Journal Home > Volume 10 , Issue 1

Previous studies have suggested that polypeptides extracted from milk, soybean, fish, eggs, and meat possess potential anti-inflammatory effects. To date, few studies have reported the anti-inflammatory function of sturgeon peptides and their underlying mechanisms are unknown. The current study was therefore to determine the anti-inflammatory potential of sturgeon peptides with lipopolysaccharide (LPS)-induced RAW264.7 inflammatory model. Pepsin hydrolysate (PeH) was purified by ultrafiltration and Sephadex G-15 gel filtration chromatography. PeH significantly reduced the inflammatory mediator (NO) and inflammatory cytokines (IL-6、TNF-α and IL-1β) expression in a dose-dependent manner. Moreover, the purified sturgeon peptide (F2) possessed strong antioxidant potential and effectively inhibited DPPH and ABTS free radicals. F2 significantly suppressed the expression of MAPKs, IκBα, and NF-κB p65, indicating that F2 exerted anti-inflammatory influence by the inhibition of MAPK and NF-κB pathways.


menu
Abstract
Full text
Outline
About this article

Peptide fraction from sturgeon muscle by pepsin hydrolysis exerts anti-inflammatory effects in LPS-stimulated RAW264.7 macrophages via MAPK and NF-κB pathways

Show Author's information Ruichang Gaoa,bWanghui ShuaYang ShenaQuancai SunaWengang JinbDajing LicYing LicLi Yuana( )
School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
Bio-resources Key Laboratory of Shaanxi Province, School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, 723001, China
Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, Jiangsu, China

Abstract

Previous studies have suggested that polypeptides extracted from milk, soybean, fish, eggs, and meat possess potential anti-inflammatory effects. To date, few studies have reported the anti-inflammatory function of sturgeon peptides and their underlying mechanisms are unknown. The current study was therefore to determine the anti-inflammatory potential of sturgeon peptides with lipopolysaccharide (LPS)-induced RAW264.7 inflammatory model. Pepsin hydrolysate (PeH) was purified by ultrafiltration and Sephadex G-15 gel filtration chromatography. PeH significantly reduced the inflammatory mediator (NO) and inflammatory cytokines (IL-6、TNF-α and IL-1β) expression in a dose-dependent manner. Moreover, the purified sturgeon peptide (F2) possessed strong antioxidant potential and effectively inhibited DPPH and ABTS free radicals. F2 significantly suppressed the expression of MAPKs, IκBα, and NF-κB p65, indicating that F2 exerted anti-inflammatory influence by the inhibition of MAPK and NF-κB pathways.

Keywords: Antioxidant, Enzymatic hydrolysis, Sturgeon, Anti-inflammation mechanism, RAW264.7 macrophages

References(49)

[1]

N. Kishore, P. Kumar, K. Shanker, et al., Human disorders associated with inflammation and the evolving role of natural products to overcome, Eur. J. Med. Chem. 179 (2019) 272–309 https://doi.org/10.1016/j.ejmech.2019.06.034.

[2]

S.T. Ahmed, L.B. Ivashkiv, Inhibition of IL-6 and IL-10 signaling and stat activation by inflammatory and stress pathways, J. Immunol. 165 (9) (2000) 5227–5237 https://doi.org/10.4049/jimmunol.165.9.5227.

[3]

M. Del Buono, A. Abbate, S. Toldo, Interplay of inflammation, oxidative stress and cardiovascular disease in rheumatoid arthritis, Heart 104 (24) (2018) 1991–1992 https://doi.org/10.1136/heartjnl-2018-313313.

[4]

Rojas-Gutierrez, G. Muñoz-Arenas, S. Treviño, et al., Alzheimer’s disease and metabolic syndrome: a link from oxidative stress and inflammation to neurodegeneration, Synapse 71 (10) (2017) e21990 https://doi.org/10.1002/syn.21990.

[5]

L.N. Dooley, K.R. Kuhlman, T.F. Robles, et al., The role of inflammation in core features of depression: insights from paradigms using exogenously-induced inflammation, Neurosci. Biobehav. R. 94 (2018) 219–237 https://doi.org/10.1016/j.neubiorev.2018.09.006.

[6]

J. Kay, E. Thadhani, L. Samson, et al., Inflammation-induced DNA damage, mutations and cancer, DNA Repair (Amst) 83 (2019) 102673 https://doi.org/10.1016/j.dnarep.2019.102673.

