Journal Home > Volume 13 , Issue 3

In order to improve the bioavailability of lutein (LUT), a novel lutein-stevioside nanoparticle (LUT-STE) were prepared previously, but the information about LUT-STE on protecting of eye health was limited. This study investigated the effect of LUT-STE on antioxidant activity of H2O2-induced human retinal pigment epithelial (ARPE) cells. LUT and LUT-STE (final concentration of 5 μg/mL) significantly enhanced cell viability from (74.84 ± 5.10)% to (81.92 ± 10.01)% (LUT) and (89.33 ± 4.34)% (LUT-STE), and inhibited the cell apoptosis (P < 0.05). After pretreatment with LUT-STE in ARPE cells, the levels of superoxide dismutase (SOD), catalase (CAT) and glutathion peroxidase (GSH-Px) in ARPE cells were significantly increased (P < 0.05), the contents of reactive oxygen species (ROS) and malondialdehyde (MDA) were decreased. In addition, the vascular endothelial growth factor (VEGF) levels were inhibited by 13.61% and 17.39%, respectively, pretreatment with LUT and LUT-STE. Western blotting results showed that the pretreatment with LUT-STE inhibited the expression of caspase-9 and caspase-3 and up-regulated Bcl-2/Bax pathway to inhibit H2O2-induced apoptosis. In summary, the novel delivery LUT-STE had more pronounced inhibitory effect on H2O2-induced damage in human ARPE cells.


menu
Abstract
Full text
Outline
About this article

Lutein-stevioside nanoparticle attenuates H2O2-induced oxidative damage in ARPE cells

Show Author's information Zhuqing Daia,1,Meimei Niea,1Ye ChenaJiangfeng SongaYayuan XuaZhongyuan Zhanga,b( )Guodong ZhangcShumo YandXing ZhangcDajing Lia( )
Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
Jiangsu Aland Nutrition Co., Ltd., Taizhou 214500, China
Aland Nutrition Taizhou Co., Ltd., Taizhou 225300, China

1 These authors contributed equally to this study.

Peer review under responsibility of Tsinghua University Press.

Abstract

In order to improve the bioavailability of lutein (LUT), a novel lutein-stevioside nanoparticle (LUT-STE) were prepared previously, but the information about LUT-STE on protecting of eye health was limited. This study investigated the effect of LUT-STE on antioxidant activity of H2O2-induced human retinal pigment epithelial (ARPE) cells. LUT and LUT-STE (final concentration of 5 μg/mL) significantly enhanced cell viability from (74.84 ± 5.10)% to (81.92 ± 10.01)% (LUT) and (89.33 ± 4.34)% (LUT-STE), and inhibited the cell apoptosis (P < 0.05). After pretreatment with LUT-STE in ARPE cells, the levels of superoxide dismutase (SOD), catalase (CAT) and glutathion peroxidase (GSH-Px) in ARPE cells were significantly increased (P < 0.05), the contents of reactive oxygen species (ROS) and malondialdehyde (MDA) were decreased. In addition, the vascular endothelial growth factor (VEGF) levels were inhibited by 13.61% and 17.39%, respectively, pretreatment with LUT and LUT-STE. Western blotting results showed that the pretreatment with LUT-STE inhibited the expression of caspase-9 and caspase-3 and up-regulated Bcl-2/Bax pathway to inhibit H2O2-induced apoptosis. In summary, the novel delivery LUT-STE had more pronounced inhibitory effect on H2O2-induced damage in human ARPE cells.

Keywords: Mechanism, Antioxidant, Lutein, Stevioside, Human retinal pigment epithelial cell

References(40)

[1]

S. Buscemi, D. Corleo, F. Di Pace, et al., The effect of lutein on eye and extra-eye health, Nutrients 10 (2018) 24. https://doi.org/10.3390/nu10091321.

[2]

I. Nwachukwu, C. Udenigwe, R. Aluko, Lutein and zeaxanthin: production technology, bioavailability, mechanisms of action, visual function, and health claim status, Trends Food Sci. Tech. 49 (2016) 74-84. https://doi.org/10.1016/j.tifs.2015.12.005.

[3]

J. Mares, Lutein and zeaxanthin isomers in eye health and disease, Annu. Rev. Nutr. 36 (2016) 571-602. https://doi.org/10.1146/annurevnutr-071715-051110.

[4]

T. Bohn, M.L. Bonet, P. Borel, et al., Mechanistic aspects of carotenoid health benefits: where are we now? Nutr. Res. Rev. 34 (2021) 276-302.https://doi.org/10.1017/S0954422421000147.

[5]

A. Ranganathan, G. Aruna, S. Paul, The macular carotenoids: a biochemical overview, BBA-Mol. Cell Biol. L. 1865 (2020) 158617. https://doi.org/10.1016/j.bbalip.2020.158617.

[6]

F. Barker, D. Snodderly, E. Johnson, et al., Nutritional manipulation of primate retinas, V: effects of lutein, zeaxanthin, and n-3 fatty acids on retinal sensitivity to blue-light-induced damage, Invest. Ophth. Vis. Sci. 52 (2011) 3934-3942. https://doi.org/10.1167/iovs.02-1233.

