J Vet Sci.  2018 Mar;19(2):179-187. 10.4142/jvs.2018.19.2.179.

Calcium-sensing receptor-mediated L-tryptophan-induced secretion of cholecystokinin and glucose-dependent insulinotropic peptide in swine duodenum

Affiliations
  • 1Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China. suqinhang69@njau.edu.cn

Abstract

This study aimed to elucidate the effect of tryptophan (Trp) on gut hormone secretion as well as the roles of the calcium-sensing receptor (CaSR) and its downstream signaling pathway in gut hormone secretion by assessing swine duodenal perfusion in vitro. Swine duodenum was perfused with Krebs-Henseleit buffer as a basal solution. Various concentrations (0, 10, and 20 mM) of Trp were applied to investigate its effect on gut hormone secretion. A CaSR antagonist was used to detect the involvement of CaSR and its signal molecules. The 20 mM Trp concentration promoted the secretion of cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP), elevated the mRNA level of CaSR, and upregulated the protein levels of CaSR, protein kinase C (PKC), and inositol trisphosphate receptor (IP3R). However, NPS 2143, an inhibitor of CaSR, attenuated the CCK and GIP release, reduced the mRNA level of CaSR, and decreased the protein levels of CaSR, PKC, and IP3R with 20 mM Trp perfusion. The results indicate that CCK and GIP secretion can be induced by Trp in swine duodenum in vitro, and the effect is mediated by CaSR and its downstream signal molecules PKC and IP3R.

Keyword

calcium-sensing receptor; gut hormone; signaling pathway; swine; tryptophan

MeSH Terms

Cholecystokinin*
Duodenum*
Gastric Inhibitory Polypeptide*
In Vitro Techniques
Inositol
Perfusion
Protein Kinase C
Receptors, Calcium-Sensing
RNA, Messenger
Swine*
Tryptophan
Cholecystokinin
Gastric Inhibitory Polypeptide
Inositol
Protein Kinase C
RNA, Messenger
Receptors, Calcium-Sensing
Tryptophan

Figure

  • Fig. 1 Schematic of the perfusion system (A) and the perfusion chamber (B). In (A): 1, oxygen bomb (mixture of 95% O2 and 5% CO2); 2, media reservoir; 3, peristaltic pump; 4, silicone tube; 5, perfusion chamber; 6, water bath (37℃); 7, collector. In (B): 1, nylon filter platform; 2, needle; 3, rubber stopper; 4, syringe; 5, rubber plunger.

  • Fig. 2 The mRNA expression of CaSR (A) and the secretion of gut hormones (B and C) regulated by different concentrations of tryptophan (Trp). The duodenal tissues were perfused with 0 (control), 10, and 20 mM Trp for 120 min. Tissues were collected at the end of perfusion. CaSR (A) mRNA expression was analyzed by quantitative polymerase chain reaction (qPCR) and glyceraldehyde phosphate dehydrogenase was used as the internal control. The concentrations of cholecystokinin (CCK, B) and glucose-dependent insulinotropic peptide (GIP, C) in the perfusate samples were detected every 20 min by using enzyme-linked immunosorbent assay kits. Values are expressed as mean ± SEM (n = 4). A statistical difference was determined by one-way ANOVA followed by Tukey's multiple comparison tests. Bars with the different letters show significant differences compared with the control group (A) (p < 0.05); * and δ (B and C); 20 mM Trp and 10 mM Trp treatments are significantly different from the control, respectively (p < 0.05). The influence of Ca2+ on the ability of Trp to induce the expression of CaSR (D) and the secretion of gut hormones (E and F). Tissue samples of duodenum were perfused with Krebs-Henseleit buffer containing 0 mM Trp without [Ca2+]e (control) and 20 mM Trp with or without 10 mM [Ca2+]e for 120 min. After perfusion, the expression of CaSR (D) was determined by qPCR. Perfusate samples were collected every 20 min to measure concentrations of CCK (E) and GIP (F). Values are expressed as mean ± SEM (n = 4). Statistical differences were determined by one-way ANOVA followed by Tukey's multiple comparison tests. Bars with the different letters showed the significant differences compared with the control group (D) (p < 0.05); γ (E and F); the combination of 20 mM Trp and 10 mM Ca2+ was significantly different from the control (p < 0.05). Effects of CaSR antagonist NPS 2143 on the expression of Trp-induced CaSR (G) and the secretion of gut hormones (H and I). Duodenal tissues were treated with 0 (control), 20, or 20 mM Trp and NPS 2143 (25 µM). At the end of perfusion (120 min), the duodenum tissues were collected to determine the mRNA expression of CaSR (G) by qPCR. The perfusate samples were obtained every 20 min to evaluate the concentration of CCK (H) and GIP (I). Values are expressed as mean ± SEM (n = 6). Statistical differences were determined by one-way ANOVA followed by Tukey's multiple comparison tests. Bars with the different letters showed the significant differences compared with the control group (G) (p < 0.05); χ (H and I); 20 mM Trp and the combination of 20 mM Trp and NPS 2143 treatments are significantly different from the control, respectively (p < 0.05).

