1
|
Lemon CH, Li J, Ali MSS, Ngum NM, Zumpano KT, Roberts CJ. Parabrachial Calca neurons influence aversive and appetitive taste function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640892. [PMID: 40093097 PMCID: PMC11908176 DOI: 10.1101/2025.02.28.640892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The parabrachial (PB) nucleus participates in taste processing and integration with other senses. PB neurons that express the Calca gene support sensory-integrative responses, albeit only limited data have addressed their influence on taste. Here we studied how chemogenetic dampening of PB-Calca neurons impacted mouse orosensory preferences for diverse taste stimuli in brief-access fluid exposure tests, which capture oral sensory/tongue control of licking behavior. Intracranial delivery of Cre-dependent viruses in female and male Calca Cre/+ mice induced expression of the inhibitory designer receptor hM4Di:mCherry (hM4Di mice) or fluorophore mCherry alone (mCherry mice) in PB-Calca neurons. Several weeks later, hM4Di and mCherry mice entered brief-access tests where they could lick taste solutions on discrete seconds-long trials. Stimuli included the behaviorally avoided, but functionally different, bitter taste stimuli quinine (0 [water], 0.1, 0.3, and 1.0 mM) and cycloheximide (0, 0.001, 0.003, and 0.01 mM), and the appetitive sugar sucrose (0, 100, 300, 500, and 1000 mM). Both hM4Di and mCherry mice received the hM4Di ligand clozapine-N-oxide (CNO, 5 mg/kg, i.p.) prior to daily tests performed by blinded experimenters. With CNO, hM4Di mice displayed greater average licking (i.e., less avoidance) of quinine (p < 0.05), but not cycloheximide (p > 0.3), than mCherry mice, implying PB-Calca neurons variably influence orosensory responses across bitter stimuli. Moreover, male hM4Di mice selectively showed reduced mean licking preferences for sucrose under CNO (p < 0.05). These data suggest that PB-Calca neurons participate in both aversive and appetitive taste-guided behaviors, with their role in appetitive taste dependent on sex.
Collapse
Affiliation(s)
- Christian H Lemon
- School of Biological Sciences, University of Oklahoma, 101 David L. Boren Blvd., Norman, OK 73019 USA
| | - Jinrong Li
- School of Biological Sciences, University of Oklahoma, 101 David L. Boren Blvd., Norman, OK 73019 USA
| | - Md Sams Sazzad Ali
- School of Biological Sciences, University of Oklahoma, 101 David L. Boren Blvd., Norman, OK 73019 USA
| | - Neville M Ngum
- School of Biological Sciences, University of Oklahoma, 101 David L. Boren Blvd., Norman, OK 73019 USA
| | - Kyle T Zumpano
- School of Biological Sciences, University of Oklahoma, 101 David L. Boren Blvd., Norman, OK 73019 USA
| | - Catori J Roberts
- School of Biological Sciences, University of Oklahoma, 101 David L. Boren Blvd., Norman, OK 73019 USA
| |
Collapse
|
2
|
Lemon CH. A non-singularity in sweet taste. Chem Senses 2024; 49:bjae006. [PMID: 38366583 DOI: 10.1093/chemse/bjae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 02/18/2024] Open
Affiliation(s)
- Christian H Lemon
- School of Biological Sciences, University of Oklahoma, Norman, OK 73019, United States
| |
Collapse
|
3
|
Lang R, Lang T, Dunkel A, Ziegler F, Behrens M. Overlapping activation pattern of bitter taste receptors affect sensory adaptation and food perception. Front Nutr 2022; 9:1082698. [PMID: 36601079 PMCID: PMC9806268 DOI: 10.3389/fnut.2022.1082698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
The composition of menus and the sequence of foodstuffs consumed during a meal underlies elaborate rules. However, the molecular foundations for the observed taste- and pleasure-raising effects of complex menus are obscure. The molecular identification and characterization of taste receptors can help to gain insight into the complex interrelationships of food items and beverages during meals. In our study, we quantified important bitter compounds in chicory and chicory-based surrogate coffee and used them to identify responsive bitter taste receptors. The two receptors, TAS2R43 and TAS2R46, are exquisitely sensitive to lactucin, lactucopicrin, and 11β,13-dihydrolactucin. Sensory testing demonstrated a profound influence of the sequence of consumption of chicory, surrogate coffee, and roasted coffee on the perceived bitterness by human volunteers. These findings pave the way for a molecular understanding of some of the mixture effects underlying empirical meal compositions.
Collapse
|
4
|
Bongers KS, McDonald RA, Winner KM, Falkowski NR, Brown CA, Baker JM, Hinkle KJ, Fergle DJ, Dickson RP. Antibiotics cause metabolic changes in mice primarily through microbiome modulation rather than behavioral changes. PLoS One 2022; 17:e0265023. [PMID: 35298489 PMCID: PMC8929607 DOI: 10.1371/journal.pone.0265023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/20/2022] [Indexed: 12/15/2022] Open
Abstract
Background The microbiome is an important and increasingly-studied mediator of organismal metabolism, although how the microbiome affects metabolism remains incompletely understood. Many investigators use antibiotics to experimentally perturb the microbiome. However, antibiotics have poorly understood yet profound off-target effects on behavior and diet, including food and water aversion, that can confound experiments and limit their applicability. We thus sought to determine the relative influence of microbiome modulation and off-target antibiotic effects on the behavior and metabolic activity of mice. Results Mice treated with oral antibiotics via drinking water exhibited significant weight loss in fat, liver, and muscle tissue. These mice also exhibited a reduction in water and food consumption, with marked variability across antibiotic regimens. While administration of bitter-tasting but antimicrobially-inert compounds caused a similar reduction in water consumption, this did not cause tissue weight loss or reduced food consumption. Mice administered intraperitoneal antibiotics (bypassing the gastrointestinal tract) exhibited reduced tissue weights and oral intake, comparable to the effects of oral antibiotics. Antibiotic-treated germ-free mice did not have reduced tissue weights, providing further evidence that direct microbiome modulation (rather than behavioral effects) mediates these metabolic changes. Conclusions While oral antibiotics cause profound effects on food and water consumption, antibiotic effects on organismal metabolism are primarily mediated by microbiome modulation. We demonstrate that tissue-specific weight loss following antibiotic administration is due primarily to microbiome effects rather than food and water aversion, and identify antibiotic regimens that effectively modulate gut microbiota while minimizing off-target behavioral effects.
