Journal Home > Volume 11 , Issue 6

Soy glycinin derived octapeptide (SGP8) is a peptide obtained from degradation of the soy glycinin, whose amino acid sequence is IAVPGEVA. To determine the effect of SGP8 on non-alcoholic fatty liver disease (NAFLD), steatosis HepG2 cells were induced by 1 mmol/L free fatty acid (FFA) and C57BL/6J mice were fed with methionine-choline deficient (MCD) diet for 3 weeks to establish NAFLD model. The results of oil red O staining and total cholesterol (TC)/triglyceride (TG) contents showed that SGP8 could significantly reduce the lipid content of steatosis HepG2 cells. In vivo, SGP8 lowered plasma alanine aminotransferase (ALT) and low density lipoprotein (LDL) content, normalized hepatic superoxide dismutase (SOD) and malondialdehyde (MDA) production, and reduced the severity of liver inflammation. The results of Western blotting showed that SGP8 increased expression of Sirtuin-1 (SIRT1) and phosphorylation level of AMP activated protein kinase (AMPK) in hepatocytes. Through activation of SIRT1/AMPK pathway, SGP8 downregulated the expression of sterol regulatory element binding protein 1c (SREBP-1c) and its target genes ACC and FAS expression levels, and increased the phosphorylation level of acetyl CoA carboxylase (ACC). Furthermore, SGP8 also upregulated the expression of transcription factor peroxisome proliferator activated receptor α (PPARα), which was regulated by SIRT1/AMPK pathway, and its target gene CPT1 level. In conclusion, SGP8 might improve NAFLD by activating the SIRT1/AMPK pathway. Our data suggest that SGP8 may act as a novel and potent therapeutic agent against NAFLD.


menu
Abstract
Full text
Outline
About this article

A soy glycinin derived octapeptide protects against MCD diet induced non-alcoholic fatty liver disease in mice

Show Author's information Peng MaRongrong HuangYu Ou( )
School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

Soy glycinin derived octapeptide (SGP8) is a peptide obtained from degradation of the soy glycinin, whose amino acid sequence is IAVPGEVA. To determine the effect of SGP8 on non-alcoholic fatty liver disease (NAFLD), steatosis HepG2 cells were induced by 1 mmol/L free fatty acid (FFA) and C57BL/6J mice were fed with methionine-choline deficient (MCD) diet for 3 weeks to establish NAFLD model. The results of oil red O staining and total cholesterol (TC)/triglyceride (TG) contents showed that SGP8 could significantly reduce the lipid content of steatosis HepG2 cells. In vivo, SGP8 lowered plasma alanine aminotransferase (ALT) and low density lipoprotein (LDL) content, normalized hepatic superoxide dismutase (SOD) and malondialdehyde (MDA) production, and reduced the severity of liver inflammation. The results of Western blotting showed that SGP8 increased expression of Sirtuin-1 (SIRT1) and phosphorylation level of AMP activated protein kinase (AMPK) in hepatocytes. Through activation of SIRT1/AMPK pathway, SGP8 downregulated the expression of sterol regulatory element binding protein 1c (SREBP-1c) and its target genes ACC and FAS expression levels, and increased the phosphorylation level of acetyl CoA carboxylase (ACC). Furthermore, SGP8 also upregulated the expression of transcription factor peroxisome proliferator activated receptor α (PPARα), which was regulated by SIRT1/AMPK pathway, and its target gene CPT1 level. In conclusion, SGP8 might improve NAFLD by activating the SIRT1/AMPK pathway. Our data suggest that SGP8 may act as a novel and potent therapeutic agent against NAFLD.

Keywords: HepG2 cells, Soy glycinin derived octapeptide (SGP8), Non-alcoholic fatty liver disease (NAFLD), Methionine-choline deficient (MCD), SIRT1/AMPK pathway

References(58)

[1]

R. Loomba, A.J. Sanyal, The global NAFLD epidemic, Nat. Rev. Gastroenterol. Hepatol. 10 (2013) 686-690. https://doi.org/10.1038/nrgastro.2013.171.

[2]

W.L. Shapiro, E.L. Yu, J.C. Arin, et al., Clinical practice approach to nonalcoholic fatty liver disease by pediatric gastroenterologists in the United States, J. Pediatr. Gastroenterol. Nutr. 68 (2019) 182-189. https://doi.org/10.1097/mpg.0000000000002194.

