DOI QR코드

DOI QR Code

Anti-inflammatory effect of enzymatic hydrolysates from Styela clava flesh tissue in lipopolysaccharide-stimulated RAW 264.7 macrophages and in vivo zebrafish model

  • Ko, Seok-Chun (Institute of Marine Biotechnology, Pukyong National University) ;
  • Jeon, You-Jin (Department of Marine Life Science, Jeju National University)
  • Received : 2014.06.05
  • Accepted : 2014.10.17
  • Published : 2015.06.01

Abstract

BACKGROUND/OBJECTIVES: In this study, potential anti-inflammatory effect of enzymatic hydrolysates from Styela clava flesh tissue was assessed via nitric oxide (NO) production in lipopolysaccahride (LPS) induced RAW 264.7 macrophages and in vivo zebrafish model. MATERIALS/METHODS: We investigated the ability of enzymatic hydrolysates from Styela clava flesh tissue to inhibit LPS-induced expression of pro-inflammatory mediators in RAW 264.7 macrophages, and the molecular mechanism through which this inhibition occurred. In addition, we evaluated anti-inflammatory effect of enzymatic hydrolysates against a LPS-exposed in in vivo zebrafish model. RESULTS: Among the enzymatic hydrolysates, Protamex-proteolytic hydrolysate exhibited the highest NO inhibitory effect and was fractionated into three ranges of molecular weight by using ultrafiltration (UF) membranes (MWCO 5 kDa and 10 kDa). The above 10 kDa fraction down-regulated LPS-induced expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), thereby reducing production of NO and prostaglandin $E_2$ ($PGE_2$) in LPS-activated RAW 264.7 macrophages. The above 10 kDa fraction suppressed LPS-induced production of pro-inflammatory cytokines, including interleukin $(IL)-1{\beta}$, IL-6, and tumor necrosis factor $(TNF)-{\alpha}$. In addition, the above 10 kDa fraction inhibited LPS-induced phosphorylation of extracellular signal-regulated kinases (ERKs), c-Jun N-terminal kinase (JNK), and p38. Furthermore, NO production in live zebrafish induced by LPS was reduced by addition of the above 10 kDa fraction from S. clava enzymatic hydrolysate. CONCLUSION: The results of this study suggested that hydrolysates derived from S. clava flesh tissue would be new anti-inflammation materials in functional resources.

