cells and taste neurons express iNOS proteins after LPS stimulation. In addition, isolated mouse taste epithelium can release NO after exposure to LPS ex vivo. In taste behavioral tests, the NO donor nitroprusside enhanced mouse aversive responses to salty, bitter, and sour taste compounds. The enhanced aversive responses were especially strong for salty taste. In conclusion, our results suggest that iNOS and NO may play a role in the inflammation-associated taste disturbances.


Subject(s)
Lipopolysaccharides , Taste Buds , Animals , Inflammation , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Taste Buds/metabolism
10.
J Oral Biosci ; 64(1): 155-158, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34979250

ABSTRACT

Taste-signaling proteins, which are expressed throughout the digestive tract, are involved in regulating metabolism and immunity. This study aimed to determine if these genes are expressed and altered in jejunal tissues from patients with extreme obesity who received bariatric surgery. Reverse transcription polymerase chain reaction revealed that phospholipase C beta 2 and transient receptor potential channel M5 expression was downregulated in the jejunum of patients with a body mass index above 50, whereas gustducin expression remained unchanged. Our data suggest that taste-signaling dysregulation might contribute to obesity.


Subject(s)
TRPM Cation Channels , Taste Buds , Humans , Jejunum/surgery , Obesity/genetics , TRPM Cation Channels/metabolism , Taste/genetics , Taste Buds/metabolism
11.
Stem Cell Reports ; 17(2): 369-383, 2022 02 08.
Article in English | MEDLINE | ID: mdl-34995498

ABSTRACT

Taste bud cells are renewed throughout life in a process requiring innervation. Recently, we reported that R-spondin substitutes for neuronal input for taste cell regeneration. R-spondin amplifies WNT signaling by interacting with stem-cell-expressed E3 ubiquitin ligases RNF43/ZNRF3 (negative regulators of WNT signaling) and G-protein-coupled receptors LGR4/5/6 (positive regulators of WNT signaling). Therefore, we hypothesized that RNF43/ZNRF3 may serve as a brake, controlled by gustatory neuron-produced R-spondin, for regulating taste tissue homeostasis. Here, we show that mice deficient for Rnf43/Znrf3 in KRT5-expressing epithelial stem/progenitor cells (RZ dKO) exhibited taste cell hyperplasia; in stark contrast, epithelial tissue on the tongue degenerated. WNT signaling blockade substantially reversed all these effects in RZ dKO mice. Furthermore, innervation becomes dispensable for taste cell renewal in RZ dKO mice. We thus demonstrate important but distinct functions of RNF43/ZNRF3 in regulating taste versus lingual epithelial tissue homeostasis.


Subject(s)
Epithelium/metabolism , Tongue/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Benzeneacetamides/pharmacology , Glossopharyngeal Nerve/surgery , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyridines/pharmacology , Stem Cells/cytology , Stem Cells/metabolism , Taste/physiology , Taste Buds/metabolism , Tongue/cytology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway/drug effects
12.
PLoS One ; 16(10): e0256989, 2021.
Article in English | MEDLINE | ID: mdl-34614010

ABSTRACT

The taste stimulus glucose comprises approximately half of the commercial sugar sweeteners used today, whether in the form of the di-saccharide sucrose (glucose-fructose) or half of high-fructose corn syrup (HFCS). Therefore, oral glucose has been presumed to contribute to the sweet taste of foods when combined with fructose. In light of recent rodent data on the role of oral metabolic glucose signaling, we examined psychopharmacologically whether oral glucose detection may also involve an additional pathway in humans to the traditional sweet taste transduction via the class 1 taste receptors T1R2/T1R3. In a series of experiments, we first compared oral glucose detection thresholds to sucralose thresholds without and with addition of the T1R receptor inhibitor Na-lactisole. Next, we compared oral detection thresholds of glucose to sucralose and to the non-metabolizable glucose analog, α-methyl-D-glucopyranoside (MDG) without and with the addition of the glucose co-transport component sodium (NaCl). Finally, we compared oral detection thresholds for glucose, MDG, fructose, and sucralose without and with the sodium-glucose co-transporter (SGLT) inhibitor phlorizin. In each experiment, psychopharmacological data were consistent with glucose engaging an additional signaling pathway to the sweet taste receptor T1R2/T1R3 pathway. Na-lactisole addition impaired detection of the non-caloric sweetener sucralose much more than it did glucose, consistent with glucose using an additional signaling pathway. The addition of NaCl had a beneficial impact on the detection of glucose and its analog MDG and impaired sucralose detection, consistent with glucose utilizing a sodium-glucose co-transporter. The addition of the SGLT inhibitor phlorizin impaired detection of glucose and MDG more than it did sucralose, and had no effect on fructose, further evidence consistent with glucose utilizing a sodium-glucose co-transporter. Together, these results support the idea that oral detection of glucose engages two signaling pathways: one that is comprised of the T1R2/T1R3 sweet taste receptor and the other that utilizes an SGLT glucose transporter.


