Wheat oligopeptides enhance the intestinal mucosal barrier and alleviate inflammation via the TLR4/Myd88/MAPK signaling pathway in aged mice

  • Yang Xian Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
  • Pan Da Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
  • Yang Chao Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
  • Xia Hui Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
  • Yang Ligang Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
  • Wang Shaokang Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
  • Sun Guiju Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China
Keywords: aging; intestinal mucosal impairment; intestinal barrier; wheat oligopeptides; anti-inflammatory;

Abstract

Background: Aging can induce oxidative stress, inflammation and mucosal impairment, and few works have been conducted to investigate the protective effects of WP on the natural intestinal aging process.

Objective: The present work aimed to examine the protective effect of wheat oligopeptides (WP) on intestine mucosal impairment in aged mice, and investigate the potential antioxidation, anti-inflammatory effects of WP.

Design: Seventy-two aged mice (24 months old) were randomly divided into six groups, 12 for each group. Twelve young mice (6 months old) were regarded as the young control group. WP (25, 50, 100, 200, or 400 mg/kg) or distilled water were administered daily by gavage for 30 days.

Results: Histological observations showed that intestinal mucosal degeneration was attenuated by WP pretreatment. WP exhibited remarkable antioxidant activity via increasing superoxide dismutase, glutathione peroxidase, total antioxidant capacity and catalase activities, and decreasing the malondialdehyde levels in small intestine mucosa. WP pretreatment significantly suppressed intestinal mucosa inflammation through the reduction of TNF-α, TGF-β, IFN-γ IL-1β and IL-6. WP markedly protect the intestinal mucosal barrier by decreasing the ICAM-1 level, and increasing ZO-1 and JAMA-A levels. WP significantly down-regulated protein expression levels of TLR4, Myd88, and MAPK, suggesting that WP have a potential effect on inhibiting aging-induced inflammatory responses by blocking TLR4/Myd88/MAPK signal transduction.

Conclusion: WP administration effectively alleviated intestinal mucosal impairment in aged mice. The potential mechanism was associated with enhancement of antioxidation and anti-inflammatory action and protection of the intestinal mucosal barrier.

Downloads

Download data is not yet available.

References


  1. Salminen A, Kauppinen A, Kaarniranta K. FGF21 activates AMPK signaling: impact on metabolic regulation and the aging process. J Mol Med 2017; 95: 123–31. doi: 10.1007/s00109-016-1477-1

  2. Morita Y, Jounai K, Sakamoto A, Tomita Y, Sugihara Y, Suzuki H, et al. Long-term intake of Lactobacillus paracasei KW3110 prevents age-related chronic inflammation and retinal cell loss in physiologically aged mice. Aging-Us 2018; 10: 2723–40. doi: 10.18632/aging.101583

  3. Donato AJ, Morgan RG, Walker AE, Lesniewski LA. Cellular and molecular biology of aging endothelial cells. J Mol Cell Cardiol 2015;89:122–35. doi: 10.1016/j.yjmcc.2015.01.021

  4. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011; 333: 1109–12. doi: 10.1126/science.1201940

  5. Piber D, Olmstead R, Cho JHJ, Witarama T, Perez C, Dietz N, et al. Inflammaging: age and systemic, cellular, and nuclear inflammatory biology in older adults. J Gerontol a-Biol 2019; 74: 1716–24. doi: 10.1093/gerona/glz130

  6. Ballak DB, Brunt VE, Sapinsley ZJ, Ziemba BP, Richey JJ, Zigler MC, et al. Short-term interleukin-37 treatment improves vascular endothelial function, endurance exercise capacity, and whole-body glucose metabolism in old mice. Aging Cell 2020; 19: e13074. doi: 10.1111/acel.13074

