51
|
|
52
|
Rønnestad I, Akiba Y, Kaji I, Kaunitz JD. Duodenal luminal nutrient sensing. Curr Opin Pharmacol 2014; 19:67-75. [PMID: 25113991 DOI: 10.1016/j.coph.2014.07.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/11/2014] [Accepted: 07/18/2014] [Indexed: 12/12/2022]
Abstract
The gastrointestinal mucosa is exposed to numerous chemical substances and microorganisms, including macronutrients, micronutrients, bacteria, endogenous ions, and proteins. The regulation of mucosal protection, digestion, absorption and motility is signaled in part by luminal solutes. Therefore, luminal chemosensing is an important mechanism enabling the mucosa to monitor luminal conditions, such as pH, ion concentrations, nutrient quantity, and microflora. The duodenal mucosa shares luminal nutrient receptors with lingual taste receptors in order to detect the five basic tastes, in addition to essential nutrients, and unwanted chemicals. The recent 'de-orphanization' of nutrient sensing G protein-coupled receptors provides an essential component of the mechanism by which the mucosa senses luminal nutrients. In this review, we will update the mechanisms of and underlying physiological and pathological roles in luminal nutrient sensing, with a main focus on the duodenal mucosa.
Collapse
Affiliation(s)
- Ivar Rønnestad
- Department of Medicine, School of Medicine, University of California, Los Angeles, USA; Department of Biology, University of Bergen, N5020 Bergen, Norway
| | - Yasutada Akiba
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, USA; Department of Medicine, School of Medicine, University of California, Los Angeles, USA; Brentwood Biomedical Research Institute, Los Angeles, CA 90073, USA
| | - Izumi Kaji
- Department of Medicine, School of Medicine, University of California, Los Angeles, USA; Brentwood Biomedical Research Institute, Los Angeles, CA 90073, USA
| | - Jonathan D Kaunitz
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, USA; Department of Medicine, School of Medicine, University of California, Los Angeles, USA; Department of Surgery, School of Medicine, University of California, Los Angeles, USA; Brentwood Biomedical Research Institute, Los Angeles, CA 90073, USA.
| |
Collapse
|
53
|
Schulze C, Bangert A, Kottra G, Geillinger KE, Schwanck B, Vollert H, Blaschek W, Daniel H. Inhibition of the intestinal sodium-coupled glucose transporter 1 (SGLT1) by extracts and polyphenols from apple reduces postprandial blood glucose levels in mice and humans. Mol Nutr Food Res 2014; 58:1795-808. [DOI: 10.1002/mnfr.201400016] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/13/2014] [Accepted: 05/07/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Christine Schulze
- Molecular Nutrition Unit; ZIEL Research Center of Nutrition and Food Sciences; Technical University of Munich; Freising-Weihenstephan Germany
| | - Adina Bangert
- Molecular Nutrition Unit; ZIEL Research Center of Nutrition and Food Sciences; Technical University of Munich; Freising-Weihenstephan Germany
| | - Gabor Kottra
- Molecular Nutrition Unit; ZIEL Research Center of Nutrition and Food Sciences; Technical University of Munich; Freising-Weihenstephan Germany
| | - Kerstin Elisabeth Geillinger
- Molecular Nutrition Unit; ZIEL Research Center of Nutrition and Food Sciences; Technical University of Munich; Freising-Weihenstephan Germany
| | - Bettina Schwanck
- Department of Pharmaceutical Biology; Pharmaceutical Institute, Christian-Albrechts-University of Kiel; Gutenbergstrasse Kiel Germany
| | | | - Wolfgang Blaschek
- Department of Pharmaceutical Biology; Pharmaceutical Institute, Christian-Albrechts-University of Kiel; Gutenbergstrasse Kiel Germany
| | - Hannelore Daniel
- Molecular Nutrition Unit; ZIEL Research Center of Nutrition and Food Sciences; Technical University of Munich; Freising-Weihenstephan Germany
| |
Collapse
|
54
|
Foster SR, Blank K, Hoe LES, Behrens M, Meyerhof W, Peart JN, Thomas WG. Bitter taste receptor agonists elicit G‐protein‐dependent negative inotropy in the murine heart. FASEB J 2014; 28:4497-508. [DOI: 10.1096/fj.14-256305] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Simon R. Foster
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Kristina Blank
- Department of Molecular GeneticsGerman Institute of Human Nutrition (DIfE) Potsdam‐RehbrückeNuthetalGermany
| | - Louise E. See Hoe
- Griffith Health InstituteGriffith UniversityGold CoastQueenslandAustralia
| | - Maik Behrens
- Department of Molecular GeneticsGerman Institute of Human Nutrition (DIfE) Potsdam‐RehbrückeNuthetalGermany
| | - Wolfgang Meyerhof
- Department of Molecular GeneticsGerman Institute of Human Nutrition (DIfE) Potsdam‐RehbrückeNuthetalGermany
| | - Jason N. Peart
- Griffith Health InstituteGriffith UniversityGold CoastQueenslandAustralia
| | - Walter G. Thomas
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
55
|
Awad WA, Aschenbach JR, Ghareeb K, Khayal B, Hess C, Hess M. Campylobacter jejuni influences the expression of nutrient transporter genes in the intestine of chickens. Vet Microbiol 2014; 172:195-201. [PMID: 24834798 DOI: 10.1016/j.vetmic.2014.04.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/25/2014] [Accepted: 04/02/2014] [Indexed: 11/27/2022]
Abstract
The gastrointestinal tract represents the first barrier against pathogens. However, the interaction of Campylobacter with intestinal epithelial cells and its effects on the intestinal function of chickens are poorly studied. Therefore, the goal of the present study was to characterize the effects of C. jejuni oral infection on the mRNA expression of nutrient transporters in the intestine. Newly hatched specific pathogen-free (SPF) chickens were orally infected with C. jejuni (NCTC 12744; 1 × 10(8)CFU/bird) at 14 days of age. Quantitative RT-PCR analyses at 14 days-post infection (dpi) revealed that the relative gene expression of the sodium/glucose cotransporter (SGLT-1) and the peptide transporter (PepT-1) was down-regulated (P<0.05) in all investigated segments (duodenum, jejunum and cecum) of Campylobacter-infected birds, while the facilitated glucose transporter (GLUT-2) was down-regulated (P<0.05) in jejunal and cecal tissues only. Furthermore, down-regulation (P<0.05) of the cationic amino acid transporter (CAT-2) and the excitatory amino acid transporter (EAAT-3) was seen in the jejunum, and down-regulation (P<0.05) of the l-type amino acid transporter (y(+)LAT-2) was noticed in the duodenum of infected birds. The decreased expression of intestinal nutrient transporters coincided with a decrease (P<0.05) in body weight and body weight gain during a 2-week post infection period. For the first time, it can be concluded that nutrient transporter expression is compromised in the small and large intestine of Campylobacter-infected birds with negative consequences on growth performance. Furthermore, the down-regulation of mRNA expression of glucose and amino acid transporters may result in accumulation of nutrients in the intestinal lumen, which may favor C. jejuni replication and colonization.
Collapse
Affiliation(s)
- Wageha A Awad
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, A-1210 Vienna, Austria; Department of Animal Hygiene, Poultry and Environment and Department of Animal Behaviour and Management, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt.
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin, 14163 Berlin, Germany
| | - Khaled Ghareeb
- Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, A-1210 Vienna, Austria; Department of Animal Hygiene, Poultry and Environment and Department of Animal Behaviour and Management, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Basel Khayal
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Claudia Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Michael Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, A-1210 Vienna, Austria
| |
Collapse
|
56
|
Awad WA, Molnár A, Aschenbach JR, Ghareeb K, Khayal B, Hess C, Liebhart D, Dublecz K, Hess M. Campylobacter infection in chickens modulates the intestinal epithelial barrier function. Innate Immun 2014; 21:151-60. [PMID: 24553586 DOI: 10.1177/1753425914521648] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Asymptomatic carriage of Campylobacter jejuni is highly prevalent in chicken flocks. Thus, we investigated whether chronic Campylobacter carriage affects chicken intestinal functions despite the absence of clinical symptoms. An experiment was carried out in which commercial chickens were orally infected with C. jejuni (1 × 10(8) CFU/bird) at 14 days of life. Changes in ion transport and barrier function were assessed by short-circuit current (I(sc)) and transepithelial ion conductance (G(t)) in Ussing chambers. G(t) increased in cecum and colon of Campylobacter-infected chicken 7 d post-infection (DPI), whereas G t initially decreased in the jejunum at 7 DPI and increased thereafter at 14 DPI. The net charge transfer across the epithelium was reduced or tended to be reduced in all segments, as evidenced by a decreased I sc. Furthermore, the infection induced intestinal histomorphological changes, most prominently including a decrease in villus height, crypt depth and villus surface area in the jejunum at 7 DPI. Furthermore, body mass gain was decreased by Campylobacter carriage. This study demonstrates, for the first time, changes in the intestinal barrier function in Campylobacter-infected chickens and these changes were associated with a decrease in growth performance in otherwise healthy-appearing birds.
