1
|
Gutierrez VA, Martin LE, Torregrossa AM. Salivary proteins rescue within-session suppression and conditioned avoidance in response to an intragastric quinine infusion. Physiol Behav 2024; 274:114430. [PMID: 38070721 PMCID: PMC11171411 DOI: 10.1016/j.physbeh.2023.114430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
A subset of salivary proteins (SPs) upregulates in response to a quinine-containing diet. The presence of these SPs then results in decreased bitter taste responding and taste nerve signaling. Bitter taste receptors in the oral cavity are also found in the stomach and intestines and contribute to behaviors that are influenced by post-oral signaling. It has been previously demonstrated that after several pairings of post-orally infused bitter stimuli and a neutral flavor, animals learn to avoid the flavor that was paired with gastric bitter, this is referred to as conditioned avoidance. Furthermore, animals will decrease licking of a neutral solution within a test session, when licking is paired with an intragastric bitter infusion; this has been described as within-session suppression. We used these paradigms to test the role of SPs in behaviors influenced by post-oral signaling. In both paradigms, the animal is given a test solution directly into the stomach (with or without quinine, and with or without SPs), and the infusions are self-administered by licking to a neutral solution (Kool-Aid). Quinine successfully conditioned a flavor avoidance, but, in a separate trial, we were unable to detect conditioning in the presence of SPs from donor animals. Likewise, quinine was able to suppress licking within the conditioned suppression paradigm, but the effect of the bitter was blocked in the presence of saliva containing SPs. Together, these data suggest that behaviors driven by post-oral signaling can be altered by SPs.
Collapse
Affiliation(s)
- V Ascencio Gutierrez
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - L E Martin
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, 97331, USA
| | - A-M Torregrossa
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, 14260, USA; University at Buffalo Center for Ingestive Behavior Research, Buffalo, New York, 14260, USA.
| |
Collapse
|
2
|
Caremoli F, Huynh J, Lagishetty V, Markovic D, Braun J, Dong TS, Jacobs JP, Sternini C. Microbiota-Dependent Upregulation of Bitter Taste Receptor Subtypes in the Mouse Large Intestine in High-Fat Diet-Induced Obesity. Nutrients 2023; 15:4145. [PMID: 37836428 PMCID: PMC10574285 DOI: 10.3390/nu15194145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Bitter taste receptors (Tas2rs in mice) detect bitterness, a warning signal for toxins and poisons, and are expressed in enteroendocrine cells. We tested the hypothesis that Tas2r138 and Tas2r116 mRNAs are modulated by microbiota alterations induced by a long-term high-fat diet (HFD) and antibiotics (ABX) (ampicillin and neomycin) administered in drinking water. Cecum and colon specimens and luminal contents were collected from C57BL/6 female and male mice for qRT-PCR and microbial luminal 16S sequencing. HFD with/without ABX significantly increased body weight and fat mass at 4, 6, and 8 weeks. Tas2r138 and Tas2r116 mRNAs were significantly increased in mice fed HFD for 8 weeks vs. normal diet, and this increase was prevented by ABX. There was a distinct microbiota separation in each experimental group and significant changes in the composition and diversity of microbiome in mice fed a HFD with/without ABX. Tas2r mRNA expression in HFD was associated with several genera, particularly with Akkermansia, a Gram-negative mucus-resident bacterium. These studies indicate that luminal bacterial composition is affected by sex, diet, and ABX and support a microbial dependent upregulation of Tas2rs in HFD-induced obesity, suggesting an adaptive host response to specific diet-induced dysbiosis.
Collapse
Affiliation(s)
- Filippo Caremoli
- Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (F.C.); (J.H.); (V.L.); (T.S.D.); (J.P.J.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Jennifer Huynh
- Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (F.C.); (J.H.); (V.L.); (T.S.D.); (J.P.J.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Venu Lagishetty
- Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (F.C.); (J.H.); (V.L.); (T.S.D.); (J.P.J.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Daniela Markovic
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Jonathan Braun
- Inflammatory Bowel and Immunobiology Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Tien S. Dong
- Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (F.C.); (J.H.); (V.L.); (T.S.D.); (J.P.J.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Jonathan P. Jacobs
- Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (F.C.); (J.H.); (V.L.); (T.S.D.); (J.P.J.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Catia Sternini
- Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (F.C.); (J.H.); (V.L.); (T.S.D.); (J.P.J.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Martin LE, Gutierrez VA, Torregrossa AM. The role of saliva in taste and food intake. Physiol Behav 2023; 262:114109. [PMID: 36740133 PMCID: PMC10246345 DOI: 10.1016/j.physbeh.2023.114109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Saliva is well-described in oral food processing, but its role in taste responsiveness remains understudied. Taste stimuli must dissolve in saliva to reach their receptor targets. This allows the constituents of saliva the opportunity to interact with taste stimuli and their receptors at the most fundamental level. Yet, despite years of correlational data suggesting a role for salivary proteins in food preference, there were few experimental models to test the role of salivary proteins in taste-driven behaviors. Here we review our experimental contributions to the hypothesis that salivary proteins can alter taste function. We have developed a rodent model to test how diet alters salivary protein expression, and how salivary proteins alter diet acceptance and taste. We have found that salivary protein expression is modified by diet, and these diet-induced proteins can, in turn, increase the acceptance of a bitter diet. The change in acceptance is in part mediated by a change in taste signaling. Critically, we have documented increased detection threshold, decreased taste nerve signaling, and decreased oromotor responding to quinine when animals have increases in a subset of salivary proteins compared to control conditions.
Collapse
Affiliation(s)
- Laura E Martin
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, 97331, USA
| | | | - Ann-Marie Torregrossa
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York, 14216, USA; University at Buffalo Center for Ingestive Behavior Research, Buffalo, New York, 14216, USA.
| |
Collapse
|
4
|
Savulescu-Fiedler I, Gurghean AL, Siliste RN. The complex involvement of the digestive tract in human defense behavior - structural and functional arguments. J Med Life 2022; 15:1081-1089. [PMID: 36415517 PMCID: PMC9635236 DOI: 10.25122/jml-2022-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
The digestive system has an innate monitoring and defense capacity, which allows the recognition and elimination of different dangerous substances. The complex analysis of the intestinal content comprises the cross-interactions between the epithelial cells, the enteroendocrine cells, the neural tissue and the cellular defense mechanisms. The enteric nervous system, also called "the enteric brain" or "the second brain" is the only neuronal network outside the central nervous system capable of autonomous reflex activity. The enteric nervous system activity is mostly independent of the central nervous system, but not in all aspects. In fact, even the enteral reflexes are a consequence of the bidirectional intestine-brain relation. The central nervous and enteric nervous systems are coupled through the sympathetic and parasympathetic branches of the autonomic nervous system. The gastrointestinal functions are regulated due to the interaction between the intrinsic neurons within the gastrointestinal wall and the extrinsic neurons outside the gastrointestinal tract. Here we provide an overview of the important role of the enteric brain in defensive behavior, as well as its structural and functional particularities that make it a special organ.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Internal Medicine and Cardiology Department, Coltea Clinical Hospital, Bucharest, Romania
- Department 1 Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Adriana Luminita Gurghean
- Internal Medicine and Cardiology Department, Coltea Clinical Hospital, Bucharest, Romania
- Department 1 Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Roxana-Nicoleta Siliste
- Internal Medicine and Cardiology Department, Coltea Clinical Hospital, Bucharest, Romania
- Department 1 Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| |
Collapse
|
5
|
Behrens M, Lang T. Extra-Oral Taste Receptors-Function, Disease, and Perspectives. Front Nutr 2022; 9:881177. [PMID: 35445064 PMCID: PMC9014832 DOI: 10.3389/fnut.2022.881177] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Taste perception is crucial for the critical evaluation of food constituents in human and other vertebrates. The five basic taste qualities salty, sour, sweet, umami (in humans mainly the taste of L-glutamic acid) and bitter provide important information on the energy content, the concentration of electrolytes and the presence of potentially harmful components in food items. Detection of the various taste stimuli is facilitated by specialized receptor proteins that are expressed in taste buds distributed on the tongue and the oral cavity. Whereas, salty and sour receptors represent ion channels, the receptors for sweet, umami and bitter belong to the G protein-coupled receptor superfamily. In particular, the G protein-coupled taste receptors have been located in a growing number of tissues outside the oral cavity, where they mediate important processes. This article will provide a brief introduction into the human taste perception, the corresponding receptive molecules and their signal transduction. Then, we will focus on taste receptors in the gastrointestinal tract, which participate in a variety of processes including the regulation of metabolic functions, hunger/satiety regulation as well as in digestion and pathogen defense reactions. These important non-gustatory functions suggest that complex selective forces have contributed to shape taste receptors during evolution.