[7]

D. Gilroy, R.D. Maeyer, New insights into the resolution of inflammation, Semin. Immunol. 27 (3) (2015) 161–168 https://doi.org/10.1016/j.smim.2015. 05.003.

[8]

S. Li, G. Chen, C. Zhang, et al., Research progress of natural antioxidants in foods for the treatment of diseases, Food Sci. Hum. Wellness 3 (3-4) (2014) 110–116 https://doi.org/10.1016/j.fshw.2014.11.002.

[9]

Y. Lin, M. Jiang, W. Chen, et al., Cancer and ER stress: mutual crosstalk between autophagy, oxidative stress and inflammatory response, Biomed. Pharmacother. 118 (2019) 109249 https://doi.org/10.1016/j.biopha.2019.109249.

[10]

M. Offengenden, S. Chakrabarti, J. Wu, Chicken collagen hydrolysates differentially mediate anti-inflammatory activity and type I collagen synthesis on human dermal fibroblasts, Food Sci. Hum. Wellness 7 (2) (2018) 138–147 https://doi.org/10.1016/j.fshw.2018.02.002.

[11]

D. Peña-Oyarzun, R. Bravo-Sagua, A. Diaz-Vega, et al., Autophagy and oxidative stress in non-communicable diseases: a matter of the inflammatory state? Free Radical Bio. Med. 124 (2018) 61–78 https://doi.org/10.1016/j.freeradbiomed.2018.05.084.

[12]

B. Fatemeh, S. Bark, C. Kwon, et al., Anti-inflammatory and antioxidant properties of peptides released from β-Lactoglobulin by high hydrostatic pressure-assisted enzymatic hydrolysis, Molecules 22 (6) (2017) 949 http://doi.org/10.3390/molecules22060949.

[13]

E.E.M. Leo, J.J.A. Fernández, M.R.S. Campos, Biopeptides with antioxidant and anti-inflammatory potential in the prevention and treatment of diabesity disease, Biomed. Pharmacother. 83 (2016) 816–826 https://doi.org/10.1016/j.biopha.2016.07.051.

[14]

E. Zielińska, B. Baraniak, M. Karaś, Antioxidant and anti-inflammatory activities of hydrolysates and peptide fractions obtained by enzymatic hydrolysis of selected heat-treated edible insects, Nutrients 9 (9) (2017) 1–14 https://doi.org/10.3390/nu9090970.

[15]

S.S.O. Hung, Recent advances in sturgeon nutrition, Anim. Nutr. 3 (3) (2017) 191–204 https://doi.org/10.1016/j.aninu.2017.05.005.

[16]

M. Nikoo, S. Benjakul, A. Ehsani, et al., Antioxidant and cryoprotective effects of a tetrapeptide isolated from Amur sturgeon skin gelatin, J. Funct. Foods 7 (2014) 609–620 https://doi.org/10.1016/j.jff.2013.12.024.

[17]

M. Nikoo, S. Benjakul, X. Xu, Antioxidant and cryoprotective effects of Amur sturgeon skin gelatin hydrolysate in unwashed fish mince, Food Chem. 181 (2015) 295–303 https://doi.org/10.1016/j.foodchem.2015.02.095.

[18]

A. Noman, Y. Xu, W. Q, et al., Influence of enzymatic hydrolysis conditions on the degree of hydrolysis and functional properties of protein hydrolysate obtained from Chinese sturgeon (Acipenser sinensis) by using papain enzyme, Process Biochem. 67 (2018) 19–28 https://doi.org/10.1016/j.procbio.2018.01.009.

[19]
Kjeldahl Method, Encyclopedia of Genetics, Genomics, Proteomics and Informatics, Kjeldahl Method, Encyclopedia of Genetics, Genomics, Proteomics and Informatics, (2008) 1063-1063.
[20]

C.B. Ahn, J.Y. Je, Y.S. Cho, Antioxidant and anti-inflammatory peptide fraction from salmon byproduct protein hydrolysates by peptic hydrolysis, Food Res. Int. 49 (1) (2012) 92–98 https://doi.org/10.1016/j.foodres.2012.08.002.

[21]

M. Mirzaei, S. Mirdamadi, M.R. Ehsani, et al., Purification and identification of antioxidant and ACE-inhibitory peptide from Saccharomyces cerevisiae protein hydrolysate, J. Funct. Foods 19 (2015) 259–268 https://doi.org/10.1016/j.jff.2015.09.031.

