EFFECT OF DIHYDROQUERCETIN ON THE TOXIC PROPERTIES OF NICKEL NANOPARTICLES
Рубрики: RESEARCH ARTICLE
Аннотация и ключевые слова
Аннотация (русский):
Dihydroquercetin (3,5,7,3',4'-pentahydroxy-flavanone) is known for its powerful antioxidant, organ-protective, and antiinflammatory activities that can be applied to heavy-metal intoxication. The present research objective was to evaluate the possible protective potential of dietary dihydroquercetin in a rat model of subacute (92 days) intoxication with nickel nanoparticles. The experiment involved five groups of twelve male Wistar rats in each. Group 1 served as control. Other groups received nickel nanoparticles as part of their diet. Groups 2 and 4 received nickel nanoparticles with an average diameter of 53.7 nm (NiNP1), while groups 3 and 5 were fed with nanoparticles with an average diameter of 70.9 nm (NiNP2). The dose was calculated as 10 mg/kg b.w. Groups 4 and 5 also received 23 mg/kg b.w. of water-soluble stabilized dihydroquercetin with drinking water. After the dihydroquercetin treatment, the group that consumed 53.7 nm nickel nanoparticles demonstrated lower blood serum glucose, triglycerides, low-density lipoprotein cholesterol, and creatinine. Dihydroquercetin prevented the increase in total protein and albumin fraction associated with nickel nanoparticles intake. The experimental rats also demonstrated lower levels of pro-inflammatory cytokines IL-1β, IL-4, IL-6, and IL-17A, as well as a lower relative spleen weight after the treatment. In the group exposed to 53.7 nm nickel nanoparticles, the dihydroquercetin treatment increased the ratio of cytokines IL-10/IL-17A and decreased the level of circulating FABP2 protein, which is a biomarker of increased intestinal barrier permeability. In the group that received 70.9 nm nickel nanoparticles, the dihydroquercetin treatment inhibited the expression of the fibrogenic Timp3 gene in the liver. In the group that received 53.7 nm nickel nanoparticles, dihydroquercetin partially improved the violated morphology indexes in liver and kidney tissue. However, dihydroquercetin restored neither the content of reduced glutathione in the liver nor the indicators of selenium safety, which were suppressed under the effect of nickel nanoparticles. Moreover, the treatment failed to restore the low locomotor activity in the elevated plus maze test. Dihydroquercetin treatment showed some signs of detoxication and anti-inflammation in rats subjected to nickel nanoparticles. However, additional preclinical studies are necessary to substantiate its prophylactic potential in cases of exposure to nanoparticles of nickel and other heavy metals.

Ключевые слова:
Nanoparticles, nickel, dihydroquercetin, rats, detoxification, cytokines, intestinal barrier permeability
Текст
Текст произведения (PDF): Читать Скачать
Список литературы

1. Zhang P, Wang L, Yang S, Schott JA, Liu X, Mahurin SM, et al. Solid-state synthesis of ordered mesoporous carbon catalysts via a mechanochemical assembly through coordination cross-linking. Nature Communications. 2017;8. https://doi.org/10.1038/ncomms15020

2. Ban I, Stergar J, Drofenik M, Ferk G, Makovec D. Synthesis of copper-nickel nanoparticles prepared by mechanical milling for use in magnetic hyperthermia. Journal of Magnetism and Magnetic Materials. 2011;323(17):2254-2258. https://doi.org/10.1016/j.jmmm.2011.04.004

3. Elango G, Roopan SM, Dhamodaran KI, Elumalai K, Al-Dhabi NA, Valan Arasu M. Spectroscopic investigation of biosynthesized nickel nanoparticles and its larvicidal, pesticidal activities. Journal of Photochemistry and Photobiology B: Biology. 2016;162:162-167. https://doi.org/10.1016/j.jphotobiol.2016.06.045

4. Borowska S, Brzóska MM. Metals in cosmetics: implications for human health. Journal of Applied Toxicology. 2015;35(6):551-572. https://doi.org/10.1002/jat.3129

5. Phillips JI, Green FY, Davies JCA, Murray J. Pulmonary and systemic toxicity following exposure to nickel nanoparticles. American Journal of Industrial Medicine. 2010;53(8):763-767. https://doi.org/10.1002/ajim.20855