[7]

J. Silvan, M. Reguero, S. de Pascual-Teresa, A protective effect of anthocyanins and xanthophylls on UVB-induced damage in retinal pigment epithelial cells, Food Funct. 7 (2016) 1067-1076. https://doi.org/10.1039/c5fo01368b.

[8]

C. Zhu, Y. Dong, H. Liu, et al., Hesperetin protects against H2O2-triggered oxidative damage via upregulation of the Keap1-Nrf2/HO-1 signal pathway in ARPE-19 cells, Biomed. Pharmacother. 88 (2017) 124-133. https://doi.org/10.1016/j.biopha.2016.11.089.

[9]

M. Giorgio, M. Trinei, E. Migliaccio, et al., Hydrogen peroxide: a metabolic by-product or a common mediator of ageing signals? Nat. Rev. Mol. cell Bio. 8 (2007) 722-728. https://doi.org/10.1038/nrm2240.

[10]

D. Kich, S. Bitencourt, B. Caye, et al., Lymphocyte genotoxicity and protective effect of Calyptranthes tricona (Myrtaceae) against H2O2-induced cell death in MCF-7 cells, Mol. Cell. Biochem. 424 (2017) 35-43. https://doi.org/10.1007/s11010-016-2840-9.

[11]

J. Cai, C. Nelson, M. Wu, et al., Oxidative damage and protection of the RPE, Prog. Retin. Eye Res. 19 (2000) 205-221. https://doi.org/10.1016/S1350-9462(99)00009-9.

[12]

S. Li, P. Wu, B. Han, et al., Deltamethrin induces apoptosis in cerebrum neurons of quail via promoting endoplasmic reticulum stress and mitochondrial dysfunction, Environ. Tox. 37 (2022) 2033-2043. https://doi.org/10.1002/tox.23548.

[13]

S. Wang, Development of fluorescent and luminescent probes for reactive oxygen species, Trac-Trend Anal. Chem. 85 (2016) 181-202. https://doi.org/10.1016/j.trac.2016.09.006.

[14]

Y. Jiao, Y. Li, Y. Niu, et al., Small-molecule fluorescent probes for imaging and detection of reactive oxygen, nitrogen, and sulfur species in biological systems, Anal. Chem. 90 (2018) 533-555. https://doi.org/10.1021/acs.analchem.7b04234.

[15]

H. Tan, P. Yu, R. Chen, et al., Lutein protects against severe traumatic brain injury through anti-inflammation and antioxidative effects via ICAM-1/Nrf-2, Mol. Med. Rep. 16 (2017) 4235-4240. https://doi.org/10.3892/mmr.2017.7040.

[16]

W. Schaffer, S. Roy, S. Mukherjee, et al., Identification of lutein, a dietary antioxidant carotenoid in guinea pig tissues, Biochem. Bioph. Res. Co. 374 (2008) 378-381. https://doi.org/10.1016/j.bbrc.2008.07.030.

[17]

S. Bhattacharyya, S. Datta, B. Mallick, et al., Lutein content and in vitro antioxidant activity of different cultivars of Indian Marigold flower (Tagetes patula L.) extracts, J. Agr. Food Chem. 58 (2010) 8259-8264. https://doi.org/10.1021/jf101262e.

[18]

C. do Nascimento, B. Cazarin, R. Marostica, et al., Microalgae carotenoids intake: Influence on cholesterol levels, lipid peroxidation and antioxidant enzymes, Int. Food Res. J. 128 (2020) 10. https://doi.org/10.1016/j.foodres.2019.108770.

[19]

X. Zhang, N. Wang, X. Zhang, et al., Polymeric micelles for pH-responsive lutein delivery, J. Drug Deliv. Sci. Tec. 45 (2018) 281-286. https://doi.org/10.1016/j.jddst.2018.03.023.

[20]

D. Cristian, A. Elham, D. Stefan, et al., Bioavailability of nutraceuticals: role of the food matrix, processing conditions, the gastrointestinal tract, and nanodelivery systems, Compr. Rev. Food Sci. F. 19 (2020) 954-994. https://doi.org/10.1111/1541-4337.12547.

[21]

I. Lacatusu, E. Mitrea, N. Badea, et al., Lipid nanoparticles based on omega-3 fatty acids as effective carriers for lutein delivery. Preparation and in vitro characterization studies, J. Funct. Foods 5 (2013) 1260-1269. https://doi.org/10.1016/j.jff.2013.04.010.

[22]

A. Kamil, D. Smith, J. Blumberg, et al., Bioavailability and biodistribution of nanodelivered lutein, Food Chem. 192 (2016) 915-923. https://doi.org/10.1016/j.foodchem.2015.07.106.

[23]

Y. Xu, F. Song, Z. Dai, et al., Study on physicochemical characteristics of lutein nanoemulsions stabilized by chickpea protein isolate-stevioside complex, J. Sci. Food Agric. 102 (2021) 1872-1882. https://doi.org/10.1002/jsfa.11524.