[3]

C.A. Onyekwere, A.O. Ogbera, B.O. Balogun, Non-alcoholic fatty liver disease and the metabolic syndrome in an urban hospital serving an African community, Ann. Hepatol. 10 (2011) 119-124. https://doi.org/10.1016/S1665-2681(19)31559-5.

[4]

L. Caballería, G. Pera, M.A. Auladell, et al., Prevalence and factors associated with the presence of nonalcoholic fatty liver disease in an adult population in Spain, Eur. J. Gastroenterol. Hepatol. 22 (2010) 24-32. https://doi.org/10.1097/MEG.0b013e32832fcdf0.

[5]

C. Xu, C. Yu, H. Ma, et al., Prevalence and risk factors for the development of nonalcoholic fatty liver disease in a nonobese Chinese population: the Zhejiang Zhenhai Study, Am. J. Gastroenterol. 108 (2013) 1299-1304. https://doi.org/10.1038/ajg.2013.104.

[6]

X. Hou, S. Xu, K.A. Maitland-Toolan, et al., SIRT1 regulates hepatocyte lipid metabolism through activating AMP-activated protein kinase, J. Biol. Chem. 283 (2008) 20015-20026. https://doi.org/10.1074/jbc.M802187200.

[7]

L. Fu, F. Li, A. Bruckbauer, et al., Interaction between leucine and phosphodiesterase 5 inhibition in modulating insulin sensitivity and lipid metabolism, Diabetes Metab. Syndr. Obes. 8 (2015) 227-239. https://doi.org/10.2147/dmso.S82338.

[8]

D.G. Hardie, F.A. Ross, S.A. Hawley, AMPK: a nutrient and energy sensor that maintains energy homeostasis, Nat. Rev. Mol. Cell Biol. 13 (2012) 251-262. https://doi.org/10.1038/nrm3311.

[9]

Y. Zhang, S. Li, W. Donelan, et al., Angiopoietin-like protein 8 (betatrophin) is a stress-response protein that down-regulates expression of adipocyte triglyceride lipase, Biochim. Biophys. Acta. 1861 (2016) 130-137. https://doi.org/10.1016/j.bbalip.2015.11.003.

[10]

M. Janevski, S. Ratnayake, S. Siljanovski, et al., Fructose containing sugars modulate mRNA of lipogenic genes ACC and FAS and protein levels of transcription factors ChREBP and SREBPIc with no effect on body weight or liver fat, Food Funct. 3 (2011) 141-149. https://doi.org/10.1039/c1fo10111k.

[11]

C. Zhang, X. Chen, R.M. Zhu, et al., Endoplasmic reticulum stress is involved in hepatic SREBP-1c activation and lipid accumulation in fructose-fed mice, Toxicol. Lett. 212 (2012) 229-240. https://doi.org/10.1016/j.toxlet.2012.06.002.

[12]

Z. Ren, Z. Xie, D. Cao, et al., C-Phycocyanin inhibits hepatic gluconeogenesis and increases glycogen synthesis via activating Akt and AMPK in insulin resistance hepatocytes, Food Funct. 9 (2018) 2829-2839. https://doi.org/10.1039/c8fo00257f.

[13]

M. Zang, A. Zuccollo, X. Hou, et al., AMP-activated protein kinase is required for the lipid-lowering effect of metformin in insulin-resistant human HepG2 cells, J. Biol. Chem. 279 (2004) 47898-47905. https://doi.org/10.1074/jbc.M408149200.

[14]

F. Jiang, Z. Zhang, Y. Zhang, et al., L-carnitine ameliorates the liver inflammatory response by regulating carnitine palmitoyltransferase I-dependent PPARγ signaling, Mol. Med. Rep. 13 (2016) 1320-1328. https://doi.org/10.3892/mmr.2015.4639.

[15]

G. Pascual, A.L. Fong, S. Ogawa, et al., A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-γ, Nature 437 (2005) 759-763. https://doi.org/10.1038/nature03988.

