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 (5.1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access

Cyanidin-3-glucoside protects the photooxidative damage of retinal pigment epithelium cells by regulating sphingolipid signaling and inhibiting MAPK pathway

Tingting Liua,bWentao QibWenting PengbJianan ZhangcYong Wangb,( )
School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Abstract

Cyanidin-3-glucoside (C3G) is the most common anthocyanin in dark grains and berries and is a food functional factor to improve visual health. However, the mechanisms of C3G on blue light-induced retinal pigment epithelial (RPE) cell photooxidative damage needs further exploration. We investigated the effects of C3G on blue light-irradiated A2E-containing RPE cells and explored whether sphingolipid, mitogen-activated protein kinase (MAPK), and mitochondria-mediated pathways are involved in this mechanism. Blue light irradiation led to mitochondria and lysosome damage in RPE cells, whereas C3G preserved mitochondrial morphology and function and maintained the lysosomal integrity. C3G suppressed the phosphorylation of JNK and p38 MAPK and mitochondria-mediated pathways to inhibit RPE cell apoptosis. Lipidomics data showed that C3G protected RPE cells against blue light-induced lipid peroxidation and apoptosis by maintaining sphingolipids balance. C3G significantly inhibited ceramide (Cer d18:0/15:0, Cer d18:0/16:0 and Cer d18:0/18:0) accumulation and elevated galactosylceramide (GalCer d18:1/15:0 and GalCer d18:1/16:0) levels in the irradiated A2E-containing RPE cells. Furthermore, C3G attenuated cell membrane damage by increasing phosphatidylcholine and phosphatidylserine levels. C3G inhibited apoptosis and preserved the structure of mitochondria and lysosome by regulating sphingolipid signaling and suppression of MAPK activation in RPE cells. Thus, dietary supplementation of C3G prevents retinal photooxidative damage.

Electronic Supplementary Material

Download File(s)
fshw-13-2-621_ESM.docx (1.4 MB)

References

[1]

X. Ouyang, J. Yang, Z. Hong, et al., Mechanisms of blue light-induced eye hazard and protective measures: a review, Biomed. Pharmacother. 130 (2020) 110577. https://doi.org/10.1016/j.biopha.2020.110577.

[2]

J.J. Hunter, J.I.W. Morgan, W.H. Merigan, et al., The susceptibility of the retina to photochemical damage from visible light, Prog. Retin. Eye Res. 31 (2012) 28-42. https://doi.org/10.1016/j.preteyeres.2011.11.001.

[3]

H.J. Kim, D. Montenegro, J. Zhao, et al., Bisretinoids of the retina: Photo-oxidation, iron-catalyzed oxidation, and disease consequences, Antioxidants 10 (2021) 1382. https://doi.org/10.3390/antiox10091382.

[4]

T. Yako, M. Nakamura, W. Otsu, et al., Mitochondria dynamics in the aged mice eye and the role in the RPE phagocytosis, Exp. Eye Res. 213 (2021) 108800. https://doi.org/10.1016/j.exer.2021.108800.

[5]

E. Pawlowska, J. Szczepanska, A. Koskela, et al., Dietary polyphenols in age-related macular degeneration: protection against oxidative stress and beyond, Oxid. Med. Cell. Longev. 2019 (2019) 9682318. https://doi.org/10.1155/2019/9682318.

[6]

Y. Wang, F. Huang, L. Zhao, et al., Protective effect of total flavones from Hippophae rhamnoides L. against visible light-induced retinal degeneration in pigmented rabbits, J. Agric. Food Chem. 64 (2016) 161-170. https://doi.org/10.1021/acs.jafc.5b04874.

[7]

J.M.T. Hyttinen, J. Viiri, K. Kaarniranta, et al., Mitochondrial quality control in AMD: does mitophagy play a pivotal role? Cell. Mol. Life Sci. 75 (2018) 2991-3008. https://doi.org/10.1007/s00018-018-2843-7.

[8]

C. Huang, P. Zhang, W. Wang, et al., Long-term blue light exposure induces RGC-5 cell death in vitro: involvement of mitochondria-dependent apoptosis, oxidative stress, and MAPK signaling pathways, Apoptosis 19 (2014) 922- 932. https://doi.org/10.1007/s10495-014-0983-2

[9]

J.R. Sparrow, B.L. Cai, Blue light-induced apoptosis of A2E-containing RPE: involvement of caspase-3 and protection by Bcl-2, Invest. Ophthalmol. Vis. Sci. 42 (2001) 1356-1362.