Collapse
Affiliation(s)
- Maik Behrens
- Leibniz Institute of Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Tatjana Lang
- Leibniz Institute of Food Systems Biology at the Technical University of Munich, Freising, Germany
| |
Collapse
|
6
|
Zhao A, Jeffery EH, Miller MJ. Is Bitterness Only a Taste? The Expanding Area of Health Benefits of Brassica Vegetables and Potential for Bitter Taste Receptors to Support Health Benefits. Nutrients 2022; 14:nu14071434. [PMID: 35406047 PMCID: PMC9002472 DOI: 10.3390/nu14071434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022] Open
Abstract
The list of known health benefits from inclusion of brassica vegetables in the diet is long and growing. Once limited to cancer prevention, a role for brassica in prevention of oxidative stress and anti-inflammation has aided in our understanding that brassica provide far broader benefits. These include prevention and treatment of chronic diseases of aging such as diabetes, neurological deterioration, and heart disease. Although animal and cell culture studies are consistent, clinical studies often show too great a variation to confirm these benefits in humans. In this review, we discuss causes of variation in clinical studies, focusing on the impact of the wide variation across humans in commensal bacterial composition, which potentially result in variations in microbial metabolism of glucosinolates. In addition, as research into host-microbiome interactions develops, a role for bitter-tasting receptors, termed T2Rs, in the gastrointestinal tract and their role in entero-endocrine hormone regulation is developing. Here, we summarize the growing literature on mechanisms of health benefits by brassica-derived isothiocyanates and the potential for extra-oral T2Rs as a novel mechanism that may in part describe the variability in response to brassica among free-living humans, not seen in research animal and cell culture studies.
Collapse
Affiliation(s)
- Anqi Zhao
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA;
| | - Elizabeth H. Jeffery
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA;
| | - Michael J. Miller
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA;
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA;
- Correspondence:
| |
Collapse
|
7
|
Abstract
Bitter taste-sensing type 2 receptors (TAS2Rs or T2Rs), belonging to the subgroup of family A G-protein coupled receptors (GPCRs), are of crucial importance in the perception of bitterness. Although in the first instance, TAS2Rs were considered to be exclusively distributed in the apical microvilli of taste bud cells, numerous studies have detected these sensory receptor proteins in several extra-oral tissues, such as in pancreatic or ovarian tissues, as well as in their corresponding malignancies. Critical points of extra-oral TAS2Rs biology, such as their structure, roles, signaling transduction pathways, extensive mutational polymorphism, and molecular evolution, have been currently broadly studied. The TAS2R cascade, for instance, has been recently considered to be a pivotal modulator of a number of (patho)physiological processes, including adipogenesis or carcinogenesis. The latest advances in taste receptor biology further raise the possibility of utilizing TAS2Rs as a therapeutic target or as an informative index to predict treatment responses in various disorders. Thus, the focus of this review is to provide an update on the expression and molecular basis of TAS2Rs functions in distinct extra-oral tissues in health and disease. We shall also discuss the therapeutic potential of novel TAS2Rs targets, which are appealing due to their ligand selectivity, expression pattern, or pharmacological profiles.
Collapse
Affiliation(s)
- Kamila Tuzim
- Department of Clinical Pathomorphology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Agnieszka Korolczuk
- Department of Clinical Pathomorphology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| |
Collapse
|
8
|
Deoxynivalenol (Vomitoxin)-Induced Anorexia Is Induced by the Release of Intestinal Hormones in Mice. Toxins (Basel) 2021; 13:toxins13080512. [PMID: 34437383 PMCID: PMC8402572 DOI: 10.3390/toxins13080512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 12/25/2022] Open
Abstract
Deoxynivalenol (DON), also known as vomitoxin, is a mycotoxin that can cause antifeeding and vomiting in animals. However, the mechanism of DON inducing anorexia is complicated. Studies have shown that intestinal hormones play a significant part in the anorexia caused by DON. We adopted the “modeling of acute antifeeding in mice” as the basic experimental model, and used two methods of gavage and intraperitoneal injection to explore the effect of intestinal hormones on the antifeedant response induced by DON in mice. We found that 1 and 2.5 mg/kg·bw of DON can acutely induce anorexia and increase the plasma intestinal hormones CCK, PYY, GIP, and GLP-1 in mice within 3 h. Direct injection of exogenous intestinal hormones CCK, PYY, GIP, and GLP-1 can trigger anorexia behavior in mice. Furthermore, the PYY receptor antagonist JNJ-31020028, GLP-1 receptor antagonist Exendin(9-39), CCK receptor antagonist Proglumide, GIP receptor antagonist GIP(3-30)NH2 attenuated both intestinal hormone and DON-induced anorectic responses. These results indicate that intestinal hormones play a critical role in the anorexia response induced by DON.
Collapse
|
9
|
Raka F, Farr S, Kelly J, Stoianov A, Adeli K. Metabolic control via nutrient-sensing mechanisms: role of taste receptors and the gut-brain neuroendocrine axis. Am J Physiol Endocrinol Metab 2019; 317:E559-E572. [PMID: 31310579 DOI: 10.1152/ajpendo.00036.2019] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nutrient sensing plays an important role in ensuring that appropriate digestive or hormonal responses are elicited following the ingestion of fuel substrates. Mechanisms of nutrient sensing in the oral cavity have been fairly well characterized and involve lingual taste receptors. These include heterodimers of G protein-coupled receptors (GPCRs) of the taste receptor type 1 (T1R) family for sensing sweet (T1R2-T1R3) and umami (T1R1-T1R3) stimuli, the T2R family for sensing bitter stimuli, and ion channels for conferring sour and salty tastes. In recent years, several studies have revealed the existence of additional nutrient-sensing mechanisms along the gastrointestinal tract. Glucose sensing is achieved by the T1R2-T1R3 heterodimer on enteroendocrine cells, which plays a role in triggering the secretion of incretin hormones for improved glycemic and lipemic control. Protein hydrolysates are detected by Ca2+-sensing receptor, the T1R1-T1R3 heterodimer, and G protein-coupled receptor 92/93 (GPR92/93), which leads to the release of the gut-derived satiety factor cholecystokinin. Furthermore, several GPCRs have been implicated in fatty acid sensing: GPR40 and GPR120 respond to medium- and long-chain fatty acids, GPR41 and GPR43 to short-chain fatty acids, and GPR119 to endogenous lipid derivatives. Aside from the recognition of fuel substrates, both the oral cavity and the gastrointestinal tract also possess T2R-mediated mechanisms of recognizing nonnutrients such as environmental contaminants, bacterial toxins, and secondary plant metabolites that evoke a bitter taste. These gastrointestinal sensing mechanisms result in the transmission of neuronal signals to the brain through the release of gastrointestinal hormones that act on vagal and enteric afferents to modulate the physiological response to nutrients, particularly satiety and energy homeostasis. Modulating these orally accessible nutrient-sensing pathways using particular foods, dietary supplements, or pharmaceutical compounds may have therapeutic potential for treating obesity and metabolic diseases.
Collapse
Affiliation(s)
- Fitore Raka
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sarah Farr
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jacalyn Kelly
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alexandra Stoianov
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Kure Liu C, Joseph PV, Feldman DE, Kroll DS, Burns JA, Manza P, Volkow ND, Wang GJ. Brain Imaging of Taste Perception in Obesity: a Review. Curr Nutr Rep 2019; 8:108-119. [PMID: 30945140 PMCID: PMC6486899 DOI: 10.1007/s13668-019-0269-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW We summarize neuroimaging findings related to processing of taste (fat, salt, umami, bitter, and sour) in the brain and how they influence hedonic responses and eating behaviors and their role in obesity. RECENT FINDINGS Neuroimaging studies in obese individuals have revealed alterations in reward/motivation, executive control/self-regulation, and limbic/affective circuits that are implicated in food and drug addiction. Psychophysical studies show that sensory properties of food ingredients may be associated with anthropometric and neurocognitive outcomes in obesity. However, few studies have examined the neural correlates of taste and processing of calories and nutrient content in obesity. The literature of neural correlated of bitter, sour, and salty tastes remains sparse in obesity. Most published studies have focused on sweet, followed by fat and umami taste. Studies on calorie processing and its conditioning by preceding taste sensations have started to delineate a dynamic pattern of brain activation associated with appetition. Our expanded understanding of taste processing in the brain from neuroimaging studies is poised to reveal novel prevention and treatment targets to help address overeating and obesity.
