51
|
Hira T, Ogasawara S, Yahagi A, Kamachi M, Li J, Nishimura S, Sakaino M, Yamashita T, Kishino S, Ogawa J, Hara H. Novel Mechanism of Fatty Acid Sensing in Enteroendocrine Cells: Specific Structures in Oxo-Fatty Acids Produced by Gut Bacteria Are Responsible for CCK Secretion in STC-1 Cells via GPR40. Mol Nutr Food Res 2018; 62:e1800146. [DOI: 10.1002/mnfr.201800146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/19/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Tohru Hira
- Research Faculty of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Shono Ogasawara
- Graduate School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Asuka Yahagi
- Graduate School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Minami Kamachi
- Graduate School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Jiaxin Li
- School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Saki Nishimura
- Fundamental Research Laboratory, Research and Development Division; J-Oil Mills, Inc.; Yokohama 230-0053 Japan
| | - Masayoshi Sakaino
- Fundamental Research Laboratory, Research and Development Division; J-Oil Mills, Inc.; Yokohama 230-0053 Japan
| | - Takatoshi Yamashita
- Fundamental Research Laboratory, Research and Development Division; J-Oil Mills, Inc.; Yokohama 230-0053 Japan
| | - Shigenobu Kishino
- Graduate School of Agriculture; Kyoto University; Kyoto 606-8502 Japan
| | - Jun Ogawa
- Graduate School of Agriculture; Kyoto University; Kyoto 606-8502 Japan
| | - Hiroshi Hara
- Research Faculty of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| |
Collapse
|
52
|
Tachibana T, Kadomoto Y, Khan MSI, Makino R, Cline MA. Effect of l-tryptophan and its metabolites on food passage from the crop in chicks. Domest Anim Endocrinol 2018; 64:59-65. [PMID: 29753195 DOI: 10.1016/j.domaniend.2018.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/09/2018] [Accepted: 03/11/2018] [Indexed: 11/27/2022]
Abstract
l-tryptophan (l-Trp), an essential amino acid, is well known as a precursor of 5-hydroxytryptamine (5-HT) and melatonin. In mammals, l-Trp itself has been reported to suppress gastric emptying in mammals. In addition, 5-HT and melatonin are found in the gastrointestinal tract and affect food passage from the digestive tract in mammals. While the function of these factors in mammals is documented, there is little knowledge on their function in the digestive tract of birds. Therefore, the purpose of the present study was to determine if l-Trp and its metabolites affect the crop emptying rate in chicks (Gallus gallus). We also investigated the effects of kynurenic acid (KYNA) and quinolinic acid (QA), which are metabolites of the kynurenine pathway for l-Trp. Oral administration of l-Trp significantly reduced the crop emptying rate in chicks. Among the metabolites, intraperitoneal injection of 5-HT and melatonin significantly reduced the crop emptying rate, whereas KYNA and QA had no effect. The present study suggests that l-Trp, 5-HT, and melatonin inhibit the movement of food in the digestive tract and thereby affect the utilization of nutrients in the diet of chicks.
Collapse
Affiliation(s)
- T Tachibana
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama 790-8566, Japan.
| | - Y Kadomoto
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama 790-8566, Japan
| | - M S I Khan
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon 791-0295, Ehime, Japan
| | - R Makino
- Department of Agrobiological Science, Faculty of Agriculture, Ehime University, Matsuyama 790-8566, Japan
| | - M A Cline
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
53
|
Abstract
Even the simplest animals possess sophisticated systems for sensing and securing nutrients. After all, ensuring adequate nutrition is essential for sustaining life. Once multicellular animals grew too large to be nourished by simple diffusion of nutrients from their environment, they required a digestive system for the absorption and digestion of food. The majority of cells in the digestive tract are enterocytes that are designed to absorb nutrients. However, the digestive tracts of animals ranging from worms to humans contain specialized cells that discriminate between nutrients and nondigestible ingestants. These cells "sense" both the environment within the gut lumen and nutrients as they cross the gut epithelium. This dual sensing is then translated into local signals that regulate the gut epithelium or distant signals through hormones or nerves. This review will discuss how sensors of the gut interact with cells of the epithelium and neurons to regulate epithelial integrity and initiate neural transmission from the gut lumen. © 2017 American Physiological Society. Compr Physiol 8:1019-1030, 2018.
Collapse
Affiliation(s)
- Rodger A Liddle
- Department of Medicine, Duke University and Durham VA Healthcare System, Durham, North Carolina, USA
| |
Collapse
|
54
|
Ueda Y, Iwakura H, Bando M, Doi A, Ariyasu H, Inaba H, Morita S, Akamizu T. Differential role of GPR142 in tryptophan-mediated enhancement of insulin secretion in obese and lean mice. PLoS One 2018; 13:e0198762. [PMID: 29889885 PMCID: PMC5995358 DOI: 10.1371/journal.pone.0198762] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/24/2018] [Indexed: 01/04/2023] Open
Abstract
Tryptophan is reportedly the most potent agonist for GPR142. Glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells are enhanced by GPR142-mediated signal. It is not clear, however, if GPR142-mediated signals is solely attributable to GSIS enhancement after tryptophan load in various pathophysiological settings. This study aims to reveal the significance of GPR142 signaling in tryptophan-mediated GSIS enhancement in normal and obese mice. Tryptophan significantly improved glucose tolerance in both lean and DIO mice, but the extent of improvement was bigger in DIO mice with augmented glucose-stimulated insulin secretion (GSIS) enhancement. The same results were obtained in ob/ob mice. GPR142 deletion almost completely blocked tryptophan actions in lean mice, suggesting that GPR142 signaling was solely responsible for the GSIS enhancement. In obese GPR142KO mice, however, a significant amount of tryptophan effects were still observed. Calcium-sensing receptors (CaSR) are also known to recognize tryptophan as ligand. Expression levels of CaSR were significantly elevated in the pancreas of DIO mice, and CaSR antagonist further blocked tryptophan’s actions in DIO mice with GPR142 deletion. Although GPR142 signaling had a major role in tryptophan recognition for the enhancement of GSIS in lean mice, other pathways including CaSR signaling also had a significant role in obese mice, which seemed to contribute to the augmented enhancement of GSIS by tryptophan in these animals.
Collapse
Affiliation(s)
- Yoko Ueda
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroshi Iwakura
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
- * E-mail:
| | - Mika Bando
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Asako Doi
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroyuki Ariyasu
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hidefumi Inaba
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shuhei Morita
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takashi Akamizu
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
55
|
Kaelberer MM, Bohórquez DV. The now and then of gut-brain signaling. Brain Res 2018; 1693:192-196. [PMID: 29580839 DOI: 10.1016/j.brainres.2018.03.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 01/06/2023]
Abstract
Since their very beginnings, animals had gut sensory epithelial cells. In one of the first multicellular animals, Trichoplax - a literal wandering gut - food sensing and feeding was coordinated by specialized ventral sensor cells. In mammals, including humans, gut epithelial sensor cells (a.k.a enteroendocrine cells) have been recognized for an array of neuropeptides, like ghrelin and cholecystokinin, that modulate hunger or satiety. Indeed, since first described as "clear cells" by Rudfolf Heidenhain (1868), research efforts increasingly focused on their hormone neuropeptides leading to the alphabetical classification of one cell-one hormone (e.g. I-cell synthesizes only cholecystokinin). A recent explosion of molecular tools to study the biology of single cells is expanding the imagination of studies and unveiling intriguing aspects of gut sensory transduction. To mention a few: multimodal sensing, one cell expressing both ghrelin and cholecystokinin-the yin and yang of appetite-, and synapses with nerves. This brief account examines recent advances on gut sensory transduction to highlight how food and bacteria in the gut alter eating.
Collapse
Affiliation(s)
- Melanie M Kaelberer
- Division of Gastroenterology, Department of Medicine, Duke University, #221A, Medical Sciences Research Building 1, 203 Research Drive, Durham, NC, USA
| | - Diego V Bohórquez
- Division of Gastroenterology, Department of Medicine, Duke University, #221A, Medical Sciences Research Building 1, 203 Research Drive, Durham, NC, USA; Department of Neurobiology, Duke University, #221A, Medical Sciences Research Building 1, 203 Research Drive, Durham, NC, USA.
| |
Collapse
|
56
|
Gwynne RM, Ly KDKN, Parry LJ, Bornstein JC. Calcium Sensing Receptors Mediate Local Inhibitory Reflexes Evoked by L-Phenylalanine in Guinea Pig Jejunum. Front Physiol 2017; 8:991. [PMID: 29255423 PMCID: PMC5722837 DOI: 10.3389/fphys.2017.00991] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/17/2017] [Indexed: 12/18/2022] Open
Abstract
Amino acids applied to the mucosa evoke inhibitory reflexes in guinea-pig jejunum, but the receptors involved in sensory transduction are still unclear. One promising candidate is the extracellular calcium sensing receptor (CaSR), which is expressed by mucosal enteroendocrine cells and is preferentially activated by aromatic L-amino acids. We tested this by applying various amino acids to the mucosa and recording the resulting inhibitory junction potentials (IJPs) in nearby circular smooth muscle via intracellular recording. The CaSR is stereospecific and L-Phenylalanine evoked a significantly larger response than D-Phenylalanine when both were applied to the same site. The same pattern was seen with L- and D-Tryptophan, another aromatic amino acid. The CaSR is preferentially activated by aromatic amino acids and responses to L-Leucine and L-Lysine were significantly lower than those to L-Phenylalanine applied to the same site. Responses to L-Phenylalanine were dose-dependently suppressed by blockade of the CaSR with NPS2143, a CaSR antagonist, and mimicked by mucosal application of cinacalcet, a CaSR agonist. Responses to cinacalcet had similar pharmacology to that of responses to L-Phenylalanine, in that each requires both P2 purinoreceptors and 5-HT receptors. L-Glutamate evoked IJPs similar to those produced by L-Phenylalanine and these were depressed by blockade of P2 receptors and 5-HT3 plus 5-HT4 receptors, but NPS2143 was ineffective. The AMPA receptor antagonists DNQX (10 μM) and CNQX (10 μM) reduced IJPs evoked by L-Glutamate by 88 and 79% respectively, but neither BAY367260 (mGluR5 antagonist) nor 2APV (NMDA antagonist) affected IJPs evoked by L-Glutamate. We conclude that local inhibitory reflexes evoked by aromatic L-amino acids in guinea pig jejunum involve activation of CaSRs which triggers release of ATP and 5-HT from the mucosa. L-Glutamate also evokes inhibitory reflexes, via a pathway that does not involve CaSRs. It is likely there are multiple receptors acting as sensory transducers for different luminal amino acids.