[10]

K. Kaarniranta, H. Uusitalo, J. Blasiak, et al., Mechanisms of mitochondrial dysfunction and their impact on age-related macular degeneration, Prog. Retin. Eye Res. 79 (2020). https://doi.org/10.1016/j.preteyeres.2020.100858.

[11]

J.R. Sparrow, J. Zhou, S. Ben-Shabat, et al., Involvement of oxidative mechanisms in blue-light-induced damage to A2E-laden RPE, Invest. Ophthalmol. Vis. Sci. 43 (2002) 1222-1227.

[12]

R. Gao, W. Shu, Y. Shen, et al., Peptide fraction from sturgeon muscle by pepsin hydrolysis exerts anti-inflammatory effects in LPS-stimulated RAW264.7 macrophages via MAPK and NF-kappa B pathways, Food Sci. Hum. Wellness 10 (2021) 103-111. https://doi.org/10.1016/j.fshw.2020.04.014.

[13]

C.X. Pan, X.Y. Liu, Y.T. Zheng, et al., The mechanisms of melanogenesis inhibition by glabridin: molecular docking, PKA/MITF and MAPK/MITF pathways, Food Sci. Hum. Wellness 12 (2023) 212-222. https://doi.org/10.1016/j.fshw.2022.07.011.

[14]

A. Kogot-Levin, A. Saada, Ceramide and the mitochondrial respiratory chain, Biochimie 100 (2014) 88-94. https://doi.org/10.1016/j.biochi.2013.07.027.

[15]

G. Kaur, L.X. Tan, G. Rathnasamy, et al., Aberrant early endosome biogenesis mediates complement activation in the retinal pigment epithelium in models of macular degeneration, Proc. Natl. Acad. Sci. U.S.A. 115 (2018) 9014-9019. https://doi.org/10.1073/pnas.1805039115.

[16]

Y. Wang, L. Zhao, F. Lu, et al., Retinoprotective effects of bilberry anthocyanins via antioxidant, anti-inflammatory, and anti-apoptotic mechanisms in a visible light-induced retinal degeneration model in pigmented rabbits, Molecules 20 (2015) 22395-22410. https://doi.org/10.3390/molecules201219785.

[17]

Z.Z. Peng, W.T. Qi, Y. Wang, et al., Recent progress in the protective effects of anthocyanins on the retina and its mechanism, Shipin Kexue 43 (2022) 249-257. https://doi.org/10.7506/spkx1002-6630-20210908-097.

[18]

Y. Wang, D. Zhang, Y.X. Liu, et al., The protective effects of berry-derived anthocyanins against visible light-induced damage in human retinal pigment epithelial cells, J. Sci. Food Agric. 95 (2015) 936-944. https://doi.org/10.1002/jsfa.6765.

[19]

Y. Wang, Y.Z. Huo, L. Zhao, et al., Cyanidin-3-glucoside and its phenolic acid metabolites attenuate visible light-induced retinal degeneration in vivo via activation of Nrf2/HO-1 pathway and NF-B suppression, Mol. Nutr. Food Res. 60 (2016) 1564-1577. https://doi.org/10.1002/mnfr.201501048.

[20]

W.T. Peng, Y.L. Wu, Z.Z. Peng, et al., Cyanidin-3-glucoside improves the barrier function of retinal pigment epithelium cells by attenuating endoplasmic reticulum stress-induced apoptosis, Food Res. Int. 157 (2022) 111313. https://doi.org/10.1016/j.foodres.2022.111313.

[21]

Y. Wang, H.J. Kim, J.R. Sparrow, Quercetin and cyanidin-3-glucoside protect against photooxidation and photodegradation of A2E in retinal pigment epithelial cells, Exp. Eye Res. 160 (2017) 45-55. https://doi.org/10.1016/j.exer.2017.04.010.

[22]

Y. Wang, W.T. Qi, Y.Z. Huo, et al., Cyanidin-3-glucoside attenuates 4-hydroxynonenal- and visible light-induced retinal damage in vitro and in vivo, Food Funct. 10 (2019) 2871-2880. https://doi.org/10.1039/C9FO00273A.

[23]

X.Y. Liu, S. Li, Z.T. Wang, et al., Ultrahigh pressure facilitates the acylation of malvidin and chlorogenic acid to increase the stability and protective effect of malvidin derivatives on H2O2-induced ARPE-19 cells, J. Agric. Food Chem. 69 (2021) 13990-14003. https://doi.org/10.1021/acs.jafc.1c03133.

