1
|
Langhans W, Watts AG, Spector AC. The elusive cephalic phase insulin response: triggers, mechanisms, and functions. Physiol Rev 2023; 103:1423-1485. [PMID: 36422994 PMCID: PMC9942918 DOI: 10.1152/physrev.00025.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/04/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
The cephalic phase insulin response (CPIR) is classically defined as a head receptor-induced early release of insulin during eating that precedes a postabsorptive rise in blood glucose. Here we discuss, first, the various stimuli that elicit the CPIR and the sensory signaling pathways (sensory limb) involved; second, the efferent pathways that control the various endocrine events associated with eating (motor limb); and third, what is known about the central integrative processes linking the sensory and motor limbs. Fourth, in doing so, we identify open questions and problems with respect to the CPIR in general. Specifically, we consider test conditions that allow, or may not allow, the stimulus to reach the potentially relevant taste receptors and to trigger a CPIR. The possible significance of sweetness and palatability as crucial stimulus features and whether conditioning plays a role in the CPIR are also discussed. Moreover, we ponder the utility of the strict classical CPIR definition based on what is known about the effects of vagal motor neuron activation and thereby acetylcholine on the β-cells, together with the difficulties of the accurate assessment of insulin release. Finally, we weigh the evidence of the physiological and clinical relevance of the cephalic contribution to the release of insulin that occurs during and after a meal. These points are critical for the interpretation of the existing data, and they support a sharper focus on the role of head receptors in the overall insulin response to eating rather than relying solely on the classical CPIR definition.
Collapse
Affiliation(s)
- Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zürich, Schwerzenbach, Switzerland
| | - Alan G Watts
- Department of Biological Sciences, USC Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Alan C Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| |
Collapse
|
2
|
Hakimi S, Kari NM, Ismail N, Ismail MN, Ahmad F. Evaluation of taste active peptides and amino acids from anchovy proteins in fish sauce by in silico approach. Food Sci Biotechnol 2022; 31:767-785. [DOI: 10.1007/s10068-022-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 11/04/2022] Open
|
3
|
Rhyu MR, Lyall V. Interaction of taste-active nutrients with taste receptors. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2020.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
4
|
Rhyu MR, Kim Y, Misaka T. Suppression of hTAS2R16 Signaling by Umami Substances. Int J Mol Sci 2020; 21:ijms21197045. [PMID: 32987926 PMCID: PMC7582725 DOI: 10.3390/ijms21197045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 01/27/2023] Open
Abstract
Interaction between umami and bitter taste has long been observed in human sensory studies and in neural responses in animal models, however, the molecular mechanism for their action has not been delineated. Humans detect diverse bitter compounds using 25-30 members of the type 2 taste receptor (TAS2R) family of G protein-coupled receptor. In this study, we investigated the putative mechanism of antagonism by umami substances using HEK293T cells expressing hTAS2R16 and two known probenecid-insensitive mutant receptors, hTAS2R16 N96T and P44T. In wild type receptor, Glu-Glu, inosine monophosphate (IMP), and l-theanine behave as partial insurmountable antagonists, and monosodium glutamate (MSG) acts as a surmountable antagonist in comparison with probenecid as a full insurmountable antagonist. The synergism with IMP of umami substances still stands in the suppression of hTAS2R16 signaling. In mutagenesis analysis, we found that Glu-Glu, MSG, and l-theanine share at least one critical binding site on N96 and P44 with probenecid. These results provide the first evidence for a direct binding of umami substances to the hTAS2R16 through the probenecid binding pocket on the receptor, resulting in the suppression of bitterness.