Collapse
Affiliation(s)
- Kale S. Bongers
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Roderick A. McDonald
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Katherine M. Winner
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Nicole R. Falkowski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Christopher A. Brown
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Institute for Research on Innovation and Science, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer M. Baker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kevin J. Hinkle
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Daniel J. Fergle
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
5
|
McCaughey SA. Variation in the gene Tas1r3 reveals complex temporal properties of mouse brainstem taste responses to sweeteners. Am J Physiol Regul Integr Comp Physiol 2021; 321:R751-R767. [PMID: 34523351 PMCID: PMC8616626 DOI: 10.1152/ajpregu.00001.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 11/22/2022]
Abstract
The gene Tas1r3 codes for the protein T1R3, which dimerizes with T1R2 to form a sweetener-binding receptor in taste cells. Tas1r3 influences sweetener preferences in mice, as shown by work with a 129.B6-Tas1r3 segregating congenic strain on a 129P3/J (129) genetic background; members of this strain vary in whether they do or do not have one copy of a donor fragment with the C57BL/6ByJ (B6) allele for Tas1r3 (B6/129 and 129/129 mice, respectively). Taste-evoked neural responses were measured in the nucleus of the solitary tract (NST), the first central gustatory relay, in B6/129 and 129/129 littermates, to examine how the activity dependent on the T1R2/T1R3 receptor is distributed across neurons and over time. Responses to sucrose were larger in B6/129 than in 129/129 mice, but only during a later, tonic response portion (>600 ms) sent to different cells than the earlier, phasic response. Similar results were found for artificial sweeteners, whose responses were best considered as complex spatiotemporal patterns. There were also group differences in burst firing of NST cells, with a significant positive correlation between bursting prevalence and sucrose response size in only the 129/129 group. The results indicate that sweetener transduction initially occurs through T1R3-independent mechanisms, after which the T1R2/T1R3 receptor initiates a separate, spatially distinct response, with the later period dominating sweet taste perceptions and driving sugar preferences. Furthermore, the current data suggest that burst firing is distributed across NST neurons nonrandomly and in a manner that may amplify weak incoming gustatory signals.
Collapse
Affiliation(s)
- Stuart A McCaughey
- Center for Medical Education, Ball State University, Muncie, Indiana
- Monell Chemical Senses Center, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Optogenetic Stimulation of Type I GAD65 + Cells in Taste Buds Activates Gustatory Neurons and Drives Appetitive Licking Behavior in Sodium-Depleted Mice. J Neurosci 2020. [PMID: 32878902 DOI: 10.1523/jneurosci.0597‐20.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mammalian taste buds are comprised of specialized neuroepithelial cells that act as sensors for molecules that provide nutrition (e.g., carbohydrates, amino acids, and salts) and those that are potentially harmful (e.g., certain plant compounds and strong acids). Type II and III taste bud cells (TBCs) detect molecules described by humans as "sweet," "bitter," "umami," and "sour." TBCs that detect metallic ions, described by humans as "salty," are undefined. Historically, type I glial-like TBCs have been thought to play a supportive role in the taste bud, but little research has been done to explore their role in taste transduction. Some evidence implies that type I cells may detect sodium (Na+) via an amiloride-sensitive mechanism, suggesting they play a role in Na+ taste transduction. We used an optogenetic approach to study type I TBCs by driving the expression of the light-sensitive channelrhodopsin-2 (ChR2) in type I GAD65+ TBCs of male and female mice. Optogenetic stimulation of GAD65+ TBCs increased chorda tympani nerve activity and activated gustatory neurons in the rostral nucleus tractus solitarius. "N neurons," whose NaCl responses were blocked by the amiloride analog benzamil, responded robustly to light stimulation of GAD65+ TBCs on the anterior tongue. Two-bottle preference tests were conducted under Na+-replete and Na+-deplete conditions to assess the behavioral impact of optogenetic stimulation of GAD65+ TBCs. Under Na+-deplete conditions GAD65-ChR2-EYFP mice displayed a robust preference for H2O illuminated with 470 nm light versus nonilluminated H2O, suggesting that type I glial-like TBCs are sufficient for driving a behavior that resembles Na+ appetite.SIGNIFICANCE STATEMENT This is the first investigation on the role of type I GAD65+ taste bud cells (TBCs) in taste-mediated physiology and behavior via optogenetics. It details the first definitive evidence that selective optogenetic stimulation of glial-like GAD65+ TBCs evokes neural activity and modulates behavior. Optogenetic stimulation of GAD65+ TBCs on the anterior tongue had the strongest effect on gustatory neurons that responded best to NaCl stimulation through a benzamil-sensitive mechanism. Na+-depleted mice showed robust preferences to "light taste" (H2O illuminated with 470 nm light vs nonilluminated H2O), suggesting that the activation of GAD65+ cells may generate a salt-taste sensation in the brain. Together, our results shed new light on the role of GAD65+ TBCs in gustatory transduction and taste-mediated behavior.
Collapse
|
7
|
Optogenetic Stimulation of Type I GAD65 + Cells in Taste Buds Activates Gustatory Neurons and Drives Appetitive Licking Behavior in Sodium-Depleted Mice. J Neurosci 2020; 40:7795-7810. [PMID: 32878902 DOI: 10.1523/jneurosci.0597-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/17/2020] [Accepted: 08/23/2020] [Indexed: 01/27/2023] Open
Abstract
Mammalian taste buds are comprised of specialized neuroepithelial cells that act as sensors for molecules that provide nutrition (e.g., carbohydrates, amino acids, and salts) and those that are potentially harmful (e.g., certain plant compounds and strong acids). Type II and III taste bud cells (TBCs) detect molecules described by humans as "sweet," "bitter," "umami," and "sour." TBCs that detect metallic ions, described by humans as "salty," are undefined. Historically, type I glial-like TBCs have been thought to play a supportive role in the taste bud, but little research has been done to explore their role in taste transduction. Some evidence implies that type I cells may detect sodium (Na+) via an amiloride-sensitive mechanism, suggesting they play a role in Na+ taste transduction. We used an optogenetic approach to study type I TBCs by driving the expression of the light-sensitive channelrhodopsin-2 (ChR2) in type I GAD65+ TBCs of male and female mice. Optogenetic stimulation of GAD65+ TBCs increased chorda tympani nerve activity and activated gustatory neurons in the rostral nucleus tractus solitarius. "N neurons," whose NaCl responses were blocked by the amiloride analog benzamil, responded robustly to light stimulation of GAD65+ TBCs on the anterior tongue. Two-bottle preference tests were conducted under Na+-replete and Na+-deplete conditions to assess the behavioral impact of optogenetic stimulation of GAD65+ TBCs. Under Na+-deplete conditions GAD65-ChR2-EYFP mice displayed a robust preference for H2O illuminated with 470 nm light versus nonilluminated H2O, suggesting that type I glial-like TBCs are sufficient for driving a behavior that resembles Na+ appetite.SIGNIFICANCE STATEMENT This is the first investigation on the role of type I GAD65+ taste bud cells (TBCs) in taste-mediated physiology and behavior via optogenetics. It details the first definitive evidence that selective optogenetic stimulation of glial-like GAD65+ TBCs evokes neural activity and modulates behavior. Optogenetic stimulation of GAD65+ TBCs on the anterior tongue had the strongest effect on gustatory neurons that responded best to NaCl stimulation through a benzamil-sensitive mechanism. Na+-depleted mice showed robust preferences to "light taste" (H2O illuminated with 470 nm light vs nonilluminated H2O), suggesting that the activation of GAD65+ cells may generate a salt-taste sensation in the brain. Together, our results shed new light on the role of GAD65+ TBCs in gustatory transduction and taste-mediated behavior.