  7. He SD, Zhang ZY, Sun HJ, Zhu YC, Cao XD, Ye YK, et al. Potential effects of rapeseed peptide Maillard reaction products on aging-related disorder attenuation and gut microbiota modulation in d-galactose induced aging mice. Food Funct 2019; 10: 4291–303. doi: 10.1039/c9fo00791a

  8. Zhang D, Han J, Li Y, Yuan B, Zhou J, Cheong L, et al. Tuna oil alleviates d-galactose induced aging in mice accompanied by modulating gut microbiota and brain protein expression. J Agric Food Chem 2018; 66: 5510–20. doi: 10.1021/acs.jafc.8b00446

  9. Mabbott NA, Kobayashi A, Sehgal A, Bradford BM, Pattison M, Donaldson DS. Aging and the mucosal immune system in the intestine. Biogerontology 2015; 16: 133–45. doi: 10.1007/s10522-014-9498-z

  10. Li J, Wu T, Li N, Wang XN, Chen GY, Lyu XL. Bilberry anthocyanin extract promotes intestinal barrier function and inhibits digestive enzyme activity by regulating the gut microbiota in aging rats. Food Funct 2019; 10: 333–43. doi: 10.1039/c8fo01962b

  11. Verburgh K. Nutrigerontology: why we need a new scientific discipline to develop diets and guidelines to reduce the risk of aging-related diseases. Aging Cell 2015; 14: 17–24. doi: 10.1111/acel.12284

  12. Mayta-Apaza AC, Pottgen E, De Bodt J, Papp N, Marasini D, Howard L, et al. Impact of tart cherries polyphenols on the human gut microbiota and phenolic metabolites in vitro and in vivo. J Nutr Biochem 2018; 59: 160–72. doi: 10.1016/j.jnutbio.2018.04.001

  13. Sharma R, Padwad Y. Perspectives of the potential implications of polyphenols in influencing the interrelationship between oxi-inflammatory stress, cellular senescence and immunosenescence during aging. Trends Food Sci Technol 2020; 98: 41–52. doi: 10.1016/j.tifs.2020.02.004

  14. Yin H, Pan X-C, Wang S-K, Yang L-G, Sun G-J. Protective effect of wheat peptides against small intestinal damage induced by non-steroidal anti-inflammatory drugs in rats. J Integr Agr 2014; 13: 2019–27. doi: 10.1016/s2095-3119(13)60619-x

  15. Niu L-Y, Jiang S-T, Pan L-J. Preparation and evaluation of antioxidant activities of peptides obtained from defatted wheat germ by fermentation. J Food Sci Technol Mysore 2013; 50: 53–61. doi: 10.1007/s13197-011-0318-z

  16. Kan JT, Hood M, Burns C, Scholten J, Chuang J, Tian F, et al. A novel combination of wheat peptides and fucoidan attenuates ethanol-induced gastric mucosal damage through anti-oxidant, anti-inflammatory, and pro-survival mechanisms. Nutrients 2017; 9: 978. doi: 10.3390/nu9090978

  17. Oita S. Extraction and enzymatic degradation of antimicrobial peptides, alpha and beta-thionins, from barley and wheat. J Japanese Soc Food Sci Technol Nippon Shokuhin Kagaku Kogaku Kaishi 2000; 47: 424–30. doi: 10.3136/nskkk.47.424

  18. Egorov TA, Odintsova TI, Pukhalsky VA, Grishin EV. Diversity of wheat anti-microbial peptides. Peptides 2005; 26: 2064–73. doi: 10.1016/j.peptides.2005.03.007

  19. Jeong HJ, Jeong JB, Kim DS, Park JH, Lee JB, Kweon D-H, et al. The cancer preventive peptide lunasin from wheat inhibits core histone acetylation. Cancer Lett 2007; 255: 42–8. doi: 10.1016/j.canlet.2007.03.022

  20. Huang L, Liu B, Ma H, Zhang X. Combined effect of ultrasound and enzymatic treatments on production of ace inhibitory peptides from wheat germ protein. J Food Process Preserv 2014; 38: 1632–40. doi: 10.1111/jfpp.12125