Keywords

References

  1. Heo SJ, Yoon WJ, Kim KN, Ahn GN, Kang SM, Kang DH, Affan A, Oh C, Jung WK, Jeon YJ. Evaluation of anti-inflammatory effect of fucoxanthin isolated from brown algae in lipopolysaccharidestimulated RAW 264.7 macrophages. Food Chem Toxicol 2010;48: 2045-51. https://doi.org/10.1016/j.fct.2010.05.003
  2. Lee SJ, Kim EK, Kim YS, Hwang JW, Lee KH, Choi DK, Kang H, Moon SH, Jeon BT, Park PJ. Purification and characterization of a nitric oxide inhibitory peptide from Ruditapes philippinarum. Food Chem Toxicol 2012;50:1660-6. https://doi.org/10.1016/j.fct.2012.02.021
  3. Kim SY, Jeong HJ, Kim DW, Kim MJ, An JJ, Sohn EJ, Kang HW, Shin MJ, Ahn EH, Kwon SW, Kim DS, Cho SW, Park J, Eum WS, Choi SY. Transduced PEP-1-FK506BP inhibits the inflammatory response in the Raw 264.7 cell and mouse models. Immunobiology 2011; 216:771-81. https://doi.org/10.1016/j.imbio.2010.12.008
  4. Schoonheim PJ, Chatzopoulou A, Schaaf MJ. The zebrafish as an in vivo model system for glucocorticoid resistance. Steroids 2010; 75:918-25. https://doi.org/10.1016/j.steroids.2010.05.010
  5. Ko SC, Cha SH, Heo SJ, Lee SH, Kang SM, Jeon YJ. Protective effect of Ecklonia cava on UVB-induced oxidative stress: in vitro and in vivo zebrafish model. J Appl Phycol 2011;23:697-708. https://doi.org/10.1007/s10811-010-9565-z
  6. Alex D, Lam IK, Lin Z, Lee SM. Indirubin shows anti-angiogenic activity in an in vivo zebrafish model and an in vitro HUVEC model. J Ethnopharmacol 2010;131:242-7. https://doi.org/10.1016/j.jep.2010.05.016
  7. Park KH, Cho KH. A zebrafish model for the rapid evaluation of pro-oxidative and inflammatory death by lipopolysaccharide, oxidized low-density lipoproteins, and glycated high-density lipoproteins. Fish Shellfish Immunol 2011;31:904-10. https://doi.org/10.1016/j.fsi.2011.08.006
  8. Jin S, Cho KH. Water extracts of cinnamon and clove exhibits potent inhibition of protein glycation and anti-atherosclerotic activity in vitro and in vivo hypolipidemic activity in zebrafish. Food Chem Toxicol 2011;49:1521-9. https://doi.org/10.1016/j.fct.2011.03.043
  9. Lee DW, You DH, Yang EK, Jang IC, Bae MS, Jeon YJ, Kim SJ, Lee SC. Antioxidant and ACE inhibitory activities of Styela clava according to harvesting time. J Korean Soc Food Sci Nutr 2010;39:331-6. https://doi.org/10.3746/jkfn.2010.39.3.331
  10. Kim JJ, Kim SJ, Kim SH, Park HR, Lee SC. Antioxidant and anticancer activities of extracts from Styela clava according to the processing methods and solvents. J Korean Soc Food Sci Nutr 2006;35:278-83. https://doi.org/10.3746/jkfn.2006.35.3.278
  11. Kim JM, Park HR, Lee SC, Park E. Ethanol induced leucocytic and hepatic DNA strand breaks are prevented by Styela clava and Styela plicata supplementation in male SD rats. J Korean Soc Food Sci Nutr 2007;36:1271-8. https://doi.org/10.3746/jkfn.2007.36.10.1271
  12. Ko SC, Lee JK, Byun HG, Lee SC, Jeon YJ. Purification and characterization of angiotensin I-converting enzyme inhibitory peptide from enzymatic hydrolysates of Styela clava flesh tissue. Process Biochem 2012;47:34-40. https://doi.org/10.1016/j.procbio.2011.10.005
  13. Ko SC, Kim DG, Han CH, Lee YJ, Lee JK, Byun HG, Lee SC, Park SJ, Lee DH, Jeon YJ. Nitric oxide-mediated vasorelaxation effects of anti-angiotensin I-converting enzyme (ACE) peptide from Styela clava flesh tissue and its anti-hypertensive effect in spontaneously hypertensive rats. Food Chem 2012;134:1141-5. https://doi.org/10.1016/j.foodchem.2012.02.210
  14. Chen C, Chi YJ, Zhao MY, Xu W. Influence of degree of hydrolysis on functional properties, antioxidant and ACE inhibitory activities of egg white protein hydrolysate. Food Sci Biotechnol 2012;21: 27-34. https://doi.org/10.1007/s10068-012-0004-6
  15. Jeon YJ, Byun HG, Kim SK. Improvement of functional properties of cod frame protein hydrolysates using ultrafiltration membranes. Process Biochem 1999;35:471-8. https://doi.org/10.1016/S0032-9592(99)00098-9