Collapse
Affiliation(s)
- Mee-Ra Rhyu
- Division of Functional Food Research, Korea Food Research Institute, Jeollabuk-do 55365, Korea;
- Correspondence: ; Tel.: +82-63-219-9268; Fax: +82-63-219-9876
| | - Yiseul Kim
- Division of Functional Food Research, Korea Food Research Institute, Jeollabuk-do 55365, Korea;
| | - Takumi Misaka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan;
| |
Collapse
|
5
|
Kalyanasundar B, Blonde GD, Spector AC, Travers SP. Electrophysiological responses to sugars and amino acids in the nucleus of the solitary tract of type 1 taste receptor double-knockout mice. J Neurophysiol 2020; 123:843-859. [PMID: 31913749 DOI: 10.1152/jn.00584.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Strong evidence supports a major role for heterodimers of the type 1 taste receptor (T1R) family in the taste transduction of sugars (T1R2+T1R3) and amino acids (T1R1+T1R3), but there are also neural and behavioral data supporting T1R-independent mechanisms. Most neural evidence for alternate mechanisms comes from whole nerve recordings in mice with deletion of a single T1R family member, limiting conclusions about the functional significance and T1R independence of the remaining responses. To clarify these issues, we recorded single-unit taste responses from the nucleus of the solitary tract in T1R double-knockout (double-KO) mice lacking functional T1R1+T1R3 [KO1+3] or T1R2+T1R3 [KO2+3] receptors and their wild-type background strains [WT; C57BL/6J (B6), 129X1/SvJ (S129)]. In both double-KO strains, responses to sugars and a moderate concentration of an monosodium glutamate + amiloride + inosine 5'-monophosphate cocktail (0.1 M, i.e., umami) were profoundly depressed, whereas a panel of 0.6 M amino acids were mostly unaffected. Strikingly, in contrast to WT mice, no double-KO neurons responded selectively to sugars and umami, precluding segregation of this group of stimuli from those representing other taste qualities in a multidimensional scaling analysis. Nevertheless, residual sugar responses, mainly elicited by monosaccharides, persisted as small "sideband" responses in double-KOs. Thus other receptors may convey limited information about sugars to the central nervous system, but T1Rs appear critical for coding the distinct perceptual features of sugar and umami stimuli. The persistence of amino acid responses supports previous proposals of alternate receptors, but because these stimuli affected multiple neuron types, further investigations are necessary.NEW & NOTEWORTHY The type 1 taste receptor (T1R) family is crucial for transducing sugars and amino acids, but there is evidence for T1R-independent mechanisms. In this study, single-unit recordings from the nucleus of the solitary tract in T1R double-knockout mice lacking T1R1+T1R3 or T1R2+T1R3 receptors revealed greatly reduced umami synergism and sugar responses. Nevertheless, residual sugar responses persisted, mainly elicited by monosaccharides and evident as "sidebands" in neurons activated more vigorously by other qualities.
Collapse
Affiliation(s)
- B Kalyanasundar
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, Ohio
| | - Ginger D Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| | - Alan C Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida
| | - Susan P Travers
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, Ohio
| |
Collapse
|
6
|
Ke X, Lin J, Li P, Wu Z, Xu R, Ci Z, Yang M, Han L, Zhang D. Transcriptional profiling of genes in tongue epithelial tissue from immature and adult rats by the RNA-Seq technique. J Cell Physiol 2019; 235:3069-3078. [PMID: 31549403 DOI: 10.1002/jcp.29211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/03/2019] [Indexed: 12/29/2022]
Abstract
Children are more sensitive than adults to bitterness and thus dislike bitter tastes more than adults do. However, why children are more sensitive to bitterness has never been revealed. To elucidate the effects of age on taste perception, a double-bottle preference test was first performed with immature and adult rats. Then, RNA-Seq analysis was performed on tongues obtained from rats of the same ages as those in the double-bottle test. The immature rats exhibited a lower consumption rate of bitter solution than the adult rats. Bioinformatics analysis yielded 1,347 differentially expressed genes (DEGs) between male adult rats (MARs, 80 days old) and male immature rats (MIRs, 20 days old) and 380 DEGs between female adult rats (FARs, 80 days old) and female immature rats (FIRs, 20 days old). These DEGs were mainly associated with growth, development, differentiation, and extracellular processes, among other mechanisms. According to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, the DEGs were enriched for bitter taste transduction. Specifically, the Gnb3 and TRPM5 genes were downregulated in FARs compared with FIRs and in MARs compared with MIRs, and the protein expression of TRPM5 was significantly downregulated in MARs compared with MIRs. The data presented herein suggest that transcriptional regulation of taste-associated signal transduction occurs differently in tongue epithelial tissue of rats at different ages, although additional analyses are needed to confirm this conclusion.
Collapse
Affiliation(s)
- Xiumei Ke
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
| | - Junzhi Lin
- Central Laboratory, The Teaching Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pan Li
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenfeng Wu
- School of pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Runchun Xu
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhimin Ci
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming Yang
- School of pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Li Han
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dingkun Zhang
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Xu Q, Singh N, Hong H, Yan X, Yu W, Jiang X, Chelikani P, Wu J. Hen protein-derived peptides as the blockers of human bitter taste receptors T2R4, T2R7 and T2R14. Food Chem 2019; 283:621-627. [PMID: 30722920 DOI: 10.1016/j.foodchem.2019.01.059] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/05/2018] [Accepted: 01/08/2019] [Indexed: 01/02/2023]
Abstract
Bitter sensation is mediated by various bitter taste receptors (T2Rs), thus T2R antagonists are actively explored. Our objective was to look for novel T2R blockers in hen protein hydrolysate (HPH). We screened the least bitter HPH fractions using electronic tongue, and analyzed their peptide sequences and calcium mobilization in HEK293T cells expressing T2Rs. The results showed that the HPH fractions with higher bitterness intensity had higher hydrophobicity, more hydrophobic amino acids, and more positively charged peptides, but fewer known umami peptides. The peptide fractions from the least bitter HPH fraction significantly inhibited quinine bitterness (P < 0.05), and also significantly inhibited quinine- or diphenhydramine-dependent calcium mobilization of HEK293T cells expressing human T2R4, T2R7, or T2R14 (P < 0.05). Among them, the first eluted (least bitter) peptide fraction showed the strongest bitter-inhibitory effect. In conclusion, HPH peptides are the blockers of T2R4, T2R7, and T2R14.