Collapse
Affiliation(s)
- Christopher Kure Liu
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
| | - Paule Valery Joseph
- Sensory Science and Metabolism Unit, Biobehavioral Branch, National Institute of Nursing Research, National Institutes of Health, 31 Center Drive, Rm 5B03, Bethesda, MD 20892-2178 USA
| | - Dana E. Feldman
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
| | - Danielle S. Kroll
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
| | - Jamie A. Burns
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
- National Institute on Drug Abuse, National Institutes of Health, 6001 Executive Blvd., Suite 5274, Bethesda, MD 20892-9581 USA
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 USA
| |
Collapse
|
11
|
Harada Y, Koseki J, Sekine H, Fujitsuka N, Kobayashi H. Role of Bitter Taste Receptors in Regulating Gastric Accommodation in Guinea Pigs. J Pharmacol Exp Ther 2019; 369:466-472. [PMID: 30967403 DOI: 10.1124/jpet.118.256008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/04/2019] [Indexed: 01/07/2023] Open
Abstract
Taste stimulants play important roles in triggering digestion and absorption of nutrients and in toxin detection, under the control of the gut-brain axis. Bitter compounds regulate gut hormone secretion and gastrointestinal motility through bitter taste receptors (TAS2Rs) located in the taste buds on the tongue and in the enteroendocrine cells. Gastric accommodation (GA) is an important physiologic function. However, the role of TAS2R agonists in regulating GA remains unclear. To clarify whether GA is influenced by bitter stimulants, we examined the effect of TAS2R agonist denatonium benzoate (DB), administered intraorally and intragastrically, by measuring the consequent intrabag pressure in the proximal stomach of guinea pigs. Effects of the Kampo medicine rikkunshito (RKT) and its bitter components liquiritigenin and naringenin on GA were also examined. Intraoral DB (0.2 nmol/ml) administration enhanced GA. Intragastric DB administration (0.1 and 1 nmol/kg) promoted GA, whereas higher DB doses (30 μmol/kg) inhibited it. Similar changes in GA were observed with intragastric (1000 mg/kg) and intraoral (200 mg/ml) RKT administration. Liquiritigenin and naringenin also promoted GA. These findings suggest that GA is affected by the stimulation of TAS2Rs in the oral cavity or gut in guinea pigs.
Collapse
Affiliation(s)
- Yumi Harada
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan (Y.H., J.K., H.S., N.F.) and Center for Advanced Kampo Medicine and Clinical Research, Juntendo Graduate School of Medicine, Tokyo, Japan (H.K.)
| | - Junichi Koseki
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan (Y.H., J.K., H.S., N.F.) and Center for Advanced Kampo Medicine and Clinical Research, Juntendo Graduate School of Medicine, Tokyo, Japan (H.K.)
| | - Hitomi Sekine
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan (Y.H., J.K., H.S., N.F.) and Center for Advanced Kampo Medicine and Clinical Research, Juntendo Graduate School of Medicine, Tokyo, Japan (H.K.)
| | - Naoki Fujitsuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan (Y.H., J.K., H.S., N.F.) and Center for Advanced Kampo Medicine and Clinical Research, Juntendo Graduate School of Medicine, Tokyo, Japan (H.K.)
| | - Hiroyuki Kobayashi
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan (Y.H., J.K., H.S., N.F.) and Center for Advanced Kampo Medicine and Clinical Research, Juntendo Graduate School of Medicine, Tokyo, Japan (H.K.)
| |
Collapse
|
12
|
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: 44] [Impact Index Per Article: 8.8] [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
|
13
|
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
|
14
|
Peptide Tyrosine Tyrosine 3-36 Reduces Meal Size and Activates the Enteric Neurons in Male Sprague-Dawley Rats. Dig Dis Sci 2017; 62:3350-3358. [PMID: 29030744 DOI: 10.1007/s10620-017-4788-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/04/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Peptide tyrosine tyrosine 3-36 (peptide YY 3-36 or PYY 3-36) reduces food intake by unknown site(s). AIM To test the hypothesis that the gastrointestinal tract contains sites of action regulating meal size (MS) and intermeal interval (IMI) length by PYY 3-36. METHODS Peptide YY 3-36 (0, 1, 5, 10 and 20 nmol/kg) was injected in the aorta, the artery that supplies the gastrointestinal tract, prior to the onset of the dark cycle in free feeding male Sprague-Dawley rats and food intake was measured. Then, PYY 3-36 (25 nmol/kg) was injected intraperitoneally in these rats and Fos-like immunoreactivity (Fos-LI, a marker for neuronal activation) was quantified in the small intestinal enteric neurons, both myenteric and submucosal, and the dorsal vagal complex (DVC) of the hindbrain. RESULTS PYY 3-36 reduced first MS, decreased IMI length, shortened duration of first meal and increased Fos-LI in enteric and DVC neurons. However, PYY 3-36 failed to change the size of the second meal, satiety ratio, latency to first meal, number of meals and 24 h intake relative to saline control. CONCLUSION The gastrointestinal tract may contain sites of action regulating MS reduction by PYY 3-36.
Collapse
|
15
|
Martin LE, Nikonova LV, Kay K, Paedae AB, Contreras RJ, Torregrossa AM. Salivary proteins alter taste-guided behaviors and taste nerve signaling in rat. Physiol Behav 2017; 184:150-161. [PMID: 29162505 DOI: 10.1016/j.physbeh.2017.11.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/31/2017] [Accepted: 11/16/2017] [Indexed: 11/24/2022]
Abstract
Taste stimuli are normally dissolved in saliva prior to interacting with their respective receptor targets. There are hundreds of proteins in saliva, and it has been hypothesized that these proteins could interact with either taste stimuli or taste receptors to alter taste signaling and diet acceptance. However, the impact of these proteins on feeding has been relatively unexplored using rodent models. We have developed a novel technique for saliva collection that allows us to link salivary protein expression with feeding behavior. First, we monitored the microstructure of rats' feeding patterns on a 0.375% quinine diet (Q-diet) while tracking changes in salivary protein expression. We found 5 protein bands were upregulated by diet exposure to Q-diet and upregulation of a subset of these bands were statistically related to increased diet acceptance, including changes in behavioral measures that are thought to represent both orosensory and postingestive signaling. In a second experiment, we measured the licking to a range of quinine solutions (0.01-1.0mM) before and after the animals were exposed to a tannic acid diet that altered salivary protein expression. Rats found the quinine solutions less aversive after salivary protein altering diets. In a third experiment we recorded the response of the chorda tympani (CT) nerve while delivering quinine solutions (0.3-30mM) to the front of the tongue dissolved in either "donor saliva" containing salivary proteins or donor saliva which has had the salivary proteins removed. Donor saliva was collected from a separate group of animals using isoproterenol and pilocarpine. The samples containing salivary proteins resulted in lower nerve responses than those without salivary proteins. Together these data suggest that salivary proteins are capable of altering taste-guided behaviors and taste nerve signaling.
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 Kay
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY 14216, USA
| | - Andrew B Paedae
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Robert J Contreras
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Ann-Marie Torregrossa
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY 14216, USA.
| |
Collapse
|
16
|
Serrano J, Casanova-Martí À, Blay MT, Terra X, Pinent M, Ardévol A. Strategy for limiting food intake using food components aimed at multiple targets in the gastrointestinal tract. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
17
|
Ekstrand B, Young JF, Rasmussen MK. Taste receptors in the gut - A new target for health promoting properties in diet. Food Res Int 2017; 100:1-8. [PMID: 28888429 DOI: 10.1016/j.foodres.2017.08.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/07/2017] [Accepted: 08/12/2017] [Indexed: 12/17/2022]
Abstract
In this review we describe a new target for food functionality, the taste receptors in the gastrointestinal tract. These receptors are involved in an intricate signalling network for monitoring of taste and nutrient intake, homeostasis and energy metabolism, and they are also an early warning system for toxic substances in our diet. Especially the receptors for bitter taste provide a new possibility to activate a number of health related signalling pathways, already at low concentrations of the active substance, without requiring uptake into the body and transport via the circulation. When ligands bind to these receptors, signalling is induced either via peptide hormones into the circulation to other organs in the body, or via nerve fibers directly to the brain.
Collapse
Affiliation(s)
- Bo Ekstrand
- Chalmers University of Technology, Department of Biology and Biological Engineering, Food and Nutrition Science, SE-412 96 Gothenburg, Sweden
| | | | | |
Collapse
|
18
|
Deloose E, Janssen P, Corsetti M, Biesiekierski J, Masuy I, Rotondo A, Van Oudenhove L, Depoortere I, Tack J. Intragastric infusion of denatonium benzoate attenuates interdigestive gastric motility and hunger scores in healthy female volunteers. Am J Clin Nutr 2017; 105:580-588. [PMID: 28148502 DOI: 10.3945/ajcn.116.138297] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/19/2016] [Indexed: 11/14/2022] Open
Abstract
Background: Denatonium benzoate (DB) has been shown to influence ongoing ingestive behavior and gut peptide secretion.Objective: We studied how the intragastric administration of DB affects interdigestive motility, motilin and ghrelin plasma concentrations, hunger and satiety ratings, and food intake in healthy volunteers.Design: Lingual bitter taste sensitivity was tested with the use of 6 concentrations of DB in 65 subjects. A placebo or 1 μmol DB/kg was given intragastrically to assess its effect on fasting gastrointestinal motility and hunger ratings, motilin and ghrelin plasma concentrations, satiety, and caloric intake.Results: Women (n = 39) were more sensitive toward a lingual bitter stimulus (P = 0.005) than men (n = 26). In women (n = 10), intragastric DB switched the origin of phase III contractions from the stomach to the duodenum (P = 0.001) and decreased hunger ratings (P = 0.04). These effects were not observed in men (n = 10). In women (n = 12), motilin (P = 0.04) plasma concentrations decreased after intragastric DB administration, whereas total and octanoylated ghrelin were not affected. The intragastric administration of DB decreased hunger (P = 0.008) and increased satiety ratings (P = 0.01) after a meal (500 kcal) in 13 women without affecting gastric emptying in 6 women. Caloric intake tended to decrease after DB administration compared with the placebo (mean ± SEM: 720 ± 58 compared with 796 ± 45 kcal; P = 0.08) in 20 women.Conclusions: Intragastric DB administration decreases both antral motility and hunger ratings during the fasting state, possibly because of a decrease in motilin release. Moreover, DB decreases hunger and increases satiety ratings after a meal and shows potential for decreasing caloric intake. This trial was registered at clinicaltrials.gov as NCT02759926.