Collapse
Affiliation(s)
- Rachel M Gwynne
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Kenny D K N Ly
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Laura J Parry
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Joel C Bornstein
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
57
|
Targeting gastrointestinal nutrient sensing mechanisms to treat obesity. Curr Opin Pharmacol 2017; 37:16-23. [DOI: 10.1016/j.coph.2017.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/20/2017] [Indexed: 12/15/2022]
|
58
|
Liu K, Shen J, Cao Y, Cai C, Yao J. Duodenal infusions of isoleucine influence pancreatic exocrine function in dairy heifers. Arch Anim Nutr 2017; 72:31-41. [DOI: 10.1080/1745039x.2017.1396144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kai Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jing Shen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chuanjiang Cai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
59
|
Abstract
PURPOSE OF REVIEW We report recently published knowledge regarding gut chemosensory mechanisms focusing on nutrient-sensing G protein-coupled receptors (GPCRs) expressed on gut enteroendocrine cells (EECs), tuft cells, and in afferent nerves in the gastroduodenal mucosa and submucosa. RECENT FINDINGS Gene profiling of EECs and tuft cells have revealed expression of a variety of nutrient-sensing GPCRs. The density of EEC and tuft cells is altered by luminal environmental changes that may occur following bypass surgery or in the presence of mucosal inflammation. Some EECs and tuft cells are directly linked to sensory nerves in the subepithelial space. Vagal afferent neurons that innervate the intestinal villi express nutrient receptors, contributing to the regulation of duodenal anion secretion in response to luminal nutrients. Nutrients are also absorbed via specific epithelial transporters. SUMMARY Gastric and duodenal epithelial cells are continually exposed to submolar concentrations of nutrients that activate GPCRs expressed on EECs, tuft cells, and submucosal afferent nerves and are also absorbed through specific transporters, regulating epithelial cell proliferation, gastrointestinal physiological function, and metabolism. The chemical coding and distribution of EECs and tuft cells are keys to the development of GPCR-targeted therapies.
Collapse
|
60
|
Petersen KU. Pepsin and Its Importance for Functional Dyspepsia: Relic, Regulator or Remedy? Dig Dis 2017; 36:98-105. [PMID: 28982106 DOI: 10.1159/000481399] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 09/06/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Functional dyspepsia is a heterogeneous disorder lacking an established therapeutic strategy. Historical treatment attempts with pepsin products were shrugged off, as a simple calculation shows that quantitative substitution is pointless. However, such attempts might have been right for the wrong reason. SUMMARY Today, the role of pepsins is primarily seen in the provision of signalling amino acids (especially phenylalanine and tryptophan) and peptides, which initiate processes promoting digestion. Proteolysis benefits from pepsin variants showing, contrary to common belief, activities of up to a pH value of 5.0. Non-clinical and clinical studies support the view that liberated amino acids produce a variety of direct and indirect effects. Signal chains stimulated by (mostly aromatic) amino acids lead to secretion of gastrin and cholecystokinin (CCK), mediated, respectively, by CCK2 (gastrin) and Ca2+-sensing receptors in the parietal cell, and Ca2+-sensing receptors in the antral and duodenal mucosa. Thus, CCK effects such as secretion of pancreatic enzymes and promotion of gastric accommodation are (also) consequential to peptic liberation of amino acids. Key Message: As functional dyspepsia represents a heterogeneous disorder, it may be intriguing to view pepsin as a potential (although still to be proven) treatment modality, distinguished by a diversity of pro-digestive effects.
Collapse
|
61
|
Whey protein-derived peptide sensing by enteroendocrine cells compared with osteoblast-like cells: Role of peptide length and peptide composition, focussing on products of β-lactoglobulin hydrolysis. Int Dairy J 2017. [DOI: 10.1016/j.idairyj.2017.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
62
|
Alamshah A, Spreckley E, Norton M, Kinsey-Jones JS, Amin A, Ramgulam A, Cao Y, Johnson R, Saleh K, Akalestou E, Malik Z, Gonzalez-Abuin N, Jomard A, Amarsi R, Moolla A, Sargent PR, Gray GW, Bloom SR, Murphy KG. l-phenylalanine modulates gut hormone release and glucose tolerance, and suppresses food intake through the calcium-sensing receptor in rodents. Int J Obes (Lond) 2017; 41:1693-1701. [PMID: 28792489 PMCID: PMC5678004 DOI: 10.1038/ijo.2017.164] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 05/26/2017] [Accepted: 06/21/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE High-protein diets (HPDs) are associated with greater satiety and weight loss than diets rich in other macronutrients. The exact mechanisms by which HPDs exert their effects are unclear. However, evidence suggests that the sensing of amino acids produced as a result of protein digestion may have a role in appetite regulation and satiety. We investigated the effects of l-phenylalanine (L-Phe) on food intake and glucose homeostasis in rodents. METHODS We investigated the effects of the aromatic amino-acid and calcium-sensing receptor (CaSR) agonist l-phenylalanine (L-Phe) on food intake and the release of the gastrointestinal (GI) hormones peptide YY (PYY), glucagon-like peptide-1 (GLP-1) and ghrelin in rodents, and the role of the CaSR in mediating these effects in vitro and in vivo. We also examined the effect of oral l-Phe administration on glucose tolerance in rats. RESULTS Oral administration of l-Phe acutely reduced food intake in rats and mice, and chronically reduced food intake and body weight in diet-induced obese mice. Ileal l-Phe also reduced food intake in rats. l-Phe stimulated GLP-1 and PYY release, and reduced plasma ghrelin, and also stimulated insulin release and improved glucose tolerance in rats. Pharmacological blockade of the CaSR attenuated the anorectic effect of intra-ileal l-Phe in rats, and l-Phe-induced GLP-1 release from STC-1 and primary L cells was attenuated by CaSR blockade. CONCLUSIONS l-Phe reduced food intake, stimulated GLP-1 and PYY release, and reduced plasma ghrelin in rodents. Our data provide evidence that the anorectic effects of l-Phe are mediated via the CaSR, and suggest that l-Phe and the CaSR system in the GI tract may have therapeutic utility in the treatment of obesity and diabetes. Further work is required to determine the physiological role of the CaSR in protein sensing in the gut, and the role of this system in humans.
Collapse
Affiliation(s)
- A Alamshah
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - E Spreckley
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - M Norton
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - J S Kinsey-Jones
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - A Amin
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - A Ramgulam
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Y Cao
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - R Johnson
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - K Saleh
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - E Akalestou
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Z Malik
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - N Gonzalez-Abuin
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - A Jomard
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - R Amarsi
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - A Moolla
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | | | | | - S R Bloom
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - K G Murphy
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| |
Collapse
|
63
|
Chandra R, Hiniker A, Kuo YM, Nussbaum RL, Liddle RA. α-Synuclein in gut endocrine cells and its implications for Parkinson's disease. JCI Insight 2017; 2:92295. [PMID: 28614796 DOI: 10.1172/jci.insight.92295] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/05/2017] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with devastating clinical manifestations. In PD, neuronal death is associated with intracellular aggregates of the neuronal protein α-synuclein known as Lewy bodies. Although the cause of sporadic PD is not well understood, abundant clinical and pathological evidence show that misfolded α-synuclein is found in enteric nerves before it appears in the brain. This suggests a model in which PD pathology originates in the gut and spreads to the central nervous system via cell-to-cell prion-like propagation, such that transfer of misfolded α-synuclein initiates misfolding of native α-synuclein in recipient cells. We recently discovered that enteroendocrine cells (EECs), which are part of the gut epithelium and directly face the gut lumen, also possess many neuron-like properties and connect to enteric nerves. In this report, we demonstrate that α-synuclein is expressed in the EEC line, STC-1, and native EECs of mouse and human intestine. Furthermore, α-synuclein-containing EECs directly connect to α-synuclein-containing nerves, forming a neural circuit between the gut and the nervous system in which toxins or other environmental influences in the gut lumen could affect α-synuclein folding in the EECs, thereby beginning a process by which misfolded α-synuclein could propagate from the gut epithelium to the brain.
Collapse
Affiliation(s)
- Rashmi Chandra
- Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| | | | - Yien-Ming Kuo
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Robert L Nussbaum
- Department of Medicine, UCSF, San Francisco, California, USA.,Invitae Corporation, San Francisco, California, USA
| | - Rodger A Liddle
- Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA.,Duke Institute for Brain Sciences, Durham, North Carolina, USA
| |
Collapse
|
64
|
Bülbül M, Sinen O, Birsen İ, Izgüt-Uysal V. Peripheral apelin-13 administration inhibits gastrointestinal motor functions in rats: The role of cholecystokinin through CCK 1 receptor-mediated pathway. Neuropeptides 2017; 63:91-97. [PMID: 28012561 DOI: 10.1016/j.npep.2016.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/25/2016] [Accepted: 12/11/2016] [Indexed: 12/31/2022]
Abstract
Apelin is the endogenous ligand of the G protein-coupled receptor APJ. The APJ receptor is widely expressed in gastrointestinal (GI) tissues including stomach and small intestine. Apelin administration was shown to induce the release of cholecystokinin (CCK) which is a well-known alimentary hormone with its inhibitory actions on GI motor functions through CCK1 receptors on vagal afferent fibers. We investigated whether; (i) peripherally injected apelin-13 alters GI motor functions, (ii) apelin-induced changes are mediated by APJ receptor or CCK1 receptor and (iii) vagal afferents are involved in inhibitory effects of apelin. Solid gastric emptying (GE) and colon transit (CT) were measured, whereas duodenal phase III-like contractions were recorded in rats administered with apelin-13 (300μg/kg, ip). CCK1 receptor antagonist lorglumide (10mg/kg, ip) or APJ receptor antagonist F13A (300μg/kg, ip) was administered 30min prior to the apelin-13 injections. Vagal afferent denervation was achieved by systemic administration of vanilloid receptor agonist capsaicin (125mg/kg, sc). Apelin-13 administration significantly (p<0.01) increased the CCK level in portal venous plasma samples. Compared with vehicle-treated rats, apelin-13 significantly delayed both GE (p<0.001) and CT (p<0.01). Pretreatment of lorglumide or F13A completely abolished the apelin-13-induced inhibitory effects on GE and CT, moreover, apelin-13 was found ineffective in rats underwent afferent denervation. F13A administration alone significantly accelerated the basal CT. Apelin-13 noticeably disturbed the duodenal fasting motor pattern by impairing phase III-like contractions while increasing the amplitudes of phase II contractions which were prevented by pretreatment of lorglumide and capsaicin. Compared with vehicle-treated rats, lorglumide and capsaicin significantly (p<0.05) reduced the apelin-13-induced increases in phase II motility index. Peripherally administered apelin-13 inhibits GI motor functions through CCK-dependent pathway which appears to be mediated by CCK1 receptors on vagal afferents. Peripheral apelin might contribute to the motility changes occurred in postprandial period.