  21. Wang L, Ding Y, Zhang X, Li Y, Wang R, Luo X, et al. Isolation of a novel calcium-binding peptide from wheat germ protein hydrolysates and the prediction for its mechanism of combination. Food Chem 2018; 239: 416–26. doi: 10.1016/j.foodchem.2017.06.090

  22. Yin H, Pan X, Song Z, Wang S, Yang L, Sun G. Protective effect of wheat peptides against indomethacin-induced oxidative stress in IEC-6 cells. Nutrients 2014; 6: 564–74. doi: 10.3390/nu6020564

  23. Tuganbaev T, Mor U, Bashiardes S, Liwinski T, Nobs SP, Leshem A, et al. Diet diurnally regulates small intestinal microbiome-epithelial-immune homeostasis and enteritis. Cell 2020; 182: 1441–+. doi: 10.1016/j.cell.2020.08.027

  24. Mabbott NA. A breakdown in communication? Understanding the effects of aging on the human small intestine epithelium. Clin Sci 2015; 129: 529–31. doi: 10.1042/Cs20150364

  25. Ames BN, Shigenaga MK. Oxidants are a major contributor to aging. Ann NY Acad Sci 1992; 663: 85–96. doi: 10.1111/j.1749-6632.1992.tb38652.x

  26. Zhang ZY, He SD, Cao XD, Ye YK, Yang L, Wang JH, et al. Potential prebiotic activities of soybean peptides Maillard reaction products on modulating gut microbiota to alleviate aging-related disorders in D-galactose-induced ICR mice. J Funct Foods 2020; 65: 103729. doi: 10.1016/j.jff.2019.103729

  27. He SD, Yu M, Sun HJ, Zhang ZY, Zhu YC, Zhao JL, et al. Potential effects of dietary Maillard reaction products derived from 1 to 3 kDa soybean peptides on the aging ICR mice. Food Chem Toxicol 2019; 125: 62–70. doi: 10.1016/j.fct.2018.12.045

  28. Li XM, Shi YH, Wang F, Wang HS, Le GW. In vitro free radical scavenging activities and effect of synthetic oligosaccharides on antioxidant enzymes and lipid peroxidation in aged mice. J Pharm Biomed Anal 2007; 43: 364–370. doi: 10.1016/j.jpba.2006.06.041

  29. Jardinaud F, Banisadr G, Noble F, Melik-Parsadaniantz S, Chen HX, Dugave C, et al. Ontogenic and adult whole body distribution of aminopeptidase N in rat investigated by in vitro autoradiography. Biochimie 2004; 86: 105–13. doi: 10.1016/j.biochi.2003.12.004

  30. Hooton D, Lentle R, Monro J, Wickham M, Simpson R. The secretion and action of brush border enzymes in the mammalian small intestine. Rev Physiol Biochem Pharmacol 2015; 168: 59–118. doi: 10.1007/112_2015_24

  31. Yanaka A, Suzuki H, Shibahara T, Matsui H, Nakahara A, Tanaka N. EGF promotes gastric mucosal restitution by activating Na+/H+ exchange of epithelial cells. Am J Physiol-Gastr L 2002; 282: G866–76. doi: 10.1152/ajpgi.00150.2001

  32. Hou YQ, Wang L, Yi D, Ding BY, Yang ZG, Li J, et al. N-acetylcysteine reduces inflammation in the small intestine by regulating redox, EGF and TLR4 signaling. Amino Acids 2013; 45: 513–22. doi: 10.1007/s00726-012-1295-x

  33. Ferrucci L, Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol 2018; 15: 505–22. doi: 10.1038/s41569-018-0064-2

  34. Koelman L, Pivovarova-Ramich O, Pfeiffer AFH, Grune T, Aleksandrova K. Cytokines for evaluation of chronic inflammatory status in ageing research: reliability and phenotypic characterisation. Immun Ageing 2019; 16: 11. doi: 10.1186/s12979-019-0151-1