  16. Ko SC, Kang M, Lee JK, Byun HG, Kim SK, Lee SC, Jeon BT, Park PJ, Jung WK, Jeon YJ. Effect of angiotensin I-converting enzyme (ACE) inhibitory peptide purified from enzymatic hydrolysates of Styela plicata. Eur Food Res Technol 2011;233:915-22. https://doi.org/10.1007/s00217-011-1585-7
  17. Byun HG, Kim SK. Purification and characterization of angiotensin I converting enzyme (ACE) inhibitory peptides from Alaska pollack (Theragra chalcogramma) skin. Process Biochem 2001;36:1155-62. https://doi.org/10.1016/S0032-9592(00)00297-1
  18. Cho JY, Baik KU, Jung JH, Park MH. In vitro anti-inflammatory effects of cynaropicrin, a sesquiterpene lactone, from Saussurea lappa. Eur J Pharmacol 2000;398:399-407. https://doi.org/10.1016/S0014-2999(00)00337-X
  19. Wijesinghe WA, Kim EA, Kang MC, Lee WW, Lee HS, Vairappan CS, Jeon YJ. Assessment of anti-inflammatory effect of 5${\beta}$-hydroxypalisadin B isolated from red seaweed Laurencia snackeyi in zebrafish embryo in vivo model. Environ Toxicol Pharmacol 2014;37:110-7. https://doi.org/10.1016/j.etap.2013.11.006
  20. Samarakoon KW, Ko JY, Shah MM, Lee JH, Kang MC, Kwon ON, Lee JB, Jeon YJ. In vitro studies of anti-inflammatory and anticancer activities of organic solvent extracts from cultured marine microalgae. Algae 2013;28:111-9. https://doi.org/10.4490/algae.2013.28.1.111
  21. Kim HK, Cheon BS, Kim YH, Kim SY, Kim HP. Effects of naturally occurring flavonoids on nitric oxide production in the macrophage cell line RAW 264.7 and their structure-activity relationships. Biochem Pharmacol 1999;58:759-65. https://doi.org/10.1016/S0006-2952(99)00160-4
  22. Kim EK, Kim YS, Hwang JW, Kang SH, Choi DK, Lee KH, Lee JS, Moon SH, Jeon BT, Park PJ. Purification of a novel nitric oxide inhibitory peptide derived from enzymatic hydrolysates of Mytilus coruscus. Fish Shellfish Immunol 2013;34:1416-20. https://doi.org/10.1016/j.fsi.2013.02.023
  23. Lee SJ, Kim EK, Kim YS, Hwang JW, Lee KH, Choi DK, Kang H, Moon SH, Jeon BT, Park PJ. Purification and characterization of a nitric oxide inhibitory peptide from Ruditapes philippinarum. Food Chem Toxicol 2012;50:1660-6. https://doi.org/10.1016/j.fct.2012.02.021
  24. Hsu KC, Li-Chan EC, Jao CL. Antiproliferative activity of peptides prepared from enzymatic hydrolysates of tuna dark muscle on human breast cancer cell line MCF-7. Food Chem 2011;126:617-22. https://doi.org/10.1016/j.foodchem.2010.11.066
  25. Chen J, Wang Y, Zhong Q, Wu Y, Xia W. Purification and characterization of a novel angiotensin-I converting enzyme (ACE) inhibitory peptide derived from enzymatic hydrolysate of grass carp protein. Peptides 2012;33:52-8. https://doi.org/10.1016/j.peptides.2011.11.006
  26. Ko SC, Kim D, Jeon YJ. Protective effect of a novel antioxidative peptide purified from a marine Chlorella ellipsoidea protein against free radical-induced oxidative stress. Food Chem Toxicol 2012;50: 2294-302. https://doi.org/10.1016/j.fct.2012.04.022
  27. Bougatef A, Nedjar-Arroume N, Manni L, Ravallec R, Barkia A, Guillochon D, Nasri M. Purification and identification of novel antioxidant peptides from enzymatic hydrolysates of sardinelle (Sardinella aurita) by-products proteins. Food Chem 2010;118: 559-65. https://doi.org/10.1016/j.foodchem.2009.05.021
  28. Park HY, Han MH, Park C, Jin CY, Kim GY, Choi IW, Kim ND, Nam TJ, Kwon TK, Choi YH. Anti-inflammatory effects of fucoidan through inhibition of NF-${\kappa}B$, MAPK and Akt activation in lipopolysaccharideinduced BV2 microglia cells. Food Chem Toxicol 2011;49:1745-52. https://doi.org/10.1016/j.fct.2011.04.020
  29. Ahmad N, Chen LC, Gordon MA, Laskin JD, Laskin DL. Regulation of cyclooxygenase-2 by nitric oxide in activated hepatic macrophages during acute endotoxemia. J Leukoc Biol 2002;71:1005-11.