[22]

S.W. Moon, C.B. Ahn, Y. Oh, et al., Lotus (Nelumbo nucifera) seed protein isolate exerts anti-inflammatory and antioxidant effects in LPS-stimulated RAW264.7 macrophages via inhibiting NF-κB and MAPK pathways, and upregulating catalase activity, Int. J. Biol. Macromol. 134 (2019) 791–797 https://doi.org/10.1016/j.ijbiomac.2019.05.094.

[23]

Y. Peng, Q. Sun, Y. Park, Chicoric acid promotes glucose uptake and Akt phosphorylation via AMP-activated protein kinase α-dependent pathway, J. Funct. Foods 59 (2019) 8–15 https://doi.org/10.1016/j.jff.2019.05.020.

[24]

J. Song, T. Li, X. Cheng, et al., Sea cucumber peptides exert anti-inflammatory activity through suppressing NF-κB and MAPK and inducing HO-1 in RAW264.7 macrophages, Food Funct. 7 (6) (2016) 2773 https://doi.org/10.1039/C5FO01622C.

[25]

Q. Sun, Z. Zhang, R. Zhang, et al., Development of functional or medical foods for oral administration of insulin for diabetes treatment: gastroprotective edible microgels, J. Agric. Food Chem. 66 (19) (2018) 4820–4826 https://doi.org/10.1021/acs.jafc.8b00233.

[26]

L. Yuan, J. Lin, Y. Xu, et al., Deltamethrin promotes adipogenesis via AMPKα and ER stress-mediated pathway in 3T3- L1 adipocytes and Caenorhabditis elegans, Food Chem. Toxicol. 134 (2019) 110791 https://doi.org/10.1016/j.fct.2019.110791.

[27]

J.E. Jin, C.B. Ahn, J.Y. Je, Purification and characterization of antioxidant peptides from enzymatically hydrolyzed ark shell (Scapharca subcrenata), Process Biochem. 72 (2018) 170–176 https://doi.org/10.1016/j.procbio.2018.06.001.

[28]

J. Cotabarren, A.M. Rosso, M. Tellechea, et al., Adding value to the chia (Salvia hispanica L.) expeller: production of bioactive peptides with antioxidant properties by enzymatic hydrolysis with Papain, Food Chem. 274 (2019) 848–856 https://doi.org/10.1016/j.foodchem.2018.09.061.

[29]

A.R. Phull, S.J. Kim, Fucoidan as bio-functional molecule: insights into the anti-inflammatory potential and associated molecular mechanisms, J. Funct. Foods 38 (2017) 415–426 https://doi.org/10.1016/j.jff.2017.09.051.

[30]

C.B. Ahn, Y.S. Cho, J.Y. Je, Purification and anti-inflammatory action of tripeptide from salmon pectoral fin byproduct protein hydrolysate, Food Chem. 168 (2015) 151–156 https://doi.org/10.1016/j.foodchem.2014.05.112.

[31]

M. Chalamaiah, W. Yu, J. Wu, Immunomodulatory and anticancer protein hydrolysates (peptides) from food proteins: a review, Food Chem. 245 (2018) 205–222 https://doi.org/10.1016/j.foodchem.2017.10.087.

[32]

Y. Ma, J. Liu, H. Shi, et al., Isolation and characterization of anti-inflammatory peptides derived from whey protein, J. Dairy Sci. 99 (9) (2016) 6902–6912 https://doi.org/10.3168/jds.2016-11186.

[33]

C. Meram, J. Wu, Anti-inflammatory effects of egg yolk livetins (α, β, and γ-livetin) fraction and its enzymatic hydrolysates in lipopolysaccharide-induced RAW 264.7 macrophages, Food Res. Int. 100 (Pt 1) (2017) 449–459 https://doi.org/10.1016/j.foodres.2017.07.032.

[34]

W. Yu, C.J. Field, J. Wu, Purification and identification of anti-inflammatory peptides from spent hen muscle proteins hydrolysate, Food Chem. 253 (2018) 101–107 https://doi.org/10.1016/j.foodchem.2018.01.093.

[35]

L. Zhao, X. Wang, X.L. Zhang, et al., Purification and identification of anti-inflammatory peptides derived from simulated gastrointestinal digests of velvet antler protein (Cervus elaphus Linnaeus), J. Food Drug Anal. 24 (2) (2016) 376–384 https://doi.org/10.1016/j.jfda.2015.10.003.