Collapse
|
8
|
Higgins MJ, Hayes JE. Discrimination of Isointense Bitter Stimuli in a Beer Model System. Nutrients 2020; 12:nu12061560. [PMID: 32471227 PMCID: PMC7352581 DOI: 10.3390/nu12061560] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 12/23/2022] Open
Abstract
Prior work suggests humans can differentiate between bitter stimuli in water. Here, we describe three experiments that test whether beer consumers can discriminate between different bitterants in beer. In Experiment 1 (n = 51), stimuli were intensity matched; Experiments 2 and 3 were a difference from control (DFC)/check-all-that-apply (CATA) test (n = 62), and an affective test (n = 81). All used a commercial non-alcoholic beer spiked with Isolone (a hop extract), quinine sulfate dihydrate, and sucrose octaacetate (SOA). In Experiment 1, participants rated intensities on general labeled magnitude scales (gLMS), which were analyzed via ANOVA. In Experiment 2, participants rated how different samples were from a reference of Isolone on a 7-point DFC scale, and endorsed 13 attributes in a CATA task. DFC data were analyzed via ANOVA with Dunnett's test to compare differences relative to a blind reference, and CATA data were analyzed via Cochran's Q test. In Experiment 3, liking was assessed on labeled affective magnitude scales, and samples were also ranked. Liking was analyzed via ANOVA and rankings were analyzed with a Cochran-Mantel-Haenszel test. Experiment 1 confirmed that samples were isointense. In Experiment 2, despite being isointense, both quinine (p = 0.04) and SOA (p = 0.03) were different from Isolone, but no significant effects were found for CATA descriptors (all p values > 0.16). In Experiment 3, neither liking (p = 0.16) or ranking (p = 0.49) differed. Collectively, these data confirm that individuals can discriminate perceptually distinct bitter stimuli in beer, as shown previously in water, but these differences cannot be described semantically, and they do not seem to influence hedonic assessments.
Collapse
Affiliation(s)
- Molly J. Higgins
- Sensory Evaluation Center, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Food Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - John E. Hayes
- Sensory Evaluation Center, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Food Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA
- Correspondence: ; Tel.: +1-814-863-7129
| |
Collapse
|
9
|
Staszko SM, Boughter JD, Fletcher ML. Taste coding strategies in insular cortex. Exp Biol Med (Maywood) 2020; 245:448-455. [PMID: 32106700 DOI: 10.1177/1535370220909096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While the cortical representation of sensory stimuli is well described for some sensory systems, a clear understanding of the cortical representation of taste stimuli remains elusive. Recent investigations have focused on both spatial and temporal organization of taste responses in the putative taste region of insular cortex. This review highlights recent literature focused on spatiotemporal coding strategies in insular cortex. These studies are examined in the context of the organization and function of the entire insular cortex, rather than a specific gustatory region of insular cortex. In regard to a taste quality-specific map, imaging studies have reported conflicting results, whereas electrophysiology studies have described a broad distribution of taste-responsive neurons found throughout insular cortex with no spatial organization. The current collection of evidence suggests that insular cortex may be organized into a hedonic or “viscerotopic” map, rather than one ordered according to taste quality. Further, it has been proposed that cortical taste responses can be separated into temporal “epochs” representing stimulus identity and palatability. This coding strategy presents a potential framework, whereby the coordinated activity of a population of neurons allows for the same neurons to respond to multiple taste stimuli or even other sensory modalities, a well-documented phenomenon in insular cortex neurons. However, these representations may not be static, as several studies have demonstrated that both spatial representation and temporal dynamics of taste coding change with experience. Collectively, these studies suggest that cortical taste representation is not organized in a spatially discrete map, but rather is plastic and spatially dispersed, using temporal information to encode multiple types of information about ingested stimuli. Impact statement The organization of taste coding in insular cortex is widely debated. While early work has focused on whether taste quality is encoded via labeled line or ensemble mechanisms, recent work has attempted to delineate the spatial organization and temporal components of taste processing in insular cortex. Recent imaging and electrophysiology studies have reported conflicting results in regard to the spatial organization of cortical taste responses, and many studies ignore potentially important temporal dynamics when investigating taste processing. This review highlights the latest research in these areas and examines them in the context of the anatomy and physiology of the insular cortex in general to provide a more comprehensive description of taste coding in insular cortex.
Collapse
Affiliation(s)
- Stephanie M Staszko
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - John D Boughter
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Max L Fletcher
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
10
|
Martin LE, Kay KE, Torregrossa AM. Rats are unable to discriminate quinine from diverse bitter stimuli. Am J Physiol Regul Integr Comp Physiol 2019; 317:R793-R802. [PMID: 31596113 DOI: 10.1152/ajpregu.00213.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Compounds described by humans as "bitter" are sensed by a family of type 2 taste receptors (T2Rs). Previous work suggested that diverse bitter stimuli activate distinct receptors, which might allow for perceptually distinct tastes. Alternatively, it has been shown that multiple T2Rs are expressed on the same taste cell, leading to the contrary suggestion that these stimuli produce a unitary perception. Behavioral work done to address this in rodent models is limited to Spector and Kopka (Spector AC, Kopka SL. J Neurosci 22: 1937-1941, 2002), who demonstrated that rats cannot discriminate quinine from denatonium. Supporting this finding, it has been shown that quinine and denatonium activate overlapping T2Rs and neurons in both the mouse and rat nucleus of the solitary tract (NTS). However, cycloheximide and 6-n-propylthiouracil (PROP) do not appear to overlap with quinine in the NTS, suggesting that these stimuli may be discriminable from quinine and the denatonium/quinine comparison is not generalizable. Using the same procedure as Spector and Kopka, we tasked animals with discriminating a range of stimuli (denatonium, cycloheximide, PROP, and sucrose octaacetate) from quinine. We replicated and expanded the findings of Spector and Kopka; rats could not discriminate quinine from denatonium, cycloheximide, or PROP. Rats showed a very weak ability to discriminate between quinine and sucrose octaacetate. All animals succeeded in discriminating quinine from KCl, demonstrating they were capable of the task. These data suggest that rats cannot discriminate this suite of stimuli, although they appear distinct by physiological measures.