  30. Hocherl K, Dreher F, Kurtz A, Bucher M. Cyclooxygenase-2 inhibition attenuates lipopolysaccharide-induced cardiovascular failure. Hypertension 2002;40:947-53. https://doi.org/10.1161/01.HYP.0000041221.13644.B9
  31. Kim KN, Heo SJ, Yoon WJ, Kang SM, Ahn G, Yi TH, Jeon YJ. Fucoxanthin inhibits the inflammatory response by suppressing the activation of NF-${\kappa}$B and MAPKs in lipopolysaccharide-induced RAW 264.7 macrophages. Eur J Pharmacol 2010;649:369-75. https://doi.org/10.1016/j.ejphar.2010.09.032
  32. Ghosh S, Karin M. Missing pieces in the NF-kappaB puzzle. Cell 2002;109 Suppl:S81-96. https://doi.org/10.1016/S0092-8674(02)00703-1
  33. Lee HS, Ryu DS, Lee GS, Lee DS. Anti-inflammatory effects of dichloromethane fraction from Orostachys japonicus in RAW 264.7 cells: suppression of NF-${\kappa}$B activation and MAPK signaling. J Ethnopharmacol 2012;140:271-6. https://doi.org/10.1016/j.jep.2012.01.016
  34. Hseu YC, Wu FY, Wu JJ, Chen JY, Chang WH, Lu FJ, Lai YC, Yang HL. Anti-inflammatory potential of Antrodia Camphorata through inhibition of iNOS, COX-2 and cytokines via the NF-kappaB pathway. Int Immunopharmacol 2005;5:1914-25. https://doi.org/10.1016/j.intimp.2005.06.013
  35. Laskin DL, Pendino KJ. Macrophages and inflammatory mediators in tissue injury. Annu Rev Pharmacol Toxicol 1995;35:655-77. https://doi.org/10.1146/annurev.pa.35.040195.003255
  36. Kyriakis JM, Avruch J. Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update. Physiol Rev 2012;92:689-737. https://doi.org/10.1152/physrev.00028.2011
  37. Ajizian SJ, English BK, Meals EA. Specific inhibitors of p38 and extracellular signal-regulated kinase mitogen-activated protein kinase pathways block inducible nitric oxide synthase and tumor necrosis factor accumulation in murine macrophages stimulated with lipopolysaccharide and interferon-gamma. J Infect Dis 1999;179:939-44. https://doi.org/10.1086/314659
  38. Hill AJ, Teraoka H, Heideman W, Peterson RE. Zebrafish as a model vertebrate for investigating chemical toxicity. Toxicol Sci 2005; 86:6-19. https://doi.org/10.1093/toxsci/kfi110
  39. Henry TR, Spitsbergen JM, Hornung MW, Abnet CC, Peterson RE. Early life stage toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in zebrafish (Danio rerio). Toxicol Appl Pharmacol 1997;142:56-68.
  40. Vo TS, Ryu B, Kim SK. Purification of novel anti-inflammatory peptides from enzymatic hydrolysate of the edible microalgal Spirulina maxima. J Funct Foods 2013;5:1336-46. https://doi.org/10.1016/j.jff.2013.05.001
  41. Ahn CB, Cho YS, Je JY. Purification and anti-inflammatory action of tripeptide from salmon pectoral fin byproduct protein hydrolysate. Food Chem 2015;168:151-6. https://doi.org/10.1016/j.foodchem.2014.05.112
  42. Kim EK, Kim YS, Hwang JW, Kang SH, Choi DK, Lee KH, Lee JS, Moon SH, Jeon BT, Park PJ. Purification of a novel nitric oxide inhibitory peptide derived from enzymatic hydrolysates of Mytilus coruscus. Fish Shellfish Immunol 2013;34:1416-20. https://doi.org/10.1016/j.fsi.2013.02.023
  43. Byun HG, Lee JK, Park HG, Jeon JK, Kim SK. Antioxidant peptides isolated from the marine rotifer, Brachionus rotundiformis. Process Biochem 2009;44:842-6. https://doi.org/10.1016/j.procbio.2009.04.003
  44. Je JY, Qian ZJ, Byun HG, Kim SK. Purification and characterization of an antioxidant peptide obtained from tuna backbone protein by enzymatic hydrolysis. Process Biochem 2007;42:840-6. https://doi.org/10.1016/j.procbio.2007.02.006
  45. Heaney RP. Factors influencing the measurement of bioavailability, taking calcium as a model. J Nutr 2001;131:1344S-1348S. https://doi.org/10.1093/jn/131.4.1344S
  46. Phelan M, Aherne A, FitzGerald RJ, O'Brien NM. Casein-derived bioactive peptides: Biological effects, industrial uses, safety aspects and regulatory status. Int Dairy J 2009;19:643-54. https://doi.org/10.1016/j.idairyj.2009.06.001