Collapse
Affiliation(s)
- Mehmet Bülbül
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey.
| | - Osman Sinen
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| | - İlknur Birsen
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| | - Vecihe Izgüt-Uysal
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| |
Collapse
|
65
|
Fakhry J, Wang J, Martins P, Fothergill LJ, Hunne B, Prieur P, Shulkes A, Rehfeld JF, Callaghan B, Furness JB. Distribution and characterisation of CCK containing enteroendocrine cells of the mouse small and large intestine. Cell Tissue Res 2017; 369:245-253. [PMID: 28413860 DOI: 10.1007/s00441-017-2612-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/20/2017] [Indexed: 01/20/2023]
Abstract
There is general consensus that enteroendocrine cells, EEC, containing the enteric hormone cholecystokinin (CCK) are confined to the small intestine and predominate in the duodenum and jejunum. Contrary to this, EEC that express the gene for CCK have been isolated from the large intestine of the mouse and there is evidence for EEC that contain CCK-like immunoreactivity in the mouse colon. However, the human and rat colons do not contain CCK cells. In the current study, we use immunohistochemistry to investigate CCK peptide presence in endocrine cells, PCR to identify cck transcripts and chromatography to identify CCK peptide forms in the mouse small and large intestine. The colocalisation of CCK and 5-HT, hormones that have been hypothesised to derive from cells of different lineages, was also investigated. CCK immunoreactivity was found in EEC throughout the mouse small and large intestine but positive cells were rare in the rectum. Immunoreactive EEC were as common in the caecum and proximal colon as they were in the duodenum and jejunum. CCK gene transcripts were found in the mucosa throughout the intestine but mRNA for gastrin, a hormone that can bind some anti-CCK antibodies, was only found in the stomach and duodenum. Characterisation of CCK peptides of the colon by extraction, chromatographic separation and radioimmunoassay revealed bioactive amidated and sulphated forms, including CCK-8 and CCK-33. Moreover, CCK-containing EEC in the large intestine bound antibodies that target the biologically active sulfated form. Colocalisation of CCK and 5-HT occurred in a proportion of EEC throughout the small intestine and in the caecum but these hormones were not colocalised in the colon, where there was CCK and PYY colocalisation. It is concluded that authentic, biologically active, CCK occurs in EEC of the mouse large intestine.
Collapse
Affiliation(s)
- Josiane Fakhry
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Joyce Wang
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Patricia Martins
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Linda J Fothergill
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Billie Hunne
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Pierre Prieur
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Arthur Shulkes
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, 3084, Australia
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Brid Callaghan
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - John B Furness
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia.
| |
Collapse
|
66
|
Abstract
The lining of the gastrointestinal tract needs to be easily accessible to nutrients and, at the same time, defend against pathogens and chemical challenges. This lining is the largest and most vulnerable surface that faces the outside world. To manage the dual problems of effective nutrient conversion and defence, the gut lining has a sophisticated system for detection of individual chemical entities, pathogenic organisms and their products, and physico-chemical properties of its contents. Detection is through specific receptors that signal to the gut endocrine system, the nervous system, the immune system and local tissue defence systems. These effectors, in turn, modify digestive functions and contribute to tissue defence. Receptors for nutrients include taste receptors for sweet, bitter and savoury, free fatty acid receptors, peptide and phytochemical receptors, that are primarily located on enteroendocrine cells. Hormones released by enteroendocrine cells act locally, through the circulation and via the nervous system, to optimise digestion and mucosal health. Pathogen detection is both through antigen presentation to T-cells and through pattern-recognition receptors (PRRs). Activation of PRRs triggers local tissue defence, for example, by causing release of antimicrobials from Paneth cells. Toxic chemicals, including plant toxins, are sensed and then avoided, expelled or metabolised. It continues to be a major challenge to develop a comprehensive understanding of the integrated responses of the gastrointestinal tract to its luminal contents.
Collapse
|
67
|
|
68
|
Beppu F, Kawamatsu T, Yamatani Y, Nagai T, Yoshinaga K, Mizobe H, Yoshida A, Kubo A, Kanda J, Gotoh N. Comparison of Catabolic Rates of <i>sn</i>-1, <i>sn</i>-2, and <i>sn</i>-3 Fatty Acids in Triacylglycerols Using <sup>13</sup>CO<sub>2</sub> Breath Test in Mice. J Oleo Sci 2017; 66:85-91. [DOI: 10.5650/jos.ess16124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fumiaki Beppu
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology
| | - Takashi Kawamatsu
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology
| | - Yoshio Yamatani
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology
| | | | | | | | | | - Atsushi Kubo
- Department of Ocean Science, Tokyo University of Marine Science and Technology
| | - Jota Kanda
- Department of Ocean Science, Tokyo University of Marine Science and Technology
| | - Naohiro Gotoh
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology
| |
Collapse
|
69
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 367] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
70
|
Stenblom EL, Weström B, Linninge C, Bonn P, Farrell M, Rehfeld JF, Montelius C. Dietary green-plant thylakoids decrease gastric emptying and gut transit, promote changes in the gut microbial flora, but does not cause steatorrhea. Nutr Metab (Lond) 2016; 13:67. [PMID: 27777602 PMCID: PMC5059972 DOI: 10.1186/s12986-016-0128-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/03/2016] [Indexed: 02/08/2023] Open
Abstract
Green-plant thylakoids increase satiety by affecting appetite hormones such as ghrelin, cholecystokinin (CCK) and glucagon-like peptide-1 (GLP-1). The objective of this study was to investigate if thylakoids also affect gastrointestinal (GI) passage and microbial composition. To analyse the effects on GI passage, 16 rats were gavage-fed a control or thylakoid-supplemented high-fat diet (HFD) 30 min before receiving Evans blue. Another 16 rats were fed a control HFD or thylakoid HFD for two weeks prior to the intragastric challenge with Evans blue. The amount of Evans blue in the stomach and the distance of migration in the intestines after 30 min were used as a measurement of gastric emptying and intestinal transit. These were reduced by thylakoid supplementation in the acute study, and however not significantly also after the two-week diet study. The second aim of the study was to investigate if thylakoid-supplementation affects the gut microbiota and amount of faecal fat in healthy human volunteers (n = 34) receiving thylakoid or placebo treatments for three months. Microbiota was analysed using 16S rRNA gene sequencing and qPCR, and faecal fat was extracted by dichloromethane. The total bacteria, and specifically the Bacteriodes fragilis group, were increased by thylakoid treatment versus placebo, while thylakoids did not cause steatorrhea. Dietary supplementation with thylakoids thus affects satiety both via appetite hormones and GI fullness, and affects the microbial composition without causing GI adverse effects such as steatorrhea. This suggests thylakoids as a novel agent in prevention and treatment of obesity.
Collapse
Affiliation(s)
- Eva-Lena Stenblom
- Department of Experimental Medical Science, Appetite Regulation Unit, BMC B11, Lund University, Sölvegatan 19, SE-221 84 Lund, Sweden
| | - Björn Weström
- Department of Biology, Lund University, Sölvegatan 35, SE-223 62 Lund, Sweden
| | - Caroline Linninge
- Department of Food Technology, Engineering and Nutrition, Lund University, Box 124, SE-221 00 Lund, Sweden
| | - Peter Bonn
- Medicinal Chemistry CVMD, AstraZeneca R&D, Mölndal, Sweden
| | - Mary Farrell
- Department of Experimental Medical Science, Appetite Regulation Unit, BMC B11, Lund University, Sölvegatan 19, SE-221 84 Lund, Sweden
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University, DK-2100 Copenhagen, Denmark
| | - Caroline Montelius
- Department of Experimental Medical Science, Appetite Regulation Unit, BMC B11, Lund University, Sölvegatan 19, SE-221 84 Lund, Sweden
| |
Collapse
|
71
|
Simulated GI digestion of dietary protein: Release of new bioactive peptides involved in gut hormone secretion. Food Res Int 2016; 89:382-390. [PMID: 28460928 DOI: 10.1016/j.foodres.2016.08.033] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 12/31/2022]
Abstract
Dietary proteins have been reported to induce a strong feeling of satiety that has been partially explained by gut hormone level increase. Up to date, various protein hydrolysates have demonstrated in vitro and in vivo their potential to stimulate gut hormone secretion related to food intake decrease and their mechanisms of action have just started to be resolved. In this context, this study aimed at identifying new peptide sequences involved in gut hormone secretion released by protein in vitro gastrointestinal digestion. Targeted gut hormones were Cholecystokinin (CCK) and Glucagon-Like Peptide 1 (GLP-1). The activity of DPP-IV was also considered as it strongly modulates GLP-1 action. In a previous study, simulated digestion of dietary protein has generated hydrolysates with enhancing effect on CCK and GLP-1 secretion in STC-1 cells as well as DPP-IV inhibitory properties. Successive purification steps were performed to isolate peptide fractions involved in these bioactivities whose sequence was determined by LC-MS-MS. Three peptide sequences ANVST, TKAVEH and KAAVT were pointed out for their stimulating effects on GLP-1 secretion. The sequence VAAA was isolated for its DPP-IV inhibitory properties. Two peptide groups were strongly involved in CCK release sharing a certain occurrence of aromatic amino acid residues.