Collapse
Affiliation(s)
- Wageha A Awad
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Andor Molnár
- Department of Animal Science and Animal Husbandry, Georgikon Faculty, University of Pannonia, Keszthely, Hungary
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin, Berlin, Germany
| | - Khaled Ghareeb
- Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria Department of Animal Hygiene, Behaviour and Management, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Basel Khayal
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Claudia Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Dieter Liebhart
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Károly Dublecz
- Department of Animal Science and Animal Husbandry, Georgikon Faculty, University of Pannonia, Keszthely, Hungary
| | - Michael Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
57
|
Xiao W, Feng Y, Holst JJ, Hartmann B, Yang H, Teitelbaum DH. Glutamate prevents intestinal atrophy via luminal nutrient sensing in a mouse model of total parenteral nutrition. FASEB J 2014; 28:2073-87. [PMID: 24497581 DOI: 10.1096/fj.13-238311] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Small intestine luminal nutrient sensing may be crucial for modulating physiological functions. However, its mechanism of action is incompletely understood. We used a model of enteral nutrient deprivation, or total parenteral nutrition (TPN), resulting in intestinal mucosal atrophy and decreased epithelial barrier function (EBF). We examined how a single amino acid, glutamate (GLM), modulates intestinal epithelial cell (IEC) growth and EBF. Controls were chow-fed mice, T1 receptor-3 (T1R3)-knockout (KO) mice, and treatment with the metabotropic glutamate receptor (mGluR)-5 antagonist MTEP. TPN significantly changed the amount of T1Rs, GLM receptors, and transporters, and GLM prevented these changes. GLM significantly prevented TPN-associated intestinal atrophy (2.5-fold increase in IEC proliferation) and was dependent on up-regulation of the protein kinase pAkt, but independent of T1R3 and mGluR5 signaling. GLM led to a loss of EBF with TPN (60% increase in FITC-dextran permeability, 40% decline in transepithelial resistance); via T1R3, it protected EBF, whereas mGluR5 was associated with EBF loss. GLM led to a decline in circulating glucagon-like peptide 2 (GLP-2) during TPN. The decline was regulated by T1R3 and mGluR5, suggesting a novel negative regulator pathway for IEC proliferation not previously described. Loss of luminal nutrients with TPN administration may widely affect intestinal taste sensing. GLM has previously unrecognized actions on IEC growth and EBF. Restoring luminal sensing via GLM could be a strategy for patients on TPN.
Collapse
Affiliation(s)
- Weidong Xiao
- 1Section of Pediatric Surgery, University of Michigan, Mott Children's Hospital F3970, Ann Arbor, MI 48109-5245, USA.
| | | | | | | | | | | |
Collapse
|
58
|
Notari L, Riera DC, Sun R, Bohl JA, McLean LP, Madden KB, van Rooijen N, Vanuytsel T, Urban JF, Zhao A, Shea-Donohue T. Role of macrophages in the altered epithelial function during a type 2 immune response induced by enteric nematode infection. PLoS One 2014; 9:e84763. [PMID: 24465430 PMCID: PMC3900397 DOI: 10.1371/journal.pone.0084763] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 11/18/2013] [Indexed: 01/10/2023] Open
Abstract
Parasitic enteric nematodes induce a type 2 immune response characterized by increased production of Th2 cytokines, IL-4 and IL-13, and recruitment of alternatively activated macrophages (M2) to the site of infection. Nematode infection is associated with changes in epithelial permeability and inhibition of sodium-linked glucose absorption, but the role of M2 in these effects is unknown. Clodronate-containing liposomes were administered prior to and during nematode infection to deplete macrophages and prevent the development of M2 in response to infection with Nippostrongylus brasiliensis. The inhibition of epithelial glucose absorption that is associated with nematode infection involved a macrophage-dependent reduction in SGLT1 activity, with no change in receptor expression, and a macrophage-independent down-regulation of GLUT2 expression. The reduced transport of glucose into the enterocyte is compensated partially by an up-regulation of the constitutive GLUT1 transporter consistent with stress-induced activation of HIF-1α. Thus, nematode infection results in a “lean” epithelial phenotype that features decreased SGLT1 activity, decreased expression of GLUT2 and an emergent dependence on GLUT1 for glucose uptake into the enterocyte. Macrophages do not play a role in enteric nematode infection-induced changes in epithelial barrier function. There is a greater contribution, however, of paracellular absorption of glucose to supply the energy demands of host resistance. These data provide further evidence of the ability of macrophages to alter glucose metabolism of neighboring cells.
Collapse
Affiliation(s)
- Luigi Notari
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Diana C. Riera
- Department of Pediatrics, Walter Reed Army Medical Center, Washington, DC, United States of America
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Rex Sun
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer A. Bohl
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Leon P. McLean
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Kathleen B. Madden
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Nico van Rooijen
- Vrije Universiteit, VUMC, Department of Molecular Cell Biology, Amsterdam, The Netherlands
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders, University Hospital Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Joseph F. Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, & Immunology Laboratory, Beltsville, Maryland, United States of America
| | - Aiping Zhao
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Terez Shea-Donohue
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
59
|
Arakawa H, Saito S, Kanagawa M, Kamioka H, Yano K, Morimoto K, Ogihara T. Evaluation of a Thiodipeptide, l-Phenylalanyl-Ψ[CS-N]-l-alanine, as a Novel Probe for Peptide Transporter 1. Drug Metab Pharmacokinet 2014; 29:470-4. [DOI: 10.2133/dmpk.dmpk-14-rg-047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
60
|
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]
|
61
|
Extrasensory perception: odorant and taste receptors beyond the nose and mouth. Pharmacol Ther 2013; 142:41-61. [PMID: 24280065 DOI: 10.1016/j.pharmthera.2013.11.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 11/04/2013] [Indexed: 12/22/2022]
Abstract
G protein-coupled receptors (GPCRs) represent the largest family of transmembrane receptors and are prime therapeutic targets. The odorant and taste receptors account for over half of the GPCR repertoire, yet they are generally excluded from large-scale, drug candidate analyses. Accumulating molecular evidence indicates that the odorant and taste receptors are widely expressed throughout the body and functional beyond the oronasal cavity - with roles including nutrient sensing, autophagy, muscle regeneration, regulation of gut motility, protective airway reflexes, bronchodilation, and respiratory disease. Given this expanding array of actions, the restricted perception of these GPCRs as mere mediators of smell and taste is outdated. Moreover, delineation of the precise actions of odorant and taste GPCRs continues to be hampered by the relative paucity of selective and specific experimental tools, as well as the lack of defined receptor pharmacology. In this review, we summarize the evidence for expression and function of odorant and taste receptors in tissues beyond the nose and mouth, and we highlight their broad potential in physiology and pathophysiology.
Collapse
|
62
|
Resnyk CW, Carré W, Wang X, Porter TE, Simon J, Le Bihan-Duval E, Duclos MJ, Aggrey SE, Cogburn LA. Transcriptional analysis of abdominal fat in genetically fat and lean chickens reveals adipokines, lipogenic genes and a link between hemostasis and leanness. BMC Genomics 2013; 14:557. [PMID: 23947536 PMCID: PMC3765218 DOI: 10.1186/1471-2164-14-557] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/12/2013] [Indexed: 12/15/2022] Open
Abstract
Background This descriptive study of the abdominal fat transcriptome takes advantage of two experimental lines of meat-type chickens (Gallus domesticus), which were selected over seven generations for a large difference in abdominal (visceral) fatness. At the age of selection (9 wk), the fat line (FL) and lean line (LL) chickens exhibit a 2.5-fold difference in abdominal fat weight, while their feed intake and body weight are similar. These unique avian models were originally created to unravel genetic and endocrine regulation of adiposity and lipogenesis in meat-type chickens. The Del-Mar 14K Chicken Integrated Systems microarray was used for a time-course analysis of gene expression in abdominal fat of FL and LL chickens during juvenile development (1–11 weeks of age). Results Microarray analysis of abdominal fat in FL and LL chickens revealed 131 differentially expressed (DE) genes (FDR≤0.05) as the main effect of genotype, 254 DE genes as an interaction of age and genotype and 3,195 DE genes (FDR≤0.01) as the main effect of age. The most notable discoveries in the abdominal fat transcriptome were higher expression of many genes involved in blood coagulation in the LL and up-regulation of numerous adipogenic and lipogenic genes in FL chickens. Many of these DE genes belong to pathways controlling the synthesis, metabolism and transport of lipids or endocrine signaling pathways activated by adipokines, retinoid and thyroid hormones. Conclusions The present study provides a dynamic view of differential gene transcription in abdominal fat of chickens genetically selected for fatness (FL) or leanness (LL). Remarkably, the LL chickens over-express a large number of hemostatic genes that could be involved in proteolytic processing of adipokines and endocrine factors, which contribute to their higher lipolysis and export of stored lipids. Some of these changes are already present at 1 week of age before the divergence in fatness. In contrast, the FL chickens have enhanced expression of numerous lipogenic genes mainly after onset of divergence, presumably directed by multiple transcription factors. This transcriptional analysis shows that abdominal fat of the chicken serves a dual function as both an endocrine organ and an active metabolic tissue, which could play a more significant role in lipogenesis than previously thought.