Collapse
Affiliation(s)
- Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan 430070, China; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Nisha Singh
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, University of Manitoba, Winnipeg, Manitoba R3E 0W2, Canada
| | - Hui Hong
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Xianghua Yan
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan 430070, China
| | - Wenlin Yu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Xu Jiang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Prashen Chelikani
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, University of Manitoba, Winnipeg, Manitoba R3E 0W2, Canada
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada.
| |
Collapse
|
8
|
Abstract
The gustatory system contributes to the flavor of foods and beverages and communicates information about nutrients and poisons. This system has evolved to detect and ultimately respond to hydrophilic molecules dissolved in saliva. Taste receptor cells, located in taste buds and distributed throughout the oral cavity, activate nerve afferents that project to the brainstem. From here, information propagates to thalamic, subcortical, and cortical areas, where it is integrated with information from other sensory systems and with homeostatic, visceral, and affective processes. There is considerable divergence, as well as convergence, of information between multiple regions of the central nervous system that interact with the taste pathways, with reciprocal connections occurring between the involved regions. These widespread interactions among multiple systems are crucial for the perception of food. For example, memory, hunger, satiety, and visceral changes can directly affect and can be affected by the experience of tasting. In this chapter, we review the literature on the central processing of taste with a specific focus on the anatomic and physiologic responses of single neurons. Emphasis is placed on how information is distributed along multiple systems with the goal of better understanding how the rich and complex sensations associated with flavor emerge from large-scale, systems-wide, interactions.
Collapse
|
9
|
Hashimoto M, Yamanaka A, Kato S, Tanifuji M, Kobayashi K, Yaginuma H. Anatomical Evidence for a Direct Projection from Purkinje Cells in the Mouse Cerebellar Vermis to Medial Parabrachial Nucleus. Front Neural Circuits 2018; 12:6. [PMID: 29467628 PMCID: PMC5808303 DOI: 10.3389/fncir.2018.00006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 01/12/2018] [Indexed: 11/28/2022] Open
Abstract
Cerebellar malformations cause changes to the sleep-wake cycle, resulting in sleep disturbance. However, it is unclear how the cerebellum contributes to the sleep-wake cycle. To examine the neural connections between the cerebellum and the nuclei involved in the sleep-wake cycle, we investigated the axonal projections of Purkinje cells in the mouse posterior vermis by using an adeno-associated virus (AAV) vector (serotype rh10) as an anterograde tracer. When an AAV vector expressing humanized renilla green fluorescent protein was injected into the cerebellar lobule IX, hrGFP and synaptophysin double-positive axonal terminals were observed in the region of medial parabrachial nucleus (MPB). The MPB is involved in the phase transition from rapid eye movement (REM) sleep to Non-REM sleep and vice versa, and the cardiovascular and respiratory responses. The hrGFP-positive axons from lobule IX went through the ventral spinocerebellar tract and finally reached the MPB. By contrast, when the AAV vector was injected into cerebellar lobule VI, no hrGFP-positive axons were observed in the MPB. To examine neurons projecting to the MPB, we unilaterally injected Fast Blue and AAV vector (retrograde serotype, rAAV2-retro) as retrograde tracers into the MPB. The cerebellar Purkinje cells in lobules VIII–X on the ipsilateral side of the Fast Blue-injected MPB were retrogradely labeled by Fast Blue and AAV vector (retrograde serotype), but no retrograde-labeled Purkinje cells were observed in lobules VI–VII and the cerebellar hemispheres. These results indicated that Purkinje cells in lobules VIII–X directly project their axons to the ipsilateral MPB but not lobules VI–VII. The direct connection between lobules VIII–X and the MPB suggests that the cerebellum participates in the neural network controlling the sleep-wake cycle, and cardiovascular and respiratory responses, by modulating the physiological function of the MPB.
