<iframe src="//www.googletagmanager.com/ns.html?id=GTM-5TSRKG" height="0" width="0" style="display: none; visibility: hidden">
Research Article
No access
Published Online: 13 April 2023

Tetrahydroxystilbene Glucoside Attenuates Oxidative Stress-Induced Aging by Regulating Oxidation Resistance and Inflammation in Larval Zebrafish

Publication: Zebrafish
Volume 20, Issue Number 2

Abstract

Population aging is a global problem worldwide, and the discovery of antiaging drugs and knowledge of their potential molecular mechanisms are research hotspots in biomedical field. Tetrahydroxystilbene glucoside (TSG) is a natural component isolated from Heshouwu (Polygonum multiflorum Thunb.). It has been widely used to treat various chronic diseases for its remarkable biological activities. In this study, we successfully established aging larval zebrafish by exposing larvae to 2 mM hydrogen peroxide (H2O2). Using this aging model, we assessed the antiaging effect of TSG with different concentrations (25–100 μg/mL). After being treated with H2O2, zebrafish showed the obvious aging-associated phenotypes characterized by higher senescence-associated β-galactosidase activity, significantly downregulated expression of sirtuin 1 (sirt1) and telomerase reverse transcriptase (tert), and upregulated serpine1 mRNA level compared to the control group. TSG pretreatment delayed the aging process of oxidative stress-induced zebrafish, indicative of the reduced positive rate of senescence-associated β-galactosidase, improved swimming velocity, and stimulus-response capacity. Further studies proved that TSG could suppress reactive oxygen species production and enhance the activity of antioxidant enzymes superoxide dismutase and catalase. TSG also inhibited the H2O2-induced expressions of inflammation-related genes il-1β, il-6, cxcl-c1c, and il-8 in aging zebrafish, but it did not affect apoptosis-related genes (bcl-2, bax, and caspase-3) of aging zebrafish. In conclusion, TSG can protect against aging by regulating the antioxidative genes and enzyme activity, as well as inflammation in larval zebrafish, providing insight into the application of TSG for clinical treatment of aging or aging-related diseases.

Get full access to this article

View all available purchase options and get full access to this article.

