DOI QR코드

DOI QR Code

Preventive effects of nano-graphene oxide against Parkinson's disease via reactive oxygen species scavenging and anti-inflammation

  • Hee-Yeong Kim (Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University) ;
  • Hyung Ho Yoon (Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Hanyu Seong (Department of Neurosurgery, Seoul Bumin Hospital) ;
  • Dong Kwang Seo (Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Soon Won Choi (Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University) ;
  • Jaechul Ryu (Institute of Bio & Nano Convergence, Biogo Co., Ltd.) ;
  • Kyung-Sun Kang (Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University) ;
  • Sang Ryong Jeon (Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine)
  • Received : 2022.09.08
  • Accepted : 2022.11.23
  • Published : 2023.03.31

Abstract

We investigated the neuroprotective effects of deca nano-graphene oxide (daNGO) against reactive oxygen species (ROS) and inflammation in the human neuroblastoma cell line SH-SY5Y and in the 6-hydroxydopamine (6-OHDA) induced Parkinsonian rat model. An MTT assay was performed to measure cell viability in vitro in the presence of 6-OHDA and/or daNGO. The intracellular ROS level was quantified using 2',7'-dichlorofluorescein diacetate. daNGO showed neuroprotective effects against 6-OHDA-induced toxicity and also displayed ROS scavenging properties. We then tested the protective effects of daNGO against 6-OHDA induced toxicity in a rat model. Stepping tests showed that the akinesia symptoms were improved in the daNGO group compared to the control group. Moreover, in an apomorphine-induced rotation test, the number of net contralateral rotations was decreased in the daNGO group compared to the control group. By immunofluorescent staining, the animals in the daNGO group had more tyrosine hydroxylase-positive cells than the controls. By anti-Iba1 staining, 6-OHDA induced microglial activation showed a significantly decrease in the daNGO group, indicating that the neuroprotective effects of graphene resulted from anti-inflammation. In conclusion, nano-graphene oxide has neuroprotective effects against the neurotoxin induced by 6-OHDA on dopaminergic neurons.

Keywords

Acknowledgement

This study was supported by an Asan Institute for Life Sciences Grant (2022IP0076, 2022IL0035) from Asan Medical Center, Seoul, Republic of Korea, and by the Basic Science Research Program through a National Research Foundation of Korea (NRF) grant of the Korean government (MSIT) (No. 2020R1A4A4078907).