Collapse
Affiliation(s)
- Mitsuhiro Hashimoto
- Department of Neuroanatomy and Embryology, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan.,Brain Interdisciplinary Research Division, Research Institute for Science and Technology, Tokyo University of Science, Noda-shi, Japan.,Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya-shi, Japan.,Laboratory for Integrative Neural Systems, RIKEN Brain Science Institute, Saitama, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya-shi, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan
| | - Manabu Tanifuji
- Laboratory for Integrative Neural Systems, RIKEN Brain Science Institute, Saitama, Japan.,Department of Life Science and Medical Bio-Science, Faculty of Science and Engineering, Waseda University, Tokyo, Japan.,Department of Complexity Science and Engineering, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan
| | - Hiroyuki Yaginuma
- Department of Neuroanatomy and Embryology, Fukushima Medical University Graduate School of Medicine, Fukushima, Japan
| |
Collapse
|
10
|
Mast TG, Breza JM, Contreras RJ. Thirst Increases Chorda Tympani Responses to Sodium Chloride. Chem Senses 2017; 42:675-681. [PMID: 28981824 DOI: 10.1093/chemse/bjx052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In nature, water is present as a low-salt solution, thus we hypothesized that thirst would increase taste responses to low-salt solutions. We investigated the effect of thirst on the 2 different salt detection mechanisms present in the rat chorda tympani (CT) nerve. The first mechanism is dependent upon the epithelial sodium channel (ENaC), is blocked by benzamil, and is specific to the cation sodium. The second mechanism, while undefined, is independent of ENaC, and detects multiple cations. We expected thirst to increase benzamil-sensitive sodium responses due to mechanistically increasing the benzamil-sensitive ENaC. We recorded CT whole-nerve electrophysiological responses to lingual application of NaCl, KCl (30, 75, 150, 300, 500, and 600 mM), and imitation rainwater in both control and 24-h water-restricted male rats. NaCl solutions were presented in artificial saliva before and after lingual application of 5µM benzamil. Water restriction significantly increased the integrated CT responses to NaCl but not to KCl or imitation rainwater. Consistent with our hypothesis, only the benzamil-sensitive, and not the benzamil-insensitive, CT sodium response significantly increased. Additionally, CT responses to salt were recorded following induction of either osmotic or volemic thirst. Both thirsts significantly enhanced the integrated CT responses to NaCl and KCl, but not imitation rainwater. Interestingly, osmotic and volemic thirsts increased CT responses by increasing both the benzamil-sensitive and benzamil-insensitive CT sodium responses. We propose that thirst increases the sensitivity of the CT nerve to sodium.
Collapse
Affiliation(s)
- Thomas G Mast
- Department of Biology, Program in Neuroscience, Eastern Michigan University, Ypsilanti, MI 48197, USA.,Department of Psychology, Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 30306, USA
| | - Joseph M Breza
- Department of Psychology, Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 30306, USA.,Department of Psychology, Program in Neuroscience, Eastern Michigan University, Ypsilanti, MI 48197, USA
| | - Robert J Contreras
- Department of Psychology, Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 30306, USA
| |
Collapse
|
11
|
Kim Y, Kim EY, Son HJ, Lee JJ, Choi YH, Rhyu MR. Identification of a key umami-active fraction in modernized Korean soy sauce and the impact thereof on bitter-masking. Food Chem 2017; 233:256-262. [PMID: 28530573 DOI: 10.1016/j.foodchem.2017.04.123] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 10/19/2022]
Abstract
Food protein hydrolysates created by natural fermentation have been used for centuries as food flavorings. The aim of this study was to define the key umami-active fraction of modernized Korean soy sauce (mJGN) and the impact thereof on bitter-masking of human sensory and bitter-taste receptor-expressing cells. We found strong correlations between taste profiles of mJGN and a contained fraction (F05). The latter contained compounds of less than 500Da, and elicits a distinct umami taste. Both free amino acids and Glu-enriched oligopeptides are suggested to be crucial in terms of the effects of F05 on taste. F05 not only reduced human-perceived bitterness, but also effectively suppressed the intracellular Ca2+ response induced by caffeine in the hTAS2R43 and hTAS2R46 human bitter-taste receptor-expressing cells. This suggests that F05, a key umami-active fraction of mJGN, contains components that at least partially modulate human bitter-taste receptor action, improving food flavor.
Collapse
Affiliation(s)
- Yiseul Kim
- Division of Functional Food Research, Korea Food Research Institute, Bundang-gu, Sungnam-si, Gyeonggi-do 13539, Republic of Korea
| | - Eun-Young Kim
- Division of Functional Food Research, Korea Food Research Institute, Bundang-gu, Sungnam-si, Gyeonggi-do 13539, Republic of Korea
| | - Hee Jin Son
- Division of Functional Food Research, Korea Food Research Institute, Bundang-gu, Sungnam-si, Gyeonggi-do 13539, Republic of Korea
| | - Jai-Jung Lee
- Sempio Fermentation Research Center 1, Sempio Foods Company, Osongup, Cheongwongun, Chungcheongbuk-do 28156, Republic of Korea
| | - Yong-Ho Choi
- Sempio Fermentation Research Center 1, Sempio Foods Company, Osongup, Cheongwongun, Chungcheongbuk-do 28156, Republic of Korea
| | - Mee-Ra Rhyu
- Division of Functional Food Research, Korea Food Research Institute, Bundang-gu, Sungnam-si, Gyeonggi-do 13539, Republic of Korea.