Collapse
Affiliation(s)
- Christopher W Resnyk
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Evidence of sugar sensitive genes in the gut of a carnivorous fish species. Comp Biochem Physiol B Biochem Mol Biol 2013; 166:58-64. [PMID: 23850750 DOI: 10.1016/j.cbpb.2013.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/01/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022]
Abstract
The ability of intestine to sense glucose in carnivorous animals (consuming minimal carbohydrate) has been partially evaluated to date only in cats. We have evaluated the expression of markers involved in the detection of simple sugars in the intestine of the strict carnivorous fish species rainbow trout (Oncorhynchus mykiss) in response to an oral glucose load and to glucose, galactose and mannose stimulation in vitro. These markers include metabolic (GLUT2 and glucokinase (hexokinase IV, GK)) and electrogenic (SGLT1) sensors, the nuclear receptor nr1h3 and the components of the G-protein-coupled taste receptors (tas1r2-like, tas1r3-like and gnat3-like). For the first time, we show that the gut of rainbow trout can detect simple sugars including glucose, galactose and mannose and respond by changing the expression levels of glucose-sensing proteins. The glucosensing response based on the metabolic and nuclear receptor systems had not been evidenced before in any carnivorous vertebrate species, whereas the responses of markers of the electrogenic mechanism and the taste receptor mechanism were different than those already described in cats. When the responses observed in rainbow trout were compared with those of omnivorous mammals, similar responses were obtained for nr1h3 whereas several differences arise in the responses of the other markers. Intestinal glucose sensing in the rainbow trout appears to be distinct from that reported for other carnivores such as cats and omnivores, revealing a novel glucose sensing mechanism not related entirely to diet in vertebrates and supports the idea that this species constitute a robust model for nutrient sensing study. Since only mRNA abundance is presented, depth studies are needed to fully understand the importance of the present findings.
Collapse
|
64
|
Wauson EM, Lorente-Rodríguez A, Cobb MH. Minireview: Nutrient sensing by G protein-coupled receptors. Mol Endocrinol 2013; 27:1188-97. [PMID: 23820899 DOI: 10.1210/me.2013-1100] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that recognize molecules in the extracellular milieu and transmit signals inside cells to regulate their behaviors. Ligands for many GPCRs are hormones or neurotransmitters that direct coordinated, stereotyped adaptive responses. Ligands for other GPCRs provide information to cells about the extracellular environment. Such information facilitates context-specific decision making that may be cell autonomous. Among ligands that are important for cellular decisions are amino acids, required for continued protein synthesis, as metabolic starting materials and energy sources. Amino acids are detected by a number of class C GPCRs. One cluster of amino acid-sensing class C GPCRs includes umami and sweet taste receptors, GPRC6A, and the calcium-sensing receptor. We have recently found that the umami taste receptor heterodimer T1R1/T1R3 is a sensor of amino acid availability that regulates the activity of the mammalian target of rapamycin. This review focuses on an array of findings on sensing amino acids and sweet molecules outside of neurons by this cluster of class C GPCRs and some of the physiologic processes regulated by them.
Collapse
Affiliation(s)
- Eric M Wauson
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9041, USA
| | | | | |
Collapse
|
65
|
Expression, regulation and putative nutrient-sensing function of taste GPCRs in the heart. PLoS One 2013; 8:e64579. [PMID: 23696900 PMCID: PMC3655793 DOI: 10.1371/journal.pone.0064579] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/15/2013] [Indexed: 01/07/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are critical for cardiovascular physiology. Cardiac cells express >100 nonchemosensory GPCRs, indicating that important physiological and potential therapeutic targets remain to be discovered. Moreover, there is a growing appreciation that members of the large, distinct taste and odorant GPCR families have specific functions in tissues beyond the oronasal cavity, including in the brain, gastrointestinal tract and respiratory system. To date, these chemosensory GPCRs have not been systematically studied in the heart. We performed RT-qPCR taste receptor screens in rodent and human heart tissues that revealed discrete subsets of type 2 taste receptors (TAS2/Tas2) as well as Tas1r1 and Tas1r3 (comprising the umami receptor) are expressed. These taste GPCRs are present in cultured cardiac myocytes and fibroblasts, and by in situ hybridization can be visualized across the myocardium in isolated cardiac cells. Tas1r1 gene-targeted mice (Tas1r1(Cre)/Rosa26(tdRFP)) strikingly recapitulated these data. In vivo taste receptor expression levels were developmentally regulated in the postnatal period. Intriguingly, several Tas2rs were upregulated in cultured rat myocytes and in mouse heart in vivo following starvation. The discovery of taste GPCRs in the heart opens an exciting new field of cardiac research. We predict that these taste receptors may function as nutrient sensors in the heart.
Collapse
|
66
|
Chaudhry RM, Garg A, Abdelfatah MM, Duenes JA, Sarr MG. Lack of functionally active sweet taste receptors in the jejunum in vivo in the rat. J Surg Res 2013; 183:606-11. [PMID: 23531453 DOI: 10.1016/j.jss.2013.02.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 02/15/2013] [Accepted: 02/19/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND When studied in enterocyte-like cell lines (Caco-2 and RIE cells), agonists and antagonists of the sweet taste receptor (STR) augment and decrease glucose uptake, respectively. We hypothesize that exposure to STR agonists and antagonists in vivo will augment glucose absorption in the rat. MATERIALS AND METHODS About 30-cm segments of jejunum in anesthetized rats were perfused with iso-osmolar solutions containing 10, 35, and 100 mM glucose solutions (n = 6 rats, each group) with and without the STR agonist 2 mM acesulfame potassium and the STR inhibitor 10 μM U-73122 (inhibitor of the phospholipase C pathway). Carrier-mediated absorption of glucose was calculated by using stereospecific and nonstereospecific (14)C-d-glucose and (3)H-l-glucose, respectively. RESULTS Addition of the STR agonist acesulfame potassium to the 10, 35, and 100 mM glucose solutions had no substantive effects on glucose absorption from 2.1 ± 0.2 to 2.0 ± 0.3, 5.8 ± 0.2 to 4.8 ± 0.2, and 15.5 ± 2.3 to 15.7 ± 2.7 μmoL/min/30-cm intestinal segment (P > 0.05), respectively. Addition of the STR inhibitor (U-73122) also had no effect on absorption in the 10, 35, and 100 mM solutions from 2.3 ± 0.1 to 2.1 ± 0.2, 7.7 ± 0.5 to 7.2 ± 0.5, and 15.7 ± 0.9 to 15.2 ± 1.1 μmoL/min/30-cm intestinal segment, respectively. CONCLUSIONS Provision of glucose directly into rat jejunum does not augment glucose absorption via STR-mediated mechanisms within the jejunum in the rat. Our experiments show either no major role of STRs in mediating postprandial augmentation of glucose absorption or that proximal gastrointestinal tract stimulation of STR or other luminal factors may be required for absorption of glucose to be augmented by STR.
Collapse
Affiliation(s)
- Rizwan M Chaudhry
- Department of Surgery and Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
67
|
Roura E, Baldwin M, Klasing K. The avian taste system: Potential implications in poultry nutrition. Anim Feed Sci Technol 2013. [DOI: 10.1016/j.anifeedsci.2012.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
68
|
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: 148] [Impact Index Per Article: 12.3] [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
|
69
|
Schiffman SS, Rother KI. Sucralose, a synthetic organochlorine sweetener: overview of biological issues. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2013; 16:399-451. [PMID: 24219506 PMCID: PMC3856475 DOI: 10.1080/10937404.2013.842523] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Sucralose is a synthetic organochlorine sweetener (OC) that is a common ingredient in the world's food supply. Sucralose interacts with chemosensors in the alimentary tract that play a role in sweet taste sensation and hormone secretion. In rats, sucralose ingestion was shown to increase the expression of the efflux transporter P-glycoprotein (P-gp) and two cytochrome P-450 (CYP) isozymes in the intestine. P-gp and CYP are key components of the presystemic detoxification system involved in first-pass drug metabolism. The effect of sucralose on first-pass drug metabolism in humans, however, has not yet been determined. In rats, sucralose alters the microbial composition in the gastrointestinal tract (GIT), with relatively greater reduction in beneficial bacteria. Although early studies asserted that sucralose passes through the GIT unchanged, subsequent analysis suggested that some of the ingested sweetener is metabolized in the GIT, as indicated by multiple peaks found in thin-layer radiochromatographic profiles of methanolic fecal extracts after oral sucralose administration. The identity and safety profile of these putative sucralose metabolites are not known at this time. Sucralose and one of its hydrolysis products were found to be mutagenic at elevated concentrations in several testing methods. Cooking with sucralose at high temperatures was reported to generate chloropropanols, a potentially toxic class of compounds. Both human and rodent studies demonstrated that sucralose may alter glucose, insulin, and glucagon-like peptide 1 (GLP-1) levels. Taken together, these findings indicate that sucralose is not a biologically inert compound.