Collapse
|
72
|
Complex morphology of gastrin-releasing G-cells in the antral region of the mouse stomach. Cell Tissue Res 2016; 366:301-310. [PMID: 27392412 DOI: 10.1007/s00441-016-2455-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/14/2016] [Indexed: 12/16/2022]
Abstract
Gastrin-releasing enteroendocrine cells (G-cells) are usually described as flask-shaped cells with a large base and a small apical pole, integrated in the epithelium lining the basal region of the antral invaginations in the stomach. By means of a transgenic mouse line in which the enhanced version of GFP is endogenously expressed under the control of a gastrin promoter, we have analyzed the spatial distribution and morphological features of G-cells. We found that G-cells were not only located at the basal region of the invagination but to a lesser extent also at the upper region. Visualization of the entire cellular morphology revealed that G-cells show complex morphologies. Basally located G-cells are roundish-shaped cells which project a prominent apical process towards the lumen and extend basal protrusions containing the hormone gastrin that were frequently found in close proximity to blood vessels and occasionally in the vicinity of nerve fibers. Inspection of G-cells in the upper region of antral invaginations disclosed a novel population of G-cells. These cells have a spindle-like contour and long apical and basal processes which extend vertically along the antral invagination, parallel to the lumen. This G-cell population seems to be in contact with a network of nerve fibers. While the functional role of these untypical G-cells is still elusive, the results of this study provide some useful indications to possible roles of these G-cells.
Collapse
|
73
|
Tang L, Cheng CY, Sun X, Pedicone AJ, Mohamadzadeh M, Cheng SX. The Extracellular Calcium-Sensing Receptor in the Intestine: Evidence for Regulation of Colonic Absorption, Secretion, Motility, and Immunity. Front Physiol 2016; 7:245. [PMID: 27458380 PMCID: PMC4914593 DOI: 10.3389/fphys.2016.00245] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/03/2016] [Indexed: 12/14/2022] Open
Abstract
Different from other epithelia, the intestinal epithelium has the complex task of providing a barrier impeding the entry of toxins, food antigens, and microbes, while at the same time allowing for the transfer of nutrients, electrolytes, water, and microbial metabolites. These molecules/organisms are transported either transcellularly, crossing the apical and basolateral membranes of enterocytes, or paracellularly, passing through the space between enterocytes. Accordingly, the intestinal epithelium can affect energy metabolism, fluid balance, as well as immune response and tolerance. To help accomplish these complex tasks, the intestinal epithelium has evolved many sensing receptor mechanisms. Yet, their roles and functions are only now beginning to be elucidated. This article explores one such sensing receptor mechanism, carried out by the extracellular calcium-sensing receptor (CaSR). In addition to its established function as a nutrient sensor, coordinating food digestion, nutrient absorption, and regulating energy metabolism, we present evidence for the emerging role of CaSR in the control of intestinal fluid homeostasis and immune balance. An additional role in the modulation of the enteric nerve activity and motility is also discussed. Clearly, CaSR has profound effects on many aspects of intestinal function. Nevertheless, more work is needed to fully understand all functions of CaSR in the intestine, including detailed mechanisms of action and specific pathways involved. Considering the essential roles CaSR plays in gastrointestinal physiology and immunology, research may lead to a translational opportunity for the development of novel therapies that are based on CaSR's unique property of using simple nutrients such as calcium, polyamines, and certain amino acids/oligopeptides as activators. It is possible that, through targeting of intestinal CaSR with a combination of specific nutrients, oral solutions that are both inexpensive and practical may be developed to help in conditioning the gut microenvironment and in maintaining digestive health.
Collapse
Affiliation(s)
- Lieqi Tang
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Catherine Y Cheng
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Xiangrong Sun
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Alexandra J Pedicone
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Mansour Mohamadzadeh
- Department of Medicine, Center for Inflammation and Mucosal Immunology, University of Florida Gainesville, FL, USA
| | - Sam X Cheng
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| |
Collapse
|
74
|
Lin HV, Efanov AM, Fang X, Beavers LS, Wang X, Wang J, Gonzalez Valcarcel IC, Ma T. GPR142 Controls Tryptophan-Induced Insulin and Incretin Hormone Secretion to Improve Glucose Metabolism. PLoS One 2016; 11:e0157298. [PMID: 27322810 PMCID: PMC4920590 DOI: 10.1371/journal.pone.0157298] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/26/2016] [Indexed: 11/19/2022] Open
Abstract
GPR142, a putative amino acid receptor, is expressed in pancreatic islets and the gastrointestinal tract, but the ligand affinity and physiological role of this receptor remain obscure. In this study, we show that in addition to L-Tryptophan, GPR142 signaling is also activated by L-Phenylalanine but not by other naturally occurring amino acids. Furthermore, we show that Tryptophan and a synthetic GPR142 agonist increase insulin and incretin hormones and improve glucose disposal in mice in a GPR142-dependent manner. In contrast, Phenylalanine improves in vivo glucose disposal independently of GPR142. Noteworthy, refeeding-induced elevations in insulin and glucose-dependent insulinotropic polypeptide are blunted in Gpr142 null mice. In conclusion, these findings demonstrate GPR142 is a Tryptophan receptor critically required for insulin and incretin hormone regulation and suggest GPR142 agonists may be effective therapies that leverage amino acid sensing pathways for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Hua V. Lin
- Lilly China Research and Development Center (LCRDC), Eli Lilly & Company, Shanghai, China
- * E-mail:
| | - Alexander M. Efanov
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly & Company, Indianapolis, Indiana, United States of America
| | - Xiankang Fang
- Lilly China Research and Development Center (LCRDC), Eli Lilly & Company, Shanghai, China
| | - Lisa S. Beavers
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly & Company, Indianapolis, Indiana, United States of America
| | - Xuesong Wang
- Lilly China Research and Development Center (LCRDC), Eli Lilly & Company, Shanghai, China
| | - Jingru Wang
- Lilly China Research and Development Center (LCRDC), Eli Lilly & Company, Shanghai, China
| | - Isabel C. Gonzalez Valcarcel
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly & Company, Indianapolis, Indiana, United States of America
| | - Tianwei Ma
- Lilly China Research and Development Center (LCRDC), Eli Lilly & Company, Shanghai, China
| |
Collapse
|
75
|
The calcium-sensing receptor and the hallmarks of cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1398-407. [DOI: 10.1016/j.bbamcr.2015.11.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
|
76
|
Alamshah A, McGavigan AK, Spreckley E, Kinsey-Jones JS, Amin A, Tough IR, O'Hara HC, Moolla A, Banks K, France R, Hyberg G, Norton M, Cheong W, Lehmann A, Bloom SR, Cox HM, Murphy KG. L-arginine promotes gut hormone release and reduces food intake in rodents. Diabetes Obes Metab 2016; 18:508-18. [PMID: 26863991 PMCID: PMC4982043 DOI: 10.1111/dom.12644] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/31/2016] [Accepted: 02/07/2016] [Indexed: 12/14/2022]
Abstract
AIMS To investigate the anorectic effect of L-arginine (L-Arg) in rodents. METHODS We investigated the effects of L-Arg on food intake, and the role of the anorectic gut hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), the G-protein-coupled receptor family C group 6 member A (GPRC6A) and the vagus nerve in mediating these effects in rodents. RESULTS Oral gavage of L-Arg reduced food intake in rodents, and chronically reduced cumulative food intake in diet-induced obese mice. Lack of the GPRC6A in mice and subdiaphragmatic vagal deafferentation in rats did not influence these anorectic effects. L-Arg stimulated GLP-1 and PYY release in vitro and in vivo. Pharmacological blockade of GLP-1 and PYY receptors did not influence the anorectic effect of L-Arg. L-Arg-mediated PYY release modulated net ion transport across the gut mucosa. Intracerebroventricular (i.c.v.) and intraperitoneal (i.p.) administration of L-Arg suppressed food intake in rats. CONCLUSIONS L-Arg reduced food intake and stimulated gut hormone release in rodents. The anorectic effect of L-Arg is unlikely to be mediated by GLP-1 and PYY, does not require GPRC6A signalling and is not mediated via the vagus. I.c.v. and i.p. administration of L-Arg suppressed food intake in rats, suggesting that L-Arg may act on the brain to influence food intake. Further work is required to determine the mechanisms by which L-Arg suppresses food intake and its utility in the treatment of obesity.