Collapse
Affiliation(s)
- Laura E Martin
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York
| | - Kristen E Kay
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York
| | - Ann-Marie Torregrossa
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York.,University at Buffalo Center for Ingestive Behavior Research, Buffalo, New York
| |
Collapse
|
11
|
Martin LE, Nikonova LV, Kay KE, Torregrossa AM. Altering salivary protein profile can increase acceptance of a novel bitter diet. Appetite 2019; 136:8-17. [PMID: 30639842 DOI: 10.1016/j.appet.2019.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 01/17/2023]
Abstract
Bitter taste is often associated with toxins, but accepting some bitter foods, such as green vegetables, can be an important part of maintaining a healthy diet. In rats and humans, repeated exposure to a bitter stimulus increases acceptance. Repeated exposure allows an individual the opportunity to learn about the food's orosensory and postingestive effects. It also alters the salivary protein (SP) profile, which in turn alters taste signaling. We have hypothesized that altering the salivary proteome plays a role in the increased acceptance after repeated exposure. Here we test this and attempt to disentangle the contribution of learning during dietary exposure from the contribution of SPs in increased acceptance of bitter diet. Dietary exposure to quinine or tannic acid and injection of isoproterenol (IPR) result in similar salivary protein profiles. Here we used either the bitter stimulus tannic acid or IPR injection to upregulate a subset of SPs before exposing animals to a novel diet containing quinine (0.375%). Control animals received either a control diet before being exposed to quinine, or a diet containing sucrose octaacetate, a compound that the animals avoid but does not alter SP profiles. The treatments that alter SP expression increased rate of feeding on the quinine diet compared to the control treatments. Additionally, tannic acid exposure altered intake and meal size of the quinine diet. These data suggest that SPs, not just learning about bitter food, increase acceptance of the bitter diet.
Collapse
Affiliation(s)
- Laura E Martin
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, 14216, USA
| | - Larissa V Nikonova
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, USA
| | - Kristen E Kay
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, 14216, USA
| | - Ann-Marie Torregrossa
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, 14216, USA.
| |
Collapse
|
12
|
Mouse Parabrachial Neurons Signal a Relationship between Bitter Taste and Nociceptive Stimuli. J Neurosci 2019; 39:1631-1648. [PMID: 30606758 DOI: 10.1523/jneurosci.2000-18.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/01/2018] [Accepted: 12/18/2018] [Indexed: 11/21/2022] Open
Abstract
Taste and somatosensation both mediate protective behaviors. Bitter taste guides avoidance of ingestion of toxins while pain sensations, such as noxious heat, signal adverse conditions to ward off harm. Although brain pathways for taste and somatosensation are typically studied independently, prior data suggest that they intersect, potentially reflecting their common protective role. To investigate this, we applied electrophysiologic and optogenetic techniques in anesthetized mice of both sexes to evaluate relationships between oral somatosensory and taste activity in the parabrachial nucleus (PbN), implicated for roles in gustation and pain. Spikes were recorded from taste-active PbN neurons tested with oral delivery of thermal and chemesthetic stimuli, including agonists of nocisensitive transient receptor potential (TRP) ion channels on somatosensory fibers. Gustatory neurons were also tested to follow electrical pulse stimulation of an oral somatosensory region of the spinal trigeminal subnucleus caudalis (Vc), which projects to the PbN. Neurons composed classic taste groups, including sodium, electrolyte, appetitive, or bitter cells. Across groups, most neurons spiked to Vc pulse stimulation, implying that trigeminal projections reach PbN gustatory neurons. Among such cells, a subpopulation responsive to the bitter taste stimuli quinine and cycloheximide, and aversive concentrations of sodium, cofired to agonists of nocisensitive TRP channels, including capsaicin, mustard oil, and noxious heat. Such neurons populated the lateral PbN. Further, nociceptive activity in PbN bitter taste neurons was suppressed during optogenetic-assisted inhibition of the Vc, implying convergent trigeminal input contributed to such activity. Our results reveal a novel role for PbN gustatory cells in cross-system signaling related to protection.SIGNIFICANCE STATEMENT Prior data suggest that gustatory and trigeminal neural pathways intersect and overlap in the parabrachial area. However, no study has directly examined such overlap and why it may exist. Here we found that parabrachial gustatory neurons can receive afferent projections from trigeminal nuclei and fire to oral nociceptive stimuli that excite somatosensory receptors and fibers. Activation to aversive nociceptive stimuli in gustatory cells was associated with responding to behaviorally avoided bitter tastants. We were further able to show that silencing trigeminal projections inhibited nociceptive activity in parabrachial bitter taste neurons. Our results imply that in the parabrachial area, there is predictable overlap between taste and somatosensory processing related to protective coding and that classically defined taste neurons contribute to this process.
Collapse
|
13
|
Schier LA, Spector AC. The Functional and Neurobiological Properties of Bad Taste. Physiol Rev 2019; 99:605-663. [PMID: 30475657 PMCID: PMC6442928 DOI: 10.1152/physrev.00044.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/18/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022] Open
Abstract
The gustatory system serves as a critical line of defense against ingesting harmful substances. Technological advances have fostered the characterization of peripheral receptors and have created opportunities for more selective manipulations of the nervous system, yet the neurobiological mechanisms underlying taste-based avoidance and aversion remain poorly understood. One conceptual obstacle stems from a lack of recognition that taste signals subserve several behavioral and physiological functions which likely engage partially segregated neural circuits. Moreover, although the gustatory system evolved to respond expediently to broad classes of biologically relevant chemicals, innate repertoires are often not in register with the actual consequences of a food. The mammalian brain exhibits tremendous flexibility; responses to taste can be modified in a specific manner according to bodily needs and the learned consequences of ingestion. Therefore, experimental strategies that distinguish between the functional properties of various taste-guided behaviors and link them to specific neural circuits need to be applied. Given the close relationship between the gustatory and visceroceptive systems, a full reckoning of the neural architecture of bad taste requires an understanding of how these respective sensory signals are integrated in the brain.
Collapse
Affiliation(s)
- Lindsey A Schier
- Department of Biological Sciences, University of Southern California , Los Angeles, California ; and Department of Psychology and Program in Neuroscience, Florida State University , Tallahassee, Florida
| | - Alan C Spector
- Department of Biological Sciences, University of Southern California , Los Angeles, California ; and Department of Psychology and Program in Neuroscience, Florida State University , Tallahassee, Florida
| |
Collapse
|
14
|
Töle J, Stolzenburg A, Tyree SM, Stähler F, Meyerhof W. Tastant-Evoked Arc Expression in the Nucleus of the Solitary Tract and Nodose/Petrosal Ganglion of the Mouse Is Specific for Bitter Compounds. Chem Senses 2018. [PMID: 29514200 DOI: 10.1093/chemse/bjy017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite long and intense research, some fundamental questions regarding representation of taste information in the brain still remain unanswered. This might in part be due to shortcomings of the established methods that limit the researcher either to thorough characterization of few elements or to analyze the response of the entirety of neurons to only one stimulus. To overcome these restrictions, we evaluate the use of the immediate early gene Arc as a neuronal activity marker in the early neural structures of the taste pathway, the nodose/petrosal ganglion (NPG) and the nucleus of the solitary tract (NTS). Responses of NPG and NTS neurons were limited to substances that taste bitter to humans and are avoided by mice. Arc-expressing cells were concentrated in the rostromedial part of the dorsal NTS suggesting a role in gustatory processing. The use of Arc as a neuronal activity marker has several advantages, primarily the possibility to analyze the response of large numbers of neurons while using more than one stimulus makes Arc an interesting new tool for research in the early stages of taste processing.