Collapse
Affiliation(s)
- Eveline Deloose
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Pieter Janssen
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Maura Corsetti
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and.,National Institute for Health Research, Nottingham Digestive Diseases Biomedical Research Unit, Nottingham University Hospitals National Health Service Trust, University of Nottingham, Nottingham, United Kingdom
| | - Jessica Biesiekierski
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Imke Masuy
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Alessandra Rotondo
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Lukas Van Oudenhove
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| |
Collapse
|
19
|
Manley KJ. Will mouth wash solutions of water, salt, sodiumbicarbonate or citric acid improve upper gastrointestinal symptoms in chronic kidney disease. Nephrology (Carlton) 2017; 22:213-219. [DOI: 10.1111/nep.12753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/07/2016] [Accepted: 02/16/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Karen Joy Manley
- Departments of Nutrition and Dietetics; Austin Health; Heidelberg Victoria Australia
| |
Collapse
|
20
|
Pham H, Hui H, Morvaridi S, Cai J, Zhang S, Tan J, Wu V, Levin N, Knudsen B, Goddard WA, Pandol SJ, Abrol R. A bitter pill for type 2 diabetes? The activation of bitter taste receptor TAS2R38 can stimulate GLP-1 release from enteroendocrine L-cells. Biochem Biophys Res Commun 2016; 475:295-300. [PMID: 27208775 DOI: 10.1016/j.bbrc.2016.04.149] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 04/28/2016] [Indexed: 02/09/2023]
Abstract
The bitter taste receptor TAS2R38 is a G protein coupled receptor (GPCR) that has been found in many extra-oral locations like the gastrointestinal (GI) system, respiratory system, and brain, though its function at these locations is only beginning to be understood. To probe the receptor's potential metabolic role, immunohistochemistry of human ileum tissues was performed, which showed that the receptor was co-localized with glucagon-like peptide 1 (GLP-1) in L-cells. In a previous study, we had modeled the structure of this receptor for its many taste-variant haplotypes (Tan et al. 2011), including the taster haplotype PAV. The structure of this haplotype was then used in a virtual ligand screening pipeline using a collection of ∼2.5 million purchasable molecules from the ZINC database. Three compounds (Z7, Z3, Z1) were purchased from the top hits and tested along with PTU (known TAS2R38 agonist) in in vitro and in vivo assays. The dose-response study of the effect of PTU and Z7 on GLP-1 release using wild-type and TAS2R38 knockout HuTu-80 cells showed that the receptor TAS2R38 plays a major role in GLP-1 release due to these molecules. In vivo studies of PTU and the three compounds showed that they each increase GLP-1 release. PTU was also chemical linked to cellulose to slow its absorption and when tested in vivo, it showed an enhanced and prolonged GLP-1 release. These results suggest that the GI lumen location of TAS2R38 on the L-cell makes it a relatively safe drug target as systemic absorption is not needed for a TAS2R38 agonist drug to effect GLP-1 release.
Collapse
Affiliation(s)
- Hung Pham
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hongxiang Hui
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA; International Center for Metabolic Diseases, Southern Medical University, Guangzhou, China
| | - Susan Morvaridi
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jiena Cai
- International Center for Metabolic Diseases, Southern Medical University, Guangzhou, China
| | - Sanqi Zhang
- Department of Medicinal Chemistry, Xi'an Jiaotong University, 710061, China
| | - Jun Tan
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA, USA; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400030, China
| | - Vincent Wu
- Veterans Affairs Greater Los Angeles Healthcare System, University of California, Los Angeles, CA, USA
| | | | - Beatrice Knudsen
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - William A Goddard
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA, USA
| | - Stephen J Pandol
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, University of California, Los Angeles, CA, USA; GIRx Metabolics Inc., Los Angeles, CA, USA
| | - Ravinder Abrol
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA, USA; GIRx Metabolics Inc., Los Angeles, CA, USA.
| |
Collapse
|
21
|
Latorre R, Sternini C, De Giorgio R, Greenwood-Van Meerveld B. Enteroendocrine cells: a review of their role in brain-gut communication. Neurogastroenterol Motil 2016; 28:620-30. [PMID: 26691223 PMCID: PMC4842178 DOI: 10.1111/nmo.12754] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/17/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Specialized endoderm-derived epithelial cells, that is, enteroendocrine cells (EECs), are widely distributed throughout the gastrointestinal (GI) tract. Enteroendocrine cells form the largest endocrine organ in the body and play a key role in the control of GI secretion and motility, the regulation of food intake, postprandial glucose levels and metabolism. EECs sense luminal content and release signaling molecules that can enter the circulation to act as classic hormones on distant targets, act locally on neighboring cells and on distinct neuronal pathways including enteric and extrinsic neurons. Recent studies have shed light on EEC sensory transmission by showing direct connections between EECs and the nervous system via axon-like processes that form a well-defined neuroepithelial circuits through which EECs can directly communicate with the neurons innervating the GI tract to initiate appropriate functional responses. PURPOSE This review will highlight the role played by the EECs in the complex and integrated sensory information responses, and discuss the new findings regarding EECs in the brain-gut axis bidirectional communication.
Collapse
Affiliation(s)
- R Latorre
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - C Sternini
- CURE Digestive Diseases Research Center, Division of Digestive Diseases and Departments of Medicine and Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - R De Giorgio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - B Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Physiology, Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
22
|
Avau B, Depoortere I. The bitter truth about bitter taste receptors: beyond sensing bitter in the oral cavity. Acta Physiol (Oxf) 2016; 216:407-20. [PMID: 26493384 DOI: 10.1111/apha.12621] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/28/2015] [Accepted: 10/14/2015] [Indexed: 11/26/2022]
Abstract
The bitter taste receptor (TAS2R)-family of G-protein-coupled receptors has been identified on the tongue as detectors of bitter taste over a decade ago. In the last few years, they have been discovered in an ever growing number of extra-oral tissues, including the airways, the gut, the brain and even the testis. In tissues that contact the exterior, protective functions for TAS2Rs have been proposed, in analogy to their function on the tongue as toxicity detector. However, TAS2Rs have also been found in internal organs, suggesting other roles for these receptors, perhaps involving as yet unidentified endogenous ligands. The current review gives an overview of the different proposed functions for TAS2Rs in tissues other than the oral cavity; from appetite regulation to the treatment of asthma, regulation of gastrointestinal motility and control of airway innate immunity.
Collapse
Affiliation(s)
- B. Avau
- Translational Research Center for Gastrointestinal Disorders (TARGID); Gut Peptide Research Lab; University of Leuven; Leuven Belgium
| | - I. Depoortere
- Translational Research Center for Gastrointestinal Disorders (TARGID); Gut Peptide Research Lab; University of Leuven; Leuven Belgium
| |
Collapse
|
23
|
Regulation of α-Transducin and α-Gustducin Expression by a High Protein Diet in the Pig Gastrointestinal Tract. PLoS One 2016; 11:e0148954. [PMID: 26871573 PMCID: PMC4752509 DOI: 10.1371/journal.pone.0148954] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/25/2016] [Indexed: 11/29/2022] Open
Abstract
Background The expression of taste receptors (TASRs) and their signalling molecules in the gastrointestinal (GI) epithelial cells, including enteroendocrine cells (EECs), suggests they participate in chemosensing mechanisms influencing GI physiology via the release of endocrine messengers. TASRs mediate gustatory signalling by interacting with different transducers, including α-gustducin (Gαgust) and α-transducin (Gαtran) G protein subunits. This study tested whether Gαtran and Gαgust immunoreactive (-IR) cells are affected by a short-term (3 days) and long-term (30 days) high protein (Hp) diet in the pig GI tract. Result In the stomach, Gαgust and Gαtran-IR cells contained serotonin (5-HT) and ghrelin (GHR), while in the small and large intestine, Gαgust and Gαtran-IR colocalized with 5-HT-, cholecystokinin (CCK)- and peptide YY (PYY)-IR. There was a significant increase in the density of Gαtran-IR cells in the pyloric mucosa in both short- and long-term Hp diet groups (Hp3 and Hp30) vs. the control group (Ctr) (P<0.05), while the increase of Gαgust-IR cells in the pyloric mucosa was significant in Hp30 group vs. Ctr and vs. Hp3 (P<0.05); these cells included Gαtran / 5HT-IR and Gαtran / GHR-IR cells (P<0.05 and P<0.001 vs. Ctr, respectively) as well as Gαgust /5-HT-IR or Gαgust / GHR-IR cells (P<0.05 and P<0.01 vs. Ctr, respectively). In the small intestine, we recorded a significant increase in Gαtran-IR cells in the duodenal crypts and a significant increase of Gαgust-IR cells in the jejunal crypts in Hp3 group compared to HP30 (P<0.05). With regard to the number of Gαtran-Gαgust IR cells colocalized with CCK or 5-HT, there was only a significant increase of Gαtran / CCK-IR cells in Hp3 group compared to Ctr (P = 0.01). Conclusion This study showed an upregulation of selected subpopulations of Gαgust / Gαtran-IR cells in distinct regions of the pig GI tract by short- and long-term Hp diet lending support to TASR-mediated effects in metabolic homeostasis and satiety mechanisms.