6. Iqbal S, Jabeen F, Peng C, Ijaz MU, Chaudhry AS. Cinnamomum cassia ameliorates Ni-NPs-induced liver and kidney damage in male Sprague Dawley rats. Human and Experimental Toxicology. 2020;39(11):1565-1581. https://doi.org/10.1177/0960327120930125

7. Zhao J, Bowman L, Zhang X, Shi X, Jiang B, Castranova V, et al. Metallic nickel nano- and fine particles induce JB6 cell apoptosis through a caspase-8/AIF mediated cytochrome c-independent pathway. Journal of Nanobiotechnology. 2009;7. https://doi.org/10.1186/1477-3155-7-2

8. Zhang Q, Chang X, Wang H, Liu Y, Wang X, Wu M, et al. TGF-β1 mediated Smad signaling pathway and EMT in hepatic fibrosis induced by Nano NiO in vivo and in vitro. Environmental Toxicology. 2020;35(4):419-429. https://doi.org/10.1002/tox.22878

9. Kong L, Hu W, Lu C, Cheng K, Tang M. Mechanisms underlying nickel nanoparticle induced reproductive toxicity and chemo-protective effects of vitamin C in male rats. Chemosphere. 2019;218:259-265. https://doi.org/10.1016/j.chemosphere.2018.11.128

10. Hansen T, Clermont G, Alves A, Eloy R, Brochhausen C, Boutrand JP, et al. Biological tolerance of different materials in bulk and nanoparticulate form in a rat model: sarcoma development by nanoparticles. Journal of the Royal Society Interface. 2006;3(11):767-775. https://doi.org/10.1098/rsif.2006.0145

11. Sutunkova MP, Privalova LI, Minigalieva IA, Gurvich VB, Panov VG, Katsnelson BA. The most important inferences from the Ekaterinburg nanotoxicology team’s animal experiments assessing adverse health effects of metallic and metal oxide nanoparticles. Toxicology Reports. 2018;5:363-376. https://doi.org/10.1016/j.toxrep.2018.03.008

12. Das A, Baidya R, Chakraborty T, Samanta AK, Roy S. Pharmacological basis and new insights of taxifolin: A comprehensive review. Biomedicine and Pharmacotherapy. 2021;142. https://doi.org/10.1016/j.biopha.2021.112004

13. Orlova SV, Tatarinov VV, Nikitina EA, Sheremeta AV, Ivlev VA, Vasil’ev VG, et al. Bioavailability and safety of dihydroquercetin (review). Pharmaceutical Chemistry Journal. 2022;55(11):1133-1137. https://doi.org/10.1007/s11094-022-02548-8

14. Zinchenko VP, Kim YuA, Tarakhovskii YuS, Bronnikov GE. Biological activity of water-soluble nanostructures of dihydroquercetin with cyclodextrins. Biophysics. 2011;56(3):418-422. https://doi.org/10.1134/S0006350911030298

15. Mzhelskaya KV, Shipelin VA, Shumakova AA, Musaeva AD, Soto JS, Riger NA, et al. Effects of quercetin on the neuromotor function and behavioral responses of Wistar and Zucker rats fed a high-fat and high-carbohydrate diet. Behavioural Brain Research. 2020;378. https://doi.org/10.1016/j.bbr.2019.112270

16. Trusov NV, Semin MO, Shipelin VA, Apryatin SA, Gmoshinski IV. Liver gene expression in normal and obese rats received resveratrol and L-carnitine. Problems of Nutrition. 2021;90(5):25-37. (In Russ.). https://doi.org/10.33029/0042-8833-2021-90-5-25-37

17. Lau E, Marques C, Pestana D, Santoalha M, Carvalho D, Freitas P, et al. The role of I-FABP as a biomarker of intestinal barrier dysfunction driven by gut microbiota changes in obesity. Nutrition and Metabolism. 2016;13. https://doi.org/10.1186/s12986-016-0089-7

18. Rosliy IM. Biochemical indicators in medicine and biology. Moscow: Meditsinskoe informatsionnoe agentstvo; 2015. 609 p. (In Russ.).