| |
Collapse
|
12
|
Hill JW, Faulkner LD. The Role of the Melanocortin System in Metabolic Disease: New Developments and Advances. Neuroendocrinology 2017; 104:330-346. [PMID: 27728914 PMCID: PMC5724371 DOI: 10.1159/000450649] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/01/2016] [Indexed: 12/17/2022]
Abstract
Obesity is increasing in prevalence across all sectors of society, and with it a constellation of associated ailments including hypertension, type 2 diabetes, and eating disorders. The melanocortin system is a critical neural system underlying the control of body weight and other functions. Deficits in the melanocortin system may promote or exacerbate the comorbidities of obesity. This system has therefore generated great interest as a potential target for treatment of obesity. However, drugs targeting melanocortin receptors are plagued by problematic side effects, including undesirable increases in sympathetic nervous system activity, heart rate, and blood pressure. Circumnavigating this roadblock will require a clearer picture of the precise neural circuits that mediate the functions of melanocortins. Recent, novel experimental approaches have significantly advanced our understanding of these pathways. We here review the latest advances in our understanding of the role of melanocortins in food intake, reward pathways, blood pressure, glucose control, and energy expenditure. The evidence suggests that downstream melanocortin-responsive circuits responsible for different physiological actions do diverge. Ultimately, a more complete understanding of melanocortin pathways and their myriad roles should allow treatments tailored to the mix of metabolic disorders in the individual patient.
Collapse
Affiliation(s)
- Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine, The University of Toledo, Toledo, OH, USA
| | | |
Collapse
|
13
|
Katagawa Y, Yasuo T, Suwabe T, Yamamura T, Gen K, Sako N. Recognition by Rats of Binary Taste Solutions and Their Components. Chem Senses 2016; 41:795-801. [PMID: 27624788 DOI: 10.1093/chemse/bjw093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This behavioral study investigated how rats conditioned to binary mixtures of preferred and aversive taste stimuli, respectively, responded to the individual components in a conditioned taste aversion (CTA) paradigm. The preference of stimuli was determined based on the initial results of 2 bottle preference test. The preferred stimuli included 5mM sodium saccharin (Sacc), 0.03M NaCl (Na), 0.1M Na, 5mM Sacc + 0.03M Na, and 5mM Sacc + 0.2mM quinine hydrochloride (Q), whereas the aversive stimuli tested were 1.0M Na, 0.2mM Q, 0.3mM Q, 5mM Sacc + 1.0M Na, and 5mM Sacc + 0.3mM Q. In CTA tests where LiCl was the unconditioned stimulus, the number of licks to the preferred binary mixtures and to all tested preferred components were significantly less than in control rats. No significant difference resulted between the number of licks to the aversive binary mixtures or to all tested aversive components. However, when rats pre-exposed to the aversive components contained of the aversive binary mixtures were conditioned to these mixtures, the number of licks to all the tested stimuli was significantly less than in controls. Rats conditioned to components of the aversive binary mixtures generalized to the binary mixtures containing those components. These results suggest that rats recognize and remember preferred and aversive taste mixtures as well as the preferred and aversive components of the binary mixtures, and that pre-exposure before CTA is an available method to study the recognition of aversive taste stimuli.
Collapse
Affiliation(s)
- Yoshihisa Katagawa
- Department of Oral Anatomy , Asahi University School of Dentistry , 1851-1 Hozumi , Mizuho , Gifu 501-0296 , Japan.,Present address: Department of Dentistry for Disability and Oral Health, Asahi University School of Dentistry , 1851-1 Hozumi, Mizuho, Gifu 501-0296 , Japan
| | - Toshiaki Yasuo
- Department of Oral Physiology , Asahi University School of Dentistry , 1851-1 Hozumi , Mizuho , Gifu 501-0296 , Japan and
| | - Takeshi Suwabe
- Department of Oral Physiology , Asahi University School of Dentistry , 1851-1 Hozumi , Mizuho , Gifu 501-0296 , Japan and
| | - Tomoki Yamamura
- Department of Oral Physiology , Asahi University School of Dentistry , 1851-1 Hozumi , Mizuho , Gifu 501-0296 , Japan and
| | - Keika Gen
- Department of Dentistry for Disability and Oral Health , Asahi University School of Dentistry , 1851-1 Hozumi , Mizuho , Gifu 501-0296 , Japan
| | - Noritaka Sako
- Department of Oral Physiology , Asahi University School of Dentistry , 1851-1 Hozumi , Mizuho , Gifu 501-0296 , Japan and
| |
Collapse
|
14
|
Formation of taste-active amino acids, amino acid derivatives and peptides in food fermentations – A review. Food Res Int 2016; 89:39-47. [DOI: 10.1016/j.foodres.2016.08.042] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 07/23/2016] [Accepted: 08/27/2016] [Indexed: 11/22/2022]
|
15
|
Beauchamp GK. Why do we like sweet taste: A bitter tale? Physiol Behav 2016; 164:432-437. [PMID: 27174610 PMCID: PMC5003684 DOI: 10.1016/j.physbeh.2016.05.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 10/21/2022]
Abstract
Sweet is widely considered to be one of a small number of basic or primary taste qualities. Liking for sweet tasting substances is innate, although postnatal experiences can shape responses. The power of sweet taste to induce consumption and to motivate behavior is profound, suggesting the importance of this sense for many species. Most investigators presume that the ability to identify sweet molecules through the sense of taste evolved to allow organisms to detect sources of readily available glucose from plants. Perhaps the best evidence supporting this presumption are recent discoveries in comparative biology demonstrating that species in the order Carnivora that do not consume plants also do not perceive sweet taste due to the pseudogenization of a component of the primary sweet taste receptor. However, arguing against this idea is the observation that the sweetness of a plant, or the amount of easily metabolizable sugars contained in the plant, provides little quantitative indication of the plant's energy or broadly conceived food value. Here it is suggested that the perceptual ratio of sweet taste to bitter taste (a signal for toxicity) may be a better gauge of a plant's broadly conceived food value than sweetness alone and that it is this ratio that helps guide selection or rejection of a potential plant food.
