DOI QR코드

DOI QR Code

Discovery and validation of PURA as a transcription target of 20(S)-protopanaxadiol: Implications for the treatment of cognitive dysfunction

  • Feiyan Chen (Research and Innovation Center, College of Traditional Chinese Medicine.Integrated Chinese and Western Medicine College, Nanjing University of Chinese Medicine) ;
  • Wenjing Zhang (Department of Physiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine) ;
  • Shuyi Xu (Department of Physiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine) ;
  • Hantao Zhang (Department of Physiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine) ;
  • Lin Chen (Department of Physiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine) ;
  • Cuihua Chen (Research and Innovation Center, College of Traditional Chinese Medicine.Integrated Chinese and Western Medicine College, Nanjing University of Chinese Medicine) ;
  • Zhu Zhu (Department of Pathology and Pathophysiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine) ;
  • Yunan Zhao (Department of Pathology and Pathophysiology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine)
  • Received : 2023.01.09
  • Accepted : 2023.04.24
  • Published : 2023.09.01

Abstract

Background: 20(S)-protopanaxadiol (PPD), a ginsenoside metabolite, has prominent benefits for the central nervous system, especially in improving learning and memory. However, its transcriptional targets in brain tissue remain unknown. Methods: In this study, we first used mass spectrometry-based drug affinity responsive target stability (DARTS) to identify the potential proteins of ginsenosides and intersected them with the transcription factor library. Second, the transcription factor PURA was confirmed as a target of PPD by biolayer interferometry (BLI) and molecular docking. Next, the effect of PPD on the transcriptional levels of target genes of PURA in brain tissues was determined by qRT-PCR. Finally, bioinformatics analysis was used to analyze the potential biological features of these target proteins. Results: The results showed three overlapping transcription factors between the proteomics of DARTS and transcription factor library. BLI analysis further showed that PPD had a higher direct interaction with PURA than parent ginsenosides. Subsequently, BLI kinetic analysis, molecular docking, and mutations in key amino acids of PURA indicated that PPD specifically bound to PURA. The results of qRT-PCR showed that PPD could increase the transcription levels of PURA target genes in brain. Finally, bioinformatics analysis showed that these target proteins were involved in learning and memory function. Conclusion: The above-mentioned findings indicate that PURA is a transcription target of PPD in brain, and PPD upregulate the transcription levels of target genes related to cognitive dysfunction by binding PURA, which could provide a chemical and biological basis for the study of treating cognitive impairment by targeting PURA.

Keywords

Acknowledgement

The study was financially supported by National Natural Science Foundation of China (No.82204647, 82003970), Supporting project of National Natural Youth Foundation of Nanjing University of Chinese Medicine (XPT82204647).