Collapse
Affiliation(s)
- Susan S. Schiffman
- Department of Electrical and Computer Engineering, College of Engineering, North Carolina State University, Raleigh, North Carolina, USA
- Address correspondence to Susan S. Schiffman, PhD, Department of Electrical and Computer Engineering, College of Engineering, North Carolina State University, Raleigh, NC 27695-7911, USA. E-mail:
| | - Kristina I. Rother
- Section on Pediatric Diabetes & Metabolism, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
70
|
Zheng Y, Sarr MG. Effect of the artificial sweetener, acesulfame potassium, a sweet taste receptor agonist, on glucose uptake in small intestinal cell lines. J Gastrointest Surg 2013; 17:153-8; discussion p. 158. [PMID: 22948835 PMCID: PMC3516624 DOI: 10.1007/s11605-012-1998-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/06/2012] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Sweet taste receptors may enhance glucose absorption. AIM This study aimed to explore the cell biology of sweet taste receptors on glucose uptake. HYPOTHESIS Artificial sweeteners increase glucose uptake via activating sweet taste receptors in the enterocyte to translocate GLUT2 to the apical membrane through the PLC βII pathway. METHODS Caco-2, RIE-1, and IEC-6 cells, starved from glucose for 1 h were pre-incubated with 10 mM acesulfame potassium (AceK). Glucose uptake was measured by incubating cells for 1 to 10 min with 0.5-50 mM glucose with or without U-73122, chelerythrine, and cytochalasin B. RESULTS In Caco-2 and RIE-1 cells, 10 mM AceK increased glucose uptake by 20-30 % at glucose >25 mM, but not in lesser glucose concentrations (<10 mM), nor at 1 min or 10 min incubations. U-73122 (PLC βII inhibitor) inhibited uptake at glucose >25 mM and for 5 min incubation; chelerythrine and cytochalasin B had similar effects. No effect occurred in IEC-6 cells. Activation of sweet taste receptors had no effect on glucose uptake in low (<25 mM) glucose concentrations but increased uptake at greater concentrations (>25 mM). CONCLUSIONS Role of artificial sweeteners on glucose uptake appears to act in part by effects on the enterocyte itself.
Collapse
|
71
|
Oya M, Kitaguchi T, Pais R, Reimann F, Gribble F, Tsuboi T. The G protein-coupled receptor family C group 6 subtype A (GPRC6A) receptor is involved in amino acid-induced glucagon-like peptide-1 secretion from GLUTag cells. J Biol Chem 2012; 288:4513-21. [PMID: 23269670 DOI: 10.1074/jbc.m112.402677] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although amino acids are dietary nutrients that evoke the secretion of glucagon-like peptide 1 (GLP-1) from intestinal L cells, the precise molecular mechanism(s) by which amino acids regulate GLP-1 secretion from intestinal L cells remains unknown. Here, we show that the G protein-coupled receptor (GPCR), family C group 6 subtype A (GPRC6A), is involved in amino acid-induced GLP-1 secretion from the intestinal L cell line GLUTag. Application of l-ornithine caused an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) in GLUTag cells. Application of a GPRC6A receptor antagonist, a phospholipase C inhibitor, or an IP(3) receptor antagonist significantly suppressed the l-ornithine-induced [Ca(2+)](i) increase. We found that the increase in [Ca(2+)](i) stimulated by l-ornithine correlated with GLP-1 secretion and that l-ornithine stimulation increased exocytosis in a dose-dependent manner. Furthermore, depletion of endogenous GPRC6A by a specific small interfering RNA (siRNA) inhibited the l-ornithine-induced [Ca(2+)](i) increase and GLP-1 secretion. Taken together, these findings suggest that the GPRC6A receptor functions as an amino acid sensor in GLUTag cells that promotes GLP-1 secretion.
Collapse
Affiliation(s)
- Manami Oya
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan
| | | | | | | | | | | |
Collapse
|
72
|
Tonack S, Tang C, Offermanns S. Endogenous metabolites as ligands for G protein-coupled receptors modulating risk factors for metabolic and cardiovascular disease. Am J Physiol Heart Circ Physiol 2012; 304:H501-13. [PMID: 23241321 DOI: 10.1152/ajpheart.00641.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During the last decade, several G protein-coupled receptors activated by endogenous metabolites have been described. These receptors respond to fatty acids, mono- and disaccharides, amino acids, or various intermediates and products of metabolism, including ketone bodies, lactate, succinate, or bile acids. Receptors of endogenous metabolites are expressed in taste cells, the gastrointestinal tract, adipose tissue, endocrine glands, immune cells, or the kidney and are therefore in a position to sense food intake in the gastrointestinal tract or to link metabolite levels to the appropriate responses of metabolic organs. Some of the receptors appear to provide a link between metabolic and neuronal or immune functions. Given that many of these metabolic processes are dysregulated under pathological conditions, including diabetes, dyslipidemia, and obesity, receptors of endogenous metabolites have also been recognized as potential drug targets to prevent and/or treat metabolic and cardiovascular diseases. This review describes G protein-coupled receptors activated by endogenous metabolites and summarizes their physiological, pathophysiological, and potential pharmacological roles.
Collapse
Affiliation(s)
- Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | |
Collapse
|
73
|
Sigoillot M, Brockhoff A, Meyerhof W, Briand L. Sweet-taste-suppressing compounds: current knowledge and perspectives of application. Appl Microbiol Biotechnol 2012; 96:619-30. [PMID: 22983596 DOI: 10.1007/s00253-012-4387-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 01/07/2023]
Abstract
Sweet-tasting compounds are recognized by a heterodimeric receptor composed of the taste receptor, type 1, members 2 (T1R2) and 3 (T1R3) located in the mouth. This receptor is also expressed in the gut where it is involved in intestinal absorption, metabolic regulation, and glucose homeostasis. These metabolic functions make the sweet taste receptor a potential novel therapeutic target for the treatment of obesity and related metabolic dysfunctions such as diabetes. Existing sweet taste inhibitors or blockers that are still in development would constitute promising therapeutic agents. In this review, we will summarize the current knowledge of sweet taste inhibitors, including a sweet-taste-suppressing protein named gurmarin, which is only active on rodent sweet taste receptors but not on that of humans. In addition, their potential applications as therapeutic tools are discussed.
Collapse
Affiliation(s)
- Maud Sigoillot
- Centre des Sciences du Goût et de l'Alimentation, UMR-1324 INRA, UMR-6265 CNRS, Université de Bourgogne, 21000, Dijon, France
| | | | | | | |
Collapse
|
74
|
Geraedts MCP, Takahashi T, Vigues S, Markwardt ML, Nkobena A, Cockerham RE, Hajnal A, Dotson CD, Rizzo MA, Munger SD. Transformation of postingestive glucose responses after deletion of sweet taste receptor subunits or gastric bypass surgery. Am J Physiol Endocrinol Metab 2012; 303:E464-74. [PMID: 22669246 PMCID: PMC3423100 DOI: 10.1152/ajpendo.00163.2012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/05/2012] [Indexed: 01/06/2023]
Abstract
The glucose-dependent secretion of the insulinotropic hormone glucagon-like peptide-1 (GLP-1) is a critical step in the regulation of glucose homeostasis. Two molecular mechanisms have separately been suggested as the primary mediator of intestinal glucose-stimulated GLP-1 secretion (GSGS): one is a metabotropic mechanism requiring the sweet taste receptor type 2 (T1R2) + type 3 (T1R3) while the second is a metabolic mechanism requiring ATP-sensitive K(+) (K(ATP)) channels. By quantifying sugar-stimulated hormone secretion in receptor knockout mice and in rats receiving Roux-en-Y gastric bypass (RYGB), we found that both of these mechanisms contribute to GSGS; however, the mechanisms exhibit different selectivity, regulation, and localization. T1R3(-/-) mice showed impaired glucose and insulin homeostasis during an oral glucose challenge as well as slowed insulin granule exocytosis from isolated pancreatic islets. Glucose, fructose, and sucralose evoked GLP-1 secretion from T1R3(+/+), but not T1R3(-/-), ileum explants; this secretion was not mimicked by the K(ATP) channel blocker glibenclamide. T1R2(-/-) mice showed normal glycemic control and partial small intestine GSGS, suggesting that T1R3 can mediate GSGS without T1R2. Robust GSGS that was K(ATP) channel-dependent and glucose-specific emerged in the large intestine of T1R3(-/-) mice and RYGB rats in association with elevated fecal carbohydrate throughout the distal gut. Our results demonstrate that the small and large intestines utilize distinct mechanisms for GSGS and suggest novel large intestine targets that could mimic the improved glycemic control seen after RYGB.