Collapse
Affiliation(s)
- Gary K Beauchamp
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, United States.
| |
Collapse
|
16
|
Raspberry marc extracts increase antioxidative potential, ellagic acid, ellagitannin and anthocyanin concentrations in fruit purees. Lebensm Wiss Technol 2016. [DOI: 10.1016/j.lwt.2015.10.069] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
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: 2.9] [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
|
18
|
Parabrachial calcitonin gene-related peptide neurons mediate conditioned taste aversion. J Neurosci 2015; 35:4582-6. [PMID: 25788675 DOI: 10.1523/jneurosci.3729-14.2015] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Conditioned taste aversion (CTA) is a phenomenon in which an individual forms an association between a novel tastant and toxin-induced gastrointestinal malaise. Previous studies showed that the parabrachial nucleus (PBN) contains neurons that are necessary for the acquisition of CTA, but the specific neuronal populations involved are unknown. Previously, we identified calcitonin gene-related peptide (CGRP)-expressing neurons in the external lateral subdivision of the PBN (PBel) as being sufficient to suppress appetite and necessary for the anorexigenic effects of appetite-suppressing substances including lithium chloride (LiCl), a compound often used to induce CTA. Here, we test the hypothesis that PBel CGRP neurons are sufficient and necessary for CTA acquisition in mice. We show that optogenetic activation of these neurons is sufficient to induce CTA in the absence of anorexigenic substances, whereas genetically induced silencing of these neurons attenuates acquisition of CTA upon exposure to LiCl. Together, these results demonstrate that PBel CGRP neurons mediate a gastrointestinal distress signal required to establish CTA.
Collapse
|
19
|
Modulation of sweet taste by umami compounds via sweet taste receptor subunit hT1R2. PLoS One 2015; 10:e0124030. [PMID: 25853419 PMCID: PMC4390298 DOI: 10.1371/journal.pone.0124030] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 02/26/2015] [Indexed: 01/17/2023] Open
Abstract
Although the five basic taste qualities—sweet, sour, bitter, salty and umami—can be recognized by the respective gustatory system, interactions between these taste qualities are often experienced when food is consumed. Specifically, the umami taste has been investigated in terms of whether it enhances or reduces the other taste modalities. These studies, however, are based on individual perception and not on a molecular level. In this study we investigated umami-sweet taste interactions using umami compounds including monosodium glutamate (MSG), 5’-mononucleotides and glutamyl-dipeptides, glutamate-glutamate (Glu-Glu) and glutamate-aspartic acid (Glu-Asp), in human sweet taste receptor hT1R2/hT1R3-expressing cells. The sensitivity of sucrose to hT1R2/hT1R3 was significantly attenuated by MSG and umami active peptides but not by umami active nucleotides. Inhibition of sweet receptor activation by MSG and glutamyl peptides is obvious when sweet receptors are activated by sweeteners that target the extracellular domain (ECD) of T1R2, such as sucrose and acesulfame K, but not by cyclamate, which interact with the T1R3 transmembrane domain (TMD). Application of umami compounds with lactisole, inhibitory drugs that target T1R3, exerted a more severe inhibitory effect. The inhibition was also observed with F778A sweet receptor mutant, which have the defect in function of T1R3 TMD. These results suggest that umami peptides affect sweet taste receptors and this interaction prevents sweet receptor agonists from binding to the T1R2 ECD in an allosteric manner, not to the T1R3. This is the first report to define the interaction between umami and sweet taste receptors.
Collapse
|
20
|
Saites LN, Goldsmith Z, Densky J, Guedes VA, Boughter JD. Mice perceive synergistic umami mixtures as tasting sweet. Chem Senses 2015; 40:295-303. [PMID: 25820205 DOI: 10.1093/chemse/bjv010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous electrophysiological investigation shows that combinations of compounds classified by humans as umami-tasting, such as glutamate salts and 5'-ribonucleotides, elicit synergistic responses in neurons throughout the rodent taste system and produce a pattern that resembles responses to sweet compounds. The current study tested the hypothesis that a synergistic mixture of monopotassium glutamate (MPG) and inositol monophosphate (IMP) possesses perceptual similarity to sucrose in mice. We estimated behavioral similarity among these tastants and the individual umami compounds using a series of conditioned taste aversion (CTA) tests, a procedure that measures whether a CTA formed to one stimulus generalizes to another. Our primary finding was that a CTA to a synergistic mixture of MPG + IMP generalizes to sucrose, and vice-versa. This indicates umami synergistic mixtures are perceived as having a sweet, or at least sucrose-like, taste to mice. Considering other recent studies, our data argue strongly in favor of multiple receptor mechanisms for umami detection, and complexity in taste perception models for rodents.