  • Fig. 3 Protein levels of calcium-sensing receptor (CaSR) and its signal molecules regulated by tryptophan (Trp). (A) The protein images of CaSR, protein kinase C (PKC), and inositol trisphosphate receptor (IP3R) at 0 and 20 mM Trp treatments. The optical densities of CaSR, PKC, and IP3R are shown in (B–D), respectively. Values are shown as mean ± SEM. **p < 0.01.

  • Fig. 4 The protein levels of calcium-sensing receptor (CaSR) and its signal molecules regulated by CaSR antagonist NPS 2143. (A) The protein images of CaSR, protein kinase C (PKC), and inositol trisphosphate receptor (IP3R) in the presence of 20 mM tryptophan and NPS 2143. The optical densities of CaSR, PKC, and IP3R are shown in (B–D), respectively. Values are shown as mean ± SEM. **p < 0.01.


Reference

1. Badger TM. Perifusion of anterior pituitary cells: release of gonadotropins and somatotropins. Methods Enzymol. 1986; 124:79–90.
2. Bronk JR, Hastewell JG. The transport and metabolism of naturally occurring pyrimidine nucleosides by isolated rat jejunum. J Physiol. 1988; 395:349–361.
Article
3. Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, Mueller R, Nolan T, Pfaffl MW, Shipley GL, Vandesompele J, Wittwer CT. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 2009; 55:611–622.
Article
4. Conigrave AD, Brown EM. Taste receptors in the gastrointestinal tract. II. L-amino acid sensing by calcium-sensing receptors: implications for GI physiology. Am J Physiol Gastrointest Liver Physiol. 2006; 291:G753–G761.
5. Conigrave AD, Franks AH, Brown EM, Quinn SJ. L-amino acid sensing by the calcium-sensing receptor: a general mechanism for coupling protein and calcium metabolism? Eur J Clin Nutr. 2002; 56:1072–1080.
Article
6. Cuber JC, Bernard C, Levenez F, Chayvialle JA. [Lipids, proteins and carbohydrates stimulate the secretion of intestinal cholecystokinin in the pig]. Reprod Nutr Dev. 1990; 30:267–275. French.
7. Hansen JR, Conn PM. Measurement of pulsatile hormone release from perifused pituitary cells immobilized on microcarriers. In : Conn PM, editor. Methods in Neurosciences. Vol. 2. Cell Culture. San Diego: Academic Press;1990. p. 181–194.
8. Hebert SC, Cheng S, Geibel J. Functions and roles of the extracellular Ca2+-sensing receptor in the gastrointestinal tract. Cell Calcium. 2004; 35:239–247.
Article
9. Hira T, Nakajima S, Eto Y, Hara H. Calcium-sensing receptor mediates phenylalanine-induced cholecystokinin secretion in enteroendocrine STC-1 cells. FEBS J. 2008; 275:4620–4626.
Article
10. Hofer AM, Brown EM. Extracellular calcium sensing and signalling. Nat Rev Mol Cell Biol. 2003; 4:530–538.
Article
11. Hou G, Xue L, Lu Z, Fan T, Tian F, Xue Y. An activated mTOR/p70S6K signaling pathway in esophageal squamous cell carcinoma cell lines and inhibition of the pathway by rapamycin and siRNA against mTOR. Cancer Lett. 2007; 253:236–248.
Article
12. Karhunen LJ, Juvonen KR, Huotari A, Purhonen AK, Herzig KH. Effect of protein, fat, carbohydrate and fibre on gastrointestinal peptide release in humans. Regul Pept. 2008; 149:70–78.
Article
