Avenanthramide supplementation reduces eccentric exercise-induced inflammation in young men and women

Tianou Zhang, Tong Zhao, Yuzi Zhang, Tao Liu, Gilles Gagnon, Jacqueline Ebrahim, Jodee Johnson, Yi-Fang Chu, Li Li Ji, Tianou Zhang, Tong Zhao, Yuzi Zhang, Tao Liu, Gilles Gagnon, Jacqueline Ebrahim, Jodee Johnson, Yi-Fang Chu, Li Li Ji

Abstract

Background: Avenanthramides (AVA) are a group of di-phenolic acids found only in oats and have shown antioxidant and anti-inflammatory effects in vitro and in vivo. Eccentric muscle contraction is intimately involved in rigorous exercise that activates systemic and local inflammatory responses. The objective of the study is to evaluate whether chronic AVA supplementation could attenuate peripheral inflammatory and immunological markers in human subjects in response to an acute bout of downhill running (DR).

Methods: Eleven male and thirteen female subjects voluntarily participated in this double-blinded, randomized controlled study and were randomly divided into AVA-supplemented (AVA) or control (C) groups. All subjects conducted a DR protocol at - 10% grade with an intensity equivalent to 75% of their maximal heart rate. Blood samples were collected at rest and various time points (0-72 h) after DR (PRE). After an 8-week washout period, participants received two cookies daily containing either 206 mg/kg (AVA) or 0 mg/kg (C) AVA for 8 weeks. Following the oat supplementation regimen, the DR and blood sampling protocols were repeated (POST). Plasma inflammatory and immunological markers were measured using Multiplex immunoassay and muscle soreness was evaluated with pain rating scale.

Results: DR increased plasma creatine kinase (CK) activity (P < 0.01) during PRE, but the response was reduced at 24 and 48 h during POST vs. PRE regardless of AVA status (P < 0.05). Neutrophil respiratory burst (NRB) levels were elevated at 4 and 24 h (P < 0.05) during PRE but were significantly decreased at 0-48 h during POST vs. PRE (P < 0.05 or 0.01). Granulocyte-colony stimulating factor (G-CSF), the neutrophil stimulating cytokine, was also increased in response to DR but showed lower levels in AVA compared to C during POST vs. PRE (P < 0.05). Plasma interleukin-6 (IL-6) content showed an increase at 0 and 4 h during PRE and 0 h during POST (P < 0.01), whereas during POST there was a trend toward a lower IL-6 level in AVA vs. C (P = 0.082). Plasma levels of anti-inflammatory agent interleukin-1 receptor antagonist (IL-1Ra) showed an increase at 4 h during PRE, and was significantly elevated in AVA vs. C during POST. Both soluble vascular cell adhesion molecule-1 (sVCAM-1) and monocyte chemoattractant protein-1 (MCP-1) contents increased at 0 and 24 h post DR during PRE as well as POST sessions, however, sVCAM-1 content was lower in AVA vs. C during POST (P < 0.05) and MCP-1 levels were below resting level at 24, 48 and 72 h during POST (P < 0.05). DR increased muscle pain at all post-DR time points (P < 0.01), but the pain level was alleviated by oat supplementation at 48 and 72 h during POST regardless of AVA treatment (P < 0.05).

Conclusions: Oat AVA supplementation reduced circulatory inflammatory cytokines and inhibited expression of chemokines and cell adhesion molecules induced by DR.

Trial registration: ClinicalTrials.gov identifier: NCT02584946 . Registered 23 October 2015.

Keywords: Avenanthramide; Chemokines; Cytokines; Downhill running; Inflammation.

Conflict of interest statement

J. J. and Y. C. are employees of PepsiCo Inc., which manufactures oatmeal products under the brand name Quaker Oats®. The views expressed in this article are those of the authors and do not necessarily reflect the opinion or policies of PepsiCo Inc.