Collapse
Affiliation(s)
- Maartje C P Geraedts
- Division of Endocrinology, Diabetes, and Nutrition, Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Merigo F, Benati D, Cristofoletti M, Amarù F, Osculati F, Sbarbati A. Glucose transporter/T1R3-expressing cells in rat tracheal epithelium. J Anat 2012; 221:138-50. [PMID: 22640462 DOI: 10.1111/j.1469-7580.2012.01522.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Glucose transport plays an important role in maintaining low sugar concentration in airway surface liquid (ASL), which is critical for mucociliary clearance and bacterial colonization. Experimental evidence indicates that glucose/hexose uptake in lung/airway cells occurs by means of two structurally distinct glucose transporter pathways: the Na(+) -dependent glucose transporters (SGLT family) and the facilitative glucose transporters (GLUT family). In this study, we examined the expression of the major glucose transporters of the intestine, GLUT2, GLUT5, SGLT1 and T1R3 taste receptor subunit, in the trachea of rats using immunohistochemistry and immunoelectron microscopy, and compared them using double-labeled confocal microscopy. We found that GLUT2, GLUT5, SGLT1 and T1R3 are selectively expressed in different cell types. T1R3 and GLUT2 are predominantly expressed in subsets of solitary chemoreceptor cells (SCCs) and ciliated cells, GLUT5 is present in subsets of SCCs and in secretory cells, and SGLT1 is exclusively expressed in a unique cell type, SCCs. Furthermore, we demonstrated that T1R3 is colocalized with SGLT1 in SCCs and with GLUT2 transporter in ciliated cells. In conclusion, these findings reveal that different cell types are associated with the uptake of glucose in ASL and that, due to their T1R3 expression, SCCs and ciliated cells are most likely to participate in the chemosensory process in ASL.
Collapse
Affiliation(s)
- Flavia Merigo
- Department of Neurological, Neuropsychological, Morphological and Movement Sciences, Human Anatomy and Histology Section, University of Verona, School of Medicine, Verona, Italy.
| | | | | | | | | | | |
Collapse
|
76
|
The intestinal epithelium tuft cells: specification and function. Cell Mol Life Sci 2012; 69:2907-17. [PMID: 22527717 PMCID: PMC3417095 DOI: 10.1007/s00018-012-0984-7] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 03/21/2012] [Accepted: 03/26/2012] [Indexed: 02/08/2023]
Abstract
The intestinal epithelium, composed of at least seven differentiated cell types, represents an extraordinary model to understand the details of multi-lineage differentiation, a question that is highly relevant in developmental biology as well as for clinical applications. This review focuses on intestinal epithelial tuft cells that have been acknowledged as a separate entity for more than 60 years but whose function remains a mystery. We discuss what is currently known about the molecular basis of tuft cell fate and differentiation and why elucidating tuft cell function has been so difficult. Finally, we summarize the current hypotheses on their potential involvement in diseases of the gastro-intestinal tract.
Collapse
|
77
|
Mace OJ, Schindler M, Patel S. The regulation of K- and L-cell activity by GLUT2 and the calcium-sensing receptor CasR in rat small intestine. J Physiol 2012; 590:2917-36. [PMID: 22495587 DOI: 10.1113/jphysiol.2011.223800] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intestinal enteroendocrine cells (IECs) secrete gut peptides in response to both nutrients and non-nutrients. Glucose and amino acids both stimulate gut peptide secretion. Our hypothesis was that the facilitative glucose transporter, GLUT2, could act as a glucose sensor and the calcium-sensing receptor, CasR, could detect amino acids in the intestine to modify gut peptide secretion. We used isolated loops of rat small intestine to study the secretion of gluco-insulinotropic peptide (GIP), glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) secretion stimulated by luminal perfusion of nutrients or bile acid. Inhibition of the sodium-dependent glucose cotransporter 1 (SGLT1) with phloridzin partially inhibited GIP, GLP-1 and PYY secretion by 45%, suggesting another glucose sensor might be involved in modulating peptide secretion. The response was completely abolished in the presence of the GLUT2 inhibitors phloretin or cytochalasin B. Given that GLUT2 modified gut peptide secretion stimulated by glucose, we investigated whether it was involved in the secretion of gut peptide by other gut peptide secretagogues. Phloretin completely abolished gut peptide secretion stimulated by artificial sweetener (sucralose), dipeptide (glycylsarcosine), lipid (oleoylethanolamine), short chain fatty acid (propionate) and major rat bile acid (taurocholate) indicating a fundamental position for GLUT2 in the gut peptide secretory mechanism. We investigated how GLUT2 was able to influence gut peptide secretion mediated by a diverse range of stimulators and discovered that GLUT2 affected membrane depolarisation through the closure of K+(ATP)-sensitive channels. In the absence of SGLT1 activity (or presence of phloridzin), the secretion of GIP, GLP-1 and PYY was sensitive to K+(ATP)-sensitive channel modulators tolbutamide and diazoxide. L-amino acids phenylalanine (Phe), tryptophan (Trp), asparagine (Asn), arginine (Arg) and glutamine (Gln) also stimulated GIP, GLP-1 and PYY secretion, which was completely abolished when extracellular Ca2+ was absent. The gut peptide response stimulated by the amino acids was also blocked by the CasR inhibitor Calhex 231 and augmented by the CasR agonist NPS-R568. GLUT2 and CasR regulate K- and L-cell activity in response to nutrient and non-nutrient stimuli.
Collapse
|
78
|
Uneyama H. Nutritional and physiological significance of luminal glutamate-sensing in the gastrointestinal functions. YAKUGAKU ZASSHI 2012; 131:1699-709. [PMID: 22129863 DOI: 10.1248/yakushi.131.1699] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent evidence indicates that free amino acids are nutrients as well as acting as chemical transmitters within the gastrointestinal tract. Gut glutamate research is the most advanced among 20 amino acids. Free glutamate carries the umami taste sensation on the tongue and a visceral sensation in the gut, especially the stomach. In the field of taste physiology, the physiological meaning of the glutamate-derived chemical sense, the umami taste, has been proposed to be a marker of protein intake. Experimental evidence in gut glutamate physiology strongly supports this hypothesis. Free glutamate is sensed by the abdominal vagus and regulates gastrointestinal functions such as secretion and emptying to accelerate protein digestion. Clinical application of glutamate has also just begun to treat gastrointestinal disorders such as dyspepsia, ulcer, dry mouth and functional dyspepsia. In this review, we introduce recent advances in gut glutamate research and consider the possible contribution of glutamate to health.
Collapse
Affiliation(s)
- Hisayuki Uneyama
- Umami Wellness Research Group, Frontier Research Laboratories, Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan.
| |
Collapse
|
79
|
Amin A, Murphy KG. Nutritional sensing and its utility in treating obesity. Expert Rev Endocrinol Metab 2012; 7:209-221. [PMID: 30764012 DOI: 10.1586/eem.12.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Obesity remains a major worldwide health problem, with current medical treatments being poorly effective. Nutrient sensing allows organs such as the GI tract and the brain to recognize and respond to fuel substrates such as carbohydrates, protein and fats. Specialized neural and hormonal pathways exist to facilitate and regulate these chemosensory mechanisms. Manipulation of factors involved in either central or peripheral chemosensory pathways may provide possible targets for the manipulation of appetite. However, further research is required to assess the utility of this approach to developing novel anti-obesity agents.
Collapse
Affiliation(s)
- Anjali Amin
- a Section of Investigative Medicine, Faculty of Medicine, Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Kevin G Murphy
- b Section of Investigative Medicine, Faculty of Medicine, Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
80
|
Hindlet P, Barraud C, Boschat L, Farinotti R, Bado A, Buyse M. Rosiglitazone and metformin have opposite effects on intestinal absorption of oligopeptides via the proton-dependent PepT1 transporter. Mol Pharmacol 2012; 81:319-27. [PMID: 22108913 DOI: 10.1124/mol.111.073874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The intestinal H(+)/peptide cotransporter 1 (PepT1) plays a major role in nitrogen supply to the body by mediating intestinal absorption of di- and tripeptides. Previous studies have reported that in animal models of type 2 diabetes/obesity, PepT1 activity and expression were markedly reduced. This prompted us to investigate the effects of two antidiabetic drugs, rosiglitazone and metformin, on PepT1 activity/expression in a murine diet-induced obesity model. C57BL/6J male mice were fed a high-fat diet (HFD) or a standard chow for 6 weeks and then were treated for 7 days with metformin (250 mg/kg/day) and/or rosiglitazone (8 mg/kg/day). For in vitro studies, Caco-2 enterocyte-like cells were treated for 7 days with metformin (10 mM) and/or rosiglitazone (10 μM). A 7-day rosiglitazone treatment increased PepT1 activity and prevented the 2-fold HFD-induced reduction in PepT1 transport. Metformin alone did not modify PepT1 activity but counteracted rosiglitazone-induced PepT1-mediated transport. As with the in vivo studies, rosiglitazone treatment up-regulated PepT1 transport activity with concomitant induction of S6 ribosomal protein activation in vitro. Furthermore, metformin decreased PepT1 expression (mRNA and protein) and its transport activity. The effect of metformin was linked to a reduction of phosphorylated S6 ribosomal protein (active form) and of phosphorylated 4E-BP1 (inactive form), a translation repressor. These data demonstrate that two antidiabetic drugs exert opposite effects on the PepT1 transport function probably through direct action on enterocytes. In our type 2 diabetes/obesity model, rosiglitazone, a peroxisome proliferator-activated receptor-γ agonist compensated for the HFD-induced PepT1 down-regulation, whereas metformin reversed rosiglitazone activity at the translational level.