Collapse
Affiliation(s)
- Jonas Töle
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee, Nuthetal, Germany
| | | | | | | | | |
Collapse
|
15
|
Di Pizio A, Ben Shoshan-Galeczki Y, Hayes JE, Niv MY. Bitter and sweet tasting molecules: It's complicated. Neurosci Lett 2018; 700:56-63. [PMID: 29679682 DOI: 10.1016/j.neulet.2018.04.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/22/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022]
Abstract
"Bitter" and "sweet" are frequently framed in opposition, both functionally and metaphorically, in regard to affective responses, emotion, and nutrition. This oppositional relationship is complicated by the fact that some molecules are simultaneously bitter and sweet. In some cases, a small chemical modification, or a chirality switch, flips the taste from sweet to bitter. Molecules humans describe as bitter are recognized by a 25-member subfamily of class A G-protein coupled receptors (GPCRs) known as TAS2Rs. Molecules humans describe as sweet are recognized by a TAS1R2/TAS1R3 heterodimer of class C GPCRs. Here we characterize the chemical space of bitter and sweet molecules: the majority of bitter compounds show higher hydrophobicity compared to sweet compounds, while sweet molecules have a wider range of sizes. Importantly, recent evidence indicates that TAS1Rs and TAS2Rs are not limited to the oral cavity; moreover, some bitterants are pharmacologically promiscuous, with the hERG potassium channel, cytochrome P450 enzymes, and carbonic anhydrases as common off-targets. Further focus on polypharmacology may unravel new physiological roles for tastant molecules.
Collapse
Affiliation(s)
- Antonella Di Pizio
- The Institute of Biochemistry, Food and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University, 76100, Rehovot, Israel; The Fritz Haber Center for Molecular Dynamics, The Hebrew University, Jerusalem, 91904, Israel
| | - Yaron Ben Shoshan-Galeczki
- The Institute of Biochemistry, Food and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University, 76100, Rehovot, Israel; The Fritz Haber Center for Molecular Dynamics, The Hebrew University, Jerusalem, 91904, Israel
| | - John E Hayes
- Department of Food Science, College of Agricultural Sciences, The Pennsylvania State University, University Park PA, USA
| | - Masha Y Niv
- The Institute of Biochemistry, Food and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University, 76100, Rehovot, Israel; The Fritz Haber Center for Molecular Dynamics, The Hebrew University, Jerusalem, 91904, Israel.
| |
Collapse
|
16
|
McCaughey SA. Characterization of mouse chorda tympani responses evoked by stimulation of anterior or posterior fungiform taste papillae. Neurosci Res 2018; 141:43-51. [PMID: 29580888 DOI: 10.1016/j.neures.2018.03.006] [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: 11/23/2017] [Revised: 02/18/2018] [Accepted: 03/22/2018] [Indexed: 11/30/2022]
Abstract
Different gustatory papilla types vary in their locations on the tongue. Distinctions have often made between types, but variation within fungiform papillae has seldom been explored. Here, regional differences in fungiform papillae were investigated by flowing solutions selectively over either an anterior fungiform (AF, tongue tip) or a posterior fungiform (PF, middle third) region as taste-evoked activity was measured in the chorda tympani nerve of C57BL/6J (B6) mice. Significantly larger responses were evoked by NaCl applied to the AF than PF region, and the ENaC blocker amiloride reduced the NaCl response size only for the former. Umami synergy, based on co-presenting MSG and IMP, was larger for the AF than PF region. The regions did not differ in response size to sour chemicals, but responses to l-lysine, l-arginine, sucrose, and tetrasodium pyrophosphate were larger for the AF than PF region. Thus, fungiform papillae on the tongue tip differed from those found further back in their transduction mechanisms for salty and umami compounds. Gustatory sensitivity also showed regional variation, albeit with a complex relationship to palatability and taste quality. Overall, the data support a regional organization for the mouse tongue, with different functional zones for the anterior, middle, and posterior thirds.
Collapse
Affiliation(s)
- Stuart A McCaughey
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States; Center for Medical Education, Ball State University, Muncie, IN, 47306, United States.
| |
Collapse
|
17
|
Abstract
The past decade has witnessed a consolidation and refinement of the extraordinary progress made in taste research. This Review describes recent advances in our understanding of taste receptors, taste buds, and the connections between taste buds and sensory afferent fibres. The article discusses new findings regarding the cellular mechanisms for detecting tastes, new data on the transmitters involved in taste processing and new studies that address longstanding arguments about taste coding.
Collapse
|
18
|
Boronat-García A, Reiter S, Sun K, Stopfer M. New Methods to Study Gustatory Coding. J Vis Exp 2017. [PMID: 28715373 PMCID: PMC5608530 DOI: 10.3791/55868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The sense of taste allows animals to detect chemicals in the environment, giving rise to behaviors critical for survival. When Gustatory Receptor Neurons (GRNs) detect tastant molecules, they encode information about the identity and concentration of the tastant as patterns of electrical activity that then propagate to follower neurons in the brain. These patterns constitute internal representations of the tastant, which then allow the animal to select actions and form memories. The use of relatively simple animal models has been a powerful tool to study basic principles in sensory coding. Here, we propose three new methods to study gustatory coding using the moth Manduca sexta. First, we present a dissection procedure for exposing the maxillary nerves and the subesophageal zone (SEZ), allowing recording of the activity of GRNs from their axons. Second, we describe the use of extracellular electrodes to record the activity of multiple GRNs by placing tetrode wires directly into the maxillary nerve. Third, we present a new system for delivering and monitoring, with high temporal precision, pulses of different tastants. These methods allow the characterization of neuronal responses in vivo directly from GRNs before, during and after tastants are delivered. We provide examples of voltage traces recorded from multiple GRNs, and present an example of how a spike sorting technique can be applied to the data to identify the responses of individual neurons. Finally, to validate our recording approach, we compare extracellular recordings obtained from GRNs with tetrodes to intracellular recordings obtained with sharp glass electrodes.