Collapse
|
24
|
Latorre R, Huynh J, Mazzoni M, Gupta A, Bonora E, Clavenzani P, Chang L, Mayer EA, De Giorgio R, Sternini C. Expression of the Bitter Taste Receptor, T2R38, in Enteroendocrine Cells of the Colonic Mucosa of Overweight/Obese vs. Lean Subjects. PLoS One 2016; 11:e0147468. [PMID: 26866366 PMCID: PMC4750998 DOI: 10.1371/journal.pone.0147468] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/03/2016] [Indexed: 12/12/2022] Open
Abstract
Bitter taste receptors (T2Rs) are expressed in the mammalian gastrointestinal mucosa. In the mouse colon, T2R138 is localized to enteroendocrine cells and is upregulated by long-term high fat diet that induces obesity. The aims of this study were to test whether T2R38 expression is altered in overweight/obese (OW/OB) compared to normal weight (NW) subjects and characterize the cell types expressing T2R38, the human counterpart of mouse T2R138, in human colon. Colonic mucosal biopsies were obtained during colonoscopy from 35 healthy subjects (20 OW/OB and 15 NW) and processed for quantitative RT-PCR and immunohistochemistry using antibodies to T2R38, chromogranin A (CgA), glucagon like peptide-1 (GLP-1), cholecystokinin (CCK), or peptide YY (PYY). T2R38 mRNA levels in the colonic mucosa of OW/OB were increased (> 2 fold) compared to NW subjects but did not reach statistical significance (P = 0.06). However, the number of T2R38 immunoreactive (IR) cells was significantly increased in OW/OB vs. NW subjects (P = 0.01) and was significantly correlated with BMI values (r = 0.7557; P = 0.001). In both OW/OB and NW individuals, all T2R38-IR cells contained CgA-IR supporting they are enteroendocrine. In both groups, T2R38-IR colocalized with CCK-, GLP1- or PYY-IR. The overall CgA-IR cell population was comparable in OW/OB and NW individuals. This study shows that T2R38 is expressed in distinct populations of enteroendocrine cells in the human colonic mucosa and supports T2R38 upregulation in OW/OB subjects. T2R38 might mediate host functional responses to increased energy balance and intraluminal changes occurring in obesity, which could involve peptide release from enteroendocrine cells.
Collapse
Affiliation(s)
- Rocco Latorre
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Huynh
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maurizio Mazzoni
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - Arpana Gupta
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Elena Bonora
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Clavenzani
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - Lin Chang
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Emeran A. Mayer
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Roberto De Giorgio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Catia Sternini
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
25
|
|
26
|
Avau B, Rotondo A, Thijs T, Andrews CN, Janssen P, Tack J, Depoortere I. Targeting extra-oral bitter taste receptors modulates gastrointestinal motility with effects on satiation. Sci Rep 2015; 5:15985. [PMID: 26541810 PMCID: PMC4635351 DOI: 10.1038/srep15985] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 10/06/2015] [Indexed: 01/01/2023] Open
Abstract
Bitter taste receptors (TAS2Rs) are present in extra-oral tissues, including gut endocrine cells. This study explored the presence and mechanism of action of TAS2R agonists on gut smooth muscle in vitro and investigated functional effects of intra-gastric administration of TAS2R agonists on gastric motility and satiation. TAS2Rs and taste signalling elements were expressed in smooth muscle tissue along the mouse gut and in human gastric smooth muscle cells (hGSMC). Bitter tastants induced concentration and region-dependent contractility changes in mouse intestinal muscle strips. Contractions induced by denatonium benzoate (DB) in gastric fundus were mediated via increases in intracellular Ca2+ release and extracellular Ca2+-influx, partially masked by a hyperpolarizing K+-efflux. Intra-gastric administration of DB in mice induced a TAS2R-dependent delay in gastric emptying. In hGSMC, bitter compounds evoked Ca2+-rises and increased ERK-phosphorylation. Healthy volunteers showed an impaired fundic relaxation in response to nutrient infusion and a decreased nutrient volume tolerance and increased satiation during an oral nutrient challenge test after intra-gastric DB administration. These findings suggest a potential role for intestinal TAS2Rs as therapeutic targets to alter gastrointestinal motility and hence to interfere with hunger signalling.
Collapse
Affiliation(s)
- Bert Avau
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Alessandra Rotondo
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Theo Thijs
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Christopher N Andrews
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Pieter Janssen
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Taste genetics and gastrointestinal symptoms experienced in chronic kidney disease. Eur J Clin Nutr 2015; 69:781-5. [PMID: 26014268 DOI: 10.1038/ejcn.2015.80] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 04/09/2015] [Accepted: 04/11/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND/OBJECTIVES It is unknown what causes uraemic symptoms in renal disease. Chronic kidney disease (CKD) patients are known to have increased levels of urea, sodium, potassium and phosphate in their saliva compared with those without renal disease. The present cross-sectional study investigated associations between known genetic traits of taste and self-reported upper gastrointestinal (GI) symptoms experienced in CKD patients with the changes in saliva composition found in renal failure. SUBJECTS/METHODS Fifty-six CKD patients (35 males, 21 females, age 67±14 years), with stages 4 and 5 renal failure, selected from a tertiary hospital renal outpatient clinic participated in this study. Subjects answered a questionnaire to assess upper GI symptoms and tested for the genetic taste recognition thresholds of thiourea, phenylthiocarbamide and sodium benzoate. Saliva samples were collected to determine biochemical composition. Possible associations between genetic taste variations, saliva composition and upper GI symptoms were investigated. RESULTS Of the 56 patients enroled, 29 (52%) reported major upper GI uraemic symptoms, whereas 27 (48%) had no symptoms or only minor complaints of dry mouth. There was a strong association between the symptomatic burden a patient experienced and the genetic ability to taste thiourea (P<0.0003). Uraemic symptoms of taste changes (P<0.004) and nausea (P<0.002) were found to be related to a patient's genetic ability to taste thiourea. CONCLUSIONS This study provides evidence that the genetic ability to taste thiourea as bitter, in combination with the increase in active compounds found in CKD patient's saliva, impacts on the uraemic upper GI symptoms experienced.
Collapse
|
28
|
Dysregulation of energy balance by trichothecene mycotoxins: Mechanisms and prospects. Neurotoxicology 2015; 49:15-27. [PMID: 25956358 DOI: 10.1016/j.neuro.2015.04.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/14/2015] [Accepted: 04/26/2015] [Indexed: 11/23/2022]
Abstract
Trichothecenes are toxic metabolites produced by fungi that constitute a worldwide hazard for agricultural production and both animal and human health. More than 40 countries have introduced regulations or guidelines for food and feed contamination levels of the most prevalent trichothecene, deoxynivalenol (DON), on the basis of its ability to cause growth suppression. With the development of analytical tools, evaluation of food contamination and exposure revealed that a significant proportion of the human population is chronically exposed to DON doses exceeding the provisional maximum tolerable daily dose. Accordingly, a better understanding of trichothecene impact on health is needed. Upon exposure to low or moderate doses, DON and other trichothecenes induce anorexia, vomiting and reduced weight gain. Several recent studies have addressed the mechanisms by which trichothecenes induce these symptoms and revealed a multifaceted action targeting gut, liver and brain and causing dysregulation in neuroendocrine signaling, immune responses, growth hormone axis, and central neurocircuitries involved in energy homeostasis. Newly identified trichothecene toxicosis biomarkers are just beginning to be exploited and already open up new questions on the potential harmful effects of chronic exposure to DON at apparently asymptomatic very low levels. This review summarizes our current understanding of the effects of DON and other trichothecenes on food intake and weight growth.
Collapse
|
29
|
Abstract
Levels of obesity have reached epidemic proportions on a global scale, which has led to considerable increases in health problems and increased risk of several diseases, including cardiovascular and pulmonary diseases, cancer and diabetes mellitus. People with obesity consume more food than is needed to maintain an ideal body weight, despite the discrimination that accompanies being overweight and the wealth of available information that overconsumption is detrimental to health. The relationship between energy expenditure and energy intake throughout an individual's lifetime is far more complicated than previously thought. An improved comprehension of the relationships between taste, palatability, taste receptors and hedonic responses to food might lead to increased understanding of the biological underpinnings of energy acquisition, as well as why humans sometimes eat more than is needed and more than we know is healthy. This Review discusses the role of taste receptors in the tongue, gut, pancreas and brain and their hormonal involvement in taste perception, as well as the relationship between taste perception, overeating and the development of obesity.