19. Salama SA, Kabel AM. Taxifolin ameliorates iron overload-induced hepatocellular injury: Modulating PI3K/AKT and p38 MAPK signaling, inflammatory response, and hepatocellular regeneration. Chemico-Biological Interactions. 2020;330. https://doi.org/10.1016/j.cbi.2020.109230

20. Moon SH, Lee CM, Nam MJ. Cytoprotective effects of taxifolin against cadmium-induced apoptosis in human keratinocytes. Human and Experimental Toxicology. 2019;38(8):992-1003. https://doi.org/10.1177/0960327119846941

21. Ding C, Zhao Y, Chen X, Zheng Y, Liu W, Liu X. Taxifolin, a novel food, attenuates acute alcohol-induced liver injury in mice through regulating the NF-κB-mediated inflammation and PI3K/Akt signalling pathways. Pharmaceutical Biology. 2021;59(1):868-879. https://doi.org/10.1080/13880209.2021.1942504

22. Liu X, Liu W, Ding C, Zhao Y, Chen X, Ling D, et al. Taxifolin, extracted from waste Larix olgensis roots, attenuates CCl4-induced liver fibrosis by regulating the PI3K/AKT/mTOR and TGF-β1/Smads signaling pathways. Drug Design, Development and Therapy. 2021;15:871-87. https://doi.org/10.2147/DDDT.S281369

23. Wang W, Ma B, Xu C, Zhou X. Dihydroquercetin protects against renal fibrosis by activating the Nrf2 pathway. Phytomedicine. 2020;69. https://doi.org/10.1016/j.phymed.2020.153185

24. Akinmoladun AC, Olaniyan OO, Famusiwa CD, Josiah SS, Olaleye MT. Ameliorative effect of quercetin, catechin, and taxifolin on rotenone-induced testicular and splenic weight gain and oxidative stress in rats. Journal of Basic and Clinical Physiology and Pharmacology. 2020;31(3). https://doi.org/10.1515/jbcpp-2018-0230

25. Hou J, Hu M, Zhang L, Gao Y, Ma L, Xu Q. Dietary taxifolin protects against dextran sulfate sodium-induced colitis via NF-κB signaling, enhancing intestinal barrier and modulating gut microbiota. Frontiers in Immunology. 2020;11. https://doi.org/10.3389/fimmu.2020.631809

26. Muramatsu D, Uchiyama H, Kida H, Iwai A. In vitro anti-inflammatory and anti-lipid accumulation properties of taxifolin-rich extract from the Japanese larch, Larix kaempferi. Heliyon. 2020;6(12). https://doi.org/10.1016/j.heliyon.2020.e05505

27. Kondo S, Adachi S, Yoshizawa F, Yagasaki K. Antidiabetic effect of taxifolin in cultured L6 myotubes and type 2 diabetic model KK-Ay/Ta mice with hyperglycemia and hyperuricemia. Current Issues in Molecular Biology. 2021;43(3):1293-1306. https://doi.org/10.3390/cimb43030092

28. Lei L, Chai Y, Lin H, Chen C, Zhao M, Xiong W, et al. Dihydroquercetin activates AMPK/Nrf2/HO-1 signaling in macrophages and attenuates inflammation in LPS-induced endotoxemic mice. Frontiers in Pharmacology. 2020;11. https://doi.org/10.3389/fphar.2020.00662

29. Pan S, Zhao X, Ji N, Shao C, Fu B, Zhang Z, et al. Inhibitory effect of taxifolin on mast cell activation and mast cell-mediated allergic inflammatory response. International Immunopharmacology. 2019;71:205-214. https://doi.org/10.1016/j.intimp.2019.03.038

30. Ye Y, Wang X, Cai Q, Zhuang J, Tan X, He W, et al. Protective effect of taxifolin on H2O2-induced H9C2 cell pyroptosis. Journal of Central South University. 2017;42(12):1367-1374. https://doi.org/10.11817/j.issn.1672-7347.2017.12.003

31. Di T, Zhai C, Zhao J, Wang Y, Chen Z, Li P. Taxifolin inhibits keratinocyte proliferation and ameliorates imiquimod-induced psoriasis-like mouse model via regulating cytoplasmic phospholipase A2 and PPAR-γ pathway. International Immunopharmacology. 2021;99. https://doi.org/10.1016/j.intimp.2021.107900


Войти или Создать
* Забыли пароль?