Figures

Fig. 1
Fig. 1
Study Design
Fig. 2
Fig. 2
Plasma CK activity. Data are shown as mean ± SEM. Exercise effect: *P < 0.05 or **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Supplementation effect: #P < 0.05 or †P = 0.094, POST vs. PRE regardless of time or AVA treatment. Interaction effect: P < 0.05, time×supplementation
Fig. 3
Fig. 3
Neutrophil Respiratory Burst (NRB). Data are shown as mean ± SEM. Exercise effect: *P < 0.05 or **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Supplementation effect: #P < 0.05 or ##P < 0.01, POST vs. PRE regardless of time or AVA treatment. Interaction effect: P < 0.05, time×AVA; P = 0.060, supplementation × AVA; P = 0.059, time×supplementation
Fig. 4
Fig. 4
Plasma G-CSF concentrations. Data are shown as mean ± SEM. Exercise effect: *P < 0.05 or **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Supplementation effect: #P = 0.056, POST vs. PRE regardless of time or AVA treatment. Interaction effect: P < 0.05, supplementation×AVA
Fig. 5
Fig. 5
Plasma IL-6 concentrations. Data are shown as mean ± SEM. Exercise effect: **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Interaction effect: P = 0.082, time×AVA
Fig. 6
Fig. 6
Plasma IL-1Ra concentrations. Data are shown as mean ± SEM. Exercise effect: *P = 0.069 or **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Interaction effect: P < 0.05, time×supplementation×AVA
Fig. 7
Fig. 7
Plasma sVCAM-1 concentrations. Data are shown as mean ± SEM. Exercise effect: **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Supplementation effect: #P = 0.079, POST vs. PRE regardless of time or AVA treatment. Interaction effect: P < 0.05, time×AVA
Fig. 8
Fig. 8
Plasma MCP-1 concentrations. Data are shown as mean ± SEM. Exercise effect: *P < 0.05 or **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Interaction effect: P = 0.064, time×supplementation
Fig. 9
Fig. 9
Pain rating scale. Data are shown as mean ± SEM. Exercise effect: **P < 0.01, 0 h/4 h/24 h/48 h/72 h post-DR vs. Rest. Supplementation effect: #P < 0.05, POST vs. PRE regardless of time or AVA treatment. Interaction effect: P < 0.05, time×supplementation