Collapse
Affiliation(s)
- Sara Santa-Cruz Calvo
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Biomedical Research Center, Room 09B133, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Biomedical Research Center, Room 09B133, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, USA
| |
Collapse
|
30
|
Wen X, Zhou J, Zhang D, Li J, Wang Q, Feng N, Zhu H, Song Y, Li H, Bai C. Denatonium inhibits growth and induces apoptosis of airway epithelial cells through mitochondrial signaling pathways. Respir Res 2015; 16:13. [PMID: 25652218 PMCID: PMC4326484 DOI: 10.1186/s12931-015-0183-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/27/2015] [Indexed: 01/11/2023] Open
Abstract
Background Denatonium, a widely used bitter agonist, activates bitter taste receptors on many cell types and plays important roles in chemical release, ciliary beating and smooth muscle relaxation through intracellular Ca2+-dependent pathways. However, the effects of denatonium on the proliferation of airway epithelial cells and on the integrity of cellular components such as mitochondria have not been studied. In this study, we hypothesize that denatonium might induce airway epithelial cell injury by damaging mitochondria. Methods Bright-field microscopy, cell counting kit-8 (CCK-8) assay and flow cytometry analysis were used to examine cellular morphology, proliferation and cell cycle, respectively. Transmission electron microscopy (TEM) was used to examine mitochondrial integrity. JC-1 dye and western blotting techniques were used to measure mitochondrial membrane potential and protein expression, respectively. Results For airway epithelial cells, we observed that denatonium significantly effects cellular morphology, decreases cell proliferation and reduces the number of cells in S phase in a dose-dependent manner. TEM analysis demonstrated that denatonium causes large amplitude swelling of mitochondria, which was confirmed by the loss of mitochondrial membrane potential, the down-regulation of Bcl-2 protein and the subsequent enhancement of the mitochondrial release of cytochrome c and Smac/DIABLO after denatonium treatment. Conclusions In this study, we demonstrated for the first time that denatonium damages mitochondria and thus induces apoptosis in airway epithelial cells. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0183-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoxing Wen
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Jian Zhou
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Dan Zhang
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Jing Li
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Qin Wang
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Nana Feng
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Haixing Zhu
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Yuanlin Song
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Huayin Li
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Chunxue Bai
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
31
|
Vegezzi G, Anselmi L, Huynh J, Barocelli E, Rozengurt E, Raybould H, Sternini C. Diet-induced regulation of bitter taste receptor subtypes in the mouse gastrointestinal tract. PLoS One 2014; 9:e107732. [PMID: 25238152 PMCID: PMC4169553 DOI: 10.1371/journal.pone.0107732] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/18/2014] [Indexed: 12/14/2022] Open
Abstract
Bitter taste receptors and signaling molecules, which detect bitter taste in the mouth, are expressed in the gut mucosa. In this study, we tested whether two distinct bitter taste receptors, the bitter taste receptor 138 (T2R138), selectively activated by isothiocyanates, and the broadly tuned bitter taste receptor 108 (T2R108) are regulated by luminal content. Quantitative RT-PCR analysis showed that T2R138 transcript is more abundant in the colon than the small intestine and lowest in the stomach, whereas T2R108 mRNA is more abundant in the stomach compared to the intestine. Both transcripts in the stomach were markedly reduced by fasting and restored to normal levels after 4 hours re-feeding. A cholesterol-lowering diet, mimicking a diet naturally low in cholesterol and rich in bitter substances, increased T2R138 transcript, but not T2R108, in duodenum and jejunum, and not in ileum and colon. Long-term ingestion of high-fat diet increased T2R138 RNA, but not T2R108, in the colon. Similarly, α-gustducin, a bitter taste receptor signaling molecule, was reduced by fasting in the stomach and increased by lowering cholesterol in the small intestine and by high-fat diet in the colon. These data show that both short and long term changes in the luminal contents alter expression of bitter taste receptors and associated signaling molecules in the mucosa, supporting the proposed role of bitter taste receptors in luminal chemosensing in the gastrointestinal tract. Bitter taste receptors might serve as regulatory and defensive mechanism to control gut function and food intake and protect the body from the luminal environment.
Collapse
Affiliation(s)
- Gaia Vegezzi
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Laura Anselmi
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Huynh
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | | | - Enrique Rozengurt
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Veterans Administration, Greater Los Angeles Health system, Los Angeles, California, United States of America
| | - Helen Raybould
- Department of Anatomy, Physiology, & Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Catia Sternini
- CURE Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Veterans Administration, Greater Los Angeles Health system, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Jung HY, Kim W, Yoo DY, Nam SM, Kim JW, Choi JH, Yoon YS, Kim HY, Hwang IK. Intragastric gavage with denatonium benzoate acutely induces neuronal activation in the solitary tract nucleus via the vagal afferent pathway. J Vet Sci 2014; 15:459-64. [PMID: 24962407 PMCID: PMC4269587 DOI: 10.4142/jvs.2014.15.4.459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 04/27/2014] [Indexed: 11/25/2022] Open
Abstract
Natural toxic substances have a bitter taste and their ingestion sends signals to the brain leading to aversive oral sensations. In the present study, we investigated chronological changes in c-Fos immunoreactivity in the nucleus tractus solitarius (NTS) to study the bitter taste reaction time of neurons in the NTS. Equal volumes (0.5 mL) of denatonium benzoate (DB), a bitter tastant, or its vehicle (distilled water) were administered to rats intragastrically. The rats were sacrificed at 0, 0.5, 1, 2, 4, 8, or 16 h after treatment. In the vehicle-treated group, the number of c-Fos-positive nuclei started to increase 0.5 h after treatment and peaked 2 h after gavage. In contrast, the number of c-Fos-positive nuclei in the DB-treated group significantly increased 1 h after gavage. Thereafter, the number of c-Fos immunoreactive nuclei decreased over time. The number of c-Fos immunoreactive nuclei in the NTS was also increased in a dose-dependent manner 1 h after gavage. Subdiaphragmatic vagotomy significantly decreased DB-induced neuronal activation in the NTS. These results suggest that intragastric DB increases neuronal c-Fos expression in the NTS 1 h after gavage and this effect is mediated by vagal afferent fibers.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wu W, Zhou HR, He K, Pan X, Sugita-Konishi Y, Watanabe M, Zhang H, Pestka JJ. Role of cholecystokinin in anorexia induction following oral exposure to the 8-ketotrichothecenes deoxynivalenol, 15-acetyldeoxynivalenol, 3-acetyldeoxynivalenol, fusarenon X, and nivalenol. Toxicol Sci 2014; 138:278-89. [PMID: 24385417 DOI: 10.1093/toxsci/kft335] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Cereal grain contamination by trichothecene mycotoxins is known to negatively impact human and animal health with adverse effects on food intake and growth being of particular concern. The head blight fungus Fusarium graminearum elaborates five closely related 8-ketotrichothecene congeners: (1) deoxynivalenol (DON), (2) 3-acetyldeoxynivalenol (3-ADON), (3) 15-acetyldeoxynivalenol (15-ADON), (4) fusarenon X (FX), and (5) nivalenol (NIV). While anorexia induction in mice exposed intraperitoneally to DON has been linked to plasma elevation of the satiety hormones cholecystokinin (CCK) and peptide YY₃₋₃₆ (PYY₃₋₃₆), the effects of oral gavage of DON or of other 8-keotrichothecenes on release of these gut peptides have not been established. The purpose of this study was to (1) compare the anorectic responses to the aforementioned 8-ketotrichothecenes following oral gavage at a common dose (2.5 mg/kg bw) and (2) relate these effects to changes plasma CCK and PYY₃₋₃₆ concentrations. Elevation of plasma CCK markedly corresponded to anorexia induction by DON and all other 8-ketotrichothecenes tested. Furthermore, the CCK1 receptor antagonist SR 27897 and the CCK2 receptor antagonist L-365,260 dose-dependently attenuated both CCK- and DON-induced anorexia, which was consistent with this gut satiety hormone being an important mediator of 8-ketotrichothecene-induced food refusal. In contrast to CCK, PYY₃₋₃₆ was moderately elevated by oral gavage with DON and NIV but not by 3-ADON, 15-ADON, or FX. Taken together, the results suggest that CCK plays a major role in anorexia induction following oral exposure to 8-ketotrichothecenes, whereas PYY₃₋₃₆ might play a lesser, congener-dependent role in this response.