Collapse
MESH Headings
- Animals
- Appetite Depressants/administration & dosage
- Appetite Depressants/adverse effects
- Appetite Depressants/pharmacology
- Appetite Depressants/therapeutic use
- Arginine/administration & dosage
- Arginine/adverse effects
- Arginine/therapeutic use
- Cells, Cultured
- Dietary Supplements/adverse effects
- Energy Intake/drug effects
- Energy Metabolism/drug effects
- Gastrointestinal Agents/administration & dosage
- Gastrointestinal Agents/adverse effects
- Gastrointestinal Agents/pharmacology
- Gastrointestinal Agents/therapeutic use
- Glucagon-Like Peptide 1/agonists
- Glucagon-Like Peptide 1/blood
- Glucagon-Like Peptide 1/metabolism
- In Vitro Techniques
- Injections, Intraperitoneal
- Injections, Intraventricular
- Intestinal Mucosa/cytology
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/diet therapy
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/pathology
- Peptide YY/agonists
- Peptide YY/blood
- Peptide YY/metabolism
- Random Allocation
- Rats, Wistar
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Weight Loss/drug effects
Collapse
Affiliation(s)
- A Alamshah
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - A K McGavigan
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - E Spreckley
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - J S Kinsey-Jones
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - A Amin
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - I R Tough
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - H C O'Hara
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - A Moolla
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - K Banks
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - R France
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - G Hyberg
- AstraZeneca R&D, Mölndal, Sweden
| | - M Norton
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - W Cheong
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - A Lehmann
- AstraZeneca R&D, Mölndal, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - S R Bloom
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - H M Cox
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - K G Murphy
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
77
|
Liu J, Yu K, Zhu W. Amino acid sensing in the gut and its mediation in gut-brain signal transduction. ACTA ACUST UNITED AC 2016; 2:69-73. [PMID: 29767064 PMCID: PMC5940996 DOI: 10.1016/j.aninu.2016.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 11/28/2022]
Abstract
Animal gastrointestinal tract is not only a digestive organ, but also a nutrient sensing organ which detects luminal nutrient and thus can regulate food intake. There are many amino acid sensing receptors and transporters in the gut. Amino acids sensing by these receptors and transporters can stimulate the intestinal endocrine cells to release a variety of gut hormones. These hormones trigger a series of physiological effects via the nerve system. This review summarized the recent advance on the amino acid sensing receptors and transporters in the gastrointestinal tract, the gut hormones released from the intestinal endocrine cells and the hormones-induced signal transduction between the gut and brain. A better understanding of these processes may help to gain further insight into the specific role of amino acids in digestion and provide guidelines in developing strategy for the better use of amino acids in the diet.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
78
|
Potential roles for calcium-sensing receptor (CaSR) and transient receptor potential ankyrin-1 (TRPA1) in murine anorectic response to deoxynivalenol (vomitoxin). Arch Toxicol 2016; 91:495-507. [PMID: 26979077 DOI: 10.1007/s00204-016-1687-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/24/2016] [Indexed: 12/26/2022]
Abstract
Food contamination by the trichothecene mycotoxin deoxynivalenol (DON, vomitoxin) has the potential to adversely affect animal and human health by suppressing food intake and impairing growth. In mice, the DON-induced anorectic response results from aberrant satiety hormone secretion by enteroendocrine cells (EECs) of the gastrointestinal tract. Recent in vitro studies in the murine STC-1 EEC model have linked DON-induced satiety hormone secretion to activation of calcium-sensing receptor (CaSR), a G-coupled protein receptor, and transient receptor potential ankyrin-1 (TRPA1), a TRP channel. However, it is unknown whether similar mechanisms mediate DON's anorectic effects in vivo. Here, we tested the hypothesis that DON-induced food refusal and satiety hormone release in the mouse are linked to activation of CaSR and TRPA1. Oral treatment with selective agonists for CaSR (R-568) or TRPA1 (allyl isothiocyanate (AITC)) suppressed food intake in mice, and the agonist's effects were suppressed by pretreatment with corresponding antagonists NPS-2143 or ruthenium red (RR), respectively. Importantly, NPS-2143 or RR inhibited both DON-induced food refusal and plasma elevations of the satiety hormones cholecystokinin (CCK) and peptide YY3-36 (PYY3-36); cotreatment with both antagonists additively suppressed both anorectic and hormone responses to DON. Taken together, these in vivo data along with prior in vitro findings support the contention that activation of CaSR and TRPA1 contributes to DON-induced food refusal by mediating satiety hormone exocytosis from EEC.
Collapse
|
79
|
Linnemann AK, Davis DB. Glucagon-like peptide-1 and cholecystokinin production and signaling in the pancreatic islet as an adaptive response to obesity. J Diabetes Investig 2016; 7 Suppl 1:44-9. [PMID: 27186355 PMCID: PMC4854504 DOI: 10.1111/jdi.12465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022] Open
Abstract
Precise control of blood glucose is dependent on adequate β‐cell mass and function. Thus, reductions in β‐cell mass and function lead to insufficient insulin production to meet demand, and result in diabetes. Recent evidence suggests that paracrine signaling in the islet might be important in obesity, and disruption of this signaling could play a role in the pathogenesis of diabetes. For example, we recently discovered a novel islet incretin axis where glucagon‐like peptide‐1 regulates β‐cell production of another classic gut hormone, cholecystokinin. This axis is stimulated by obesity, and plays a role in enhancing β‐cell survival. In the present review, we place our observations in the wider context of the literature on incretin regulation in the islet, and discuss the potential for therapeutic targeting of these pathways.
Collapse
Affiliation(s)
- Amelia K Linnemann
- Department of Medicine Division of Endocrinology University of Wisconsin-Madison Madison Wisconsin USA
| | - Dawn Belt Davis
- Department of MedicineDivision of EndocrinologyUniversity of Wisconsin-MadisonMadisonWisconsinUSA; William S Middleton Memorial Veterans HospitalMadisonWisconsinUSA
| |
Collapse
|
80
|
Cheng SX. Calcium-sensing receptor: A new target for therapy of diarrhea. World J Gastroenterol 2016; 22:2711-2724. [PMID: 26973410 PMCID: PMC4777994 DOI: 10.3748/wjg.v22.i9.2711] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/18/2015] [Accepted: 12/08/2015] [Indexed: 02/06/2023] Open
Abstract
Management of acute diarrhea remains a global challenge, particularly in resource-limiting countries. Oral rehydration solution (ORS), a passive rehydrating therapy developed approximately 40 years ago, remains the mainstay treatment. Although ORS is effective for hydration, since it does not inhibit enterotoxin-mediated excessive secretion, reduced absorption and compromised barrier function - the primary mechanisms of diarrhea, ORS does not offer a rapid relief of diarrhea symptom. There are a few alternative therapies available, yet the use of these drugs is limited by their expense, lack of availability and/or safety concerns. Novel anti-diarrheal therapeutic approaches, particularly those simple affordable therapies, are needed. This article explores intestinal calcium-sensing receptor (CaSR), a newly uncovered target for therapy of diarrhea. Unlike others, targeting this host antidiarrheal receptor system appears “all-inclusive”: it is anti-secretory, pro-absorptive, anti-motility, and anti-inflammatory. Thus, activating CaSR reverses changes of both secretory and inflammatory diarrheas. Considering its unique property of using simple nutrients such as calcium, polyamines, and certain amino acids/oligopeptides as activators, it is possible that through targeting of CaSR with a combination of specific nutrients, novel oral rehydrating solutions that are inexpensive and practical to use in all countries may be developed.
Collapse
|
81
|
Abstract
Glucagon like peptide-1 is an insulinotropic hormone released from intestinal L-cells in response to food ingestion. Here, we investigated mechanisms underlying the sensing of peptones by primary small intestinal L-cells. Meat, casein and vegetable-derived peptones (5 mg/ml), the L-amino acids Phe, Trp, Gln and Ala (20 mM each), and the dipeptide glycine-sarcosine (20 mM) stimulated GLP-1 secretion from primary cultures prepared from the small intestine. Further mechanistic studies were performed with meat peptone, and revealed the elevation of intracellular calcium in L-cells. Inhibition of the calcium sensing receptor (CaSR), transient receptor potential (TRP) channels and Q-type voltage gated calcium channels (VGCC) significantly attenuated peptone-stimulated GLP-1 release and reduced intracellular Ca(2+) responses. CaSR inhibition also attenuated the GLP-1 secretory response to Gln. Targeting these pathways in L-cells could be used to increase endogenous production of GLP-1 and offer exploitable avenues for the development of therapeutics to treat diabetes and obesity.
Collapse
Affiliation(s)
- Ramona Pais
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK.
| |
Collapse
|
82
|
Abstract
The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon like peptide-1 (GLP-1) are secreted from enteroendocrine cells in the gut and regulate physiological and homeostatic functions related to glucose control, metabolism and food intake. This review provides a systematic summary of the molecular mechanisms underlying secretion from incretin cells, and an understanding of how they sense and interact with lumen and vascular factors and the enteric nervous system through transporters and G-protein coupled receptors (GPCRs) present on their surface to ultimately culminate in hormone release. Some of the molecules described below such as sodium coupled glucose transporter 1 (SGLT1), G-protein coupled receptor (GPR) 119 and GPR40 are targets of novel therapeutics designed to enhance endogenous gut hormone release. Synthetic ligands at these receptors aimed at treating obesity and type 2 diabetes are currently under investigation.
Collapse
Affiliation(s)
- Ramona Pais
- The Wellcome Trust–MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Fiona M. Gribble
- The Wellcome Trust–MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes’s Hospital, Box 289, Hills Road, Cambridge, CB2 0QQ, UK
| | | |
Collapse
|
83
|
Posovszky C, Wabitsch M. Regulation of appetite, satiation, and body weight by enteroendocrine cells. Part 1: characteristics of enteroendocrine cells and their capability of weight regulation. Horm Res Paediatr 2015; 83:1-10. [PMID: 25471008 DOI: 10.1159/000368898] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 09/23/2014] [Indexed: 11/19/2022] Open
Abstract
The gastrointestinal tract is the gateway for food in our body. Food ingestion and the ensuing digestive processes depend on the composition and amount of ingested nutrients. This complex process of nutrient digestion and absorption is effectively regulated by the enteroendocrine system. Enteroendocrine cells (EECs) reside scattered throughout the intestinal epithelium. They express nutrient receptors that face the lumen and secrete peptide hormones in response to food. Besides regulating digestion, gastrointestinal endocrine cells are involved in the regulation of appetite and satiety. The first part of this review describes the anatomical and biological characteristics of EECs and discusses the capability of their hormones to influence appetite, satiety, and body weight. In the second part, we then discuss the therapeutic potential of EECs in the treatment of obesity.