References

    1. Armstrong RB. Mechanisms of exercise-induced delayed onset muscular soreness: a brief review. Med Sci Sports Exerc. 1984;16(6):529–538.
    1. Cheung K, Hume P, Maxwell L. Delayed onset muscle soreness : treatment strategies and performance factors. Sports Med. 2003;33(2):145–164.
    1. Gulick DT, Kimura IF, Sitler M, Paolone A, Kelly JD. Various treatment techniques on signs and symptoms of delayed onset muscle soreness. J Athl Train. 1996;31(2):145–152.
    1. Proske U, Morgan DL. Muscle damage from eccentric exercise: mechanism, mechanical signs, adaptation and clinical applications. J Physiol Lond. 2001;537(Pt 2):333–345.
    1. Paulsen G, Mikkelsen UR, Raastad T, Peake JM. Leucocytes, cytokines and satellite cells: what role do they play in muscle damage and regeneration following eccentric exercise? Exerc Immunol Rev. 2012;18:42–97.
    1. Butterfield TA, Best TM, Merrick MA. The dual roles of neutrophils and macrophages in inflammation: a critical balance between tissue damage and repair. J Athl Train. 2006;41(4):457–465.
    1. Paulsen G, Crameri R, Benestad HB, Fjeld JG, Mørkrid L, Hallén J, et al. Time course of leukocyte accumulation in human muscle after eccentric exercise. Med Sci Sports Exerc. 2010;42(1):75–85.
    1. Pyne DB, Smith JA, Baker MS, Telford RD, Weidemann MJ. Neutrophil oxidative activity is differentially affected by exercise intensity and type. J Sci Med Sport. 2000;3(1):44–54.
    1. Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Phys Regul Integr Comp Phys. 2010;298(5):R1173–R1187.
    1. Howatson G, van Someren KA. The prevention and treatment of exercise-induced muscle damage. Sports Med. 2008;38(6):483–503.
    1. Collins FW. Oat phenolics: avenanthramides, novel substituted N-cinnamoylanthranilate alkaloids from oat groats and hulls. J Agric Food Chem. 1989;37(1):60–66.
    1. Ji LL, Lay D, Chung E, Fu Y, Peterson DM. Effects of avenanthramides on oxidant generation and antioxidant enzyme activity in exercised rats. Nutr Res. 2003;23(11):1579–1590.
    1. Koenig R, Dickman JR, Kang C, Zhang T, Chu Y-F, Ji LL. Avenanthramide supplementation attenuates exercise-induced inflammation in postmenopausal women. Nutr J. 2014;13:21.
    1. Koenig RT, Dickman JR, Kang C-H, Zhang T, Chu Y-F, Ji LL. Avenanthramide supplementation attenuates eccentric exercise-inflicted blood inflammatory markers in women. Eur J Appl Physiol. 2016;116(1):67–76.
    1. Kang C, Shin WS, Yeo D, Lim W, Zhang T, Ji LL. Anti-inflammatory effect of avenanthramides via NF-κB pathways in C2C12 skeletal muscle cells. Free Radic Biol Med. 2018;117:30–36.
    1. Yeo D, Kang C, Zhang T, Ji LL. Avenanthramides attenuate inflammation and atrophy in muscle cells. J Sport Health Sci. 2019;8(2):189–195.
    1. Zhang T, Ji LL. Avenanthramides. In: Whole grains and their bioactives: Wiley; 2019. p. 307–38. [cited 2019 May 13]. Available from: .
    1. Koenig RT, Dickman JR, Wise ML, Ji LL. Avenanthramides are bioavailable and accumulate in hepatic, cardiac, and skeletal muscle tissue following oral gavage in rats. J Agric Food Chem. 2011;59(12):6438–6443.
    1. Chen C-YO, Milbury PE, Collins FW, Blumberg JB. Avenanthramides are bioavailable and have antioxidant activity in humans after acute consumption of an enriched mixture from oats. J Nutr. 2007;137(6):1375–1382.
    1. Zhang T, Shao J, Gao Y, Chen C, Yao D, Chu YF, et al. Absorption and elimination of oat avenanthramides in humans after acute consumption of oat cookies. Oxid Med Cell Longev. 2017; [cited 2019 Aug 26]. Available from: .
    1. Liu L, Zubik L, Collins FW, Marko M, Meydani M. The antiatherogenic potential of oat phenolic compounds. Atherosclerosis. 2004;175(1):39–49.
    1. Guo W, Wise ML, Collins FW, Meydani M. Avenanthramides, polyphenols from oats, inhibit IL-1β-induced NF-κB activation in endothelial cells. Free Radic Biol Med. 2008;44(3):415–429.
    1. Nie L, Wise ML, Peterson DM, Meydani M. Avenanthramide, a polyphenol from oats, inhibits vascular smooth muscle cell proliferation and enhances nitric oxide production. Atherosclerosis. 2006;186(2):260–266.
    1. Garber CE, Blissmer B, Deschenes MR, Franklin BA, Lamonte MJ, Lee I-M, et al. American College of Sports Medicine position stand. Quantity and quality of exercise for developing and maintaining cardiorespiratory, musculoskeletal, and neuromotor fitness in apparently healthy adults: guidance for prescribing exercise. Med Sci Sports Exerc. 2011;43(7):1334–1359.
    1. Ji LL. Exercise-induced modulation of antioxidant defense. Ann N Y Acad Sci. 2002;959:82–92.
    1. Ji LL, Gomez-Cabrera M-C, Vina J. Role of free radicals and antioxidant signaling in skeletal muscle health and pathology. Infect Disord Drug Targets. 2009;9(4):428–444.
    1. Pedersen BK, Toft AD. Effects of exercise on lymphocytes and cytokines. Br J Sports Med. 2000;34(4):246–251.
    1. Proske U, Allen TJ. Damage to skeletal muscle from eccentric exercise. Exerc Sport Sci Rev. 2005;33(2):98–104.