[24]

C.A. Parish, M. Hashimoto, K. Nakanishi, et al., Isolation and one-step preparation of A2E and iso-A2E, fluorophores from human retinal pigment epithelium, Proc. Natl. Acad. Sci. U.S.A. 95 (1998) 14609-14613. https://doi.org/10.1073/pnas.95.25.14609.

[25]

S.G. Wang, L. Zheng, T.T. Zhao, et al., The neuroprotective effect of walnut-derived peptides against glutamate-induced damage in PC12 cells: mechanism and bioavailability, Food Sci. Hum. Wellness 11 (2022) 933-942. https://doi.org/10.1016/j.fshw.2022.03.021.

[26]

J.W. Song, S.M. Lam, X. Fan, et al., Omics-driven systems interrogation of metabolic dysregulation in COVID-19 pathogenesis, Cell Metab. 32 (2020) 188-202. https://doi.org/10.1016/j.cmet.2020.06.016.

[27]

S.M. Lam, C. Zhang, Z.H. Wang, et al., A multi-omics investigation of the composition and function of extracellular vesicles along the temporal trajectory of COVID-19, Nat. Metab. 3 (2021) 909-922. https://doi.org/10.1038/s42255-021-00425-4.

[28]

G.H. Shui, W.F. Cheong, I.A. Jappar, et al., Derivatization-independent cholesterol analysis in crude lipid extracts by liquid chromatography/mass spectrometry: applications to a rabbit model for atherosclerosis, J. Chromatogr. A 1218 (2011) 4357-4365. https://doi.org/10.1016/j.chroma.2011.05.011.

[29]

Y.S. Cheng, M. Linetsky, X. Gu, et al., Light-induced generation and toxicity of docosahexaenoate-derived oxidation products in retinal pigmented epithelial cells, Exp. Eye Res. 181 (2019) 325-345. https://doi.org/10.1016/j.exer.2018.09.012.

[30]

J.L. Zhou, B.L. Cai, Y.P. Jang, et al., Mechanisms for the induction of HNE-MDA-and AGE-adducts, RAGE and VEGF in retinal pigment epithelial cells, Exp. Eye Res. 80 (2005) 567-580. https://doi.org/10.1016/j.exer.2004.11.009.

[31]

Y. Shi, J. Ren, B. Zhao, et al., Photoprotective mechanism of fucoxanthin in ultraviolet B irradiation-induced retinal müller cells based on lipidomics analysis, J. Agric. Food Chem. 70 (2022) 3181-3193. https://doi.org/10.1021/acs.jafc.1c07980.

[32]

A. Alaimo, G.G. Linares, J.M. Bujjamer, et al., Toxicity of blue led light and A2E is associated to mitochondrial dynamics impairment in ARPE-19 cells: implications for age-related macular degeneration, Arch. Toxicol. 93 (2019) 1401-1415. https://doi.org/10.1007/s00204-019-02409-6.

[33]

S.Y. Jeong, X. Gu, K.W. Jeong, Photoactivation of N-retinylidene-N-retinylethanolamine compromises autophagy in retinal pigmented epithelial cells, Food Chem. Toxicol. 131 (2019) 110555. https://doi.org/10.1016/j.fct.2019.06.002.

[34]

E. Afsar, E. Kirimlioglu, T. Ceker, et al., Effect of ER stress on sphingolipid levels and apoptotic pathways in retinal pigment epithelial cells, Redox Biol. 30 (2020) 101430. https://doi.org/10.1016/j.redox.2020.101430.

[35]

C.H. Lin, M.R. Wu, W.J. Huang, et al., Low-luminance blue light-enhanced phototoxicity in A2E-laden RPE cell cultures and rats, Int. J. Mol. Sci. 20 (2019) 1799. https://doi.org/10.3390/ijms20071799.

[36]

S.X. Zhang, E. Sanders, S.J. Fliesler, et al., Endoplasmic reticulum stress and the unfolded protein responses in retinal degeneration, Exp. Eye Res. 125 (2014) 30-40. https://doi.org/10.1016/j.exer.2014.04.015.

[37]

K. Ogawa, Y. Kuse, K. Tsuruma, et al., Protective effects of bilberry and lingonberry extracts against blue light-emitting diode light-induced retinal photoreceptor cell damage in vitro, BMC Complem. Altern. Med. 14 (2014) 120. https://doi.org/10.1186/1472-6882-14-120.