Collapse
Affiliation(s)
- Carsten Posovszky
- University Outpatient Clinic for Pediatric Gastroenterology, and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | |
Collapse
|
84
|
Mine Y, Zhang H. Calcium-sensing receptor (CaSR)-mediated anti-inflammatory effects of L-amino acids in intestinal epithelial cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:9987-9995. [PMID: 26551350 DOI: 10.1021/acs.jafc.5b03749] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Calcium-sensing receptor (CaSR) plays an essential role in sensing nutrients and monitoring ion balance in the human gut. However, no discovery of CaSR-mediated anti-inflammatory effect of l-amino acids (l-AAs) on the gut system has been reported. The aim of this study is to screen and identify the anti-inflammatory activity of various l-AAs in intestinal epithelial cells (IECs) and stepwise illustrate a possible molecular mechanism for anti-inflammation. We used Caco-2 and HT-29 cell lines to evaluate the anti-inflammatory activity of l-AAs and revealed that l-tryptophan (l-Trp) and l-valine (l-Val) have strong anti-inflammatory activity consistent in both cell lines. l-Trp treatment (5 mM) reduced TNF-α-induced IL-8 secretion from HT-29 or Caco-2 cells to about 50 or 40%, respectively. l-Trp also significantly inhibited the expression of phosphorylation of JNK or IκBα to around 50% in HT-29 cells. However, the above inhibitory effects of l-Trp on inflammatory responses in TNF-α-induced HT-29 cells were abrogated by NPS-2143. The result of CaSR antagonist NPS-2143 pretreatment study suggests l-Trp exerts anti-inflammatory effects on IECs through CaSR activation. The involvement of β-arrestin2 was then found to block tumor necrosis factor (TNF)-α-induced signaling pathways after CaSR activated by l-Trp. These results validate a novel mechanism underlying CaSR agonistic l-AAs exerting anti-inflammatory effects on human intestinal epithelia.
Collapse
Affiliation(s)
- Yoshinori Mine
- Department of Food Science, University of Guelph , Guelph, Ontario N1G 2W1, Canada
| | - Hua Zhang
- Department of Food Science, University of Guelph , Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
85
|
Bohórquez D, Haque F, Medicetty S, Liddle RA. Correlative Confocal and 3D Electron Microscopy of a Specific Sensory Cell. J Vis Exp 2015:e52918. [PMID: 26273796 PMCID: PMC4544857 DOI: 10.3791/52918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Delineation of a cell's ultrastructure is important for understanding its function. This can be a daunting project for rare cell types diffused throughout tissues made of diverse cell types, such as enteroendocrine cells of the intestinal epithelium. These gastrointestinal sensors of food and bacteria have been difficult to study because they are dispersed among other epithelial cells at a ratio of 1:1,000. Recently, transgenic reporter mice have been generated to identify enteroendocrine cells by means of fluorescence. One of those is the peptide YY-GFP mouse. Using this mouse, we developed a method to correlate confocal and serial block-face scanning electron microscopy. We named the method cocem3D and applied it to identify a specific enteroendocrine cell in tissue and unveil the cell's ultrastructure in 3D. The resolution of cocem3D is sufficient to identify organelles as small as secretory vesicles and to distinguish cell membranes for volume rendering. Cocem3D can be easily adapted to study the 3D ultrastructure of other specific cell types in their native tissue.
Collapse
Affiliation(s)
| | - Fariha Haque
- Department of Chemistry, University of North Carolina - Chapel Hill
| | | | | |
Collapse
|
86
|
Kinsey-Jones JS, Alamshah A, McGavigan AK, Spreckley E, Banks K, Cereceda Monteoliva N, Norton M, Bewick GA, Murphy KG. GPRC6a is not required for the effects of a high-protein diet on body weight in mice. Obesity (Silver Spring) 2015; 23:1194-200. [PMID: 25958858 PMCID: PMC4692088 DOI: 10.1002/oby.21083] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/19/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The G-protein coupled receptor family C group 6 member A (GPRC6A) is activated by proteinogenic amino acids and may sense amino acids in the gastrointestinal tract and the brain. The study investigated whether GPRC6A was necessary for the effects of low- and high-protein diets on body weight and food intake in mice. METHODS The role of GPRC6A in mediating the effects of a low-protein diet on body weight was investigated in GPRC6a knockout (GPRC6a-KO) and wild-type (WT) mice fed a control diet (18% protein) or a low-protein diet (6% protein) for 9 days. The role of GPRC6A in mediating the effects of a high-protein diet on body weight was investigated in GPRC6a-KO and WT mice fed a control diet (18% protein) or a high-protein diet (50% protein) for 5 weeks. RESULTS A high-protein diet reduced body weight gain and food intake compared with a control diet in both WT and GPRC6a-KO mice. A low-protein diet decreased body weight gain in GPRC6a-KO mice. CONCLUSIONS GPRC6A was not necessary for the effects of a low- or high-protein diet on body weight and likely does not play a role in protein-induced satiety.
Collapse
Affiliation(s)
- James S Kinsey-Jones
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Amin Alamshah
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Anne K McGavigan
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Eleanor Spreckley
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Katherine Banks
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Nicholas Cereceda Monteoliva
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Mariana Norton
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Gavin A Bewick
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
- Division of Diabetes and Nutritional Sciences, King's College London, Guy's Campus, London, UK
| | - Kevin G Murphy
- Section of Investigative Medicine, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
87
|
Linnemann AK, Neuman JC, Battiola TJ, Wisinski JA, Kimple ME, Davis DB. Glucagon-Like Peptide-1 Regulates Cholecystokinin Production in β-Cells to Protect From Apoptosis. Mol Endocrinol 2015; 29:978-87. [PMID: 25984632 DOI: 10.1210/me.2015-1030] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cholecystokinin (CCK) is a classic gut hormone that is also expressed in the pancreatic islet, where it is highly up-regulated with obesity. Loss of CCK results in increased β-cell apoptosis in obese mice. Similarly, islet α-cells produce increased amounts of another gut peptide, glucagon-like peptide 1 (GLP-1), in response to cytokine and nutrient stimulation. GLP-1 also protects β-cells from apoptosis via cAMP-mediated mechanisms. Therefore, we hypothesized that the activation of islet-derived CCK and GLP-1 may be linked. We show here that both human and mouse islets secrete active GLP-1 as a function of body mass index/obesity. Furthermore, GLP-1 can rapidly stimulate β-cell CCK production and secretion through direct targeting by the cAMP-modulated transcription factor, cAMP response element binding protein (CREB). We find that cAMP-mediated signaling is required for Cck expression, but CCK regulation by cAMP does not require stimulatory levels of glucose or insulin secretion. We also show that CREB directly targets the Cck promoter in islets from obese (Leptin(ob/ob)) mice. Finally, we demonstrate that the ability of GLP-1 to protect β-cells from cytokine-induced apoptosis is partially dependent on CCK receptor signaling. Taken together, our work suggests that in obesity, active GLP-1 produced in the islet stimulates CCK production and secretion in a paracrine manner via cAMP and CREB. This intraislet incretin loop may be one mechanism whereby GLP-1 protects β-cells from apoptosis.
Collapse
Affiliation(s)
- Amelia K Linnemann
- Department of Medicine (A.K.L., T.J.B., J.A.W., M.E.K., D.B.D.), Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Nutritional Sciences (J.C.N.), University of Wisconsin-Madison, Madison, Wisconsin 53706; and William S. Middleton Memorial Veterans Hospital (D.B.D.), Madison, Wisconsin 53705
| | - Joshua C Neuman
- Department of Medicine (A.K.L., T.J.B., J.A.W., M.E.K., D.B.D.), Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Nutritional Sciences (J.C.N.), University of Wisconsin-Madison, Madison, Wisconsin 53706; and William S. Middleton Memorial Veterans Hospital (D.B.D.), Madison, Wisconsin 53705
| | - Therese J Battiola
- Department of Medicine (A.K.L., T.J.B., J.A.W., M.E.K., D.B.D.), Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Nutritional Sciences (J.C.N.), University of Wisconsin-Madison, Madison, Wisconsin 53706; and William S. Middleton Memorial Veterans Hospital (D.B.D.), Madison, Wisconsin 53705
| | - Jaclyn A Wisinski
- Department of Medicine (A.K.L., T.J.B., J.A.W., M.E.K., D.B.D.), Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Nutritional Sciences (J.C.N.), University of Wisconsin-Madison, Madison, Wisconsin 53706; and William S. Middleton Memorial Veterans Hospital (D.B.D.), Madison, Wisconsin 53705
| | - Michelle E Kimple
- Department of Medicine (A.K.L., T.J.B., J.A.W., M.E.K., D.B.D.), Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Nutritional Sciences (J.C.N.), University of Wisconsin-Madison, Madison, Wisconsin 53706; and William S. Middleton Memorial Veterans Hospital (D.B.D.), Madison, Wisconsin 53705
| | - Dawn Belt Davis
- Department of Medicine (A.K.L., T.J.B., J.A.W., M.E.K., D.B.D.), Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Department of Nutritional Sciences (J.C.N.), University of Wisconsin-Madison, Madison, Wisconsin 53706; and William S. Middleton Memorial Veterans Hospital (D.B.D.), Madison, Wisconsin 53705
| |
Collapse
|
88
|
Tang L, Peng M, Liu L, Chang W, Binder HJ, Cheng SX. Calcium-sensing receptor stimulates Cl(-)- and SCFA-dependent but inhibits cAMP-dependent HCO3(-) secretion in colon. Am J Physiol Gastrointest Liver Physiol 2015; 308:G874-83. [PMID: 25792563 PMCID: PMC4437021 DOI: 10.1152/ajpgi.00341.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 03/10/2015] [Indexed: 02/07/2023]
Abstract
Colonic bicarbonate (HCO3(-)) secretion is a well-established physiological process that is closely linked to overall fluid and electrolyte movement in the mammalian colon. These present studies show that extracellular calcium-sensing receptor (CaSR), a fundamental mechanism for sensing and regulating ionic and nutrient compositions of extracellular milieu in the small and large intestine, regulates HCO3(-) secretion. Basal and induced HCO3(-) secretory responses to CaSR agonists were determined by pH stat techniques used in conjunction with short-circuit current measurements in mucosa from rat distal colon mounted in Ussing chambers. R568, a specific CaSR activator, stimulated lumen Cl(-)- and short-chain fatty acid (SCFA)-dependent HCO3(-) secretion but inhibited cyclic nucleotide-activated HCO3(-) secretion. Consequently, at physiological conditions (either at basal or during lumen acid challenge) when electroneutral Cl(-)/HCO3(-) and SCFA/HCO3(-) exchangers dominate, CaSR stimulates HCO3(-) secretion; in contrast, in experimental conditions that stimulate fluid and HCO3(-) secretion, e.g., when forskolin activates electrogenic cystic fibrosis transmembrane conductance regulator-mediated HCO3(-) conductance, CaSR activation inhibits HCO3(-) secretion. Corresponding changes in JHCO3 (μeq·h(-1)·cm(-2), absence vs. presence of R568) were 0.18 ± 0.03 vs. 0.31 ± 0.08 under basal nonstimulated conditions and 1.85 ± 0.23 vs. 0.45 ± 0.06 under forskolin-stimulated conditions. Similarly, activation of CaSR by R568 stimulated Cl(-)- and SCFA-dependent HCO3(-) secretion and inhibited cAMP-dependent HCO3(-) secretion in colon mucosa of wild-type mice; such effects were abolished in CaSR-null mice. These results suggest a new paradigm for regulation of intestinal ion transport in which HCO3(-) secretion may be fine-tuned by CaSR in accordance with nutrient availability and state of digestion and absorption. The ability of CaSR agonists to inhibit secretagogue-induced intestinal HCO3(-) secretion suggests that modulation of CaSR activity may provide a new therapeutic approach to correct HCO3(-) deficit and metabolic acidosis, a primary cause of morbidity and mortality in acute infectious diarrheal illnesses.