Subject(s)
Glucose/metabolism , Receptors, G-Protein-Coupled/metabolism , Sodium-Glucose Transport Proteins/metabolism , Taste , Adult , Female , Glucose/analysis , Humans , Male , Middle Aged , Signal Transduction
13.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34290141

ABSTRACT

"Taste-like" tuft cells in the intestine trigger type 2 immunity in response to worm infection. The secretion of interleukin-13 (IL-13) from type 2 innate lymphoid cells (ILC2) represents a key step in the tuft cell-ILC2 cell-intestinal epithelial cell circuit that drives the clearance of worms from the gut via type 2 immune responses. Hallmark features of type 2 responses include tissue remodeling, such as tuft and goblet cell expansion, and villus atrophy, yet it remains unclear if additional molecular changes in the gut epithelium facilitate the clearance of worms from the gut. Using gut organoids, we demonstrated that IL-4 and IL-13, two type 2 cytokines with similar functions, not only induced the classical type 2 responses (e.g., tuft cell expansion) but also drastically up-regulated the expression of gasdermin C genes (Gsdmcs). Using an in vivo worm-induced type 2 immunity model, we confirmed the up-regulation of Gsdmcs in Nippostrongylus brasiliensis-infected wild-type C57BL/6 mice. Consistent with gasdermin family members being principal effectors of pyroptosis, overexpression of Gsdmc2 in human embryonic kidney 293 (HEK293) cells triggered pyroptosis and lytic cell death. Moreover, in intestinal organoids treated with IL-4 or IL-13, or in wild-type mice infected with N. brasiliensis, lytic cell death increased, which may account for villus atrophy observed in worm-infected mice. Thus, we propose that the up-regulated Gsdmc family may be major effectors for type 2 responses in the gut and that Gsdmc-mediated pyroptosis may provide a conduit for the release of antiparasitic factors from enterocytes to facilitate the clearance of worms.


Subject(s)
Cell Death , DNA-Binding Proteins/metabolism , Enterocytes/pathology , Immunity, Innate/immunology , Intestine, Small/pathology , Strongylida Infections/complications , Th2 Cells/immunology , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Enterocytes/immunology , Enterocytes/metabolism , Enterocytes/parasitology , Female , Interleukin-13/metabolism , Interleukin-4/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/parasitology , Male , Mice , Mice, Inbred C57BL , Nippostrongylus/physiology , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
14.
Dev Biol ; 477: 232-240, 2021 09.
Article in English | MEDLINE | ID: mdl-34097879

ABSTRACT

In mammals, multiple cell-signaling pathways and transcription factors regulate development of the embryonic taste system and turnover of taste cells in the adult stage. Using single-cell RNA-Seq of mouse taste cells, we found that the homeobox-containing transcription factor Nkx2-2, a target of the Sonic Hedgehog pathway and a key regulator of the development and regeneration of multiple cell types in the body, is highly expressed in type III taste cells but not in type II or taste stem cells. Using in situ hybridization and immunostaining, we confirmed that Nkx2-2 is expressed specifically in type III taste cells in the endoderm-derived circumvallate and foliate taste papillae but not in the ectoderm-derived fungiform papillae. Lineage tracing revealed that Nkx2-2-expressing cells differentiate into type III, but not type II or type I cells in circumvallate and foliate papillae. Neonatal Nkx2-2-knockout mice did not express key type III taste cell marker genes, while the expression of type II and type I taste cell marker genes were unaffected in these mice. Our findings indicate that Nkx2-2-expressing cells are committed to the type III lineage and that Nkx2-2 may be critical for the development of type III taste cells in the posterior tongue, thus illustrating a key difference in the mechanism of type III cell lineage specification between ectoderm- and endoderm-derived taste fields.