    1. Liao P, Zhou J, Ji LL, Zhang Y. Eccentric contraction induces inflammatory responses in rat skeletal muscle: role of tumor necrosis factor-alpha. Am J Phys Regul Integr Comp Phys. 2010;298(3):R599–R607.
    1. Ji LL, Kang C, Zhang Y. Exercise-induced hormesis and skeletal muscle health. Free Radic Biol Med. 2016;98:113–122.
    1. Ristow M, Zarse K, Oberbach A, Klöting N, Birringer M, Kiehntopf M, et al. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc Natl Acad Sci U S A. 2009;106(21):8665–8670.
    1. Fusco D, Colloca G, Lo Monaco MR, Cesari M. Effects of antioxidant supplementation on the aging process. Clin Interv Aging. 2007;2(3):377–387.
    1. Pham-Huy LA, He H, Pham-Huy C. Free radicals, antioxidants in disease and health. Int J Biomed Sci. 2008;4(2):89–96.
    1. Willcox JK, Ash SL, Catignani GL. Antioxidants and prevention of chronic disease. Crit Rev Food Sci Nutr. 2004;44(4):275–295.
    1. Mayama S, Matsuura Y, Iida H, Tani T. The role of avenalumin in the resistance of oat to crown rust, Puccinia coronata f. sp. avenae. Physiol Plant Pathol. 1982;20(2):189–199.
    1. Miyagawa H, Ishihara A, Nishimoto T, Ueno T, Mayama S. Induction of avenanthramides in oat leaves inoculated with crown rust fungus, Puccinia coronata f. sp. avenae. Biosci Biotechnol Biochem. 1995;59(12):2305–2306.
    1. Dahlgren C, Karlsson A. Respiratory burst in human neutrophils. J Immunol Methods. 1999;232(1–2):3–14.
    1. Pizza FX, Cavender D, Stockard A, Baylies H, Beighle A. Anti-inflammatory doses of ibuprofen: effect on neutrophils and exercise-induced muscle injury. Int J Sports Med. 1999;20(2):98–102.
    1. Petersen EW, Ostrowski K, Ibfelt T, Richelle M, Offord E, Halkjaer-Kristensen J, et al. Effect of vitamin supplementation on cytokine response and on muscle damage after strenuous exercise. Am J Phys Cell Phys. 2001;280(6):C1570–C1575.
    1. Pizza FX, Mitchell JB, Davis BH, Starling RD, Holtz RW, Bigelow N. Exercise-induced muscle damage: effect on circulating leukocyte and lymphocyte subsets. Med Sci Sports Exerc. 1995;27(3):363–370.
    1. Smith LL, Anwar A, Fragen M, Rananto C, Johnson R, Holbert D. Cytokines and cell adhesion molecules associated with high-intensity eccentric exercise. Eur J Appl Physiol. 2000;82(1–2):61–67.
    1. Paulsen G, Benestad HB, Strøm-Gundersen I, Mørkrid L, Lappegård KT, Raastad T. Delayed leukocytosis and cytokine response to high-force eccentric exercise. Med Sci Sports Exerc. 2005;37(11):1877–1883.
    1. Hirose L, Nosaka K, Newton M, Laveder A, Kano M, Peake J, et al. Changes in inflammatory mediators following eccentric exercise of the elbow flexors. Exerc Immunol Rev. 2004;10:75–90.
    1. Peake JM, Suzuki K, Wilson G, Hordern M, Nosaka K, Mackinnon L, et al. Exercise-induced muscle damage, plasma cytokines, and markers of neutrophil activation. Med Sci Sports Exerc. 2005;37(5):737–745.
    1. Peake JM, Suzuki K, Hordern M, Wilson G, Nosaka K, Coombes JS. Plasma cytokine changes in relation to exercise intensity and muscle damage. Eur J Appl Physiol. 2005;95(5–6):514–521.
    1. Pereira Panza VS, Diefenthaeler F, da Silva EL. Benefits of dietary phytochemical supplementation on eccentric exercise-induced muscle damage: is including antioxidants enough? Nutrition. 2015;31(9):1072–1082.
    1. Peake J, Nosaka K, Suzuki K. Characterization of inflammatory responses to eccentric exercise in humans. Exerc Immunol Rev. 2005;11:64–85.
    1. Pedersen BK, Steensberg A, Schjerling P. Muscle-derived interleukin-6: possible biological effects. J Physiol. 2001;536(Pt 2):329–337.
    1. Croisier JL, Camus G, Venneman I, Deby-Dupont G, Juchmès-Ferir A, Lamy M, et al. Effects of training on exercise-induced muscle damage and interleukin 6 production. Muscle Nerve. 1999;22(2):208–212.
    1. Toft AD, Jensen LB, Bruunsgaard H, Ibfelt T, Halkjaer-Kristensen J, Febbraio M, et al. Cytokine response to eccentric exercise in young and elderly humans. Am J Phys Cell Phys. 2002;283(1):C289–C295.
    1. Arend WP, Malyak M, Guthridge CJ, Gabay C. Interleukin-1 receptor antagonist: role in biology. Annu Rev Immunol. 1998;16:27–55.
    1. Prame Kumar K, Nicholls AJ, Wong CHY. Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease. Cell Tissue Res. 2018;371(3):551–565.
    1. Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules. Blood. 1994;84(7):2068–2101.
    1. Edelman GM. Cell adhesion molecules. Science. 1983;219(4584):450–457.
    1. Elangbam CS, Qualls CW, Dahlgren RR. Cell adhesion molecules--update. Vet Pathol. 1997;34(1):61–73.
    1. Pigott R, Dillon LP, Hemingway IH, Gearing AJ. Soluble forms of E-selectin, ICAM-1 and VCAM-1 are present in the supernatants of cytokine activated cultured endothelial cells. Biochem Biophys Res Commun. 1992;187(2):584–589.
    1. Nakai K, Itoh C, Kawazoe K, Miura Y, Sotoyanagi H, Hotta K, et al. Concentration of soluble vascular cell adhesion molecule-1 (VCAM-1) correlated with expression of VCAM-1 mRNA in the human atherosclerotic aorta. Coron Artery Dis. 1995;6(6):497–502.
    1. Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interf Cytokine Res. 2009;29(6):313–326.

Source: PubMed

3
Předplatit