Collapse
Affiliation(s)
- Patrick Hindlet
- Clinical Pharmacy Department (EA4123), Paris-Sud 11 University, Châtenay-Malabry, France.
| | | | | | | | | | | |
Collapse
|
81
|
Zhang CL, Yuan J, Wang Q, Wang YH, Fang XT, Lei CZ, Yang DY, Chen H. The association of bovine T1R family of receptors polymorphisms with cattle growth traits. Res Vet Sci 2012; 93:783-7. [PMID: 22336349 DOI: 10.1016/j.rvsc.2012.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 12/25/2011] [Accepted: 01/20/2012] [Indexed: 11/27/2022]
Abstract
The three members of the T1R class of taste-specific G protein-coupled receptors have been proven to function in combination with heterodimeric sweet and umami taste receptors in many mammals that affect food intake. This may in turn affect growth traits of livestock. We performed a comprehensive evaluation of single-nucleotide polymorphisms (SNPs) in the bovine TAS1R gene family, which encodes receptors for umami and sweet tastes. Complete DNA sequences of TAS1R1-, TAS1R2-, and TAS1R3-coding regions, obtained from 436 unrelated female cattle, representing three breeds (Qinchuan, Jiaxian Red, Luxi), revealed substantial coding and noncoding diversity. A total of nine SNPs in the TAS1R1 gene were identified, among which seven SNPs were in the coding region, and two SNPs were in the introns. All five SNPs in the TAS1R2 gene and all three SNPs in the TAS1R3 gene were identified in the coding region. Four SNPs (TAS1R1 g.5081C>T, TAS1R1 g.5110C>A, TAS1R2 g.288A>G, TAS1R2 g.2552T>C) were significantly associated with body height of Qinchuan cattle (P<0.05). The heterozygous genotypes of the four SNPs showed a molecular heterosis on cattle heights at hip cross and sacra. The individuals with different genotypic combinations of the four SNPs had significant association with heights at hip cross and sacra (P<0.05).
Collapse
Affiliation(s)
- C L Zhang
- Institute of Cellular and Molecular Biology, Xuzhou Normal University, Xuzhou, Jiangsu, PR China
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Kitamura A, Tsurugizawa T, Uematsu A, Torii K, Uneyama H. New therapeutic strategy for amino acid medicine: effects of dietary glutamate on gut and brain function. J Pharmacol Sci 2012; 118:138-44. [PMID: 22293294 DOI: 10.1254/jphs.11r06fm] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
The gustatory and visceral stimulation from food regulates digestion and nutrient utilization, and free glutamate (Glu) release from food is responsible for the umami taste perception that increases food palatability. The results of recent studies reveal a variety of physiological roles for Glu. For example, luminal applications of Glu into the mouth, stomach, and intestine increase the afferent nerve activities of the glossopharyngeal nerve, the gastric branch of the vagus nerve, and the celiac branch of the vagus nerve, respectively. Additionally, luminal Glu evokes efferent nerve activation of each branch of the abdominal vagus nerve. The intragastric administration of Glu activates several brain areas (e.g., insular cortex, limbic system, and hypothalamus) and has been shown to induce flavor-preference learning in rats. Functional magnetic resonance imaging of rats has shown that the intragastric administration of Glu activates the nucleus tractus solitarius, amygdala, and lateral hypothalamus. In addition, Glu may increase flavor preference as a result of its postingestive effect. Considering these results, we propose that dietary Glu functions as a signal for the regulation of the gastrointestinal tract via the gut-brain axis and contributes to the maintenance of a healthy life.
Collapse
|
83
|
Nerveless and gutsy: intestinal nutrient sensing from invertebrates to humans. Semin Cell Dev Biol 2012; 23:614-20. [PMID: 22248674 PMCID: PMC3712190 DOI: 10.1016/j.semcdb.2012.01.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 12/27/2011] [Accepted: 01/04/2012] [Indexed: 12/22/2022]
Abstract
The increasingly recognized role of gastrointestinal signals in the regulation of food intake, insulin production and peripheral nutrient storage has prompted a surge of interest in studying how the gastrointestinal tract senses and responds to nutritional information. Identification of metabolically important intestinal nutrient sensors could provide potential new drug targets for the treatment of diabetes, obesity and gastrointestinal disorders. From a more fundamental perspective, the study of intestinal chemosensation is revealing novel, non-neuronal modes of communication involving differentiated epithelial cells. It is also identifying signalling mechanisms downstream of not only canonical receptors but also nutrient transporters, thereby supporting a chemosensory role for “transceptors” in the intestine. This review describes known and proposed mechanisms of intestinal carbohydrate, protein and lipid sensing, best characterized in mammalian systems. It also highlights the potential of invertebrate model systems such as C. elegans and Drosophila melanogaster by summarizing known examples of molecular evolutionary conservation. Recently developed genetic tools in Drosophila, an emerging model system for the study of physiology and metabolism, allow the temporal, spatial and high-throughput manipulation of putative intestinal sensors. Hence, fruit flies may prove particularly suited to the study of the link between intestinal nutrient sensing and metabolic homeostasis.
Collapse
|
84
|
Abstract
Ingestion of a meal triggers a range of physiological responses both within and outside the gut, and results in the remote modulation of appetite and glucose homeostasis. Luminal contents are sensed by specialised chemosensitive cells scattered throughout the intestinal epithelium. These enteroendocrine and tuft cells make direct contact with the gut lumen and release a range of chemical mediators, which can either act in a paracrine fashion interacting with neighbouring cells and nerve endings or as classical circulating hormones. At the molecular level, the chemosensory machinery involves multiple and complex signalling pathways including activation of G-protein-coupled receptors and solute carrier transporters. This chapter will discuss our current knowledge of the molecular mechanisms underlying intestinal chemosensation with a particular focus on the relatively well-characterised nutrient-triggered secretion from the enteroendocrine system.
Collapse
Affiliation(s)
- Gwen Tolhurst
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0XY, UK
| | | | | |
Collapse
|
85
|
Chevrot M, Martin C, Passilly-Degrace P, Besnard P. Role of CD36 in oral and postoral sensing of lipids. Handb Exp Pharmacol 2012:295-307. [PMID: 22249820 DOI: 10.1007/978-3-642-24716-3_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Obesity and associated plethora of diseases constitute a major public health challenge worldwide. The conjunction of profound changes in our lifestyle and a thrifty genetic that evolved in an environment of food scarcity largely explains this epidemic situation. Food abundance promotes our specific appetite for the more palatable food generally rich in lipids. It is noteworthy that this attraction for fatty food is not specific to humans. Rats and mice also spontaneously prefer lipid-rich food in a free-choice situation. Detection of lipids in food requires the presence of specific sensors located in strategic places (e.g., oral cavity, small intestine, brain) whose activation results in a modulation of the eating behavior. Recent data strongly suggest that the glycoprotein CD36 plays a significant role in this sensing system.
Collapse
Affiliation(s)
- M Chevrot
- Physiologie de la Nutrition, UMR U866 INSERM/Université de Bourgogne, AgroSup Dijon, 1, Esplanade Erasme, 21000 Dijon, France
| | | | | | | |
Collapse
|
86
|
Short-term consumption of sucralose, a nonnutritive sweetener, is similar to water with regard to select markers of hunger signaling and short-term glucose homeostasis in women. Nutr Res 2011; 31:882-8. [DOI: 10.1016/j.nutres.2011.10.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 10/13/2011] [Accepted: 10/14/2011] [Indexed: 01/18/2023]
|
87
|
Treesukosol Y, Smith KR, Spector AC. The functional role of the T1R family of receptors in sweet taste and feeding. Physiol Behav 2011; 105:14-26. [PMID: 21376068 PMCID: PMC3186843 DOI: 10.1016/j.physbeh.2011.02.030] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 02/17/2011] [Accepted: 02/17/2011] [Indexed: 01/02/2023]
Abstract
The discovery of the T1R family of Class C G protein-coupled receptors in the peripheral gustatory system a decade ago has been a tremendous advance for taste research, and its conceptual reach has extended to other organ systems. There are three proteins in the family, T1R1, T1R2, and T1R3, encoded by their respective genes, Tas1r1, Tas1r2, and Tas1r3. T1R2 combines with T1R3 to form a heterodimer that binds with sugars and other sweeteners. T1R3 also combines with T1R1 to form a heterodimer that binds with l-amino acids. These proteins are expressed not only in taste bud cells, but one or more of these T1Rs have also been identified in the nasal epithelium, gut, pancreas, liver, kidney, testes and brain in various mammalian species. Here we review current perspectives regarding the functional role of these receptors, concentrating on sweet taste and feeding. We also discuss behavioral findings suggesting that a glucose polymer mixture, Polycose, which rodents avidly prefer, appears to activate a receptor that does not depend on the combined expression of T1R2 and T1R3. In addition, although the T1Rs have been implicated as playing a role in glucose sensing, T1R2 knock-out (KO) and T1R3 KO mice display normal chow and fluid intake as well as normal body weight compared with same-sex littermate wild type (WT) controls. Moreover, regardless of whether they are fasted or not, these KO mice do not differ from their WT counterparts in their Polycose intake across a broad range of concentrations in 30-minute intake tests. The functional implications of these results and those in the literature are considered.