Cited by

  1. B Signaling Pathways in LPS-Stimulated RAW 264.7 Cells vol.2015, pp.1741-4288, 2015, https://doi.org/10.1155/2015/474509
  2. Immunomodulatory effects of polysaccharide compounds in macrophages revealed by high resolution AFM vol.38, pp.6, 2016, https://doi.org/10.1002/sca.21329
  3. Comparison of the Biological Activities of Electrodialysis-desalted Bioactive Compounds from the Halophyte Suaeda japonica vol.49, pp.2, 2016, https://doi.org/10.5657/KFAS.2016.0124
  4. Peptides, Peptidomimetics, and Polypeptides from Marine Sources: A Wealth of Natural Sources for Pharmaceutical Applications vol.15, pp.4, 2017, https://doi.org/10.3390/md15040124
  5. Antioxidant and anti-inflammatory functionality of ten Sri Lankan seaweed extracts obtained by carbohydrase assisted extraction pp.2092-6456, 2018, https://doi.org/10.1007/s10068-018-0406-1
  6. Spermidine Protects against Oxidative Stress in Inflammation Models Using Macrophages and Zebrafish vol.26, pp.2, 2018, https://doi.org/10.4062/biomolther.2016.272
  7. Evaluation of bioactivity of phenolic compounds from the brown seaweed of Sargassum fusiforme and development of their stable emulsion vol.30, pp.3, 2018, https://doi.org/10.1007/s10811-017-1383-0
  8. Physical and functional properties of tunicate (Styela clava) hydrolysate obtained from pressurized hydrothermal process vol.20, pp.7, 2015, https://doi.org/10.1186/s41240-017-0060-1
  9. The pepsinolytic hydrolysate from Johnius belengerii frame inhibited LPS-stimulated production of pro-inflammatory mediators via the inactivating of JNK and NF-κB pathways in RAW 264.7 macrophage vol.21, pp.5, 2018, https://doi.org/10.1186/s41240-018-0091-2
  10. Anti-inflammatory effect of ozonated krill (Euphausia superba) oil in lipopolysaccharide-stimulated RAW 264.7 macrophages vol.21, pp.6, 2015, https://doi.org/10.1186/s41240-018-0092-1
  11. Phylogenetic Analysis and Screening of Antimicrobial and Antiproliferative Activities of Culturable Bacteria Associated with the Ascidian Styela clava from the Yellow Sea, China vol.2019, pp.None, 2015, https://doi.org/10.1155/2019/7851251
  12. Anti-inflammatory effect of water extracts obtained from doenjang in LPS-stimulated RAW 264.7 cells vol.39, pp.4, 2015, https://doi.org/10.1590/fst.15918
  13. 건조 방법에 따른 홍해삼(Stipchopus japonicus) 효소 가수분해물의 지방 축적 억제 효과 vol.53, pp.5, 2015, https://doi.org/10.5657/kfas.2020.0707
  14. Anti-Inflammatory Effects of Antarctic Lichen Umbilicaria antarctica Methanol Extract in Lipopolysaccharide-Stimulated RAW 264.7 Macrophage Cells and Zebrafish Model vol.2021, pp.None, 2015, https://doi.org/10.1155/2021/8812090
  15. Blue Whiting Protein Hydrolysates Exhibit Antioxidant and Immunomodulatory Activities in Stimulated Murine RAW264.7 Cells vol.11, pp.20, 2021, https://doi.org/10.3390/app11209762
  16. Diverse Krill Lipid Fractions Differentially Reduce LPS-Induced Inflammatory Markers in RAW264.7 Macrophages In Vitro vol.10, pp.11, 2015, https://doi.org/10.3390/foods10112887
  17. Anti-inflammatory activity of a new compound from Vernonia amygdalina vol.35, pp.23, 2015, https://doi.org/10.1080/14786419.2020.1788556
  18. Isolation and Characterization of Polysaccharides from the Ascidian Styela clava vol.14, pp.1, 2015, https://doi.org/10.3390/polym14010016