Relationship between plasma free amino acid profiles and changes in Crohn’s disease activity index after administration of an elemental diet in patients with Crohn’s disease

Main Article Content

Toshimi Chiba

Abstract

Purpose: We aimed to elucidate the relationship between plasma free amino acid profiles and changes in the Crohn’s disease activity index (CDAI) among patients with Crohn’s disease (CD) after administration of an elemental diet (ED).


Patients and methods: We measured fasting plasma concentrations of free amino acids in 5 patients with CD and evaluated the relationship between amino acid concentration and disease activity at 4 weeks after administration of an ED.


Results: Concentrations of valine, lysine, tyrosine, and glutamic acid were significantly increased after administration of an ED. Significant correlations were noted between changes in CDAI scores and valine concentrations. Tryptophan concentrations also exhibited a strong correlation with CDAI scores.


Conclusion: In patients with CD, plasma concentrations of valine and tryptophan appear to be associated with disease activity following treatment with an ED.

Keywords: Crohn’s disease, amino acids, elemental diet, CDAI

Article Details

How to Cite
CHIBA, Toshimi. Relationship between plasma free amino acid profiles and changes in Crohn’s disease activity index after administration of an elemental diet in patients with Crohn’s disease. Medical Research Archives, [S.l.], v. 9, n. 10, oct. 2021. ISSN 2375-1924. Available at: <https://esmed.org/MRA/mra/article/view/2559>. Date accessed: 29 mar. 2024. doi: https://doi.org/10.18103/mra.v9i10.2559.
Section
Research Articles