[36]

L. Koch, D. Frommhold, K. Buschmann, et al., LPS- and LTA-induced expression of IL-6 and TNF-α in neonatal and adult blood: role of MAPKs and NF-κB, Mediators Inflamm. (2014) 283126 https://doi.org/10.1155/2014/283126.

[37]

Z. Zeng, Y. Zhang, X. Liang, et al., Qingnao dripping pills mediate immune-inflammatory response and MAPK signaling pathway after acute ischemic stroke in rats, J. Pharmacol. Sci. 139 (3) (2019) 143–150 https://doi.org/10.1016/j.jphs.2018.12.009.

[38]

D. Capece, D. Verzella, B.D. Francesco, et al., NF-κB and mitochondria cross paths in cancer: mitochondrial metabolism and beyond, Semin. Cell Dev. Biol. 98 (2020) 118–128 https://doi.org/10.1016/j.semcdb.2019.05.021.

[39]

Z.A. Ibrahim, C.L. Armour, S. Phipps, et al., RAGE and TLRs: relatives, friends or neighbours? Mol. Immunol. 56 (4) (2013) 739–744 https://doi.org/10.1016/j.molimm.2013.07.008.

[40]

Y.S. Kim, C.B. Ahn, J.Y. Je, Anti-inflammatory action of high molecular weight Mytilus edulis hydrolysates fraction in LPS-induced RAW264.7 macrophage via NF-κB and MAPK pathways, Food Chem. 202 (2016) 9–14 https://doi.org/10.1016/j.foodchem.2016.01.114.

[41]

E.A. Barbosa, A. Oliveira, A. Plácido, et al., Structure and function of a novel antioxidant peptide from the skin of tropical frogs, Free Radical Bio. Med. 115 (2018) 68–79 https://doi.org/10.1016/j.freeradbiomed.2017.11.001.

[42]

K. Neha, M.R. Haider, A. Pathak, et al., Medicinal prospects of antioxidants: a review, Eur. J. Med. Chem. 178 (2019) 687–704 https://doi.org/10.1016/j.ejmech.2019.06.010.

[43]

S. Chakrabarti, J. Wu, Bioactive peptides on endothelial function, Food Sci. Hum. Wellness 5 (1) (2016) 1–7 https://doi.org/10.1016/j.fshw.2015.11.004.

[44]

I. Cruz-Chamorro, N. Álvarez-Sánchez, Md.C. Millán-Linares, et al., Lupine protein hydrolysates decrease the inflammatory response and improve the oxidative status in human peripheral lymphocytes, Food Res. Int. 126 (2019) 108585 https://doi.org/10.1016/j.foodres.2019.108585.

[45]

P. Indiano-Romacho, S. Fernández-Tomé, L. Amigo, et al., Multifunctionality of lunasin and peptides released during its simulated gastrointestinal digestion, Food Res. Int. 125 (2019) 108513 https://doi.org/10.1016/j.foodres.2019.108513.

[46]

I.C. Sheih, T.K. Wu, T.J. Fang, Antioxidant properties of a new antioxidative peptide from algae protein waste hydrolysate in different oxidation systems, Bioresour. Technol. Rep. 100 (13) (2009) 3419–3425 https://doi.org/10.1016/j.biortech.2009.02.014.

[47]

M. González-Montoya, B. Hernández-Ledesma, J.M. Silván, et al., Peptides derived from in vitro gastrointestinal digestion of germinated soybean proteins inhibit human colon cancer cells proliferation and inflammation, Food Chem. 242 (2018) 75–82 https://doi.org/10.1016/j.foodchem.2017.09. 035.

[48]

S. Guha, K. Majumder, Structural-features of food-derived bioactive peptides with anti-inflammatory activity: a brief review, J. Food Biochem. 43 (2018) e12531 https://doi.org/10.1111/jfbc.12531.

[49]

N.Y. Hai, P.K. Hyon, P. Yoonkyung, et al., Investigating the effects of positive charge and hydrophobicity on the cell selectivity, mechanism of action and anti-inflammatory activity of a Trp-rich antimicrobial peptide indolicidin, FEMS Microbiol. Lett. 292 (1) (2009) 134–140 https://doi.org/10.1111/j.1574-6968.2008.01484.x.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 23 December 2019
Revised: 15 March 2020
Accepted: 25 April 2020
Published: 08 July 2020
Issue date: January 2021

Copyright

© 2021 Beijing Academy of Food Sciences. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

Acknowledgements

There is no conflict of interest associated with authors in this manuscript. This project was supported by China Agriculture Research System (CARS-46).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return