13. Liddle RA, Green GM, Conrad CK, Williams JA. Proteins but not amino acids, carbohydrates, or fats stimulate cholecystokinin secretion in the rat. Am J Physiol. 1986; 251:G243–G248.
Article
14. Liou AP, Sei Y, Zhao X, Feng J, Lu X, Thomas C, Pechhold S, Raybould HE, Wank SA. The extracellular calcium-sensing receptor is required for cholecystokinin secretion in response to L-phenylalanine in acutely isolated intestinal I cells. Am J Physiol Gastrointest Liver Physiol. 2011; 300:G538–G546.
Article
15. Mace OJ, Schindler M, Patel S. The regulation of K- and L-cell activity by GLUT2 and the calcium-sensing receptor CasR in rat small intestine. J Physiol. 2012; 590:2917–2936.
Article
16. Macleod RJ. CaSR function in the intestine: hormone secretion, electrolyte absorption and secretion, paracrine non-canonical Wnt signaling and colonic crypt cell proliferation. Best Pract Res Clin Endocrinol Metab. 2013; 27:385–402.
Article
17. Miazza B, Palma R, Lachance JR, Chayvialle JA, Jonard PP, Modigliani R. Jejunal secretory effect of intraduodenal food in humans. A comparison of mixed nutrients, proteins, lipids, and carbohydrates. Gastroenterology. 1985; 88:1215–1222.
Article
18. Moran-Ramos S, Tovar AR, Torres N. Diet: friend or foe of enteroendocrine cells--how it interacts with enteroendocrine cells. Adv Nutr. 2012; 3:8–20.
Article
19. Murphy KG, Bloom SR. Gut hormones and the regulation of energy homeostasis. Nature. 2006; 444:854–859.
Article
20. Neary MT, Batterham RL. Gut hormones: implications for the treatment of obesity. Pharmacol Ther. 2009; 124:44–56.
Article
21. Nemeth EF, Steffey ME, Hammerland LG, Hung BC, Van Wagenen BC, DelMar EG, Balandrin MF. Calcimimetics with potent and selective activity on the parathyroid calcium receptor. Proc Natl Acad Sci U S A. 1998; 95:4040–4045.
Article
22. Rasoamanana R, Darcel N, Fromentin G, Tomé D. Nutrient sensing and signalling by the gut. Proc Nutr Soc. 2012; 71:446–455.
Article
23. Steinert RE, Beglinger C. Nutrient sensing in the gut: interactions between chemosensory cells, visceral afferents and the secretion of satiation peptides. Physiol Behav. 2011; 105:62–70.
Article
24. Steinert RE, Luscombe-Marsh ND, Little TJ, Standfield S, Otto B, Horowitz M, Feinle-Bisset C. Effects of intraduodenal infusion of L-tryptophan on ad libitum eating, antropyloroduodenal motility, glycemia, insulinemia, and gut peptide secretion in healthy men. J Clin Endocrinol Metab. 2014; 99:3275–3284.
Article
25. Tan YF, Chen WH, Zou SX. The effect of β Casomorphin on piglets' stomach antrum under superfusion. J Nanjing Agric Univ. 2000; 23:72–75.
26. Wang Y, Chandra R, Samsa LA, Gooch B, Fee BE, Cook JM, Vigna SR, Grant AO, Liddle RA. Amino acids stimulate cholecystokinin release through the Ca2+-sensing receptor. Am J Physiol Gastrointest Liver Physiol. 2011; 300:G528–G537.
Article
27. Zhou HR, Pestka JJ. Deoxynivalenol (vomitoxin)-induced cholecystokinin and glucagon-like peptide-1 release in the STC-1 enteroendocrine cell model is mediated by calcium-sensing receptor and transient receptor potential ankyrin-1 channel. Toxicol Sci. 2015; 145:407–417.
Article
Full Text Links
  • JVS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr