AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (5.9 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Procyanidin A1 and its digestive products alleviate acrylamide-induced IPEC-J2 cell damage through regulating Keap1/Nrf2 pathway

Fangfang YanaQun Lua,bChengming Wanga,b( )Rui Liua,b,c,( )
College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
Key Laboratory of Urban Agriculture in Central China, Ministry of Agriculture and Rural Affairs, Wuhan 430070, China

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Highlights

• Acrylamide slightly activates Nrf2 pathway, which cannot prevent cell damage.

• Procyanidin A1 and its digestive products can equally prevent cell damage.

• Nrf2 pathway plays a critical role in preventing cell damage.

• Procyanidin A1 and its digestive products activate Nrf2 by interacting with Keap1.

Graphical Abstract

Abstract

Our previous study has revealed that procyanidin A1 (A1) and its simulated digestive product (D-A1) can alleviate acrylamide (ACR)-induced intestine cell damage. However, the underlying mechanism remains unknown. In this study, we elucidated the molecular mechanism for A1 and D-A1 to alleviate ACR-stimulated IPEC-J2 cell damage. ACR slightly activated nuclear factor erythroid 2-related factor 2 (Nrf2) signaling and its target genes, but this activation could not reduce intestine cell damage. A1 and D-A1 could alleviate ACR-induced cell damage, but the effect was abrogated in cells transiently transfected with Nrf2 small interfering RNA (siRNA). Further investigation confirmed that A1 and D-A1 interacted with Kelch-like ECH-associated protein 1 (Keap1), which boosted the stabilization of Nrf2, subsequently promoted the translocation of Nrf2 into the nucleus, and further increased the expression of antioxidant proteins, thereby inhibiting glutathione (GSH) consumption, maintaining redox balance and eventually alleviating ACR-induced cell damage. Importantly, there was no difference between A1 and D-A1 treated groups, indicating that A1 can tolerate gastrointestinal digestion and may be a potential compound to limit the toxicity of ACR.

References

[1]

L. Rifai, F.A. Saleh, A review on acrylamide in food: occurrence, toxicity, and mitigation strategies, Int. J. Toxicol. 39 (2020) 93-102. https://doi.org/10.1177/1091581820902405.

[2]

A.A. Maan, M.A. Anjum, M.K. Iqbal Khan, et al., Acrylamide formation and different mitigation strategies during food processing: a review, Food Rev. Int. 38 (2022) 70-87. https://doi.org/10.1080/87559129.2020.1719505.

[3]

R. Pernice, J. Hauder, P. Koehler, et al., Effect of sulforaphane on glutathione-adduct formation and on glutathione-S-transferase-dependent detoxification of acrylamide in Caco-2 cells, Mol. Nutr. Food Res. 53 (2009) 1540-1550. https://doi.org/10.1002/mnfr.200900447.

[4]

Z. Cai, H. Rieka, U. Makoto, et al., Role of microglial activation and neuroinflammation in neurotoxicity of acrylamide in vivo and in vitro, Arch. Toxicol. 93 (2019) 2007-2019. https://doi.org/10.1007/s00204-019-02471-0.

[5]

X. Tan, J. Ye, W. Liu, et al., Acrylamide aggravates cognitive deficits at night period via the gut-brain axis by reprogramming the brain circadian clock, Arch. Toxicol. 93 (2019) 467-486. https://doi.org/10.1007/s00204-018-2340-7.

[6]

E. Sengul, V. Gelen, S. Yildirim, et al., The effects of selenium in acrylamide-induced nephrotoxicity in rats: roles of oxidative stress, inflammation, apoptosis, and DNA damage, Biol. Trace Elem. Res. 199 (2021) 173-184. https://doi.org/10.1007/s12011-020-02111-0.

[7]

Q. Bu, Y. Huang, M. Li, et al., Acrylamide exposure represses neuronal differentiation, induces cell apoptosis and promotes tau hyperphosphorylation in hESC-derived 3D cerebral organoids, Food Chem. Toxicol. 144 (2020) 111643. https://doi.org/10.1016/j.fct.2020.111643.

[8]

K. Sedat, S. Varol, K.H. Mehtap, Effect of acrylamide on BEAS-2B normal human lung cells: cytotoxic, oxidative, apoptotic and morphometric analysis, Acta Histochem. 121 (2019) 595-603. https://doi.org/10.1016/j.acthis.2019.05.005.

[9]

M.J. Kerins, A. Ooi, A catalogue of somatic Nrf2 gain-of-function mutations in cancer, Sci. Rep. 8 (2018) 12846. https://doi.org/10.1038/s41598-018-31281-0.

[10]

R.M. LoPachin, T. Gavin, Acrylamide-induced nerve terminal damage:relevance to neurotoxic and neurodegenerative mechanisms, J. Agr. Food Chem. 56 (2008) 5994-6003. https://doi.org/10.1021/jf703745t.

[11]

X. Sui, J. Yang, G. Zhang, et al., NLRP3 inflammasome inhibition attenuates subacute neurotoxicity induced by acrylamide in vitro and in vivo, Toxicology 432 (2020) 152392. https://doi.org/10.1016/j.tox.2020.152392.

[12]

L.H. Zhang, T. Gavin, D.S. Barber, et al., Role of the Nrf2-ARE pathway in acrylamide neurotoxicity, Toxicol. Lett. 205 (2011) 1-7. https://doi.org/10.1016/j.toxlet.2011.04.011.

[13]

X.T. Tan, L. Li, J. Wang, et al., Resveratrol prevents acrylamide-induced mitochondrial dysfunction and inflammatory responses via targeting circadian regulator Bmal1 and Cry1 in hepatocytes, J. Agric. Food Chem. 67 (2019) 8510-8519. https://doi.org/10.1021/acs.jafc.9b03368.

[14]

M. Krishnan, S.C. Kang, Vitexin inhibits acrylamide-induced neuroinflammation and improves behavioral changes in zebrafish larvae, Neurotoxicol. Teratol. 74 (2019) 106811. https://doi.org/10.1016/j.ntt.2019.106811.

[15]

I. Rodriguez-Ramiro, S. Ramos, L. Bravo, et al., Procyanidin B2 and a cocoa polyphenolic extract inhibit acrylamide-induced apoptosis in human Caco-2 cells by preventing oxidative stress and activation of JNK pathway, J. Nutr. Biochem. 22 (2011) 1186-1194. https://doi.org/10.1016/j.jnutbio.2010.10.005.

[16]

C. Xiao, X. Jia, H. Lingxiao, et al., Effects of in vitro digestion on the content and biological activity of polyphenols from Acacia mearnsii bark, Molecules 23 (2018) 1804. https://doi.org/10.3390/molecules23071804.

[17]

F. Yan, W. Chen, L. Zhao, et al., Procyanidin A1 and its digestive products prevent acrylamide-induced intestinal barrier dysfunction via the MAPKmediated MLCK pathway, Food Funct. 12 (2021) 11956-11965. https://doi.org/10.1039/d1fo01918j.

[18]

F. Yan, L. Zhao, W. Chen, et al., Comparison of the inhibitory effects of procyanidins with different structures and their digestion products against acrylamide-induced cytotoxicity in IPEC-J2 cells, J. Funct. Foods 72 (2020) 104073. https://doi.org/10.1016/j.jff.2020.104073.

[19]

L. Zheng, H. Wei, H. Yu, et al., Fish skin gelatin hydrolysate production by ginger powder induces glutathione synthesis to prevent hydrogen peroxide induced intestinal oxidative stress via the Pept1-p62-Nrf2 cascade, J. Agr. Food Chem. 66 (2018) 11601-11611. https://doi.org/10.1021/acs.jafc.8b02840.

[20]

S. Miwa, S. Kashyap, E. Chini, et al., Mitochondrial dysfunction in cell senescence and aging, J. Clin. Invest. 132 (2022). https://doi.org/10.1172/jci158447.

[21]

S. Lee, L. Hu, Nrf2 activation through the inhibition of Keap1-Nrf2 protein-protein interaction, Med. Chem. Res. 29 (2020) 846-867. https://doi.org/10.1007/s00044-020-02539-y.

[22]

G.C. Tong, W.K. Cornwelll, G.E. Means, Reactions of acrylamide with glutathione and serum albumin, Toxicol. Lett. 147 (2004) 127-131. https://doi.org/10.1016/j.toxlet.2003.10.021.

[23]

J.M. Latzin, B.K. Schindler, T. Weiss, et al., Determination of 2,3-dihydroxypropionamide, an oxidative metabolite of acrylamide, in human urine by gas chromatography coupled with mass spectrometry, Anal. Bioanal. Chem. 402 (2012) 2431-2438. https://doi.org/10.1007/s00216-011-5692-x.

[24]

R.M. LoPachin, D.S. Barber, Synaptic cysteine sulfhydryl groups as targets of electrophilic neurotoxicants, Toxicol. Sci. 94 (2006) 240-255. https://doi.org/10.1093/toxsci/kfl066.

[25]

H.A. Edres, N.M. Taha, M.A. Lebda, et al., The potential neuroprotective effect of allicin and melatonin in acrylamide-induced brain damage in rats, Environ. Sci. Pollut. Res. 28 (2021) 58768-58780. https://doi.org/10.1007/s11356-021-14800-x.

[26]

D.B. Donmez, S. Kacar, R. Bagci, et al., Protective effect of carnosic acid on acrylamide-induced liver toxicity in rats: mechanistic approach over Nrf2-Keap1 pathway, J. Biochem. Mol. Toxicol. 34 (2020) e22524. https://doi.org/10.1002/jbt.22524.

[27]

M. McMahon, K. Itoh, M. Yamamoto, et al., Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression, J. Biol. Chem. 278 (2003) 21592-21600. https://doi.org/10.1074/jbc.M300931200.

[28]

M. Kobayashi, M. Yamamoto, Nrf2-Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species, Adv. Enzyme Regul. 46 (2006) 113-140. https://doi.org/10.1016/j.advenzreg.2006.01.007.

[29]

D.S. Barber, S. Stevens, R.M. LoPachin, Proteomic analysis of rat striatal synaptosomes during acrylamide intoxication at a low dose rate, Toxicol. Sci. 100 (2007) 156-167. https://doi.org/10.1093/toxsci/kfm210.

[30]

Z.G. Liu, S. Ge, Z. Chen, et al., Acrylamide induces mitochondrial dysfunction and apoptosis in BV-2 microglial cells, Free Radical Biol. Med.84 (2015) 42-53. https://doi.org/10.1016/j.freeradbiomed.2015.03.013.

[31]

S. Kacar, V. Sahinturk, O. Tomsuk, et al., The effects of thymoquinone and quercetin on the toxicity of acrylamide in rat glioma cells, J. Biochem. Mol. Toxicol. 36 (2022) e22992. https://doi.org/10.1002/jbt.22992.

[32]

M. Zhao, F.S.L. Wang, X. Hu, et al., Acrylamide-induced neurotoxicity in primary astrocytes and microglia: roles of the Nrf2-ARE and NF-κB pathways, Food Chem. Toxicol. 106 (2017) 25-35. https://doi.org/10.1016/j.fct.2017.05.007.

[33]

F.A. Ekuban, C. Zong, M. Takikawa, et al., Genetic ablation of Nrf2 exacerbates neurotoxic effects of acrylamide in mice, Toxicology 456 (2021) 152785. https://doi.org/10.1016/j.tox.2021.152785.

[34]

L. Li, H.Y. Sun, W. Liu, et al., Silymarin protects against acrylamideinduced neurotoxicity via Nrf2 signalling in PC12 cells, Food Chem. Toxicol. 102 (2017) 93-101. https://doi.org/10.1016/j.fct.2017.01.021.

[35]

S. Ge, Z.G. Liu, L.F. Wang, et al., Protective effects of lipoic acid against acrylamide-induced neurotoxicity: involvement of mitochondrial energy metabolism and autophagy, Food Funct. 8 (2017) 4657-4667. https://doi.org/10.1039/c7fo01429e.

[36]

X.Y. Wang, B.R. Zhu, Q. Jia, et al., Cinnamtannin D1 protects pancreatic beta-cells from glucolipotoxicity-induced apoptosis by enhancement of autophagy in vitro and in vivo, J. Agr. Food Chem. 68 (2020) 12617-12630. https://doi.org/10.1021/acs.jafc.0c04898.

[37]

C.C. Franklin, D.S. Backos, I. Mohar, et al., Structure, function, and posttranslational regulation of the catalytic and modifier subunits of glutamate cysteine ligase, Mol. Aspects Med. 30 (2009) 86-98. https://doi.org/10.1016/j.mam.2008.08.009.

[38]

T. Mohan, K.K.S. Narasimhan, D.B. Ravi, et al., Role of Nrf2 dysfunction in the pathogenesis of diabetic nephropathy: therapeutic prospect of epigallocatechin-3-gallate, Free Radical Biol. Med. 160 (2020) 227-238. https://doi.org/10.1016/j.freeradbiomed.2020.07.037.

[39]

Y. Cui, Y. Li, N. Huang, et al., Structure based modification of chalcone analogue activates Nrf2 in the human retinal pigment epithelial cell line ARPE-19, Free Radical Biol. Med. 148 (2020) 52-59. https://doi.org/10.1016/j.freeradbiomed.2019.12.033.

[40]

N. Robledinos-Anton, R. Fernandez-Gines, G. Manda, et al., Activators and inhibitors of Nrf2: a review of their potential for clinical development, Oxid. Med. Cell. Longevity 2019 (2019) 9372182. https://doi.org/10.1155/2019/9372182.

[41]

I. Rodriguez-Ramiro, S. Ramos, L. Bravo, et al., Procyanidin B2 induces Nrf2 translocation and glutathione S-transferase P1 expression via ERKs and p38-MAPK pathways and protect human colonic cells against oxidative stress, Eur. J. Nutr. 51 (2012) 881-892. https://doi.org/10.1007/s00394-011-0269-1.

[42]

J.J. Lu, H.J. Jiang, B.Y. Liu, et al., Grape seed procyanidin extract protects against Pb-induced lung toxicity by activating the AMPK/Nrf2/p62 signaling axis, Food Chem. Toxicol. 116 (2018) 59-69. https://doi.org/10.1016/j.fct.2018.03.034.

Food Science and Human Wellness
Pages 1475-1484
Cite this article:
Yan F, Lu Q, Wang C, et al. Procyanidin A1 and its digestive products alleviate acrylamide-induced IPEC-J2 cell damage through regulating Keap1/Nrf2 pathway. Food Science and Human Wellness, 2024, 13(3): 1475-1484. https://doi.org/10.26599/FSHW.2022.9250124

873

Views

207

Downloads

1

Crossref

0

Web of Science

1

Scopus

0

CSCD

Altmetrics

Received: 06 October 2022
Revised: 29 October 2022
Accepted: 15 November 2022
Published: 08 February 2024
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

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

Return