[38]

L. Lei, J.A. Zhang, E.A. Decker, et al., Roles of lipid peroxidation-derived electrophiles in pathogenesis of colonic inflammation and colon cancer, Front. Cell Dev. Biol. 9 (2021) 665591. https://doi.org/10.3389/fcell.2021.665591.

[39]

L. Lei, J. Yang, J.N. Zhang, et al., The lipid peroxidation product EKODE exacerbates colonic inflammation and colon tumorigenesis, Redox Biol. 42 (2021) 101880. https://doi.org/10.1016/j.redox.2021.101880.

[40]

D.H. Zhu, P.G. Sreekumar, D.R. Hinton, et al., Expression and regulation of enzymes in the ceramide metabolic pathway in human retinal pigment epithelial cells and their relevance to retinal degeneration, Vision Res. 50 (2010) 643-651. https://doi.org/10.1016/j.visres.2009.09.002.

[41]

A. Barak, L.S. Morse, T. Goldkorn, Ceramide: a potential mediator of apoptosis in human retinal pigment epithelial cells, Invest. Ophthalmol. Visual Sci. 42 (2001) 247-254.

[42]

C.L. Chen, C.F. Lin, W.T. Chang, et al., Ceramide induces p38 MAPK and JNK activation through a mechanism involving a thioredoxin-interacting protein-mediated pathway, Blood 111 (2008) 4365-4374. https://doi.org/10.1182/blood-2007-08-106336.

[43]

S. Dadsena, S. Bockelmann, J.G.M. Mina, et al., Ceramides bind VDAC2 to trigger mitochondrial apoptosis, Nat. Commun. 10 (2019) 1832. https://doi.org/10.1038/s41467-019-09654-4.

[44]

J. Yao, H.E. Bi, Y. Sheng, et al., Ultraviolet (UV) and hydrogen peroxide activate ceramide-ER stress-AMPK signaling axis to promote retinal pigment epithelium (RPE) cell apoptosis, Int. J. Mol. Sci. 14 (2013) 10355-10368. https://doi.org/10.3390/ijms140510355.

[45]

D. Lewandowski, A.T. Foik, R. Smidak, et al., Inhibition of ceramide accumulation in adipoR1(-/-) mice increases photoreceptor survival and improves vision, JCI Insight 7 (2022) e156301. https://doi.org/10.1172/jci. insight.156301.

[46]

Y.M. Zhao, H.H. Xu, Z.Z. Tian, et al., Dose-dependent reductions in plasma ceramides after anthocyanin supplementation are associated with improvements in plasma lipids and cholesterol efflux capacity in dyslipidemia: a randomized controlled trial, Clin. Nutr. 40 (2021) 1871-1878. https://doi.org/10.1016/j.clnu.2020.10.014.

[47]

X. Si, J.L. Tian, C. Shu, et al., Serum ceramide reduction by blueberry anthocyanin-rich extract alleviates insulin resistance in hyperlipidemia mice, J. Agric. Food Chem. 68 (2020) 8185-8194. https://doi.org/10.1021/acs. jafc.0c01931.

[48]

K. Doffek, X. Chen, S.L. Sugg, et al., Phosphatidylserine inhibits NF-κB and p38 MAPK activation in human monocyte derived dendritic cells, Mol. Immunol. 48 (2011) 1771-1777. https://doi.org/10.1016/j.molimm.2011.04.021.

[49]

G. Paradies, V. Paradies, F.M. Ruggiero, et al., Role of cardiolipin in mitochondrial function and dynamics in health and disease: molecular and pharmacological aspects, Cells 8 (2019) 728. https://doi.org/10.3390/cells8070728.

[50]

M.Q. Xiao, H.Q. Zhong, L. Xia, et al., Pathophysiology of mitochondrial lipid oxidation: role of 4-hydroxynonenal (4-HNE) and other bioactive lipids in mitochondria, Free Radical Biol. Med. 111 (2017) 316-327. https://doi.org/10.1016/j.freeradbiomed.2017.04.363.

Food Science and Human Wellness
Pages 621-632
Cite this article:
Liu T, Qi W, Peng W, et al. Cyanidin-3-glucoside protects the photooxidative damage of retinal pigment epithelium cells by regulating sphingolipid signaling and inhibiting MAPK pathway. Food Science and Human Wellness, 2024, 13(2): 621-632. https://doi.org/10.26599/FSHW.2022.9250053

1508

Views

278

Downloads

1

Crossref

1

Web of Science

2

Scopus

0

CSCD

Altmetrics

Received: 23 October 2022
Revised: 06 November 2022
Accepted: 23 November 2022
Published: 25 September 2023
© 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