Collapse
Affiliation(s)
- Alejandra Boronat-García
- National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH)
| | - Sam Reiter
- National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH); Max Planck Institute for Brain Research
| | - Kui Sun
- National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH)
| | - Mark Stopfer
- National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH);
| |
Collapse
|
19
|
Qing-Hua Granule induces GLP-1 secretion via bitter taste receptor in db/db mice. Biomed Pharmacother 2017; 89:10-17. [PMID: 28213324 DOI: 10.1016/j.biopha.2017.01.168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 12/22/2022] Open
Abstract
Qing-Hua Granule (QHG), the modified formulation of a classical Chinese prescription named Gegen Qinlian Decoction, was clinically employed to treat type 2 diabetes mellitus (T2DM) through regulation of glucagon-like peptide-1 (GLP-1). However, the potential mechanism is unknown. We investigate whether QHG induces GLP-1 secretion via activation of bitter taste receptor (TAS2R) pathway in the gastrointestinal tract of db/db mice. The db/db mice were intragastrically (i.g.) administered QHG (low/medium/high dose) once daily for 8 weeks. GLP-1 secretion was evaluated. The bitter receptor signaling pathway, which regulates GLP-1 secretion, including TAS2R5 (a subtype of TAS2R), α-gustducin (Gαgust), 1-phosphatidylinositol-4, 5-bisphosphate phosphodiesterase beta-2 (PLCβ2), transient receptor potential cation channel subfamily M member 5 (TRPM5), was assessed by quantitative real-time polymerase chain reaction (qRT-PCR), Western blot and immunohistochemistry (IHC). The biochemical observations of ileum and pancreas tissue were detected histopathologically. Acquity Ultra Performance LCTM - Micromass ZQ 2000 (UPLC-MS) was used for the phytochemical analysis. QHG exhibited significant and dose-dependent effect on GLP-1 secretion in db/db mice, along with significant up-regulation of TAS2R5 mRNA level and activation of TAS2R pathway (p<0.05). In addition, QHG improved the histopathological structure of ileum and pancreatic tissue. Seventeen compounds were identified in QHG. In conclusion, QHG induces GLP-1 secretion in db/db mice, most likely through the bitter taste receptor pathway.
Collapse
|
20
|
|
21
|
Lemon CH, Kang Y, Li J. Separate functions for responses to oral temperature in thermo-gustatory and trigeminal neurons. Chem Senses 2016; 41:457-71. [PMID: 26976122 PMCID: PMC4910675 DOI: 10.1093/chemse/bjw022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oral temperature is a component and modifier of taste perception. Both trigeminal (V) and taste-sensitive cells, including those in the nucleus of the solitary tract (NTS), can respond to oral temperature. However, functional associations in thermal sensitivity between V and gustatory neurons are poorly understood. To study this we recorded electrophysiological responses to oral stimulation with cool (9, 15, 25, 32, and 34 °C) and warm (40 and 45 °C) temperatures from medullary V (n = 45) and taste-sensitive NTS (n = 27) neurons in anesthetized mice. Results showed temperatures below 34 °C activated the majority of V neurons but only a minority of NTS units. V neurons displayed larger responses to cooling and responded to temperatures that poorly stimulated NTS cells. Multivariate analyses revealed different temperatures induced larger differences in responses across V compared with NTS neurons, indicating V pathways possess greater capacity to signal temperature. Conversely, responses to temperature in NTS units associated with gustatory tuning. Further analyses identified two types of cooling-sensitive V neurons oriented toward innocuous or noxious cooling. Multivariate analyses indicated the combined response of these cells afforded distinction among a broad range of cool temperatures, suggesting multiple types of V neurons work together to represent oral cooling.
Collapse
Affiliation(s)
- Christian H Lemon
- Department of Biology, The University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA
| | - Yi Kang
- Department of Biology, The University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA
| | - Jinrong Li
- Department of Biology, The University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, USA
| |
Collapse
|
22
|
Tokita K, Boughter JD. Topographic organizations of taste-responsive neurons in the parabrachial nucleus of C57BL/6J mice: An electrophysiological mapping study. Neuroscience 2015; 316:151-66. [PMID: 26708748 DOI: 10.1016/j.neuroscience.2015.12.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 11/26/2022]
Abstract
The activities of 178 taste-responsive neurons were recorded extracellularly from the parabrachial nucleus (PbN) in the anesthetized C57BL/6J mouse. Taste stimuli included those representative of five basic taste qualities, sweet, salty, sour, bitter and umami. Umami synergism was represented by all sucrose-best and sweet-sensitive sodium chloride-best neurons. Mediolaterally the PbN was divided into medial, brachium conjunctivum (BC) and lateral subdivisions while rostrocaudally the PbN was divided into rostral and caudal subdivisions for mapping and reconstruction of recording sites. Neurons in the medial and BC subdivisions had a significantly greater magnitude of response to sucrose and to the mixture of monopotassium glutamate and inosine monophosphate than those found in the lateral subdivision. In contrast, neurons in the lateral subdivision possessed a more robust response to quinine hydrochloride. Rostrocaudally no difference was found in the mean magnitude of response. Analysis on the distribution pattern of neuron types classified by their best stimulus revealed that the proportion of neuron types in the medial vs. lateral and BC vs. lateral subdivisions was significantly different, with a greater amount of sucrose-best neurons found medially and within the BC, and a greater amount of sodium chloride-, citric acid- and quinine hydrochloride-best neurons found laterally. There was no significant difference in the neuron-type distribution between rostral and caudal PbN. We also assessed breadth of tuning in these neurons by calculating entropy (H) and noise-to-signal (N/S) ratio. The mean N/S ratio of all neurons (0.43) was significantly lower than that of H value (0.64). Neurons in the caudal PbN had a significantly higher H value than in the rostral PbN. In contrast, mean N/S ratios were not different both mediolaterally and rostrocaudally. These results suggest that although there is overlap in taste quality representation in the mouse PbN, taste-responsive neurons still possessed a topographic organization.
Collapse
Affiliation(s)
- K Tokita
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA.
| | - J D Boughter
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA
| |
Collapse
|
23
|
Abstract
UNLABELLED Four of the five major sensory systems (vision, olfaction, somatosensation, and audition) are thought to use different but partially overlapping sets of neurons to form unique representations of vast numbers of stimuli. The only exception is gustation, which is thought to represent only small numbers of basic taste categories. However, using new methods for delivering tastant chemicals and making electrophysiological recordings from the tractable gustatory system of the moth Manduca sexta, we found chemical-specific information is as follows: (1) initially encoded in the population of gustatory receptor neurons as broadly distributed spatiotemporal patterns of activity; (2) dramatically integrated and temporally transformed as it propagates to monosynaptically connected second-order neurons; and (3) observed in tastant-specific behavior. Our results are consistent with an emerging view of the gustatory system: rather than constructing basic taste categories, it uses a spatiotemporal population code to generate unique neural representations of individual tastant chemicals. SIGNIFICANCE STATEMENT Our results provide a new view of taste processing. Using a new, relatively simple model system and a new set of techniques to deliver taste stimuli and to examine gustatory receptor neurons and their immediate followers, we found no evidence for labeled line connectivity, or basic taste categories such as sweet, salty, bitter, and sour. Rather, individual tastant chemicals are represented as patterns of spiking activity distributed across populations of receptor neurons. These representations are transformed substantially as multiple types of receptor neurons converge upon follower neurons, leading to a combinatorial coding format that uniquely, rapidly, and efficiently represents individual taste chemicals. Finally, we found that the information content of these neurons can drive tastant-specific behavior.