Collapse
Affiliation(s)
- Wenda Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Taste Receptor Gene Expression Outside the Gustatory System. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_79] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
McMullen MK. Evidence does not support the suggestion that gustatory exposure to bitter tastants reduces cardiac activity. JOURNAL OF ETHNOPHARMACOLOGY 2013; 149:838-839. [PMID: 23933498 DOI: 10.1016/j.jep.2013.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/02/2013] [Accepted: 08/02/2013] [Indexed: 06/02/2023]
|
36
|
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
|
37
|
Janssen S, Depoortere I. Nutrient sensing in the gut: new roads to therapeutics? Trends Endocrinol Metab 2013; 24:92-100. [PMID: 23266105 DOI: 10.1016/j.tem.2012.11.006] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/20/2012] [Accepted: 11/22/2012] [Indexed: 01/12/2023]
Abstract
The release of gut hormones involved in the control of food intake is dependent on the acute nutritional status of the body, suggesting that chemosensory mechanisms are involved in the control of their release. G protein-coupled taste receptors similar to those in the lingual system, that respond to sweet, bitter, umami, and fatty acids, are expressed in endocrine cells within the gut mucosa, and coordinate, together with other chemosensory signaling elements, the release of hormones that regulate energy and glucose homeostasis. In health, these nutrient sensors are likely to function as inhibitors to excessive nutrient exposure, and their malfunction may be responsible for a variety of metabolic dysfunctions associated with obesity; they may thus be considered as new therapeutic targets.
Collapse
Affiliation(s)
- Sara Janssen
- Translational Research Center for Gastrointestinal Disorders, Gut Peptide Research Lab, Catholic University of Leuven, 3000 Leuven, Belgium
| | | |
Collapse
|
38
|
Golden GJ, Hussey AM, Kimball BA. Do gastrointestinal taste receptors contribute to associative learning and foraging behavior? J Anim Sci 2012; 90:4297-307. [PMID: 22829611 DOI: 10.2527/jas.2012-5089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Foraging behavior is an expression of learning, context, and experience arising from integration of sensory information obtained during feeding with postingestive consequences of food ingestion. Although it has been well established that gustatory and olfactory systems of the mouth and nose provide sensory information to the consumer (in the form of flavor), sweet and bitter taste receptors have recently been identified in the intestinal tract of humans and rodents. It remains possible that sensory information generated in the gut could contribute to the learning process. Thus, a series of experiments was conducted to determine if classical associative learning occurs when the conditional stimulus circumvents oronasal presentation via direct delivery to the gut or peritoneal cavity. Mice receiving an intragastric infusion of 5 mM sodium saccharin immediately followed by LiCl administration demonstrated a significant decrease in preference for 5 mM saccharin in 4 consecutive 23 h, 2-bottle preference tests versus water (P = 0.0053). Saccharin was highly preferred in mice receiving intragastric (IG) saccharin only or interperitoneal (i.p.) injection of LiCl only. This reduced preference indicated that mice "tasted" saccharin infused into the gut. However, efforts to replicate with a reduced infusion volume failed to result in decreased preference. To understand if there were alternative pathways for oral detection of infused saccharin, mice received intragastric infusions (5.4 mM) and i.p. injections (10.8 mM) of sodium fluorescein. Fluorescence was observed from the tongues and esophagi of mice infused with volumes of 0.5 mL or more or injected with volumes of 0.25 mL or greater. Interperitoneal injections of 5 mM saccharin in mice resulted in reduced preference for 5 mM saccharin presented orally in 2-bottle preference tests (P = 0.0287). Oral delivery of a 500-fold less concentration of saccharin (0.01 mM) during conditioning resulted in a similar preference expression as shown in the initial IG experiment. These results demonstrate that although compounds may be tasted in the mouth absent of oral contact, associative learning is attenuated. Therefore, intestinal taste receptors are unlikely to participate directly in learning and recognition of foods during foraging events.
Collapse
Affiliation(s)
- G J Golden
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the presence and possible roles of peripheral taste/nutrient sensors, particularly taste receptors. RECENT FINDINGS Recent studies have demonstrated that taste signaling molecules are distributed not only in the gustatory epithelium, but also in other tissues, including the gastrointestinal tract, airways, testes and brain. Taste signaling mechanisms in the gastrointestinal tract were reported to participate in detecting sweet, umami and bitter compounds. Several research groups have suggested that tastant/nutrient detection by other systems contributes to the behavioral responses to food intake. SUMMARY Taste-like cells expressing taste signaling components are distributed in multiple tissues. Investigation of their potential roles in chemosensing has just begun. Researchers have identified at least two chemosensory pathways in the gastrointestinal tract for detecting tastants/nutrients. One is the taste receptor signaling pathway and the other is the currently unknown nutrient-sensing pathway that elicits postingestive effects. The former system utilizes a mechanism similar to taste sensing in the oral cavity. By understanding how tastants/nutrients are sensed and regulated through both systems, we may be able to more effectively control food intake in the future.
Collapse
Affiliation(s)
- Ken Iwatsuki
- Institute for Innovation, Ajinomoto Co. Inc., Kawasaki-ku, Kawasaki, Japan
| | | |
Collapse
|
40
|
Abstract
Recent advances in molecular biology have led to the investigation of the molecular mechanism by which chemicals such as odors and tastants are perceived by specific chemosensory organs. For example, G protein-coupled receptors expressed within the nasal epithelium and taste receptors in the oral cavity have been identified as odorant and taste receptors, respectively. However, there is much evidence to indicate that these chemosensory receptors are not restricted to primary chemosensory cells; they are also expressed and have function in other cells such as those in the airways and gastrointestinal (GI) tract. This short review describes the possible mechanisms by which taste signal transduction occurs in the oral cavity and tastants/nutrients are sensed in the GI tract by taste-like cells, mainly enteroendocrine and brush cells. Furthermore, it discusses the future perspectives of chemosensory studies.
Collapse
Affiliation(s)
- Ken Iwatsuki
- Institute for Innovation, Ajinomoto Co., Inc., Japan.
| | | |
Collapse
|
41
|
Green BG. Chemesthesis and the chemical senses as components of a "chemofensor complex". Chem Senses 2011; 37:201-6. [PMID: 22210122 DOI: 10.1093/chemse/bjr119] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
An important function of the chemical senses is to warn against dangerous biological and chemical agents in the environment. The discovery in recent years of "taste" receptor cells outside the oral cavity that appear to have protective functions has raised new questions about the nature and scope of the chemical senses in general and of chemesthesis in particular. The present paper briefly reviews these findings within the context of what is currently known about the body's chemically sensitive protective mechanisms, including nonsensory processes that help to expel or neutralize threatening agents once they have been encountered. It is proposed that this array of defense mechanisms constitutes a "chemofensor complex" in which chemesthesis is the most ubiquitous, functionally diverse, and interactive chemosensory component.
Collapse
Affiliation(s)
- Barry G Green
- The John B. Pierce Laboratory, New Haven, CT 06519, USA.
| |
Collapse
|
42
|
|
43
|
Gustatory and extragustatory functions of mammalian taste receptors. Physiol Behav 2011; 105:4-13. [DOI: 10.1016/j.physbeh.2011.02.010] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 02/01/2011] [Accepted: 02/07/2011] [Indexed: 01/05/2023]
|
44
|
Abstract
Taste is a discriminative sense involving specialized receptor cells of the oral cavity (taste buds) and at least two distinct families of G protein-coupled receptor molecules that detect nutritionally important substances or potential toxins. Yet the receptor mechanisms that drive taste also are utilized by numerous systems throughout the body. How and why these so-called taste receptors are used to regulate digestion and respiration is now a matter of intense study. In this article we provide a historical perspective and an overview of these systems, leading to speculations on directions for further research.
Collapse
Affiliation(s)
- Thomas E. Finger
- Anschutz Medical Campus, University of Colorado Denver, School of Medicine, Rocky Mountain Taste & Smell Center, Department of Cell and Developmental BiologyRC-1 South, Room 11118, PO Box 6511, Mail Stop 8108, Aurora, CO 80045USA
| | - Sue C. Kinnamon
- Anschutz Medical Campus, University of Colorado Denver, School of Medicine, Rocky Mountain Taste & Smell Center, Department of Otolaryngology12700 E 19th Avenue, MS 8606, Aurora, CO 80045USA
| |
Collapse
|
45
|
Horn CC, Murat C, Rosazza M, Still L. Effects of gastric distension and infusion of umami and bitter taste stimuli on vagal afferent activity. Brain Res 2011; 1419:53-60. [PMID: 21925651 DOI: 10.1016/j.brainres.2011.08.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/22/2011] [Accepted: 08/22/2011] [Indexed: 11/24/2022]
Abstract
Until recently, sensory nerve pathways from the stomach to the brain were thought to detect distension and play little role in nutritional signaling. Newer data have challenged this view, including reports on the presence of taste receptors in the gastrointestinal lumen and the stimulation of multi-unit vagal afferent activity by glutamate infusions into the stomach. However, assessing these chemosensory effects is difficult because gastric infusions typically evoke a distension-related vagal afferent response. In the current study, we recorded gastric vagal afferent activity in the rat to investigate the possibility that umami (glutamate, 150 mM) and bitter (denatonium, 10 mM) responses could be dissociated from distension responses by adjusting the infusion rate and opening or closing the drainage port in the stomach. Slow infusions of saline (5 ml over 2 min, open port) produced no significant effects on vagal activity. Using the same infusion rate, glutamate or denatonium solutions produced little or no effects on vagal afferent activity. In an attempt to reproduce a prior report that showed distention and glutamate responses, we produced a distension response by closing the exit port. Under this condition, response to the infusion of glutamate or denatonium was similar to saline. In summary, we found little or no effect of gastric infusion of glutamate or denatonium on gastric vagal afferent activity that could be distinguished from distension responses. The current results suggest that sensitivity to umami or bitter stimuli is not a common property of gastric vagal afferent fibers.