References

1. Partridge L, Fuentealba M, Kennedy BK. The quest to slow ageing through drug discovery. Nat Rev Drug Discov 2020;19(8):513–532;
2. López-Otín C, Blasco MA, Partridge L, et al. The hallmarks of aging. Cell 2013;153(6):1194–1217;
3. Palmer AK, Xu M, Zhu Y, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 2019;18(3):e12950;
4. Villeda SA, Plambeck KE, Middeldorp J, et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med 2014;20(6):659–663;
5. Smith P, Willemsen D, Popkes M, et al. Regulation of life span by the gut microbiota in the short-lived African turquoise killifish. Elife 2017;6:e27014;
6. Callaway E. ‘Young poo’ makes aged fish live longer. Nature 2017;544(7649):147;
7. Omodei D, Fontana L. Calorie restriction and prevention of age-associated chronic disease. FEBS Lett 2011;585(11):1537–1542;
8. Howe K, Clark MD, Torroja CF, et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature 2013;496(7446):498–503;
9. Van Houcke J, De Groef L, Dekeyster E, et al. The zebrafish as a gerontology model in nervous system aging, disease, and repair. Ageing Res Rev 2015;24(Pt B):358–368;
10. Henriques CM, Carneiro MC, Tenente IM, et al. Telomerase is required for zebrafish lifespan. PLoS Genet 2013;9(1):e1003214;
11. Gilbert MJ, Zerulla TC, Tierney KB. Zebrafish (Danio rerio) as a model for the study of aging and exercise: Physical ability and trainability decrease with age. Exp Gerontol 2014;50:106–113;
12. Tsai SB, Tucci V, Uchiyama J, et al. Differential effects of genotoxic stress on both concurrent body growth and gradual senescence in the adult zebrafish. Aging Cell 2007;6(2):209–224;
13. Hayes AJ, Reynolds S, Nowell MA, et al. Spinal deformity in aged zebrafish is accompanied by degenerative changes to their vertebrae that resemble osteoarthritis. PLoS One 2013;8(9):e75787;
14. Liu Y, Wang Q, Yang J, et al. Polygonum multiflorum thunb.: A review on chemical analysis, processing mechanism, quality evaluation, and hepatotoxicity. Front Pharmacol 2018;9:364;
15. Bounda GA, Feng YU. Review of clinical studies of Polygonum multiflorum thunb. and its isolated bioactive compounds. Pharmacognosy Res 2015;7(3):225–236;
16. Chen HS, Liu Y, Lin LQ, et al. Anti-proliferative effect of an extract of the root of Polygonum multiflorum thunb. on MCF-7 human breast cancer cells and the possible mechanisms. Molecular medicine reports 2011;4(6):1313–1319;
17. Hu M, Zeng W, Tomlinson B. Evaluation of a crataegus-based multiherb formula for dyslipidemia: A randomized, double-blind, placebo-controlled clinical trial. Evidence-based complementary and alternative medicine: eCAM 2014;2014:365742;
18. Sun ML, Chen XY, Cao JJ, et al. Polygonum multiflorum thunb extract extended the lifespan and healthspan of Caenorhabditis elegans via DAF-16/SIR-2.1/SKN-1. Food Funct 2021;12(18):8774–8786;
19. Li X, Matsumoto K, Murakami Y, et al. Neuroprotective effects of Polygonum multiflorum on nigrostriatal dopaminergic degeneration induced by paraquat and maneb in mice. Pharmacol Biochem Behav 2005;82(2):345–352;
20. Wang C, Zhou Y, Gong X, et al. In vitro and in situ study on characterization and mechanism of the intestinal absorption of 2,3,5,4′-tetrahydroxy-stilbene-2-O-β-D-glucoside. BMC Pharmacol Toxicol 2020;21(1):7;
21. Zhang L, Chen J. Biological effects of tetrahydroxystilbene glucoside: An active component of a rhizome extracted from Polygonum multiflorum. Oxid Med Cell Longev 2018;2018:3641960;
22. Wang C, Dai S, Gong L, et al. A review of pharmacology, toxicity and pharmacokinetics of 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside. Front Pharmacol 2022;12:791214;
23. Robert G, Wagner JR. ROS-induced DNA damage as an underlying cause of aging. Adv Geriatr Med Res 2020;2(4):e200024;
24. Davalli P, Mitic T, Caporali A, et al. ROS, cell senescence, and novel molecular mechanisms in aging and age-related diseases. Oxidat Med Cell Longev 2016;2016:3565127;
25. Li E, Wang Y, Li Q, et al. Protective effects of sal b on oxidative stress-induced aging by regulating the Keap1/Nrf2 signaling pathway in zebrafish. Molecules 2021;26(17):5239;
26. Kishi S, Bayliss PE, Uchiyama J, et al. The identification of zebrafish mutants showing alterations in senescence-associated biomarkers. PLoS Genet 2008;4(8):e1000152;
27. Yu L, Tucci V, Kishi S, et al. Cognitive aging in zebrafish. PLoS One 2006;1(1):e14;
28. Kregel KC, Zhang HJ. An integrated view of oxidative stress in aging: Basic mechanisms, functional effects, and pathological considerations. Am J Physiol Regul Integr Comp Physiol 2007;292(1):R18–R36;
29. Chung HY, Kim DH, Lee EK, et al. Redefining chronic inflammation in aging and age-related diseases: Proposal of the senoinflammation concept. Aging Dis 2019;10(2):367–382;
30. Lee HC, Wei YH. Oxidative stress, mitochondrial DNA mutation, and apoptosis in aging. Exp Biol Med (Maywood, NJ) 2007;232(5):592–606.
31. Wang S, Chi Q, Hu X, et al. Hydrogen sulfide-induced oxidative stress leads to excessive mitochondrial fission to activate apoptosis in broiler myocardia. Ecotoxicol Environ Safety 2019;183:109578;
32. Mitchell SJ, Scheibye-Knudsen M, Longo DL, et al. Animal models of aging research: Implications for human aging and age-related diseases. Annu Rev Anim Biosci 2015;3:283–303;
33. Kishi S, Uchiyama J, Baughman AM, et al. The zebrafish as a vertebrate model of functional aging and very gradual senescence. Exp Gerontol 2003;38(7):777–786;
34. Zhao T, Ye S, Tang Z, et al. Loss-of-function of p53 isoform Δ113p53 accelerates brain aging in zebrafish. Cell Death Dis 2021;12(2):151;
35. Kim DH, Jung IH, Kim DH, et al. Knockout of longevity gene Sirt1 in zebrafish leads to oxidative injury, chronic inflammation, and reduced life span. PLoS One 2019;14(8):e0220581;
36. Bonomini F, Rodella LF, Rezzani R. Metabolic syndrome, aging and involvement of oxidative stress. Aging Dis 2015;6(2):109–120;
37. Singh A, Kukreti R, Saso L, et al. Oxidative stress: A key modulator in neurodegenerative diseases. Molecules 2019;24(8):1583;
38. Jiang Z, Wang W, Guo C. Tetrahydroxy stilbene glucoside ameliorates H2O2-induced human brain microvascular endothelial cell dysfunction in vitro by inhibiting oxidative stress and inflammatory responses. Mol Med Rep 2017;16(4):5219–5224;
39. Xie M, Zhang G, Yin W, et al. Cognitive enhancing and antioxidant effects of tetrahydroxystilbene glucoside in Aβ1-42-induced neurodegeneration in mice. J Integrat Neurosci 2018;17(3–4):355–365;
40. Tao L, Li X, Zhang L, et al. Protective effect of tetrahydroxystilbene glucoside on 6-OHDA-induced apoptosis in PC12 cells through the ROS-NO pathway. PLoS One 2011;6(10):e26055;
41. Wu TY, Lin JN, Luo ZY, et al. 2,3,4′,5-tetrahydroxystilbene-2-O-β-D-glucoside (THSG) activates the Nrf2 antioxidant pathway and attenuates oxidative stress-induced cell death in mouse cochlear UB/OC-2 cells. Biomolecules 2020;10(3):465;
42. Büchter C, Zhao L, Havermann S, et al. TSG (2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside) from the Chinese herb Polygonum multiflorum increases life span and stress resistance of Caenorhabditis elegans. Oxidat Med Cell Longev 2015;2015:124357;
43. Lv L, Gu X, Tang J, et al. Antioxidant activity of stilbene glycoside from Polygonum multiflorum thunb in vivo. Food Chem 2007;104(4):1678–1681;
44. Draganidis D, Jamurtas AZ, Chondrogianni N, et al. Low-grade systemic inflammation interferes with anabolic and catabolic characteristics of the aged human skeletal muscle. Oxidat Med Cell Longev 2021;2021:8376915;
45. Sanada F, Taniyama Y, Muratsu J, et al. Source of chronic inflammation in aging. Front Cardiovasc Med 2018;5:12;
46. Singh T, Newman AB. Inflammatory markers in population studies of aging. Ageing Res Rev 2011;10(3):319–329;
47. Yu S, Xia H, Guo Y, et al. Ginsenoside Rb1 retards aging process by regulating cell cycle, apoptotic pathway and metabolism of aging mice. J Ethnopharmacol 2020;255:112746;
48. Chin YT, Hsieh MT, Lin CY, et al. 2,3,5,4′-tetrahydroxystilbene-2-O-β-glucoside isolated from polygoni multiflori ameliorates the development of periodontitis. Mediators Inflamm 2016;2016:6953459;
49. Zeng C, Xiao JH, Chang MJ, et al. Beneficial effects of THSG on acetic acid-induced experimental colitis: Involvement of upregulation of PPAR-γ and inhibition of the Nf-Κb inflammatory pathway. Molecules 2011;16(10):8552–8568;
50. Fan YF, Guan SY, Luo L, et al. Tetrahydroxystilbene glucoside relieves the chronic inflammatory pain by inhibiting neuronal apoptosis, microglia activation, and GluN2B overexpression in anterior cingulate cortex. Mol Pain 2018;14:1744806918814367;
51. Zhang R, Sun F, Zhang L, et al. Tetrahydroxystilbene glucoside inhibits α-synuclein aggregation and apoptosis in A53T α-synuclein-transfected cells exposed to MPP. Can J Physiol Pharmacol 2017;95(6):750–758;
52. Zhang L, Huang L, Li X, et al. Potential molecular mechanisms mediating the protective effects of tetrahydroxystilbene glucoside on MPP(+)-induced PC12 cell apoptosis. Mol Cell Biochem 2017;436(1–2):203–213;