Collapse
Affiliation(s)
- Yada Treesukosol
- Department of Psychology and Program in Neuroscience, Florida State University 1107 West Call Street, P.O. Box 3064301, Tallahassee FL 32306 USA
| | - Kimberly R. Smith
- Department of Psychology and Program in Neuroscience, Florida State University 1107 West Call Street, P.O. Box 3064301, Tallahassee FL 32306 USA
| | - Alan C. Spector
- Department of Psychology and Program in Neuroscience, Florida State University 1107 West Call Street, P.O. Box 3064301, Tallahassee FL 32306 USA
| |
Collapse
|
88
|
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: 12.3] [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]
|
89
|
Abstract
The sweet taste receptor is expressed in taste cells located in taste buds of the tongue. This receptor senses sweet substances in the oral cavity, activates taste cells, and transmits the taste signals to adjacent neurons. The sweet taste receptor is a heterodimer of two G protein-coupled receptors, T1R2 and T1R3. Recent studies have shown that this receptor is also expressed in the extragustatory system, including the gastrointestinal tract, pancreatic β-cells, and glucose-responsive neurons in the brain. In the intestine, the sweet taste receptor regulates secretion of incretin hormones and glucose uptake from the lumen. In β-cells, activation of the sweet taste receptor leads to stimulation of insulin secretion. Collectively, the sweet taste receptor plays an important role in recognition and metabolism of energy sources in the body.
Collapse
Affiliation(s)
- Itaru Kojima
- Laboratory of Cell Physiology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Yuko Nakagawa
- Laboratory of Cell Physiology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
90
|
Zheng Y, Scow JS, Duenes JA, Sarr MG. Mechanisms of glucose uptake in intestinal cell lines: role of GLUT2. Surgery 2011; 151:13-25. [PMID: 21943636 DOI: 10.1016/j.surg.2011.07.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 07/06/2011] [Indexed: 12/25/2022]
Abstract
BACKGROUND GLUT2 is translocated to the apical membrane of enterocytes exposed to glucose concentrations >∼50 mM. Mechanisms of GLUT2-mediated glucose uptake in cell culture models of enterocytes have not been studied. AIM To explore mechanism(s) of glucose uptake in 3 enterocyte-like cell lines. METHODS Glucose uptake was measured in Caco-2, RIE-1, and IEC-6 cell lines using varying concentrations of glucose (0.5-50 mM). Effects of phlorizin (SGLT1 inhibitor), phloretin (GLUT2 inhibitor), nocodazole and cytochalasin B (disrupters of cytoskeleton), calphostin C and chelerythrine (PKC inhibitors), and phorbol 12-myristate 13-acetate (PKC activator) were evaluated. RESULTS Phlorizin inhibited glucose uptake in all 3 cell lines. Phloretin inhibited glucose uptake in Caco-2 and RIE-1 cells. Starving cells decreased glucose uptake in Caco-2 and RIE-1 cells. Glucose uptake was saturated at >10 mM glucose in all 3 cell lines when exposed briefly (<1 min) to glucose. After exposure for >5 min in Caco-2 and RIE-1 cells, glucose uptake did not saturate and K(m) and V(max) increased. This increase in glucose uptake was inhibited by phloretin, nocodazole, cytochalasin B, calphostin C, and chelerythrine. PMA enhanced glucose uptake by 20%. Inhibitors and PMA had little or no effect in the IEC-6 cells. CONCLUSION Constitutive expression of GLUT2 in the apical membrane along with additional translocation of cytoplasmic GLUT2 to the apical membrane via an intact cytoskeleton and activated PKC appears responsible for enhanced carrier-mediated glucose uptake at greater glucose concentrations (>20 mM) in Caco-2 and RIE-1 cells. IEC-6 cells do not appear to express functional GLUT2.
Collapse
Affiliation(s)
- Ye Zheng
- Department of Surgery and the Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
91
|
Otsubo H, Kondoh T, Shibata M, Torii K, Ueta Y. Induction of Fos expression in the rat forebrain after intragastric administration of monosodium L-glutamate, glucose and NaCl. Neuroscience 2011; 196:97-103. [PMID: 21930190 DOI: 10.1016/j.neuroscience.2011.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 08/29/2011] [Accepted: 09/02/2011] [Indexed: 11/24/2022]
Abstract
l-glutamate, an umami taste substance, is a key molecule coupled to a food intake signaling pathway. Furthermore, recent studies have unveiled new roles for dietary glutamate on gut-brain axis communication via activation of gut glutamate receptors and subsequent vagus nerve. In the present study, we mapped activation sites of the rat forebrain after intragastric load of 60 mM monosodium l-glutamate (MSG) by measurement of Fos protein, a functional marker of neuronal activation. The same concentration of d-glucose (sweet) and NaCl (salty) was used as controls. MSG administration exclusively produced enhanced Fos expression in four hypothalamic regions (the medial preoptic area, lateral hypothalamic area, dorsomedial nucleus, and arcuate nucleus). On the other hand, glucose administration exclusively enhanced Fos induction in the nucleus accumbens. Both MSG and glucose enhanced Fos induction in three brain regions (the habenular nucleus, paraventricular nucleus, and central nucleus of the amygdala). However, MSG induced Fos inductions were more potent than those of glucose in the habenular nucleus and paraventricular nucleus. Importantly, the present study identified for the first time two brain areas (the paraventricular and arcuate hypothalamic nuclei) that are more potently activated by intragastric MSG loads compared with glucose and NaCl. Overall, our results suggest significant activation of a neural network comprising the habenular nucleus, amygdala, and the hypothalamic subnuclei following intragastric load with glutamate.
Collapse
Affiliation(s)
- H Otsubo
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | | | | | | | | |
Collapse
|
92
|
Awad WA, Vahjen W, Aschenbach JR, Zentek J. A diet naturally contaminated with the Fusarium mycotoxin deoxynivalenol (DON) downregulates gene expression of glucose transporters in the intestine of broiler chickens. Livest Sci 2011. [DOI: 10.1016/j.livsci.2011.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
93
|
Mourad FH, Saadé NE. Neural regulation of intestinal nutrient absorption. Prog Neurobiol 2011; 95:149-62. [PMID: 21854830 DOI: 10.1016/j.pneurobio.2011.07.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 04/28/2011] [Accepted: 07/20/2011] [Indexed: 12/17/2022]
Abstract
The nervous system and the gastrointestinal (GI) tract share several common features including reciprocal interconnections and several neurotransmitters and peptides known as gut peptides, neuropeptides or hormones. The processes of digestion, secretion of digestive enzymes and then absorption are regulated by the neuro-endocrine system. Luminal glucose enhances its own absorption through a neuronal reflex that involves capsaicin sensitive primary afferent (CSPA) fibres. Absorbed glucose stimulates insulin release that activates hepatoenteric neural pathways leading to an increase in the expression of glucose transporters. Adrenergic innervation increases glucose absorption through α1 and β receptors and decreases absorption through activation of α2 receptors. The vagus nerve plays an important role in the regulation of diurnal variation in transporter expression and in anticipation to food intake. Vagal CSPAs exert tonic inhibitory effects on amino acid absorption. It also plays an important role in the mediation of the inhibitory effect of intestinal amino acids on their own absorption at the level of proximal or distal segment. However, chronic extrinsic denervation leads to a decrease in intestinal amino acid absorption. Conversely, adrenergic agonists as well as activation of CSPA fibres enhance peptides uptake through the peptide transporter PEPT1. Finally, intestinal innervation plays a minimal role in the absorption of fat digestion products. Intestinal absorption of nutrients is a basic vital mechanism that depends essentially on the function of intestinal mucosa. However, intrinsic and extrinsic neural mechanisms that rely on several redundant loops are involved in immediate and long-term control of the outcome of intestinal function.