References

  1. Lambert SA, Jolma A, Campitelli LF, Das PK, Yin Y, Albu M, et al. The human transcription factors. Cell 2018;172:650-65. https://doi.org/10.1016/j.cell.2018.01.029
  2. Goswami S, Kareem O, Goyal RK, Mumtaz SM, Tonk RK, Gupta R, et al. Role of forkhead transcription factors of the O class (FoxO) in development and progression of Alzheimer's disease. CNS & Neurological Disorders Drug Targets 2020;19:709-21. https://doi.org/10.2174/1871527319666201001105553
  3. Vargas DM, De Bastiani MA, Zimmer ER, Klamt F. Alzheimer's disease master regulators analysis: search for potential molecular targets and drug repositioning candidates. Alzheimer's Research & Therapy 2018;10:59.
  4. Dard RF, Dahan L, Rampon C. Targeting hippocampal adult neurogenesis using transcription factors to reduce Alzheimer's disease-associated memory impairments. Hippocampus 2019;29:579-86. https://doi.org/10.1002/hipo.23052
  5. Catala-Solsona J, Minano-Molina AJ, Rodriguez-Alvarez J. Nr4a2 transcription factor in hippocampal synaptic plasticity, memory and cognitive dysfunction: a perspective review. Frontiers in Molecular Neuroscience 2021;14:786226.
  6. Chen F, Li C, Cao H, Zhang H, Lu C, Li R, et al. Identification of adenylate kinase 5 as a protein target of ginsenosides in brain tissues using mass spectrometry-based drug affinity responsive target stability (DARTS) and cellular thermal shift assay (CETSA) techniques. Journal of Agricultural and Food Chemistry 2022;70:2741-51. https://doi.org/10.1021/acs.jafc.1c07819
  7. Pan W, Xue B, Yang C, Miao L, Zhou L, Chen Q, et al. Biopharmaceutical characters and bioavailability improving strategies of ginsenosides. Fitoterapia 2018;129:272-82. https://doi.org/10.1016/j.fitote.2018.06.001
  8. Xu ZL, Chen G, Liu X, Xie D, Zhang J, Ying Y. Effects of ginsenosides on memory impairment in propofol-anesthetized rats. Bioengineered 2022;13:617-23. https://doi.org/10.1080/21655979.2021.2012407
  9. Wang Q, Sun LH, Jia W, Liu XM, Dang HX, Mai WL, et al. Comparison of ginsenosides Rg1 and Rb1 for their effects on improving scopolamine-induced learning and memory impairment in mice. Phytotherapy Research : PTR 2010;24:1748-54. https://doi.org/10.1002/ptr.3130
  10. Yang R, Jiang X, He X, Liang D, Sun S, Zhou G. Ginsenoside Rb1 improves cognitive impairment induced by insulin resistance through cdk5/p35-NMDAR-IDE pathway. BioMed Research International 2020;2020:3905719.
  11. Cui L, Wu SQ, Zhao CA, Yin CR. Microbial conversion of major ginsenosides in ginseng total saponins by Platycodon grandiflorum endophytes. Journal of Ginseng Research 2016;40:366-74. https://doi.org/10.1016/j.jgr.2015.11.004
  12. Chen F, Chen L, Liang W, Zhang Z, Li J, Zheng W, et al. Identification and confirmation of 14-3-3 z as a novel target of ginsenosides in brain tissues. Journal of Ginseng Research 2021;45:465-72. https://doi.org/10.1016/j.jgr.2020.12.007
  13. Musende AG, Eberding A, Wood CA, Adomat H, Fazli L, Hurtado-Coll A, et al. A novel oral dosage formulation of the ginsenoside aglycone protopanaxadiol exhibits therapeutic activity against a hormone-insensitive model of prostate cancer. Anti-cancer Drugs 2012;23:543-52. https://doi.org/10.1097/CAD.0b013e32835006f5
  14. Chen C, Wang L, Cao F, Miao X, Chen T, Chang Q, et al. Formulation of 20(S)-protopanaxadiol nanocrystals to improve oral bioavailability and brain delivery. International Journal of Pharmaceutics 2016;497:239-47. https://doi.org/10.1016/j.ijpharm.2015.12.014
  15. Lu C, Dong L, Lv J, Wang Y, Fan B, Wang F, et al. 20(S)-protopanaxadiol (PPD) alleviates scopolamine-induced memory impairment via regulation of cholinergic and antioxidant systems, and expression of Egr-1, c-Fos and c-Jun in mice. Chemico-biological Interactions 2018;279:64-72. https://doi.org/10.1016/j.cbi.2017.11.008
  16. Lu C, Lv J, Dong L, Jiang N, Wang Y, Fan B, et al. The protective effect of 20(S)-protopanaxadiol (PPD) against chronic sleep deprivation (CSD)-induced memory impairments in mice. Brain Research Bulletin 2018;137:249-56. https://doi.org/10.1016/j.brainresbull.2017.12.012
  17. Ren YS, Li HL, Piao XH, Yang ZY, Wang SM, Ge YW. Drug affinity responsive target stability (DARTS) accelerated small molecules target discovery: principles and application. Biochemical Pharmacology 2021;194:114798.
  18. Lomenick B, Hao R, Jonai N, Chin RM, Aghajan M, Warburton S, et al. Target identification using drug affinity responsive target stability (DARTS). Proceedings of the National Academy of Sciences of the United States of America 2009;106:21984-9. https://doi.org/10.1073/pnas.0910040106