Collapse
|
24
|
Sollai G, Tomassini Barbarossa I, Solari P, Crnjar R. Taste discriminating capability to different bitter compounds by the larval styloconic sensilla in the insect herbivore Papilio hospiton (Géné). JOURNAL OF INSECT PHYSIOLOGY 2015; 74:45-55. [PMID: 25702827 DOI: 10.1016/j.jinsphys.2015.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 06/04/2023]
Abstract
Herbivorous animals may benefit from the capability to discriminate the taste of bitter compounds since plants produce noxious compounds, some of which toxic, while others are only unpalatable. Our goal was to investigate the contribution of the peripheral taste system in the discrimination of different bitter compounds by an herbivorous insect using the larvae of Papilio hospiton Géné as the experimental model, showing a narrow choice range of host plants. The spike activity from the lateral and medial styloconic sensilla, housing two and one bitter-sensitive gustatory receptor neurons (GRNs), respectively, was recorded following stimulation with nicotine, caffeine, salicin and quercitrin and the time course of the discharges was analyzed. Nicotine and caffeine activated all three bitter-sensitive GRNs, while salicin and quercitrin affected only two of them. In feeding behavior bioassays, intact larvae ate glass-fiber disks moistened with salicin and quercitrin, but rejected those with nicotine and caffeine, while lateral sensillum-ablated insects also ate the disks with the two latter compounds. The capability to discriminate bitter taste stimuli and the neural codes involved are discussed.
Collapse
Affiliation(s)
- Giorgia Sollai
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, 09042 Monserrato, CA, Italy
| | - Iole Tomassini Barbarossa
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, 09042 Monserrato, CA, Italy
| | - Paolo Solari
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, 09042 Monserrato, CA, Italy
| | - Roberto Crnjar
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, 09042 Monserrato, CA, Italy.
| |
Collapse
|
25
|
Li J, Lemon CH. Influence of stimulus and oral adaptation temperature on gustatory responses in central taste-sensitive neurons. J Neurophysiol 2015; 113:2700-12. [PMID: 25673737 DOI: 10.1152/jn.00736.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 02/05/2015] [Indexed: 01/19/2023] Open
Abstract
The temperature of taste stimuli can modulate gustatory processing. Perceptual data indicate that the adapted temperature of oral epithelia also influences gustation, although little is known about the neural basis of this effect. Here, we electrophysiologically recorded orosensory responses (spikes) to 25°C (cool) and 35°C (warm) solutions of sucrose (0.1 and 0.3 M), NaCl (0.004, 0.1, and 0.3 M), and water from taste-sensitive neurons in the nucleus of the solitary tract in mice under varied thermal adaptation of oral epithelia. Conditions included presentation of taste stimuli isothermal to adaptation temperatures of 25°C (constant cooling) and 35°C (constant warming), delivery of 25°C stimuli following 35°C adaptation (relative cooling), and presentation of 35°C stimuli following 25°C adaptation (relative warming). Responses to sucrose in sucrose-oriented cells (n = 15) were enhanced under the constant and relative warming conditions compared with constant cooling, where contiguous cooling across adaptation and stimulus periods induced the lowest and longest latency responses to sucrose. Yet compared with constant warming, cooling sucrose following warm adaptation (relative cooling) only marginally reduced activity to 0.1 M sucrose and did not alter responses to 0.3 M sucrose. Thus, warmth adaptation counteracted the attenuation in sucrose activity associated with stimulus cooling. Analysis of sodium-oriented (n = 25) neurons revealed adaptation to cool water, and cooling taste solutions enhanced unit firing to 0.004 M (perithreshold) NaCl, whereas warmth adaptation and stimulus warming could facilitate activity to 0.3 M NaCl. The concentration dependence of this thermal effect may reflect a dual effect of temperature on the sodium reception mechanism that drives sodium-oriented cells.
Collapse
Affiliation(s)
- Jinrong Li
- Department of Biology, University of Oklahoma, Norman, Oklahoma
| | | |
Collapse
|
26
|
Coupland JN, Hayes JE. Physical approaches to masking bitter taste: lessons from food and pharmaceuticals. Pharm Res 2014; 31:2921-39. [PMID: 25205460 PMCID: PMC4898047 DOI: 10.1007/s11095-014-1480-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/15/2014] [Indexed: 01/08/2023]
Abstract
Many drugs and desirable phytochemicals are bitter, and bitter tastes are aversive. Food and pharmaceutical manufacturers share a common need for bitterness-masking strategies that allow them to deliver useful quantities of the active compounds in an acceptable form and in this review we compare and contrast the challenges and approaches by researchers in both fields. We focus on physical approaches, i.e., micro- or nano-structures to bind bitter compounds in the mouth, yet break down to allow release after they are swallowed. In all of these methods, the assumption is the degree of bitterness suppression depends on the concentration of bitterant in the saliva and hence the proportion that is bound. Surprisingly, this hypothesis has only rarely been fully tested using a combination of adequate human sensory trials and measurements of binding. This is especially true in pharmaceutical systems, perhaps due to the greater experimental challenges in sensory analysis of drugs.
Collapse
Affiliation(s)
- John N Coupland
- Department of Food Science, The Pennsylvania State University, 337 Food Science Building, University Park, Pennsylvania, 16802, USA,
| | | |
Collapse
|
27
|
Brasser SM, Castro N, Feretic B. Alcohol sensory processing and its relevance for ingestion. Physiol Behav 2014; 148:65-70. [PMID: 25304192 DOI: 10.1016/j.physbeh.2014.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 10/24/2022]
Abstract
Alcohol possesses complex sensory attributes that are first detected by the body via sensory receptors and afferent fibers that promptly transmit signals to brain areas involved in mediating ingestive motivation, reinforcement, and addictive behavior. Given that the chemosensory cues accompanying alcohol consumption are among the most intimate, consistent, and immediate predictors of alcohol's postabsorptive effects, with experience these stimuli also gain powerful associative incentive value to elicit craving and related physiologic changes, maintenance of ongoing alcohol use, and reinstatement of drug seeking after periods of abstinence. Despite the above, preclinical research has traditionally dichotomized alcohol's taste and postingestive influences as independent regulators of motivation to drink. The present review summarizes current evidence regarding alcohol's ability to directly activate peripheral and central oral chemosensory circuits, relevance for intake of the drug, and provides a framework for moving beyond a dissociation between the sensory and postabsorptive effects of alcohol to understand their neurobiological integration and significance for alcohol addiction.