References

1. Hanauer SB, Feagan BG, Lichtenstein GR, et al. Maintenance infliximab for Crohn’s disease: the ACCENT 1 randomised trial. Lancet. 2002;359:1541-1549.
2. Lichtenstein GR, Feagan BG, Cohen RD, et al. Serious Infections and Mortality in Association With Therapies for Crohn’s disease: TREAT Registry. Clin Gastroentrol Hepatol. 2006;4:621-630.
3. Coeffier M, Marion-Letellier R, Dechelotte P. Potential for amino acids supplementation during Inflammatory Bowel Diseases. Inflamm Bowel Dis. 2010;16:518-524.
4. Coeffier M, Dechelotte P. The role of glutamine in intensive care unit patients: mechanisms of action and clinical outcome. Nutr Rev. 2005;63:65-69.
5. Wu G, Morris SM Jr. Arginine metabolism: nitric oxide and beyond. Biochem J. 1998;336:1-17.
6. Andou A, Hisamatsu T, Okamoto S, et al. Dietary histidine ameliorates murine colitis by inhibition of proinflammatory cytokine production from macrophages. Gastroenterology. 2009;136:564-574.
7. Yamamoto T, Nakahigashi M, Saniabadi AR. Review article: diet and inflammatory bowel disease-epidemiology and treatment. Aliment Pharmacol Ther. 2009;30:99-112.
8. Matsui T, Sakurai T, Yao T. Nutritional therapy for Crohn’s disease in Japan. J Gastroenterol. 2005;40:25-31.
9. Ameho CK, Adjei AA, Harrison EK, et al. Prophylactic effect of dietary glutamine supplementation on interleukin and tumor necrosis factor alpha production in trinitrobenzene sulphonic acid induced colitis. Gut. 1997;41:487-493.
10. Israeli E, Berenshtein E, Wengrower D, et al. Prophylactic administration of topical glutamine enhances the capability of the rat colon to resist inflammatory damage. Dig Dis Sci. 2004;49:1705-1712.
11. Giris¸ M, Erbil Y, Dogru-Abbasoglu S, et al. The effect of heme oxygenase-1 induction by glutamine on TNBS-induced colitis. The effect of glutamine on TNBS colitis. Int J Colorectal Dis. 2007;22:591-599.
12. Vicario M, Amat C, Rivero M, et al. Dietary glutamine affects mucosal functions in rats with mild DSS induced colitis. J Nutr. 2007;137:1931-1937.
13. Arndt H, Kullmann F, Reuss F, et al. Glutamine attenuates leukocyte endothelial cell adhesion in indomethacin-induced intestinal inflammation in the rat. JPEN. 1999;23:12-18.
14. Basivireddy J, Jakob M, Balasubramanian KA. Oral glutamine attenuates indomethacin-induced small intestinal damage. Clin Sci. 2004;107:281-289.
15. Fillmann H, Kretzmann NA, San-Miguel B, et al. Glutamine inhibits over-expression of pro-inflammatory genes and down-regulates the nuclear factor kappa B pathway in an experimental model of colitis in the rat. Toxicology. 2007;236:217-226.
16. Scoville EA, Allaman MM, Brown CT, et al. Alterations in lipid, amino acid, and energy metabolism distinguish Crohn's disease from ulcerative colitis and control subjects by serum metabolomic profiling. Metabolomics. 2018;14:1-20.
17. Chiba T, Suzuki K, Matsumoto T. Plasma-free amino acid profiles in Crohn's disease: relationship with the Crohn Disease Activity Index. Clin Med Insights Gastroenterol. 2018;11:1-7.
18. Grimble RF. Nutritional modulation of immune function. Froc Nutr Soc. 2001;60:389-397.
19. Ockenga J, Borchert K, Stüber E, et al. Glutamine-enriched total parenteral nutrition in patients with inflammatory bowel disease. Eur J Clin Nutr. 2005;59:1302-1309.
20. Hisamatsu T, Okamoto S, Hashimoto M, et al. Novel, objective, multivariate biomarkers composed of plasma amino acid profiles for the diagnosis and assessment of inflammatory bowel disease. PLoS One. 2012;7:e31131.
21. Best WR, Becktel JM, Singleton JW, et al. Development of a Crohn’s disease activity index. National Cooperative Crohn’s Disease Study. Gastroenterology. 1976;70:439-444.
22. Teahon K, Smethurst P, Pearson M, et al. The effect of elemental diet on intestinal permeability and inflammation in Crohn’s disease. Gastronenterology. 1991;101:84-89.
23. Zhang H, Hu CAA, Kovacs-Nolan J, Mine Y. Bioactive dietary peptides and amino acids in inflammatory bowel disease. Amino Acids. 2015;47:2127-2141.
24. Faure M, Mettraux C, Moennoz D, et al. Specific amino acids increase mucin synthesis and microbiota in dextran sulfate sodium-treated rats. J Nutr. 2006;136:1558-1564.
25. Ockenga GJ. Glutamine-enriched total parenteral nutrition in patients with inflammatory bowel disease. Eur J Clin Nutri. 2005;59:1302-1309.
26. Elias RJ, Kellerby SS, Decker EA. Antioxidant activity of proteins and peptides. Crit Rev Food Sci Nutr. 2008;48:430-441.
27. Son DO, Satsu H, Shimizu M. Histidine inhibits oxidative stress- and TNF-alpha-induced interleukin-8 secretion in intestinal epithelial cells. FEBS Lett. 2005;579:4671-4677.
28. Katayama S, Mine Y. Antioxidative activity of amino acids on tissue oxidative stress in human intestinal epithelial cell model. J Agric Food Chem. 2007;17:8458-8464.
29. Nakano M, Tominaga K, Hoshino A, et al. Therapeutic efficacy of an elemental diet for patients with crohn's disease and its association with amino acid metabolism. Saudi J Gastroenterol. 2017;23:20-27.
30. Faure M, Mettraux C, Moennoz D, et al. Specific amino acids increase mucin synthesis and microbiota in dextran sulfate sodium-treated rats. J Nutr. 2006;136:1558-1564.
31. Shizuma T, Mori H, Fukuyama N. Protective effect of tryptophan against dextran sulfate sodium- induced experimental colitis. Turk J Gastroenterol. 2013;24:30-35.
32. Shiomi Y, Nishiumi S, Ooi M, et al. GCMS-based metabolomic study in mice with colitis induced by dextran sulfate sodium. Inflamm Bowel Dis. 2011;17:2261-2274.
33. Gupta NK, Thaker AI, Kanuri N, et al. Serum analysis of tryptophan catabolism pathway: correlation with Crohn’s disease activity. Inflamm Bowel Dis. 2012;18:1214-1220.
34. Puccetti P, Grohmann U. IDO and regulatory T cells: a role for reverse signalling and non-canonical NF-kappa B activation. Nat Rev Immunol. 2007;7:817-823.
35. Munn DH, Zhou M, Attwood JT, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281:1191-1193.
36. Romani L, Fallarino F, De Luca A, et al. Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature. 2008;451:211-215.
37. Jiang Z M, Cao JD, Zhu XG, et al. The impact of alanyl-glutamine on clinical safety, nitrogen balance, intestinal permeability, and clinical outcome in postoperative patients: a randomized, double-blind, controlled study of 120 patients. J Parenteral Enteral Nutr. 1999;23:S62-S66.
38. Fernández-Banares F, Cabré E, Esteve-Comas M, et al. How effective is enteral nutrition in inducing clinical remission in active Crohn’s disease? A meta-analysis of the randomized clinical trials. JPEN. 1995;19:356-364.
39. Yamamoto T, Nakahigashi M, Umegae S, et al. Impact of elemental diet on mucosal inflammation in patients with active Crohn’s Disease: cytokine production and endoscopic and histological findings. Inflamm Bowel Dis. 2005;11:580-588.
40. Wang Y, Chandra R, Samsa LA, Gooch B, Fee BE, Cook JM et al. Amino acids stimulate cholecystokinin release through the Ca2+-sensing receptor. Am J Physiol Gastrointest Liver Physiol. 2011;300:G528-G537.
41. Moffett JR, Namboodiri MA. Tryptophan and the immune response. Immunol Cell Biol. 2003;81:247-265
42. Kim CJ, Kovacs-Nolan JA, Yang C, et al. L-Tryptophan exhibits therapeutic function in a porcine model of dextran sodium sulfate (DSS)-induced colitis. J Nutr Biochem. 2010;21:468-475.
43. Schröcksnadel K, Wirleitner B, Winkler C, Fuchs D. Monitoring tryptophan metabolism in chronic immune activation. Clin Chim Acta. 2006;364:82-90.
44. Islam J, Sato S, Watanabe K, et al. Dietary tryptophan alleviates dextran sodium sulfate-induced colitis through aryl hydrocarbon receptor in mice. J Nutr Biochem. 2017;42:43–50.
45. Tapiero H, Mathé G, Couvreur P, Tew KD. Glutamine and glutamate. Biomed Pharmacother. 2002;56:446-457.
46. Blachier F, Boutry C, Bos C, Tomé D. Metabolism and functions of L-glutamate in the epithelial cells of the small and large intestines. Am J Clin Nutr. 2009;90:814S-821S.
47. Jiao N, Wu Z, Ji Y, et al. L-Glutamate enhances barrier and antioxidative functions in intestinal porcine epithelial cells. J Nutr. 2015;145:2258-2264.
48. Wu M, Xiao H, Ren W, et al. Therapeutic effects of glutamic acid in piglets challenged with deoxynivalenol. PLoS ONE. 2014;9:e100591.
49. Li TT, Zhang JF, Fei SJ, et al. Glutamate microinjection into the hypothalamic paraventricular nucleus attenuates ulcerative colitis in rats. Acta Pharmacol Sin. 2014;35:185-194.