[16]

A.K.S. Silva, C.A. Peixoto, Role of peroxisome proliferator-activated receptors in non-alcoholic fatty liver disease inflammation, Cell Mol. Life. Sci. 75 (2018) 2951-2961. https://doi.org/10.1007/s00018-018-2838-4.

[17]

C.J. Villanueva, P. Tontonoz, Licensing PPARγ to work in macrophages, Immunity. 33 (2010) 647-649. https://doi.org/10.1016/j.immuni.2010.11.017.

[18]

A. Mahzari, S. Li, X. Zhou, et al., Matrine protects against MCD-induced development of NASH via upregulating HSP72 and downregulating mTOR in a manner distinctive from metformin, Front. Pharmacol. 10 (2019) 405. https://doi.org/10.3389/fphar.2019.00405.

[19]

X.L. Liu, Y.N. Ming, J.Y. Zhang, et al., Gene-metabolite network analysis in different nonalcoholic fatty liver disease phenotypes, Exp. Mol. Med. 49 (2017) e283. https://doi.org/10.1038/emm.2016.123.

[20]

Q.M. Anstee, R.D. Goldin, Mouse models in non-alcoholic fatty liver disease and steatohepatitis research, Int. J. Exp. Pathol. 87 (2006) 1-16. https://doi.org/10.1111/j.0959-9673.2006.00465.x.

[21]

R. Kumar, S. Mohan, Non-alcoholic fatty liver disease in lean subjects: characteristics and implications, J. Clin. Transl. Hepatol. 5 (2017) 216-223. https://doi.org/10.14218/jcth.2016.00068.

[22]

K.M. Schneider, A. Mohs, K. Kilic, et al., Intestinal microbiota protects against MCD diet-induced steatohepatitis, Int. J. Mol. Sci. 20(2) (2019) 308. https://doi.org/10.3390/ijms20020308.

[23]

M.V. Machado, G.A. Michelotti, G. Xie, et al., Mouse models of diet-induced nonalcoholic steatohepatitis reproduce the heterogeneity of the human disease, PLoS ONE 10 (2015) e0127991. https://doi.org/10.1371/journal.pone.0127991.

[24]

Z.M. Younossi, M. Stepanova, F. Negro, et al., Nonalcoholic fatty liver disease in lean individuals in the United States, Medicine (Baltimore) 91 (2012) 319-327. https://doi.org/10.1097/MD.0b013e3182779d49.

[25]

H. Korhonen, A. Pihlanto, Bioactive peptides: production and functionality, Int. Dairy J. 16 (2006) 945-960. https://doi.org/10.1016/j.idairyj.2005.10.012.

[26]
L. Mora, M. Gallego, M.C. Aristoy, et al., 12-Bioactive peptides, in: F.J. Barba, J.M.A. Saraiva, G. Cravotto, et al. (Eds. ), Innovative Thermal and Non-thermal Processing, Bioaccessibility and Bioavailability of Nutrients and Bioactive Compounds, Woodhead Publishing, 2019, pp. 333-345. https://doi.org/10.1016/B978-0-12-814174-8.00012-3.
DOI
[27]

C. Lammi, C. Zanoni, A. Arnoldi, IAVPGEVA, IAVPTGVA, and LPYP, three peptides from soy glycinin modulates cholesterol metabolism in HepG2 cells through the activation of the LDLR-SREBP2 pathway, J. Funct. Foods 14 (2015) 469-478. https://doi.org/10.1016/j.jff.2015.02.021.

[28]

C. Lammi, C. Zanoni, A. Arnoldi, Three peptides from soy glycinin modulate glucose metabolism in human hepatic HepG2 cells, Int. J. Mol. Sci. 16 (2015) 27362-27370. https://doi.org/10.3390/ijms161126029.

[29]

F. Li, Y. Yang, L. Yang, et al., Resveratrol alleviates FFA and CCl4 induced apoptosis in HepG2 cells via restoring endoplasmic reticulum stress, Oncotarget 8 (2017) 43799-43809. https://doi.org/10.18632/oncotarget.16460.

[30]

Y.L. Liu, L.C. Lin, Y.T. Tung, et al., Rhododendron oldhamii leaf extract improves fatty liver syndrome by increasing lipid oxidation and decreasing the lipogenesis pathway in mice, Int. J. Med. Sci. 14 (2017) 862-870. https://doi.org/10.7150/ijms.19553.