Collapse
Affiliation(s)
- Louis N Saites
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA
| | - Zachary Goldsmith
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA
| | - Jaron Densky
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA
| | - Vivian A Guedes
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA
| | - John D Boughter
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Suite 515, Memphis, TN 38163, USA
| |
Collapse
|
21
|
Grone BP, Maruska KP. A second corticotropin-releasing hormone gene (CRH2) is conserved across vertebrate classes and expressed in the hindbrain of a basal neopterygian fish, the spotted gar (Lepisosteus oculatus). J Comp Neurol 2015; 523:1125-43. [PMID: 25521515 DOI: 10.1002/cne.23729] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 12/31/2022]
Abstract
To investigate the origins of the vertebrate stress-response system, we searched sequenced vertebrate genomes for genes resembling corticotropin-releasing hormone (CRH). We found that vertebrate genomes possess, in addition to CRH, another gene that resembles CRH in sequence and syntenic environment. This paralogous gene was previously identified only in the elephant shark (a holocephalan), but we find it also in marsupials, monotremes, lizards, turtles, birds, and fishes. We examined the relationship of this second vertebrate CRH gene, which we name CRH2, to CRH1 (previously known as CRH) and urocortin1/urotensin1 (UCN1/UTS1) in primitive fishes, teleosts, and tetrapods. The paralogs CRH1 and CRH2 likely evolved via duplication of CRH during a whole-genome duplication early in the vertebrate lineage. CRH2 was subsequently lost in both teleost fishes and eutherian mammals but retained in other lineages. To determine where CRH2 is expressed relative to CRH1 and UTS1, we used in situ hybridization on brain tissue from spotted gar (Lepisosteus oculatus), a neopterygian fish closely related to teleosts. In situ hybridization revealed widespread distribution of both crh1 and uts1 in the brain. Expression of crh2 was restricted to the putative secondary gustatory/secondary visceral nucleus, which also expressed calcitonin-related polypeptide alpha (calca), a marker of parabrachial nucleus in mammals. Thus, the evolutionary history of CRH2 includes restricted expression in the brain, sequence changes, and gene loss, likely reflecting release of selective constraints following whole-genome duplication. The discovery of CRH2 opens many new possibilities for understanding the diverse functions of the CRH family of peptides across vertebrates.
Collapse
Affiliation(s)
- Brian P Grone
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, 94143
| | | |
Collapse
|
22
|
Kim MJ, Son HJ, Kim Y, Misaka T, Rhyu MR. Umami–bitter interactions: The suppression of bitterness by umami peptides via human bitter taste receptor. Biochem Biophys Res Commun 2015; 456:586-90. [DOI: 10.1016/j.bbrc.2014.11.114] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 11/28/2014] [Indexed: 11/16/2022]
|
23
|
Abstract
TRP channels are expressed in taste buds, nerve fibers, and keratinocytes in the oronasal cavity. These channels play integral roles in transducing chemical stimuli, giving rise to sensations of taste, irritation, warmth, coolness, and pungency. Specifically, TRPM5 acts downstream of taste receptors in the taste transduction pathway. TRPM5 channels convert taste-evoked intracellular Ca(2+) release into membrane depolarization to trigger taste transmitter secretion. PKD2L1 is expressed in acid-sensitive (sour) taste bud cells but is unlikely to be the transducer for sour taste. TRPV1 is a receptor for pungent chemical stimuli such as capsaicin and for several irritants (chemesthesis). It is controversial whether TRPV1 is present in the taste buds and plays a direct role in taste. Instead, TRPV1 is expressed in non-gustatory sensory afferent fibers and in keratinocytes of the oronasal cavity. In many sensory fibers and epithelial cells lining the oronasal cavity, TRPA1 is also co-expressed with TRPV1. As with TRPV1, TRPA1 transduces a wide variety of irritants and, in combination with TRPV1, assures that there is a broad response to noxious chemical stimuli. Other TRP channels, including TRPM8, TRPV3, and TRPV4, play less prominent roles in chemesthesis and no known role in taste, per se. The pungency of foods and beverages is likely highly influenced by the temperature at which they are consumed, their acidity, and, for beverages, their carbonation. All these factors modulate the activity of TRP channels in taste buds and in the oronasal mucosa.