  35. Ye L, Mauro TM, Dang E, Wang G, Hu LZ, Yu C, et al. Topical applications of an emollient reduce circulating pro-inflammatory cytokine levels in chronically aged humans: a pilot clinical study. J Eur Acad Dermatol 2019; 33: 2197–201. doi: 10.1111/jdv.15540

  36. Gonzalez OA, Kirakodu S, Novak MJ, Stromberg AJ, Orraca L, Gonzalez-Martinez J, et al. Comparative analysis of microbial sensing molecules in mucosal tissues with aging. Immunobiology 2018; 223: 279–87. doi: 10.1016/j.imbio.2017.10.034

  37. Sanjeewa KKA, Jayawardena TU, Kim H-S, Kim S-Y, Fernando IPS, Wang L, et al. Fucoidan isolated from Padina commersonii inhibit LPS-induced inflammation in macrophages blocking TLR/NF-kappa B signal pathway. Carbohydr Polym 2019; 224: 115195. doi: 10.1016/j.carbpol.2019.115195

  38. Bailey KL, Smith LM, Heires AJ, Katafiasz DM, Romberger DJ, LeVan TD. Aging leads to dysfunctional innate immune responses to TLR2 and TLR4 agonists. Aging Clin Exp Res 2019; 31: 1185–93. doi: 10.1007/s40520-018-1064-0

  39. Lan J, Dou XJ, Li JW, Yang Y, Xue CY, Wang CX, et al. L-arginine ameliorates lipopolysaccharide-induced intestinal inflammation through inhibiting the TLR4/NF-kappa B and MAPK pathways and stimulating beta-defensin expression in vivo and in vitro. J Agricult Food Chem 2020; 68: 2648–63. doi: 10.1021/acs.jafc.9b07611

  40. Man AL, Bertelli E, Rentini S, Regoli M, Briars G, Marini M, et al. Age-associated modifications of intestinal permeability and innate immunity in human small intestine. Clin Sci 2015; 129: 515–27. doi: 10.1042/cs20150046

  41. Sumagin R, Robin AZ, Nusrat A, Parkos CA. Transmigrated neutrophils in the intestinal lumen engage ICAM-1 to regulate the epithelial barrier and neutrophil recruitment. Mucosal Immunol 2014; 7: 905–15. doi: 10.1038/mi.2013.106

  42. Danese S, Semeraro S, Marini M, Roberto I, Armuzzi A, Papa A, et al. Adhesion molecules in inflammatory bowel disease: therapeutic implications for gut inflammation. Digest Liver Dis 2005; 37: 811–18. doi: 10.1016/j.dld.2005.03.013

  43. Laukoetter MG, Nava P, Lee WY, Severson EA, Capaldo CT, Babbin BA, et al. JAM-A regulates permeability and inflammation in the intestine in vivo. J Exp Med 2007; 204: 3067–76. doi: 10.1084/jem.20071416

  44. Mandell KJ, Babbin BA, Nusrat A, Parkos CA. Junctional adhesion molecule 1 regulates epithelial cell morphology through effects on beta 1 Integrins and Rap1 activity. J Biol Chem 2005; 280: 11665–74. doi: 10.1074/jbc.M412650200

  45. Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Bio 2016; 17: 564–80. doi: 10.1038/nrm.2016.80

Published
2022-02-14
How to Cite
Xian Y., Da P., Chao Y., Hui X., Ligang Y., Shaokang W., & Guiju S. (2022). Wheat oligopeptides enhance the intestinal mucosal barrier and alleviate inflammation via the TLR4/Myd88/MAPK signaling pathway in aged mice. Food & Nutrition Research, 66. https://doi.org/10.29219/fnr.v66.5690
Section
Original Articles