Collapse
Affiliation(s)
- Lieqi Tang
- 1Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, Florida;
| | - Minzhi Peng
- 1Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, Florida;
| | - Li Liu
- 2Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, People's Republic of China;
| | - Wenhan Chang
- 3Endocrine Research, VA Medical Center, University of California at San Francisco, San Francisco, California;
| | - Henry J. Binder
- 4Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Sam X. Cheng
- 1Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, Florida;
| |
Collapse
|
89
|
Abstract
The gastrointestinal (GI) tract comprises a large endocrine organ that regulates not only nutrient sensing and metabolising but also satiety and energy homeostasis. More than 20 hormones secreted from the stomach, intestine, and pancreas as well as signaling mediators of the gut microbiome are involved in this process. A better understanding of how related pathways affect body weight and food intake will help us to find new strategies and drugs to treat obesity. For example, weight loss secondary to lifestyle intervention is often accompanied by unfavorable changes in multiple GI hormones, which may cause difficulties in maintaining a lower body weight status. Conversely, bariatric surgery favorably changes the hormone profile to support improved satiety and metabolic function. This partially explains stronger sustained body weight reduction resulting in better long-term results of improved metabolic functions. This review focuses on GI hormones and signaling mediators of the microbiome involved in satiety regulation and energy homeostasis and summarizes their changes following weight loss. Furthermore, the potential role of GI hormones as anti-obesity drugs is discussed.
Collapse
Affiliation(s)
- Thomas Reinehr
- Vestische Hospital for Children and Adolescents Datteln, Institute for Pediatric Endocrinology, Diabetes and Nutrition Medicine, University of Witten/Herdecke, Datteln, Germany,
| | | |
Collapse
|
90
|
Leach K, Conigrave AD, Sexton PM, Christopoulos A. Towards tissue-specific pharmacology: insights from the calcium-sensing receptor as a paradigm for GPCR (patho)physiological bias. Trends Pharmacol Sci 2015; 36:215-25. [PMID: 25765207 DOI: 10.1016/j.tips.2015.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 12/25/2022]
Abstract
The calcium-sensing receptor (CaSR) is a widely expressed G protein-coupled receptor (GPCR) that mediates numerous tissue-specific functions. Its multiple ligands and diverse roles attest to the need for exquisite control over the signaling pathways that mediate its effects. 'Biased signaling' is the phenomenon by which distinct ligands stabilize preferred receptor signaling states. The CaSR is subject to biased signaling in response to its endogenous ligands. Interestingly, the 'natural' bias of the CaSR is altered in disease states, and small molecule drugs engender biased allosteric modulation of downstream signaling pathways. Thus, biased signaling from the CaSR also has important implications pathophysiologically and therapeutically. As outlined in this review, this novel paradigm extends to other GPCRs, making the CaSR a model for studies of ligand-biased signaling and for understanding how it may be used to foster selective drug activity in different tissues.
Collapse
Affiliation(s)
- Katie Leach
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville VIC, Australia.
| | - Arthur D Conigrave
- School of Molecular Bioscience, Charles Perkins Centre, University of Sydney, NSW 2006, Australia
| | - Patrick M Sexton
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville VIC, Australia
| | - Arthur Christopoulos
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville VIC, Australia
| |
Collapse
|
91
|
Abstract
The intestinal production of lipoproteins is one of the key processes by which the body prepares dietary lipid for dissemination to locations throughout the body where they are required. Paramount to this is the relationship between dietary lipid and the enterocytes that line the gut, along with the processes which prepare this lipid for efficient uptake by these cells. These include those which occur in the mouth and stomach along with those which occur within the intestinal lumen itself. Additionally, the interplay between digested lipid, dual avenues for lipid uptake by enterocytes (passive and lipid transporter proteins), a system of intercellular lipid resynthesis and transport, and a complex system of lipoprotein synthesis yield a system open to significant modulation. In this review, we will attempt to outline the processes of lipid digestion, lipoprotein synthesis and the exogenous and endogenous factors which exert their influence.
Collapse
Affiliation(s)
- Alan A Hennessy
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland,
| | | | | | | | | |
Collapse
|
92
|
Bohórquez DV, Shahid RA, Erdmann A, Kreger AM, Wang Y, Calakos N, Wang F, Liddle RA. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. J Clin Invest 2015; 125:782-6. [PMID: 25555217 DOI: 10.1172/jci78361] [Citation(s) in RCA: 311] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 11/20/2014] [Indexed: 12/20/2022] Open
Abstract
Satiety and other core physiological functions are modulated by sensory signals arising from the surface of the gut. Luminal nutrients and bacteria stimulate epithelial biosensors called enteroendocrine cells. Despite being electrically excitable, enteroendocrine cells are generally thought to communicate indirectly with nerves through hormone secretion and not through direct cell-nerve contact. However, we recently uncovered in intestinal enteroendocrine cells a cytoplasmic process that we named neuropod. Here, we determined that neuropods provide a direct connection between enteroendocrine cells and neurons innervating the small intestine and colon. Using cell-specific transgenic mice to study neural circuits, we found that enteroendocrine cells have the necessary elements for neurotransmission, including expression of genes that encode pre-, post-, and transsynaptic proteins. This neuroepithelial circuit was reconstituted in vitro by coculturing single enteroendocrine cells with sensory neurons. We used a monosynaptic rabies virus to define the circuit's functional connectivity in vivo and determined that delivery of this neurotropic virus into the colon lumen resulted in the infection of mucosal nerves through enteroendocrine cells. This neuroepithelial circuit can serve as both a sensory conduit for food and gut microbes to interact with the nervous system and a portal for viruses to enter the enteric and central nervous systems.
Collapse
|
93
|
Zhang H, Hu CAA, Kovacs-Nolan J, Mine Y. Bioactive dietary peptides and amino acids in inflammatory bowel disease. Amino Acids 2014; 47:2127-41. [DOI: 10.1007/s00726-014-1886-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 11/27/2014] [Indexed: 12/21/2022]
|
94
|
Kendig DM, Hurst NR, Bradley ZL, Mahavadi S, Kuemmerle JF, Lyall V, DeSimone J, Murthy KS, Grider JR. Activation of the umami taste receptor (T1R1/T1R3) initiates the peristaltic reflex and pellet propulsion in the distal colon. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1100-7. [PMID: 25324508 PMCID: PMC4254958 DOI: 10.1152/ajpgi.00251.2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intraluminal nutrients in the gut affect the peristaltic reflex, although the mechanism is not well defined. Recent evidence supports the presence of taste receptors and their signaling components in enteroendocrine cells, although their function is unclear. This study aimed to determine if nutrients modify colonic motility through activation of taste receptors. Colonic sections were immunostained for the umami taste receptor T1R1/T1R3, which mediates the response to umami ligands, such as monosodium glutamate (MSG), in taste cells. Ascending contraction, descending relaxation, and calcitonin gene-related peptide release were measured in three-chamber flat-sheet preparations of rat colon in response to MSG alone or with inosine 5'-monophosphate (IMP). Velocity of artificial fecal pellet propulsion was measured by video recording in guinea pig distal colon. T1R1/T1R3 receptors were present in enteroendocrine cells of colonic sections from human, rat, mouse, and guinea pig. MSG initiated ascending contraction and descending relaxation components of the peristaltic reflex and calcitonin gene-related peptide release in flat-sheet preparations. IMP augmented the MSG-induced effects, suggesting activation of T1R1/T1R3 receptors. In T1R1(-/-) mice, mucosal stroking, but not MSG, elicited a peristaltic reflex. Intraluminal perfusion of MSG enhanced the velocity of artificial fecal pellet propulsion, which was also augmented by IMP. Propulsion was also increased by l-cysteine, but not l-tryptophan, supporting a role of T1R1/T1R3 receptors. We conclude that T1R1/T1R3 activation by luminal MSG or l-cysteine elicits a peristaltic reflex and CGRP release and increases the velocity of pellet propulsion in distal colon. This mechanism may explain how nutrients regulate colonic propulsion.
Collapse
Affiliation(s)
- Derek M. Kendig
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Norman R. Hurst
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Zachary L. Bradley
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Sunila Mahavadi
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - John F. Kuemmerle
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and ,2Department of Medicine, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Vijay Lyall
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - John DeSimone
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Karnam S. Murthy
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and ,2Department of Medicine, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - John R. Grider
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and ,2Department of Medicine, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
95
|
Leach K, Sexton PM, Christopoulos A, Conigrave AD. Engendering biased signalling from the calcium-sensing receptor for the pharmacotherapy of diverse disorders. Br J Pharmacol 2014; 171:1142-55. [PMID: 24111791 DOI: 10.1111/bph.12420] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 12/14/2022] Open
Abstract
The human calcium-sensing receptor (CaSR) is widely expressed in the body, where its activity is regulated by multiple orthosteric and endogenous allosteric ligands. Each ligand stabilizes a unique subset of conformational states, which enables the CaSR to couple to distinct intracellular signalling pathways depending on the extracellular milieu in which it is bathed. Differential signalling arising from distinct receptor conformations favoured by each ligand is referred to as biased signalling. The outcome of CaSR activation also depends on the cell type in which it is expressed. Thus, the same ligand may activate diverse pathways in distinct cell types. Given that the CaSR is implicated in numerous physiological and pathophysiological processes, it is an ideal target for biased ligands that could be rationally designed to selectively regulate desired signalling pathways in preferred cell types.