Information & Authors

Information

Published In

cover image Zebrafish
Zebrafish
Volume 20Issue Number 2April 2023
Pages: 55 - 66

History

Published online: 13 April 2023
Published in print: April 2023

Permissions

Request permissions for this article.

Topics

Authors

Affiliations

Hui Xia*
College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China.
Xue Cheng*
College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China.
Mengxi Cao
Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, China.
Xiongjie Sun
College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China.
Fuyi He
College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China.
Xiaowei Yao
College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China.
College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China.

Notes

*
These authors contributed equally to this work and should be considered co-first authors.
Address correspondence to: Hongtao Liu, PhD, College of Basic Medicine, Hubei University of Chinese Medicine, 16 Huangjiahu West Road, Hongshan Disctrict, Wuhan 430065, China [email protected]

Authors' Contributions

All authors contributed to the study conception and design. H.L. and X.Z. conceived and designed research. H.X. and X.C. conducted experiments and wrote the article. X.Y. and X.S. contributed new reagents or analytical tools. F.H. and M.C. analyzed data.

Disclosure Statement

No competing financial interests exist.

Funding Information

This work was supported by Hubei Provincial Department of Education (no. Q20192006).

Ethical Approval

All animal experiments were approved by the animal ethics committee of Hubei University of Chinese Medicine and conducted following the Guiding Principles for the Care and Use of Laboratory Animals.

Metrics & Citations

Metrics

Citations

Export citation

Select the format you want to export the citations of this publication.

View Options

Get Access

Access content

To read the fulltext, please use one of the options below to sign in or purchase access.

Society Access

If you are a member of a society that has access to this content please log in via your society website and then return to this publication.

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

View options

PDF/EPUB

View PDF/ePub

Full Text

View Full Text

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media

Back to Top