[31]

K.J. Oh, J. Park, S.Y. Lee, et al., A typical antipsychotic drugs perturb AMPK-dependent regulation of hepatic lipid metabolism, Am. J. Physiol. Endocrinol. Metab. 300 (2011) E624-E632. https://doi.org/10.1152/ajpendo.00502.2010.

[32]

A.R. Lee, S.N. Han, Pinolenic acid downregulates lipid anabolic pathway in HepG2 cells, Lipids 51 (2016) 847-855. https://doi.org/10.1007/s11745-016-4149-6.

[33]

N. Gao, X. Qu, J. Yan, et al., L-FABP T94A decreased fatty acid uptake and altered hepatic triglyceride and cholesterol accumulation in Chang liver cells stably transfected with L-FABP, Mol. Cell. Biochem. 345 (2010) 207-214. https://doi.org/10.1007/s11010-010-0574-7.

[34]

S. Del Guerra, M. Bugliani, V. D'Aleo, et al., G-protein-coupled receptor 40 (GPR40) expression and its regulation in human pancreatic islets: the role of type 2 diabetes and fatty acids, Nutr. Metab. Cardiovasc. Dis. 20 (2010) 22-25. https://doi.org/10.1016/j.numecd.2009.02.008.

[35]

Y. Zhang, M.L. Chen, Y. Zhou, et al., Resveratrol improves hepatic steatosis by inducing autophagy through the cAMP signaling pathway, Mol. Nutr. Food. Res. 59 (2015) 1443-1457. https://doi.org/10.1002/mnfr.201500016.

[36]

P. Nguyen, V. Leray, M. Diez, et al., Liver lipid metabolism, J. Anim. Physiol. Anim. Nutr (Berl). 92 (2008) 272-283. https://doi.org/10.1111/j.1439-0396.2007.00752.x.

[37]

Y. Wang, J. Viscarra, S.J. Kim, et al., Transcriptional regulation of hepatic lipogenesis, Nat. Rev. Mol. Cell Biol. 16 (2015) 678-689. https://doi.org/10.1038/nrm4074.

[38]

X. Li, H. Chen, Y. Guan, et al., Acetic acid activates the AMP-activated protein kinase signaling pathway to regulate lipid metabolism in bovine hepatocytes, PLoS ONE 8 (2013) e67880. https://doi.org/10.1371/journal.pone.0067880.

[39]

M. Bajaj, S. Suraamornkul, L.J. Hardies, et al., Effects of peroxisome proliferator-activated receptor (PPAR)-α and PPAR-γ agonists on glucose and lipid metabolism in patients with type 2 diabetes mellitus, Diabetologia 50 (2007) 1723-1731. https://doi.org/10.1007/s00125-007-0698-9.

[40]

K. Toriguchi, E. Hatano, K. Tanabe, et al., Attenuation of steatohepatitis, fibrosis, and carcinogenesis in mice fed a methionine-choline deficient diet by CCAAT/enhancer-binding protein homologous protein deficiency, J. Gastroenterol. Hepatol. 29 (2014) 1109-1118. https://doi.org/10.1111/jgh.12481.

[41]

Y.A. Jung, Y.K. Choi, G.S. Jung, et al., Sitagliptin attenuates methionine/choline-deficient diet-induced steatohepatitis, Diabetes Res. Clin. Pract. 105 (2014) 47-57. https://doi.org/10.1016/j.diabres.2014.04.028.

[42]

L.M. Jiménez-Flores, S. López-Briones, M.H. Macías-Cervantes, et al., A PPARγ, NF-κB and AMPK-dependent mechanism may be involved in the beneficial effects of curcumin in the diabetic db/db mice liver, Molecules 19 (2014) 8289-8302. https://doi.org/10.3390/molecules19068289.

[43]

L. Wu, C. Yan, M. Czader, et al., Inhibition of PPARγ in myeloid-lineage cells induces systemic inflammation, immunosuppression, and tumorigenesis, Blood 119 (2012) 115-126. https://doi.org/10.1182/blood-2011-06-363093.

[44]

N.L. Price, A.P. Gomes, A.J. Ling, et al., SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function, Cell Metab. 15 (2012) 675-690. https://doi.org/10.1016/j.cmet.2012.04.003.