Collapse
Affiliation(s)
- K Leach
- Pharmaceutical Sciences, Monash University, Melbourne, Vic., Australia
| | | | | | | |
Collapse
|
96
|
Sommer CA, Mostoslavsky G. RNA-Seq analysis of enteroendocrine cells reveals a role for FABP5 in the control of GIP secretion. Mol Endocrinol 2014; 28:1855-65. [PMID: 25268051 DOI: 10.1210/me.2014-1194] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In response to fat intake, enteroendocrine K cells release the hormone glucose-dependent insulinotropic polypeptide (GIP). GIP acts on adipocytes to increase lipid uptake and enhance adipokine secretion, promoting weight gain and insulin resistance. Modulation of intestinal GIP release could therefore represent a therapeutic strategy for the treatment and prevention of obesity and diabetes. However, the prospects of using drugs to effectively target specific enteroendocrine cell types have been tempered by the realization that these cells share similar transcriptional programs and frequently employ common mechanisms of hormone secretion. To gain novel insights into the regulation of GIP release, we generated knock-in mice expressing green fluorescent protein (GFP) under the control of the endogenous GIP promoter that enable the isolation of a purified population of small intestine K cells. Using RNA sequencing, we comprehensively characterized the transcriptomes of GIP(GFP) cells as well as the entire enteroendocrine lineage derived from Neurogenin3-expressing progenitors. Among the genes differentially expressed in GIP(GFP) cells, we identified and validated fatty acid-binding protein 5 (FABP5) as a highly expressed marker of GIP-producing cells that is absent in other enteroendocrine cell types. FABP5 promotes intracellular transport and inactivation of endocannabinoids, including anandamide, which inhibits GIP release. Remarkably, we found that circulating levels of GIP were significantly decreased in FABP5-deficient mice in the fasting state and in response to acute, oral fat diet administration. Our findings highlight the power of RNA sequencing to uncover molecular signatures of specific enteroendocrine cell types that can potentially be exploited for therapeutic purposes in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Cesar A Sommer
- Section of Gastroenterology, Department of Medicine, and Center for Regenerative Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | | |
Collapse
|
97
|
Emerging roles of the extracellular calcium-sensing receptor in nutrient sensing: control of taste modulation and intestinal hormone secretion. Br J Nutr 2014; 111 Suppl 1:S16-22. [PMID: 24382107 DOI: 10.1017/s0007114513002250] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The extracellular Ca-sensing receptor (CaSR) is a sensor for a number of key nutrients within the body, including Ca ions (Ca²⁺) and L-amino acids. The CaSR is expressed in a number of specialised cells within the gastrointestinal (GI) tract, and much work has been done to examine CaSR's role as a nutrient sensor in this system. This review article examines two emerging roles for the CaSR within the GI tract--as a mediator of kokumi taste modulation in taste cells and as a regulator of dietary hormone release in response to L-amino acids in the intestine.
Collapse
|
98
|
Baruffol C, Jordi J, Camargo S, Radovic T, Herzog B, Fried M, Schwizer W, Verrey F, Lutz TA, Steingoetter A. L-lysine dose dependently delays gastric emptying and increases intestinal fluid volume in humans and rats. Neurogastroenterol Motil 2014; 26:999-1009. [PMID: 24890878 DOI: 10.1111/nmo.12354] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/31/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Novel sensory inputs for the control of food intake and gastrointestinal (GI) function are of increasing interest due to the rapid increase in nutrition-related diseases. The essential amino acid L-lysine was demonstrated to have a selective impact on food intake, gastric emptying, and intestinal transit in rats, thus indicating a potential novel direct sensory input to assess dietary protein content and quality. The aim of this study was to assess translational aspects of this finding and to investigate the dose-dependent effect of L-lysine on human and rat GI function. METHODS L-lysine doses from 0-800 mg in rats and 0.5-7.5 g in humans were analyzed for their effect on gastric emptying and GI secretion. Human GI function was assessed non-invasively using magnetic resonance imaging (MRI), rat data were acquired using standard lethal measurement methods. L-lysine dose dependently delayed gastric emptying and stimulated GI secretion in rats as reflected by residual phenol red content and increased gastric wet weight. KEY RESULTS The dose-dependent delay in gastric emptying observed in rats was confirmed in humans with an increase in halftime of gastric emptying of 4 min/g L-lysine, p < 0.01. Moreover, a dose-dependent increase in intestinal fluid accumulation was observed (0.4 mL/min/g L-lysine, p < 0.0001). No effect on alkaline tide, glucose concentration, hematocrit, or visceral sensations was detected. CONCLUSIONS & INFERENCES This translational study demonstrates comparable dose-dependent effects of intragastric L-lysine on GI function in humans and rats and suggests a broader role for individual amino acids in the control of GI motility and secretion in vivo.
Collapse
Affiliation(s)
- C Baruffol
- Division of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Muramatsu M, Hira T, Mitsunaga A, Sato E, Nakajima S, Kitahara Y, Eto Y, Hara H. Activation of the gut calcium-sensing receptor by peptide agonists reduces rapid elevation of plasma glucose in response to oral glucose load in rats. Am J Physiol Gastrointest Liver Physiol 2014; 306:G1099-107. [PMID: 24812056 DOI: 10.1152/ajpgi.00155.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The calcium-sensing receptor (CaSR) is expressed in various tissues, including the gastrointestinal tract. To investigate the role of gut CaSR on glycemic control, we examined whether single oral administration of CaSR agonist peptides affected the glycemic response in rats. Glucose tolerance tests were performed under oral or duodenal administration of various CaSR agonist peptides (γGlu-Cys, protamine, and poly-d-lysine hydrobromide) in conscious rats. Involvement of CaSR was determined by using a CaSR antagonist. Signaling pathways underlying CaSR agonist-modified glycemia were investigated using gut hormone receptor antagonists. The gastric emptying rate after the administration of CaSR agonist peptides was measured by the phenol red recovery method. Oral and duodenal administration of CaSR agonist peptides attenuated glycemic responses under the oral glucose tolerance test, but the administration of casein did not. The promotive effect on glucose tolerance was weakened by luminal pretreatment with a CaSR antagonist. Treatment with a 5-HT3 receptor antagonist partially diminished the glucose-lowering effect of peptides. Furthermore, the gastric emptying rate was decreased by duodenal administration of CaSR agonist peptides. These results demonstrate that activation of the gut CaSR by peptide agonists promotes glucose tolerance in conscious rats. 5-HT3 receptor and the delayed gastric emptying rate appear to be involved in the glucose-lowering effect of CaSR agonist peptides. Thus, activation of gut CaSR by dietary peptides reduces glycemic responses so that gut CaSR may be a potential target for the improvement of postprandial glycemia.
Collapse
Affiliation(s)
- Maya Muramatsu
- Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Tohru Hira
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan;
| | - Arimi Mitsunaga
- Faculty of Agriculture, Hokkaido University, Sapporo, Japan; and
| | - Eri Sato
- Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Shingo Nakajima
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Yoshiro Kitahara
- Faculty of Agriculture, Hokkaido University, Sapporo, Japan; and
| | - Yuzuru Eto
- Faculty of Agriculture, Hokkaido University, Sapporo, Japan; and
| | - Hiroshi Hara
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
100
|
Bohórquez DV, Samsa LA, Roholt A, Medicetty S, Chandra R, Liddle RA. An enteroendocrine cell-enteric glia connection revealed by 3D electron microscopy. PLoS One 2014; 9:e89881. [PMID: 24587096 PMCID: PMC3935946 DOI: 10.1371/journal.pone.0089881] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/28/2014] [Indexed: 11/25/2022] Open
Abstract
The enteroendocrine cell is the cornerstone of gastrointestinal chemosensation. In the intestine and colon, this cell is stimulated by nutrients, tastants that elicit the perception of flavor, and bacterial by-products; and in response, the cell secretes hormones like cholecystokinin and peptide YY – both potent regulators of appetite. The development of transgenic mice with enteroendocrine cells expressing green fluorescent protein has allowed for the elucidation of the apical nutrient sensing mechanisms of the cell. However, the basal secretory aspects of the enteroendocrine cell remain largely unexplored, particularly because a complete account of the enteroendocrine cell ultrastructure does not exist. Today, the fine ultrastructure of a specific cell can be revealed in the third dimension thanks to the invention of serial block face scanning electron microscopy (SBEM). Here, we bridged confocal microscopy with SBEM to identify the enteroendocrine cell of the mouse and study its ultrastructure in the third dimension. The results demonstrated that 73.5% of the peptide-secreting vesicles in the enteroendocrine cell are contained within an axon-like basal process. We called this process a neuropod. This neuropod contains neurofilaments, which are typical structural proteins of axons. Surprisingly, the SBEM data also demonstrated that the enteroendocrine cell neuropod is escorted by enteric glia – the cells that nurture enteric neurons. We extended these structural findings into an in vitro intestinal organoid system, in which the addition of glial derived neurotrophic factors enhanced the development of neuropods in enteroendocrine cells. These findings open a new avenue of exploration in gastrointestinal chemosensation by unveiling an unforeseen physical relationship between enteric glia and enteroendocrine cells.
Collapse
Affiliation(s)
- Diego V. Bohórquez
- Departments of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| | - Leigh A. Samsa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Andrew Roholt
- Renovo Neural Incorporation, Cleveland, Ohio, United States of America
| | - Satish Medicetty
- Renovo Neural Incorporation, Cleveland, Ohio, United States of America
| | - Rashmi Chandra
- Departments of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Rodger A. Liddle
- Departments of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Veterans Affairs Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|