Subject(s)
Cell Lineage/physiology , Homeodomain Proteins/physiology , Taste Buds/embryology , Zebrafish Proteins/physiology , Animals , Animals, Newborn , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/physiology , Cell Count , Cell Lineage/genetics , Female , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/biosynthesis , Male , Mice , RNA-Seq , Taste Buds/cytology , Taste Buds/metabolism , Zebrafish Proteins/biosynthesis
15.
Mol Pharmacol ; 99(5): 319-327, 2021 05.
Article in English | MEDLINE | ID: mdl-33824185

ABSTRACT

Children have difficulty swallowing capsules. Yet, when presented with liquid formulations, children often reject oral medications due to their intense bitterness. Presently, effective strategies to identify methods, reagents, and tools to block bitterness remain elusive. For a specific bitter-tasting drug, identification of the responsible bitter receptors and discovery of antagonists for those receptors can provide a method to block perceived bitterness. We have identified a compound (6-methylflavone) that can block responses to an intensely bitter-tasting anti-human immunodeficiency virus (HIV) drug, tenofovir alafenamide (TAF), using a primary human taste bud epithelial cell culture as a screening platform. Specifically, TAS2R39 and TAS2R1 are the main type 2 taste receptors responding to TAF observed via heterologously expressing specific TAS2R receptors into HEK293 cells. In this assay, 6-methylflavone blocked the responses of TAS2R39 to TAF. In human sensory testing, 8 of 16 subjects showed reduction in perceived bitterness of TAF after pretreating (or "prerinsing") with 6-methylflavone and mixing 6-methylflavone with TAF. Bitterness was completely and reliably blocked in two of these subjects. These data demonstrate that a combined approach of human taste cell culture-based screening, receptor-specific assays, and human psychophysical testing can successfully discover molecules for blocking perceived bitterness of pharmaceuticals, such as the HIV therapeutic TAF. Our hope is to use bitter taste blockers to increase medical compliance with these vital medicines. SIGNIFICANCE STATEMENT: Identification of a small molecule that inhibits bitter taste from tenofovir alafenamide may increase the compliance in treating children with human immunodeficiency virus infections.


Subject(s)
Adenine/analogs & derivatives , Flavoring Agents/administration & dosage , Flavoring Agents/chemistry , Taste Buds/drug effects , Taste/drug effects , Adenine/adverse effects , Adenine/chemistry , Adult , Alanine , Antiviral Agents/adverse effects , Antiviral Agents/chemistry , Cell Line , Female , Flavones/administration & dosage , Flavones/chemistry , HEK293 Cells , Humans , Male , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Taste Buds/metabolism , Tenofovir/analogs & derivatives
16.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33443181

ABSTRACT

Taste bud cells regenerate throughout life. Taste bud maintenance depends on continuous replacement of senescent taste cells with new ones generated by adult taste stem cells. More than a century ago it was shown that taste buds degenerate after their innervating nerves are transected and that they are not restored until after reinnervation by distant gustatory ganglion neurons. Thus, neuronal input, likely via neuron-supplied factors, is required for generation of differentiated taste cells and taste bud maintenance. However, the identity of such a neuron-supplied niche factor(s) remains unclear. Here, by mining a published RNA-sequencing dataset of geniculate ganglion neurons and by in situ hybridization, we demonstrate that R-spondin-2, the ligand of Lgr5 and its homologs Lgr4/6 and stem-cell-expressed E3 ligases Rnf43/Znrf3, is expressed in nodose-petrosal and geniculate ganglion neurons. Using the glossopharyngeal nerve transection model, we show that systemic delivery of R-spondin via adenovirus can promote generation of differentiated taste cells despite denervation. Thus, exogenous R-spondin can substitute for neuronal input for taste bud cell replenishment and taste bud maintenance. Using taste organoid cultures, we show that R-spondin is required for generation of differentiated taste cells and that, in the absence of R-spondin in culture medium, taste bud cells are not generated ex vivo. Thus, we propose that R-spondin-2 may be the long-sought neuronal factor that acts on taste stem cells for maintaining taste tissue homeostasis.