Collapse
Affiliation(s)
- Stephen D Roper
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Ave., Miami, FL, 33136, USA,
| |
Collapse
|
24
|
Magableh A, Lundy R. Somatostatin and corticotrophin releasing hormone cell types are a major source of descending input from the forebrain to the parabrachial nucleus in mice. Chem Senses 2014; 39:673-82. [PMID: 25086873 DOI: 10.1093/chemse/bju038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The pontine parabrachial nucleus (PBN) receives substantial descending input from higher order forebrain regions that exerts inhibitory and excitatory influences on taste-evoked responses. Somatostatin (Sst) and corticotrophin releasing hormone (Crh) reporter mice were used in conjunction with injection of the retrograde tracer CTb-488 into the caudal PBN to determine the extent to which Sst and Crh cell types contribute to the descending pathways originating in the lateral hypothalamus (LH), central nucleus of the amygdala (CeA), bed nucleus of the stria terminalis (BNST), and insular cortex (IC). Five to 7 days following injections, the animals were euthanized and tissue sections prepared for confocal microscopy. Crh cell types in each forebrain site except IC project to the PBN with the greatest percentage originating in the BNST. For Sst cell types, the largest percentage of double-labeled cells was found in the CeA followed by the BNST. Few retrogradely labeled cells in the LH coexpressed Sst, whereas no double-labeled cells were observed in IC. The present results suggest that Sst and Crh cell types are a substantial component of the descending pathways from the amygdala and/or BNST to the PBN and are positioned to exert neuromodulatory effects on central taste processing.
Collapse
Affiliation(s)
- Ali Magableh
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Robert Lundy
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
25
|
Tokita K, Armstrong WE, St John SJ, Boughter JD. Activation of lateral hypothalamus-projecting parabrachial neurons by intraorally delivered gustatory stimuli. Front Neural Circuits 2014; 8:86. [PMID: 25120438 PMCID: PMC4114292 DOI: 10.3389/fncir.2014.00086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 07/04/2014] [Indexed: 11/13/2022] Open
Abstract
The present study investigated a subpopulation of neurons in the mouse parabrachial nucleus (PbN), a gustatory and visceral relay area in the brainstem, that project to the lateral hypothalamus (LH). We made injections of the retrograde tracer Fluorogold (FG) into LH, resulting in fluorescent labeling of neurons located in different regions of the PbN. Mice were stimulated through an intraoral cannula with one of seven different taste stimuli, and PbN sections were processed for immunohistochemical detection of the immediate early gene c-Fos, which labels activated neurons. LH projection neurons were found in all PbN subnuclei, but in greater concentration in lateral subnuclei, including the dorsal lateral subnucleus (dl). Fos-like immunoreactivity (FLI) was observed in the PbN in a stimulus-dependent pattern, with the greatest differentiation between intraoral stimulation with sweet (0.5 M sucrose) and bitter (0.003 M quinine) compounds. In particular, sweet and umami-tasting stimuli evoked robust FLI in cells in the dl, whereas quinine evoked almost no FLI in cells in this subnucleus. Double-labeled cells were also found in the greatest quantity in the dl. Overall, these results support the hypothesis that the dl contains direct a projection to the LH that is activated preferentially by appetitive compounds; this projection may be mediated by taste and/or postingestive mechanisms.
Collapse
Affiliation(s)
- Kenichi Tokita
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center Memphis, TN, USA
| | - William E Armstrong
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center Memphis, TN, USA
| | | | - John D Boughter
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center Memphis, TN, USA
| |
Collapse
|
26
|
Genetic identification of a neural circuit that suppresses appetite. Nature 2013; 503:111-4. [PMID: 24121436 PMCID: PMC3878302 DOI: 10.1038/nature12596] [Citation(s) in RCA: 422] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/20/2013] [Indexed: 12/03/2022]
Abstract
Appetite suppression occurs following a meal and also during conditions when it is unfavorable to eat, such as during illness or exposure to toxins. A brain region hypothesized to play a role in appetite suppression is the parabrachial nucleus (PBN)1-3, a heterogeneous population of neurons surrounding the superior cerebellar peduncle in the brainstem. The PBN is thought to mediate the suppression of appetite induced by the anorectic hormones amylin and cholecystokinin, as well as lithium chloride and lipopolysaccharide, compounds that mimic the effects of toxic foods and bacterial infections, respectively4-6. Hyperactivity of the PBN is also thought to cause starvation following ablation of orexigenic agouti-related peptide (AgRP) neurons in adult mice1,7. However, the identities of PBN neurons that regulate feeding are unknown, as are the functionally relevant downstream projections. Here we identify calcitonin gene-related peptide (CGRP)-expressing neurons in the outer external lateral subdivision of the PBN that project to the laterocapsular division of the central nucleus of the amygdala (CeAlc) as forming a functionally important circuit for the suppression of appetite. Using genetically-encoded anatomical, optogenetic8, and pharmacogenetic9 tools, we demonstrate that activation of PBelo CGRP neurons projecting to the CeAlc suppresses appetite. In contrast, inhibition of these neurons increases food intake in circumstances when mice do not normally eat and prevents starvation in adult AgRP neuron-ablated mice. Taken together, our data demonstrate that this neural circuit from the PBN to CeAlc mediates appetite suppression in conditions when it is unfavorable to eat. This neural circuit may provide targets for therapeutic intervention to overcome or promote appetite.
Collapse
|