Collapse
Affiliation(s)
- Fadi H Mourad
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | | |
Collapse
|
94
|
Kellett GL. Alternative perspective on intestinal calcium absorption: proposed complementary actions of Ca(v)1.3 and TRPV6. Nutr Rev 2011; 69:347-70. [PMID: 21729089 DOI: 10.1111/j.1753-4887.2011.00395.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcellular models of dietary Ca(2+) absorption by the intestine assign essential roles to TRPV6 and calbindin-D(9K) . However, studies with gene-knockout mice challenge this view. Something fundamental is missing. The L-type channel Ca(v) 1.3 is located in the apical membrane from the duodenum to the ileum. In perfused rat jejunum in vivo and in Caco-2 cells, Ca(v) 1.3 mediates sodium glucose transporter 1 (SGLT1)-dependent and prolactin-induced active, transcellular Ca(2+) absorption, respectively. TRPV6 is activated by hyperpolarization and is vitamin D dependent; in contrast, Ca(v) 1.3 is activated by depolarization and is independent of calbindin-D(9K) and vitamin D. This review considers evidence supporting the idea that Ca(v) 1.3 and TRPV6 have complementary roles in the regulation of intestinal Ca(2+) absorption as depolarization and repolarization of the apical membrane occur during and between digestive periods, respectively, and as chyme moves from one intestinal segment to another and food transit times increase. Reassessment of current arguments for paracellular flow reveals that key phenomena have alternative explanations within the integrated Ca(v) 1.3/TRPV6 view of transcellular Ca(2+) absorption.
Collapse
Affiliation(s)
- George L Kellett
- Department of Biology, University of York, Heslington, United Kingdom.
| |
Collapse
|
95
|
Association between TAS2R38 gene polymorphisms and colorectal cancer risk: a case-control study in two independent populations of Caucasian origin. PLoS One 2011; 6:e20464. [PMID: 21674048 PMCID: PMC3107225 DOI: 10.1371/journal.pone.0020464] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 04/25/2011] [Indexed: 01/01/2023] Open
Abstract
Molecular sensing in the lingual mucosa and in the gastro-intestinal tract play a role in the detection of ingested harmful drugs and toxins. Therefore, genetic polymorphisms affecting the capability of initiating these responses may be critical for the subsequent efficiency of avoiding and/or eliminating possible threats to the organism. By using a tagging approach in the region of Taste Receptor 2R38 (TAS2R38) gene, we investigated all the common genetic variation of this gene region in relation to colorectal cancer risk with a case-control study in a German population (709 controls and 602 cases) and in a Czech population (623 controls and 601 cases). We found that there were no significant associations between individual SNPs of the TAS2R38 gene and colorectal cancer in the Czech or in the German population, nor in the joint analysis. However, when we analyzed the diplotypes and the phenotypes we found that the non-taster group had an increased risk of colorectal cancer in comparison to the taster group. This association was borderline significant in the Czech population, (OR = 1.28, 95% CI 0.99–1.67; Pvalue = 0.058) and statistically significant in the German population (OR = 1.36, 95% CI 1.06–1.75; Pvalue = 0.016) and in the joint analysis (OR = 1.34, 95% CI 1.12–1.61; Pvalue = 0.001). In conclusion, we found a suggestive association between the human bitter tasting phenotype and the risk of CRC in two different populations of Caucasian origin.
Collapse
|
96
|
Abstract
This review discusses the role of enteroendocrine cells in the gastrointestinal tract as chemoreceptors that sense intraluminal contents and induce changes in food intake through the release of signalling substances, such as satiety hormones. Recent evidence supports the concept that chemosensing in the gut involves G protein-coupled receptors (GPCRs) that are known to mediate gustatory signals in the oral cavity. GPCRs can be grouped into several families, depending on the stimuli to which they respond, e.g. proteins, amino acids, carbohydrates, fatty acids, or tastants. Sensing of these stimuli by GPCRs results in hormone secretions of enteroendocrine cells, which participate in the control of food intake. A better understanding of the stimuli that induce the strongest binding with these receptors, and thus induce a strong release of hormones, can be a very useful strategy for the development of novel foods in the treatment of obesity.
Collapse
Affiliation(s)
- M C P Geraedts
- Department of Human Biology, Maastricht University Medical Centre +, PO Box 616, 6200 MD Maastricht, The Netherlands.
| | | | | |
Collapse
|
97
|
Scow JS, Tavakkolizadeh A, Zheng Y, Sarr MG. Acute "adaptation" by the small intestinal enterocyte: a posttranscriptional mechanism involving apical translocation of nutrient transporters. Surgery 2011; 149:601-5. [PMID: 21496564 PMCID: PMC3081107 DOI: 10.1016/j.surg.2011.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 02/10/2011] [Indexed: 11/25/2022]
Affiliation(s)
- Jeffrey S. Scow
- Department of Surgery and the Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905
| | - Ali Tavakkolizadeh
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Ye Zheng
- Department of Surgery and the Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905
| | - Michael G. Sarr
- Department of Surgery and the Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
98
|
Powley TL, Spaulding RA, Haglof SA. Vagal afferent innervation of the proximal gastrointestinal tract mucosa: chemoreceptor and mechanoreceptor architecture. J Comp Neurol 2011; 519:644-60. [PMID: 21246548 DOI: 10.1002/cne.22541] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The vagus nerve supplies low-threshold chemo- and mechanosensitive afferents to the mucosa of the proximal gastrointestinal (GI) tract. The absence of a full characterization of the morphology and distributions of these projections has hampered comprehensive functional analyses. In the present experiment, dextran (10K) conjugated with tetramethylrhodamine and biotin was injected into the nodose ganglion and used to label the terminal arbors of individual vagal afferents of both rats and mice. Series of serial 100-μm thick sections of the initial segment of the duodenum as well as the pyloric antrum were collected and processed with diaminobenzidine for permanent tracer labeling. Examination of over 400 isolated afferent fibers, more than 200 from each species, indicated that three vagal afferent specializations, each distinct in morphology and in targets, innervate the mucosa of the proximal GI tract. One population of fibers, the villus afferents, supplies plates of varicose endings to the apical tips of intestinal villi, immediately subjacent to the epithelial wall. A second type of afferent, the crypt afferent, forms subepithelial rings of varicose processes encircling the intestinal glands or crypts, immediately below the crypt-villus junction. Statistical assessment of the isolated fibers indicated that the villus arbors and the crypt endings are independent, issued by different vagal afferents. A third vagal afferent specialization, the antral gland afferent, arborizes along the gastric antral glands and forms terminal concentrations immediately below the luminal epithelial wall. The terminal locations, morphological features, and regional distributions of these three specializations provide inferences about the sensitivities of the afferents.
Collapse
Affiliation(s)
- Terry L Powley
- Purdue University Life Sciences Program and Department of Psychological Sciences, Purdue University, West Lafayette, Indiana 47907-2081, USA.
| | | | | |
Collapse
|
99
|
Young RL. Sensing via intestinal sweet taste pathways. Front Neurosci 2011; 5:23. [PMID: 21519398 PMCID: PMC3080736 DOI: 10.3389/fnins.2011.00023] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Accepted: 02/10/2011] [Indexed: 12/15/2022] Open
Abstract
The detection of nutrients in the gastrointestinal (GI) tract is of fundamental significance to the control of motility, glycemia and energy intake, and yet we barely know the most fundamental aspects of this process. This is in stark contrast to the mechanisms underlying the detection of lingual taste, which have been increasingly well characterized in recent years, and which provide an excellent starting point for characterizing nutrient detection in the intestine. This review focuses on the form and function of sweet taste transduction mechanisms identified in the intestinal tract; it does not focus on sensors for fatty acids or proteins. It examines the intestinal cell types equipped with sweet taste transduction molecules in animals and humans, their location, and potential signals that transduce the presence of nutrients to neural pathways involved in reflex control of GI motility.
Collapse
Affiliation(s)
- Richard L Young
- Discipline of Medicine, School of Medicine, University of Adelaide Adelaide, SA, Australia
| |
Collapse
|
100
|
Abstract
The duodenal mucosa is exposed to endogenous and exogenous chemicals, including acid, CO(2), bile acids and nutrients. Mucosal chemical sensors are necessary to exert physiological responses such as secretion, digestion, absorption, and motility. We propose a mucosal chemosensing system by which luminal chemicals are sensed via mucosal acid sensors and G-protein-coupled receptors. Luminal acid/CO(2) sensing consists of ecto- and cytosolic carbonic anhydrases, epithelial ion transporters, and acid sensors expressed on the afferent nerves in the duodenum. Furthermore, a luminal L-glutamate signal is mediated via mucosal L-glutamate receptors, including metabotropic glutamate receptors and taste receptor 1 family heterodimers, with activation of afferent nerves and cyclooxygenase, whereas luminal Ca(2+) is differently sensed via the calcium-sensing receptor in the duodenum. Recent studies also show the involvement of enteroendocrine G-protein-coupled receptors in bile acid and fatty acid sensing in the duodenum. These luminal chemosensors help activate mucosal defense mechanisms in or- der to maintain the mucosal integrity and physiological responses. Stimulation of luminal chemosensing in the duodenal mucosa may prevent mucosal injury, affect nutrient metabolism, and modulate sensory nerve activity.
Collapse
Affiliation(s)
- Yasutada Akiba
- *Yasutada Akiba, MD, PhD, Bldg 114, Suite 217, West Los Angeles VA Medical Center, 11301 Wilshire Blvd, Los Angeles, CA 90073 (USA), Tel. +1 310 478 3711, Fax +1 310 268 4811, E-Mail
| | | |
Collapse
|