Subject(s)
Regeneration , Taste Buds/physiology , Thrombospondins/metabolism , Animals , Cell Differentiation , Mice , Organoids , Taste Buds/cytology
17.
Cell Mol Gastroenterol Hepatol ; 11(2): 349-369, 2021.
Article in English | MEDLINE | ID: mdl-32882403

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDA) initiation and progression are accompanied by an immunosuppressive inflammatory response. Here, we evaluated the immunomodulatory role of chemosensory signaling in metaplastic tuft cells (MTCs) by analyzing the role of GNAT3, a gustatory pathway G-protein expressed by MTCs, during PDA progression. METHODS: Gnat3-null (Gnat3-/-) mice were crossbred with animals harboring a Cre-inducible KrasLSL-G12D/+ allele with either Ptf1aCre/+ (KC) or tamoxifen-inducible Ptf1aCreERT/+ (KCERT) mice to drive oncogenic KRAS expression in the pancreas. Ex vivo organoid conditioned medium generated from KC and Gnat3-/-;KC acinar cells was analyzed for cytokine secretion. Experimental pancreatitis was induced in KCERT and Gnat3-/-;KCERT mice to accelerate tumorigenesis, followed by analysis using mass cytometry and single-cell RNA sequencing. To study PDA progression, KC and Gnat3-/-;KC mice were aged to morbidity or 52 weeks. RESULTS: Ablation of Gnat3 in KC organoids increased release of tumor-promoting cytokines in conditioned media, including CXCL1 and CXCL2. Analysis of Gnat3-/-;KCERT pancreata found altered expression of immunomodulatory genes in Cxcr2 expressing myeloid-derived suppressor cells (MDSCs) and an increased number of granulocytic MDSCs, a subset of tumor promoting MDSCs. Importantly, expression levels of CXCL1 and CXCL2, known ligands for CXCR2, were also elevated in Gnat3-/-;KCERT pancreata. Consistent with the tumor-promoting role of MDSCs, aged Gnat3-/-;KC mice progressed more rapidly to metastatic carcinoma compared with KC controls. CONCLUSIONS: Compromised gustatory sensing, achieved by Gnat3 ablation, enhanced the CXCL1/2-CXCR2 axis to alter the MDSC population and promoted the progression of metastatic PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Heterotrimeric GTP-Binding Proteins/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/immunology , Animals , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cells, Cultured , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Culture Media, Conditioned/metabolism , Disease Models, Animal , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells , Organoids , Pancreatic Ducts/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Primary Cell Culture , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/immunology
18.
J Neurochem ; 158(2): 233-245, 2021 07.
Article in English | MEDLINE | ID: mdl-33319361

ABSTRACT

Leptin is known to selectively suppress neural and taste cell responses to sweet compounds. The sweet suppressive effect of leptin is mediated by the leptin receptor Ob-Rb, and the ATP-gated K+ (KATP ) channel expressed in some sweet-sensitive, taste receptor family 1 member 3 (T1R3)-positive taste cells. However, the intracellular transduction pathway connecting Ob-Rb to KATP channel remains unknown. Here we report that phosphoinositide 3-kinase (PI3K) mediates leptin's suppression of sweet responses in T1R3-positive taste cells. In in situ taste cell recording, systemically administrated leptin suppressed taste cell responses to sucrose in T1R3-positive taste cells. Such leptin's suppression of sucrose responses was impaired by co-administration of PI3K inhibitors (wortmannin or LY294002). In contrast, co-administration of signal transducer and activator of transcription 3 inhibitor (Stattic) or Src homology region 2 domain-containing phosphatase-2 inhibitor (SHP099) had no effect on leptin's suppression of sucrose responses, although signal transducer and activator of transcription 3 and Src homology region 2 domain-containing phosphatase-2 were expressed in T1R3-positive taste cells. In peeled tongue epithelium, phosphatidylinositol (3,4,5)-trisphosphate production and phosphorylation of AKT by leptin were immunohistochemically detected in some T1R3-positive taste cells but not in glutamate decarboxylase 67-positive taste cells. Leptin-induced phosphatidylinositol (3,4,5)-trisphosphate production was suppressed by LY294002. Thus, leptin suppresses sweet responses of T1R3-positive taste cells by activation of Ob-Rb-PI3K-KATP channel pathway.