Collapse
Affiliation(s)
- Charles C Horn
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA.
| | | | | | | |
Collapse
|
46
|
Schier LA, Davidson TL, Powley TL. Ongoing ingestive behavior is rapidly suppressed by a preabsorptive, intestinal "bitter taste" cue. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1557-68. [PMID: 21865540 DOI: 10.1152/ajpregu.00344.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The discovery that cells in the gastrointestinal (GI) tract express the same molecular receptors and intracellular signaling components known to be involved in taste has generated great interest in potential functions of such post-oral "taste" receptors in the control of food intake. To determine whether taste cues in the GI tract are detected and can directly influence behavior, the present study used a microbehavioral analysis of intake, in which rats drank from lickometers that were programmed to simultaneously deliver a brief yoked infusion of a taste stimulus to the intestines. Specifically, in daily 30-min sessions, thirsty rats with indwelling intraduodenal catheters were trained to drink hypotonic (0.12 M) sodium chloride (NaCl) and simultaneously self-infuse a 0.12 M NaCl solution. Once trained, in a subsequent series of intestinal taste probe trials, rats reduced licking during a 6-min infusion period, when a bitter stimulus denatonium benzoate (DB; 10 mM) was added to the NaCl vehicle for infusion, apparently conditioning a mild taste aversion. Presentation of the DB in isomolar lithium chloride (LiCl) for intestinal infusions accelerated the development of the response across trials and strengthened the temporal resolution of the early licking suppression in response to the arrival of the DB in the intestine. In an experiment to evaluate whether CCK is involved as a paracrine signal in transducing the intestinal taste of DB, the CCK-1R antagonist devazepide partially blocked the response to intestinal DB. In contrast to their ability to detect and avoid the bitter taste in the intestine, rats did not modify their licking to saccharin intraduodenal probe infusions. The intestinal taste aversion paradigm developed here provides a sensitive and effective protocol for evaluating which tastants-and concentrations of tastants-in the lumen of the gut can control ingestion.
Collapse
Affiliation(s)
- Lindsey A Schier
- Dept. of Psychological Sciences, 703 Third St., Purdue Univ., West Lafayette, IN 47907, USA.
| | | | | |
Collapse
|
47
|
Loney GC, Torregrossa AM, Smith JC, Sclafani A, Eckel LA. Rats display a robust bimodal preference profile for sucralose. Chem Senses 2011; 36:733-45. [PMID: 21653913 DOI: 10.1093/chemse/bjr048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Female Sprague-Dawley rats display considerable variability in their preference for the artificial sweetener sucralose over water. While some rats can be classified as sucralose preferrers (SP), as they prefer sucralose across a broad range of concentrations, others can be classified as sucralose avoiders (SA), as they avoid sucralose at concentrations above 0.1 g/L. Here, we expand on a previous report of this phenomenon by demonstrating, in a series of 2-bottle 24-h preference tests involving water and an ascending series of sucralose concentrations, that this variability in sucralose preference is robust across sex, stage of the estrous cycle, and 2 rat strains (Long-Evans and Sprague-Dawley). In a second experiment involving a large sample of rats (n = 50), we established that the ratio of SP to SA is approximately 35-65%. This bimodal behavioral response to sucralose appears to be driven by taste because rats display a similar bimodal licking response to a range of sucralose solutions presented during brief-access tests. Finally, we have shown that sucralose avoidance is extremely robust as 23-h water-deprived SA continue to avoid sucralose in 1-h single-bottle intake tests. Based on their reduced licking responses to sucralose during brief-access (taste driven) tests, and the fact that their distaste for sucralose cannot be overcome by the motivation to rehydrate, we conclude that SA detect a negative taste quality of sucralose that SP are relatively insensitive to.
Collapse
Affiliation(s)
- Gregory C Loney
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, USA
| | | | | | | | | |
Collapse
|
48
|
Stengel A, Taché Y. Interaction between gastric and upper small intestinal hormones in the regulation of hunger and satiety: ghrelin and cholecystokinin take the central stage. Curr Protein Pept Sci 2011; 12:293-304. [PMID: 21428875 PMCID: PMC3670092 DOI: 10.2174/138920311795906673] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 03/23/2011] [Indexed: 12/13/2022]
Abstract
Several peptides are produced and released from endocrine cells scattered within the gastric oxyntic and the small intestinal mucosa. These peptide hormones are crucially involved in the regulation of gastrointestinal functions and food intake by conveying their information to central regulatory sites located in the brainstem as well as in the forebrain, such as hypothalamic nuclei. So far, ghrelin is the only known hormone that is peripherally produced in gastric X/A-like cells and centrally acting to stimulate food intake, whereas the suppression of feeding seems to be much more redundantly controlled by a number of gut peptides. Cholecystokinin produced in the duodenum is a well established anorexigenic hormone that interacts with ghrelin to modulate food intake indicating a regulatory network located at the first site of contact with nutrients in the stomach and upper small intestine. In addition, a number of peptides including leptin, urocortin 2, amylin and glucagon-like peptide 1 interact synergistically with CCK to potentiate its satiety signaling effect. New developments have led to the identification of additional peptides in X/A-like cells either derived from the pro-ghrelin gene by alternative splicing and posttranslational processing (obestatin) or a distinct gene (nucleobindin2/nesfatin-1) which have been investigated for their influence on food intake.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yvette Taché
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
49
|
Jahan-Mihan A, Luhovyy BL, El Khoury D, Anderson GH. Dietary proteins as determinants of metabolic and physiologic functions of the gastrointestinal tract. Nutrients 2011; 3:574-603. [PMID: 22254112 PMCID: PMC3257691 DOI: 10.3390/nu3050574] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 04/29/2011] [Accepted: 05/09/2011] [Indexed: 02/07/2023] Open
Abstract
Dietary proteins elicit a wide range of nutritional and biological functions. Beyond their nutritional role as the source of amino acids for protein synthesis, they are instrumental in the regulation of food intake, glucose and lipid metabolism, blood pressure, bone metabolism and immune function. The interaction of dietary proteins and their products of digestion with the regulatory functions of the gastrointestinal (GI) tract plays a dominant role in determining the physiological properties of proteins. The site of interaction is widespread, from the oral cavity to the colon. The characteristics of proteins that influence their interaction with the GI tract in a source-dependent manner include their physico-chemical properties, their amino acid composition and sequence, their bioactive peptides, their digestion kinetics and also the non-protein bioactive components conjugated with them. Within the GI tract, these products affect several regulatory functions by interacting with receptors releasing hormones, affecting stomach emptying and GI transport and absorption, transmitting neural signals to the brain, and modifying the microflora. This review discusses the interaction of dietary proteins during digestion and absorption with the physiological and metabolic functions of the GI tract, and illustrates the importance of this interaction in the regulation of amino acid, glucose, lipid metabolism, and food intake.
Collapse
Affiliation(s)
- Alireza Jahan-Mihan
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | |
Collapse
|
50
|
Bitter taste receptors and α-gustducin regulate the secretion of ghrelin with functional effects on food intake and gastric emptying. Proc Natl Acad Sci U S A 2011; 108:2094-9. [PMID: 21245306 DOI: 10.1073/pnas.1011508108] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a hunger hormone with gastroprokinetic properties but the factors controlling ghrelin secretion from the stomach are unknown. Bitter taste receptors (T2R) and the gustatory G proteins, α-gustducin (gust) and α-transducin, are expressed in the gut and are involved in the chemosensation of nutrients. This study aimed to investigate whether T2R-agonists affect (i) ghrelin release via α-gustducin and (ii) food intake and gastric emptying via the release of ghrelin. The mouse stomach contains two ghrelin cell populations: cells containing octanoyl and desoctanoyl ghrelin, which were colocalized with α-gustducin and α-transducin, and cells staining for desoctanoyl ghrelin. Gavage of T2R-agonists increased plasma octanoyl ghrelin levels in WT mice but the effect was partially blunted in gust(-/-) mice. Intragastric administration of T2R-agonists increased food intake during the first 30 min in WT but not in gust(-/-) and ghrelin receptor knockout mice. This increase was accompanied by an increase in the mRNA expression of agouti-related peptide in the hypothalamus of WT but not of gust(-/-) mice. The temporary increase in food intake was followed by a prolonged decrease (next 4 h), which correlated with an inhibition of gastric emptying. The delay in emptying, which was partially counteracted by ghrelin, was not mediated by cholecystokinin and GLP-1 but involved a direct inhibitory effect of T2R-agonists on gastric contractility. This study is unique in providing functional evidence that activation of bitter taste receptors stimulates ghrelin secretion. Modulation of endogenous ghrelin levels by tastants may provide novel therapeutic applications for the treatment of weight -and gastrointestinal motility disorders.
Collapse
|