[45]

A. Sandoval-Rodriguez, H.C. Monroy-Ramirez, A. Meza-Rios, et al., Pirfenidone is an agonistic ligand for PPARα and improves NASH by activation of SIRT1/LKB1/pAMPK, Hepatol. Commun. 4 (2020) 434-449. https://doi.org/10.1002/hep4.1474.

[46]

A. Stacchiotti, I. Grossi, R. García-Gómez, et al., Melatonin effects on non-alcoholic fatty liver disease are related to microRNA-34a-5p/Sirt1 axis and autophagy, Cells 8(9) (2019) 1053. https://doi.org/10.3390/cells8091053.

[47]

N. Ruderman, M. Prentki, AMP kinase and malonyl-CoA: targets for therapy of the metabolic syndrome, Nat. Rev. Drug. Discov. 3 (2004) 340-351. https://doi.org/10.1038/nrd1344.

[48]

S.C. Stein, A. Woods, N.A. Jones, et al., The regulation of AMP-activated protein kinase by phosphorylation, Biochem. J. 345(3) (2000) 437-443. https://doi.org/10.1042/0264-6021:3450437.

[49]

E. Yuan, X. Duan, L. Xiang, et al., Aged oolong tea reduces high-fat diet-induced fat accumulation and dyslipidemia by regulating the AMPK/ACC signaling pathway, Nutrients 10(2) (2018) 187. https://doi.org/10.3390/nu10020187.

[50]

E. Robinson, D.J. Grieve, Significance of peroxisome proliferator-activated receptors in the cardiovascular system in health and disease, Pharmacol. Ther. 122 (2009) 246-263. https://doi.org/10.1016/j.pharmthera.2009.03.003.

[51]

M.S. Rao, J.K. Reddy, PPARα in the pathogenesis of fatty liver disease, Hepatology 40 (2004) 783-786. https://doi.org/10.1002/hep.20453.

[52]

T. Venäläinen, F. Molnár, C. Oostenbrink, et al., Molecular mechanism of allosteric communication in the human PPARα-RXRα heterodimer, Proteins 78 (2010) 873-887. https://doi.org/10.1002/prot.22613.

[53]

J. Mun, S. Kim, H.G. Yoon, et al., Water extract of Curcuma longa L. ameliorates non-alcoholic fatty liver disease, Nutrients 11(10) (2019) 2536. https://doi.org/10.3390/nu11102536.

[54]

R.H. Wong, I. Chang, C.S. Hudak, et al., A role of DNA-PK for the metabolic gene regulation in response to insulin, Cell 136 (2009) 1056-1072. https://doi.org/10.1016/j.cell.2008.12.040.

[55]

M.J. Griffin, R.H. Wong, N. Pandya, et al., Direct interaction between USF and SREBP-1c mediates synergistic activation of the fatty-acid synthase promoter, J. Biol. Chem. 282 (2007) 5453-5467. https://doi.org/10.1074/jbc.M610566200.

[56]

F. Wen, C. An, X. Wu, et al., MiR-34a regulates mitochondrial content and fat ectopic deposition induced by resistin through the AMPK/PPARα pathway in HepG2 cells, Int. J. Biochem. Cell Biol. 94 (2018) 133-145. https://doi.org/10.1016/j.biocel.2017.11.008.

[57]

J.W. Lim, J. Dillon, M. Miller, Proteomic and genomic studies of non-alcoholic fatty liver disease--clues in the pathogenesis, World J. Gastroenterol. 20 (2014) 8325-8340. https://doi.org/10.3748/wjg.v20.i26.8325.

[58]

G. Tarantino, V. Citro, D. Capone, Nonalcoholic fatty liver disease: a challenge from mechanisms to therapy, J. Clin. Med. 9(1) (2019) 15. https://doi.org/10.3390/jcm9010015.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 09 November 2020
Revised: 01 February 2021
Accepted: 31 March 2021
Published: 18 July 2022
Issue date: November 2022

Copyright

© 2022 Beijing Academy of Food Sciences.

Acknowledgements

Acknowledgements

This work was funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). The authors also thank Dr. Zhu Zhou (City University of New York) for her assistance in reviewing this manuscript and valuable discussion.

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