Subject(s)
Leptin/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Receptors, G-Protein-Coupled/drug effects , Sweetening Agents/pharmacology , Taste Buds/drug effects , Taste/drug effects , Animals , Female , Male , Mice , Mice, Transgenic , Phosphatidylinositols/metabolism , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Tongue/cytology , Tongue/drug effects
19.
Acta Physiol (Oxf) ; 230(4): e13529, 2020 12.
Article in English | MEDLINE | ID: mdl-32599649

ABSTRACT

AIM: We investigated potential neuron types that code sugar information and how sodium-glucose cotransporters (SGLTs) and T1Rs are involved. METHODS: Whole-nerve recordings in the chorda tympani (CT) and the glossopharyngeal (GL) nerves and single-fibre recordings in the CT were performed in T1R3-KO and wild-type (WT) mice. Behavioural response measurements were conducted in T1R3-KO mice using phlorizin (Phl), a competitive inhibitor of SGLTs. RESULTS: Results indicated that significant enhancement occurred in responses to sucrose and glucose (Glc) by adding 10 mmol/L NaCl but not in responses to KCl, monopotassium glutamate, citric acid, quinine sulphate, SC45647(SC) or polycose in both CT and GL nerves. These enhancements were abolished by lingual application of Phl. In single-fibre recording, fibres showing maximal response to sucrose could be classified according to responses to SC and Glc with or without 10 mmol/L NaCl in the CT of WT mice, namely, Phl-insensitive type, Phl-sensitive Glc-type and Mixed (Glc and SC responding)-type fibres. In T1R3-KO mice, Phl-insensitive-type fibres disappeared. Results from behavioural experiments showed that the number of licks and amount of intake for Glc with or without 10 mmol/L NaCl were significantly suppressed by Phl. CONCLUSION: We found evidence for the contribution of SGLTs in sugar sensing in taste cells of mouse tongue. Moreover, we found T1R-dependent (Phl-insensitive) type, Glc-type and Mixed (SGLTs and T1Rs)-type fibres. SGLT1 may be involved in the latter two types and may play important roles in the glucose-specific cephalic phase of digestion and palatable food intake.


Subject(s)
Sugars , Taste , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Sodium-Glucose Transporter 1 , Tongue
20.
Chem Senses ; 45(3): 187-194, 2020 04 17.
Article in English | MEDLINE | ID: mdl-31993633

ABSTRACT

Inflammatory cytokines are signaling molecules that regulate numerous physiological processes, from tissue homeostasis to metabolism and food intake. Expression of certain cytokines can be markedly induced in subsets of taste bud cells under acute and chronic inflammation. This may contribute to altered taste perception and preference associated with many diseases. Although the pathways of cytokine induction are well studied in immune cells, they remain poorly characterized in taste cells, in part due to the difficulties of performing biochemical analyses with a limited number of taste cells. The recently developed taste organoid model provides an opportunity to carry out these mechanistic studies in vitro. However, it was unknown whether taste organoids respond to inflammatory stimuli as do in vivo native taste buds. Here we analyze lipopolysaccharide (LPS)-induced expression and secretion of two inflammatory cytokines, tumor necrosis factor (TNF), and interleukin-6 (IL-6). We show that, similarly to native mouse taste epithelia, organoids derived from mouse circumvallate stem cells express several toll-like receptors (TLRs), including TLR4-the primary receptor for LPS. Organoids and native taste epithelia express all five genes in the nuclear factor-κb (Nfkb) family that encode the transcription factor NF-κB, a critical regulator of inflammatory responses. LPS stimulates fast induction of TNF and IL-6 with similar induction kinetics in organoids and native taste epithelia. These results show that taste epithelial cells possess necessary components for inflammatory cytokine induction and secretion and suggest that the organoid model can be a useful tool to dissect the underlying mechanisms.


Subject(s)
Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Organoids/drug effects , Taste/drug effects , Tumor Necrosis Factors/biosynthesis , Animals , Cells, Cultured , Female , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C57BL , Organoids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...