References

  1. Poewe W, Seppi K, Tanner CM et al (2017) Parkinson disease. Nat Rev Dis Primers 3, 1-21  https://doi.org/10.1038/nrdp.2017.13
  2. Fabbrini G, Brotchie JM, Grandas F, Nomoto M and Goetz CG (2007) Levodopa-induced dyskinesias. Mov Disord 22, 1379-1389  https://doi.org/10.1002/mds.21475
  3. Benabid AL (2003) Deep brain stimulation for Parkinson's disease. Curr Opin Neurobiol 13, 696-706  https://doi.org/10.1016/j.conb.2003.11.001
  4. Kalia LV, Kalia SK and Lang AE (2015) Disease-modifying strategies for Parkinson's disease. Mov Disord 30, 1442-1450  https://doi.org/10.1002/mds.26354
  5. Lotharius J and Brundin P (2002) Pathogenesis of Parkinson's disease: dopamine, vesicles and alpha-synuclein. Nat Rev Neurosci 3, 932-942  https://doi.org/10.1038/nrn983
  6. Jenner P (2003) Oxidative stress in Parkinson's disease. Ann Neurol 53, S26-S38  https://doi.org/10.1002/ana.10483
  7. Lee BC, Lee JY, Kim J et al (2020) Graphene quantum dots as anti-inflammatory therapy for colitis. Sci Adv 6, eaaz2630 
  8. Han J, Kim YS, Lim MY et al (2018) Dual roles of graphene oxide to attenuate inflammation and elicit timely polarization of macrophage phenotypes for cardiac repair. Acs Nano 12, 1959-1977  https://doi.org/10.1021/acsnano.7b09107
  9. Bacon M, Bradley SJ and Nann T (2014) Graphene quantum dots. Part Part Syst Charact 31, 415-428  https://doi.org/10.1002/ppsc.201300252
  10. Kim D, Yoo JM, Hwang H et al (2018) Graphene quantum dots prevent alpha-synucleinopathy in Parkinson's disease. Nat Nanotechnol 13, 812-818  https://doi.org/10.1038/s41565-018-0179-y
  11. Jeong JK, Lee YJ, Jeong SY, Jeong S, Lee GW and Park SY (2017) Autophagic flux induced by graphene oxide has a neuroprotective effect against human prion protein fragments. Int J Nanomedicine 12, 8143 
  12. Xiaoli F, Yaqing Z, Ruhui L et al (2021) Graphene oxide disrupted mitochondrial homeostasis through inducing intracellular redox deviation and autophagy-lysosomal network dysfunction in SH-SY5Y cells. J Hazard Mater 416, 126158 
  13. Lee BC, Lee JY, Kim J et al (2020) Graphene quantum dots as anti-inflammatory therapy for colitis. Sci Adv 6, eaaz2630 
  14. Ma J, Liu R, Wang X et al (2015) Crucial role of lateral size for graphene oxide in activating macrophages and stimulating pro-inflammatory responses in cells and animals. ACS Nano 9, 10498-10515  https://doi.org/10.1021/acsnano.5b04751
  15. Yue H, Wei W, Yue Z et al (2012) The role of the lateral dimension of graphene oxide in the regulation of cellular responses. Biomaterials 33, 4013-4021  https://doi.org/10.1016/j.biomaterials.2012.02.021
  16. Markovic ZM, Jovanovic SP, Maskovic PZ et al (2019) Graphene oxide size and structure pro-oxidant and antioxidant activity and photoinduced cytotoxicity relation on three cancer cell lines. J Photochem Photobiol B 200, 111647 
  17. Guo S, Bezard E and Zhao B (2005) Protective effect of green tea polyphenols on the SH-SY5Y cells against 6-OHDA induced apoptosis through ROS-NO pathway. Free Radic Biol Med 39, 682-695  https://doi.org/10.1016/j.freeradbiomed.2005.04.022
  18. Shahriar SMS, Nafiujjaman M, An JM et al (2022) Graphene: a promising theranostic agent. Adv Exp Med Biol 1351, 149-176  https://doi.org/10.1007/978-981-16-4923-3_8
  19. Huang C, Wu J, Jiang W, Liu R, Li Z and Luan Y (2018) Amphiphilic prodrug-decorated graphene oxide as a multifunctional drug delivery system for efficient cancer therapy. Mater Sci Eng C Mater Biol Appl 89, 15-24  https://doi.org/10.1016/j.msec.2018.03.017
  20. Jiang W, Chen J, Gong C, Wang Y, Gao Y and Yuan Y (2020) Intravenous delivery of enzalutamide based on high drug loading multifunctional graphene oxide nanoparticles for castration-resistant prostate cancer therapy. J Nanobiotechnology 18, 50 
  21. Tabish TA and Narayan RJ (2021) Crossing the bloodbrain barrier with graphene nanostructures. Mater Today 51, 393-401  https://doi.org/10.1016/j.mattod.2021.08.013
  22. Mittal M, Siddiqui MR, Tran K, Reddy SP and Malik AB (2014) Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal 20, 1126-1167  https://doi.org/10.1089/ars.2012.5149
  23. Moore DJ, West AB, Dawson VL and Dawson TM (2005) Molecular pathophysiology of Parkinson's disease. Annu Rev Neurosci 28, 57-87  https://doi.org/10.1146/annurev.neuro.28.061604.135718
  24. Ren C, Hu X and Zhou Q (2018) Graphene oxide quantum dots reduce oxidative stress and inhibit neurotoxicity in vitro and in vivo through catalase-like activity and metabolic regulation. Adv Sci (Weinh) 5, 1700595 
  25. Srikanth K, Sundar LS, Pereira E and Duarte AC (2018) Graphene oxide induces cytotoxicity and oxidative stress in bluegill sunfish cells. J Appl Toxicol 38, 504-513  https://doi.org/10.1002/jat.3557
  26. Nanda SS, An SSA and Yi DK (2015) Oxidative stress and antibacterial properties of a graphene oxide-cystamine nanohybrid. Int J Nanomedicine 10, 549 
  27. Wang A, Pu K, Dong B et al (2013) Role of surface charge and oxidative stress in cytotoxicity and genotoxicity of graphene oxide towards human lung fibroblast cells. J Appl Toxicol 33, 1156-1164  https://doi.org/10.1002/jat.2877
  28. Elkon H, Melamed E and Offen D (2004) Oxidative stress, induced by 6-hydroxydopamine, reduces proteasome activities in PC12 cells: implications for the pathogenesis of Parkinson's disease. J Mol Neurosci 24, 387-400  https://doi.org/10.1385/JMN:24:3:387
  29. Xicoy H, Wieringa B and Martens GJ (2017) The SH-SY5Y cell line in Parkinson's disease research: a systematic review. Mol Neurodegener 12, 10 
  30. Schober A (2004) Classic toxin-induced animal models of Parkinson's disease: 6-OHDA and MPTP. Cell Tissue Res 318, 215-224  https://doi.org/10.1007/s00441-004-0938-y
  31. Olsson M, Nikkhah G, Bentlage C and Bjorklund A (1995) Forelimb akinesia in the rat Parkinson model: differential effects of dopamine agonists and nigral transplants as assessed by a new stepping test. J Neurosci 15, 3863-3875  https://doi.org/10.1523/JNEUROSCI.15-05-03863.1995
  32. Miyanishi K, Choudhury ME, Watanabe M et al (2019) Behavioral tests predicting striatal dopamine level in a rat hemi-Parkinson's disease model. Neurochem Int 122, 38-46  https://doi.org/10.1016/j.neuint.2018.11.005
  33. Mahmoudi M, Akhavan O, Ghavami M, Rezaee F and Ghiasi SM (2012) Graphene oxide strongly inhibits amyloid beta fibrillation. Nanoscale 4, 7322-7325  https://doi.org/10.1039/c2nr31657a
  34. Ghaeidamini M, Bernson D, Sasanian N, Kumar R and Esbjorner EK (2020) Graphene oxide sheets and quantum dots inhibit alpha-synuclein amyloid formation by different mechanisms. Nanoscale 12, 19450-19460  https://doi.org/10.1039/d0nr05003b
  35. Alimohammadi E, Khedri M, Miri Jahromi A, Maleki R and Rezaian M (2020) Graphene-based nanoparticles as potential treatment options for Parkinson's disease: a molecular dynamics study. Int J Nanomedicine 15, 6887-6903  https://doi.org/10.2147/IJN.S265140
  36. Park WK, Kim H, Kim T et al (2015) Facile synthesis of graphene oxide in a Couette-Taylor flow reactor. Carbon 83, 217-223 https://doi.org/10.1016/j.carbon.2014.11.024