Collapse
Affiliation(s)
- Susan M Brasser
- Department of Psychology, College of Sciences, San Diego State University, San Diego, CA 92120, USA.
| | - Norma Castro
- Department of Psychology, College of Sciences, San Diego State University, San Diego, CA 92120, USA
| | - Brian Feretic
- Department of Psychology, College of Sciences, San Diego State University, San Diego, CA 92120, USA
| |
Collapse
|
28
|
Wilson DM, Lemon CH. Temperature systematically modifies neural activity for sweet taste. J Neurophysiol 2014; 112:1667-77. [PMID: 24966301 DOI: 10.1152/jn.00368.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Temperature can modify neural and behavioral responses to taste stimuli that elicit "sweetness," a perception linked to intake of calorie-laden foods. However, the role of temperature in the neural representation of sweet taste is poorly understood. Here we made electrophysiological recordings from gustatory neurons in the medulla of inbred mice to study how adjustments in taste solution temperature to cool (18°C), ambient (22°C), and warm (30°C and 37°C) values changed the magnitude and latency of gustatory activity to sucrose (0, 0.05, 0.1, 0.17, 0.31, and 0.56 M). Analysis of 22 sucrose-best neurons revealed that temperature markedly influenced responses to sucrose, which, across concentrations, were largest when solutions were warmed to 30°C. However, reducing solution temperature from warm to ambient to cool progressively steepened the slope of the sucrose concentration-response function computed across cells (P < 0.05), indicating that mean activity to sucrose increased more rapidly with concentration steps under cooling than with warming. Thus the slope of the sucrose concentration-response function shows an inverse relation with temperature. Temperature also influenced latency to the first spike of the sucrose response. Across neurons, latencies were shorter when sucrose solutions were warmed and longer, by hundreds of milliseconds, when solutions were cooled (P < 0.05), indicating that temperature is also a temporal parameter of sucrose activity. Our findings reveal that temperature systematically modifies the timing of gustatory activity to sucrose in the mammalian brain and how this activity changes with concentration. Results further highlight how oral somatosensory cues function as physiological modulators of gustatory processing.
Collapse
Affiliation(s)
- David M Wilson
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri; and
| | | |
Collapse
|
29
|
Lin XB, Pierce DR, Light KE, Hayar A. The fine temporal structure of the rat licking pattern: what causes the variabiliy in the interlick intervals and how is it affected by the drinking solution? Chem Senses 2013; 38:685-704. [PMID: 23902635 DOI: 10.1093/chemse/bjt038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Licking is a repetitive behavior controlled by a central pattern generator. Even though interlick intervals (ILIs) within bursts of licks are considered fairly regular, the conditions that affect their variability are unknown. We analyzed the licking pattern in rats that licked water, 10% sucrose solution, or 10% ethanol solution, in 90-min recording sessions after 4h of water deprivation. The histograms of ILIs indicate that licking typically occurred at a preferred ILI of about 130-140ms with evidence of bimodal or multimodal distributions due to occasional licking failures. We found that the longer the pause between bursts of licks, the shorter was the first ILI of the burst. When bursts of licks were preceded by a pause >4 s, the ILI was the shortest (~110ms) at the beginning of the burst, and then it increased rapidly in the first few licks and slowly in subsequent licks. Interestingly, the first ILI of a burst of licks was not significantly different when licking any of the 3 solutions, but subsequent licks exhibited a temporal pattern characteristic of each solution. The rapid deceleration in intraburst licking rate was due to an increase from ~27ms to ~56ms in the tongue-spout contact duration while the intercontact interval was only slightly changed (80-90ms). Therefore, the contact duration seems to be the major factor that increases the variability in the ILIs and could be another means for the rat to adjust the amount of fluid ingested in each individual lick.
Collapse
Affiliation(s)
- Xiong Bin Lin
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301W. Markham Street Slot# 847, Little Rock, AR 72205, USA.
| | | | | | | |
Collapse
|
30
|
Sugita M, Yamamoto K, Hirono C, Shiba Y. Information processing in brainstem bitter taste-relaying neurons defined by genetic tracing. Neuroscience 2013; 250:166-80. [PMID: 23850686 DOI: 10.1016/j.neuroscience.2013.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 06/07/2013] [Accepted: 06/17/2013] [Indexed: 11/19/2022]
Abstract
Bitter reception is mediated by taste receptor cells that coexpress multiple T2Rs, a family of G-protein-coupled receptors. However, it remains elusive how bitter taste information is translated in the brain into appropriate behavioral responses. Here we used a combination of genetic tracing and electrophysiological and immunohistochemical analyses in mice to functionally characterize the neurons in the solitary tract nuclei of the medulla, which receive input from mT2R5-expressing cells. The neurons defined by a transneuronal tracer originating from mT2R5-expressing cells receive glutamatergic synaptic input via the AMPA receptor. The satiety peptide cholecystokinin increases glutamatergic transmission, suggesting an interaction between information processing of taste and the homeostatic control of feeding. Nevertheless, the tracer-labeled neuron types are heterogeneous, and can be classified into catecholamine and pro-opiomelanocortin neurons. Our data reveal that the architectural solution in the first-order central relay that processes information from mT2R5-expressing cells uses unique ensembles of neurons with different neurotransmitters.
Collapse
Affiliation(s)
- M Sugita
- Department of Physiology and Oral Physiology, Institute of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan.
| | | | | | | |
Collapse
|
31
|
Abstract
Changes in oral temperature can influence taste perception, indicating overlap among mechanisms for taste and oral somesthesis. Medullary gustatory neurons can show cosensitivity to temperature, albeit how these cells process combined taste and thermal input is poorly understood. Here, we electrophysiologically recorded orosensory responses (spikes) from 39 taste-sensitive neurons in the nucleus tractus solitarii of anesthetized mice during oral delivery of tastants adjusted to innocuous cool (16 and 18°C), room (22°C, baseline), and warm (30 and 37°C) oral temperatures. Stimuli included (in mM) 100 sucrose, 30 NaCl, 3 HCl, 3 quinine, an umami mixture, and water. Although cooled water excited few cells, water warmed to 30 and 37°C significantly excited 33% and 64% of neurons, respectively. Warmth induced responses of comparable magnitude to room temperature tastants. Furthermore, warming taste solutions influenced the distribution of gustatory responses among neurons and increased (P < 0.05) neuronal breadth of tuning across taste qualities. The influence of warmth on response magnitude was stimulus specific. Across neurons, warming facilitated responses to sucrose and umami in a superadditive manner, as these responses exceeded (P < 0.05) the arithmetic sum of activity to warming alone and the taste stimulus tested at room temperature. Superadditive increases (P < 0.05) in responding were also noted in some cells for warmed HCl. Yet warming induced only simple additive or subtractive effects on responses to quinine and NaCl. Data show temperature is a parameter of gustatory processing, like taste quality and concentration, in medullary circuits for taste.
Collapse
Affiliation(s)
- David M Wilson
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|