  19. Friman T. Mass spectrometry-based Cellular Thermal Shift Assay (CETSA®) for target deconvolution in phenotypic drug discovery. Bioorganic & Medicinal Chemistry 2020;28:115174.
  20. Kanai Y, Dohmae N, Hirokawa N. Kinesin transports RNA: isolation and characterization of an RNA-transporting granule. Neuron 2004;43:513-25. https://doi.org/10.1016/j.neuron.2004.07.022
  21. Chen F, Zhu K, Chen L, Ouyang L, Chen C, Gu L, et al. Protein target identification of ginsenosides in skeletal muscle tissues: discovery of natural small-molecule activators of muscle-type creatine kinase. Journal of Ginseng Research 2020;44:461-74. https://doi.org/10.1016/j.jgr.2019.02.005
  22. Pai MY, Lomenick B, Hwang H, Schiestl R, McBride W, Loo JA, et al. Drug affinity responsive target stability (DARTS) for small-molecule target identification. Methods in Molecular Biology (Clifton, NJ) 2015;1263:287-98. https://doi.org/10.1007/978-1-4939-2269-7_22
  23. Lu C, Fan L, Zhang PF, Tao WW, Yang CB, Shang EX, et al. A novel P38a MAPK activator Bruceine A exhibits potent anti-pancreatic cancer activity. Computational and Structural Biotechnology Journal 2021;19:3437-50. https://doi.org/10.1016/j.csbj.2021.06.011
  24. Molitor L, Bacher S, Burczyk S, Niessing D. The molecular function of PURA and its implications in neurological diseases. Frontiers in Genetics 2021;12:638217.
  25. Lomenick B, Jung G, Wohlschlegel JA, Huang J. Target identification using drug affinity responsive target stability (DARTS). Current Protocols in Chemical Biology 2011;3:163-80. https://doi.org/10.1002/9780470559277.ch110180
  26. Yang Z, Kuboyama T, Tohda C. A systematic strategy for discovering a therapeutic drug for Alzheimer's disease and its target molecule. Frontiers in Pharmacology 2017;8:340.
  27. Sun J, Prabhu N, Tang J, Yang F, Jia L, Guo J, et al. Recent advances in proteome-wide label-free target deconvolution for bioactive small molecules. Medicinal Research Reviews 2021;41:2893-926. https://doi.org/10.1002/med.21788
  28. Johnson EM, Daniel DC, Gordon J. The pur protein family: genetic and structural features in development and disease. Journal of Cellular Physiology 2013;228:930-7. https://doi.org/10.1002/jcp.24237
  29. Daniel DC, Johnson EM. PURA, the gene encoding Pur-alpha, member of an ancient nucleic acid-binding protein family with mammalian neurological functions. Gene 2018;643:133-43. https://doi.org/10.1016/j.gene.2017.12.004
  30. Barbe MF, Krueger JJ, Loomis R, Otte J, Gordon J. Memory deficits, gait ataxia and neuronal loss in the hippocampus and cerebellum in mice that are heterozygous for Pur-alpha. Neuroscience 2016;337:177-90. https://doi.org/10.1016/j.neuroscience.2016.09.018
  31. Khalili K, Del Valle L, Muralidharan V, Gault WJ, Darbinian N, Otte J, et al. Puralpha is essential for postnatal brain development and developmentally coupled cellular proliferation as revealed by genetic inactivation in the mouse. Molecular and Cellular Biology 2003;23:6857-75. https://doi.org/10.1128/MCB.23.19.6857-6875.2003
  32. Tanaka AJ, Bai R, Cho MT, Anyane-Yeboa K, Ahimaz P, Wilson AL, et al. De novo mutations in PURA are associated with hypotonia and developmental delay. Cold Spring Harbor Molecular Case Studies 2015;1:a000356.
  33. Xu Z, Poidevin M, Li X, Li Y, Shu L, Nelson DL, et al. Expanded GGGGCC repeat RNA associated with amyotrophic lateral sclerosis and frontotemporal dementia causes neurodegeneration. Proceedings of the National Academy of Sciences of the United States of America 2013;110:7778-83. https://doi.org/10.1073/pnas.1219643110
  34. Shi X, Ren S, Zhang B, Guo S, He W, Yuan C, et al. Analysis of the role of Pura in the pathogenesis of Alzheimer's disease based on RNA-seq and ChIP-seq. Scientific Reports 2021;11:12178.
  35. Cheung HN, Dunbar C, Morotz GM, Cheng WH, Chan HY, Miller CC, et al. FE65 interacts with ADP-ribosylation factor 6 to promote neurite outgrowth. official publication of the Federation of American Societies for Experimental Biology FASEB Journal 2014;28:337-49. https://doi.org/10.1096/fj.13-232694
  36. Borroni V, Barrantes FJ. Homomeric and heteromeric α7 nicotinic acetylcholine receptors in health and some central nervous system diseases. Membranes 2021:11.
  37. Elitt MS, Barbar L, Shick HE, Powers BE, Maeno-Hikichi Y, Madhavan M, et al. Suppression of proteolipid protein rescues Pelizaeus-Merzbacher disease. Nature 2020;585:397-403. https://doi.org/10.1038/s41586-020-2494-3
  38. Wang Y, Zhang M, Moon C, Hu Q, Wang B, Martin G, et al. The APP-interacting protein FE65 is required for hippocampus-dependent learning and long-term potentiation. Learning & Memory (Cold Spring Harbor, NY) 2009;16:537-44. https://doi.org/10.1101/lm.1499309