[24]

H. Nguyen, J. Si, C. Kang, et al., Facile preparation of water soluble curcuminoids extracted from turmeric (Curcuma longa L.) powder by using steviol glucosides, Food Chem. 214 (2017) 366-373. https://doi.org/10.1016/j.foodchem.2016.07.102.

[25]
Z. Dai, J. Song, Y. Chen, et al., Study on the bioavailability of steviosideencapsulized lutein and its mechanism, Food Chem. 354 (2021) 129528.https://doi.org/110.1016/j.foodchem.2021.129528.
DOI
[26]

W. Huang, H. Wu, D. Li, et al., Protective effects of blueberry anthocyanins against H2O2-induced oxidative injuries in human retinal pigment epithelial cells, J. Agr. Food Chem. 66 (2018) 1638-1648. https://doi.org/10.1021/acs.jafc.7b06135.

[27]

Y. Chong, C. Mai, C. Leong, et al., Lutein improves cell viability and reduces Alu RNA accumulation in hydrogen peroxide challenged retinal pigment epithelial cells, Cutan. Ocu. Toxicol. 37 (2018) 52-60. https://doi.org/10.1080/15569527.2017.1335748.

[28]

Y. Liu, M. Liu. X. Zhang, et al., Protective effect of fucoxanthin isolated from Laminaria japonica against visible light-induced retinal damage both in vitro and in vivo, J. Agr. Food Chem. 64 (2016) 416-424. https://doi.org/10.1021/acs.jafc.5b05436.

[29]

L. Li, J. Lee, H. Leung, et al., Lutein supplementation for eye diseases, Nutrients 12 (2020) 1721. https://doi.org/10.3390/nu12061721.

[30]

C. Marazita, A. Duguor, D. Marquioni-Ramella, et al., Oxidative stressinduced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: Implications for age-related macular degeneration, Redox Biol. 7 (2016) 78-87. https://doi.org/10.1016/j.redox.2015.11.011.

[31]

H. Ku, J. Park. Downregulation of IDH2 exacerbates H2O2-mediated cell death and hypertrophy, Redox Rep. 1 (2017) 35-41. https://doi.org/10.1080/13510002.2015.1135581.

[32]

T. Ni, W. Yang, Y. Xing, Protective effects of delphinidin against H2O2-induced oxidative injuries in human retinal pigment epithelial cells, Biosci. Rep. 39 (2019) BSR20190689. https://doi.org/10.1042/BSR20190689.

[33]

A. Sene, D. Chin-Yee, R. S. Apte, Seeing through VEGF: innate and adaptive immunity in pathological angiogenesis in the eye, Trends Mol. Med. 21(2015) 43-51. https://doi.org/10.1016/j.molmed.2014.10.005.

[34]

M. Karkkainen, T. Petrova, Vascular endothelial growth factor receptors in the regulation of angiogenesis and lymphangiogenesis, Oncogene 19 (2000) 5598-5605. https://doi.org/10.1038/sj.onc.1203855.

[35]

M. Brentnall, L. Rodriguez-Menocal, R. de Guevara, et al., Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis, BMC Cell Biol. 14(1) (2013) 23834359.

[36]

R. Murthy, K. Ravi, S. Balaiya, et al., Lutein protects retinal pigment epithelium from cytotoxic oxidative stress, Cutan. Ocul. Toxicol. 33 (2014) 132-137. https://doi.org/10.3109/15569527.2013.812108.

[37]

H. Liu, W. Liu, X. Zhou, et al., Protective effect of lutein on ARPE-19 cells upon H2O2-induced G2/M arrest, Mole. Med. Rep. 16 (2017) 2069. https://doi.org/10.3892/mmr.2017.6838.

[38]

N. Singh, Apoptosis in health and disease and modulation of apoptosis for therapy: an overview, Ind. J. Clini. Biochem. 22 (2007) 6-16. https://doi.org/10.1089/neu.2000.17.801.

[39]

D.R. Green, Apoptotic pathways: paper wraps stone blunts scissors, Cell 102 (2000) 1-4. https://doi.org/10.1016/s0092-8674(00)00003-9.

[40]

E. Khodapasand, N. Jafarzadeh, F. Farrokhi, et al., Is Bax/Bcl-2 ratio considered as a prognostic marker with age and tumor location in colorectal cancer? Iran. Biomed. J. 19 (2015) 69-75. https://doi.org/10.6091/ibj.1366.2015.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 16 November 2022
Revised: 07 December 2022
Accepted: 19 December 2022
Published: 08 February 2024
Issue date: May 2024

Copyright

© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

Acknowledgements

Acknowledgement

The present work received research funding from the National Natural Science Foundation of China (31801541), the Independent Innovation Fund Project of Agricultural Science and Technology in Jiangsu Province (CX(22)3065), Major Scientific and Technological Achievements Transformation Project of Taizhou (SCG 202105), and the Taizhou Science and Technology Support Plan (TN202106).

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