Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The marine-derived compound TAG alleviates Parkinson’s disease by restoring RUBCN-mediated lipid metabolism homeostasis

Abstract

Parkinson’s disease (PD) is the second most common neurodegenerative disease, and its prevalence is increasing. Currently, no effective therapies for PD exist. Marine-derived natural compounds are considered important resources for the discovery of new drugs due to their distinctive structures and diverse activities. In this study, tetrahydroauroglaucin (TAG), a polyketide isolated from a marine sponge, was found to have notable neuroprotective effects on MPTP/MPP+-induced neurotoxicity. RNA sequencing analysis and metabolomics revealed that TAG significantly improved lipid metabolism disorder in PD models. Further investigation indicated that TAG markedly decreased the accumulation of lipid droplets (LDs), downregulated the expression of RUBCN, and promoted autophagic flux. Moreover, conditional knockdown of Rubcn notably attenuated PD-like symptoms and the accumulation of LDs, accompanied by blockade of the neuroprotective effect of TAG. Collectively, our results first indicated that TAG, a promising PD therapeutic candidate, could suppress the accumulation of LDs through the RUBCN-autophagy pathway, which highlighted a novel and effective strategy for PD treatment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: TAG improved mitochondrial dysfunction and reduced apoptosis in MPP+-treated SH-SY5Y cells.
Fig. 2: Neuroprotective effects of TAG on MPP+-induced primary neurons.
Fig. 3: TAG protected against MPTP-induced pathology in vivo.
Fig. 4: TAG ameliorated MPP+/MPTP-induced lipid metabolic disorder in vivo and in vitro.
Fig. 5: TAG downregulated the expression of RUBCN.
Fig. 6: Knockdown of Rubcn abolished the protective effect of TAG against MPP+-induced toxicity.
Fig. 7: The effects of TAG on MPTP-induced PD model mice were abrogated after sh-Rubcn transfection.
Fig. 8: The inhibitory effect of TAG on the accumulation of LDs was mediated by the Rubcn-autophagy axis.

Similar content being viewed by others

References

  1. Samii A, Nutt JG, Ransom BR. Parkinson’s disease. Lancet. 2004;363:1783–93.

    Article  CAS  PubMed  Google Scholar 

  2. Balestrino R, Schapira AHV. Parkinson disease. Eur J Neurol. 2020;27:27–42.

    Article  CAS  PubMed  Google Scholar 

  3. Shulman JM, De Jager PL, Feany MB. Parkinson’s disease: genetics and pathogenesis. Annu Rev Pathol. 2011;6:193–222.

    Article  CAS  PubMed  Google Scholar 

  4. Moustafa AA, Chakravarthy S, Phillips JR, Gupta A, Keri S, Polner B, et al. Motor symptoms in Parkinson’s disease: a unified framework. Neurosci Biobehav Rev. 2016;68:727–40.

    Article  PubMed  Google Scholar 

  5. Ko YF, Kuo PH, Wang CF, Chen YJ, Chuang PC, Li SZ, et al. Quantification analysis of sleep based on smartwatch sensors for Parkinson’s disease. Biosensors. 2022;12:74.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Schrag A, Horsfall L, Walters K, Noyce A, Petersen I. Prediagnostic presentations of Parkinson’s disease in primary care: a case-control study. Lancet Neurol. 2015;14:57–64.

    Article  PubMed  Google Scholar 

  7. Day JO, Mullin S. The genetics of Parkinson’s disease and implications for clinical practice. Genes. 2021;12:1006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Barreto GE, Iarkov A, Moran VE. Beneficial effects of nicotine, cotinine and its metabolites as potential agents for Parkinson’s disease. Front Aging Neurosci. 2014;6:340.

    PubMed  Google Scholar 

  9. Aryal B, Lee Y. Disease model organism for Parkinson disease: Drosophila melanogaster. BMB Rep. 2019;52:250–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cacabelos R. Parkinson’s disease: from pathogenesis to pharmacogenomics. Int J Mol Sci. 2017;18:551.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs. 2010;8:2619–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang B, Zhang T, Xu J, Lu J, Qiu P, Wang T, et al. Marine sponge-associated Fungi as potential novel bioactive natural product sources for drug discovery: a review. Mini Rev Med Chem. 2020;20:1966–2010.

    Article  CAS  PubMed  Google Scholar 

  13. Lin Z, Phadke S, Lu Z, Beyhan S, Abdel Aziz MH, Reilly C, et al. Onydecalins, fungal polyketides with anti- Histoplasma and anti-TRP activity. J Nat Prod. 2018;81:2605–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shah M, Sun C, Sun Z, Zhang G, Che Q, Gu Q, et al. Antibacterial polyketides from Antarctica sponge-derived fungus Penicillium sp. HDN151272. Mar Drugs. 2020;18:71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liu Z, Qiu P, Liu H, Li J, Shao C, Yan T, et al. Identification of anti-inflammatory polyketides from the coral-derived fungus Penicillium sclerotiorin: In vitro approaches and molecular-modeling. Bioorg Chem. 2019;88:102973.

    Article  CAS  PubMed  Google Scholar 

  16. Zhang Q, Su G, Yang Q, Wei Z, Wang J, Zheng L, et al. Round scad-derived octapeptide WCPFSRSF confers neuroprotection by regulating Akt/Nrf2/NFκB signaling. J Agric Food Chem. 2021;69:10606–16.

    Article  CAS  PubMed  Google Scholar 

  17. Catanesi M, Caioni G, Castelli V, Benedetti E, d’Angelo M, Cimini A. Benefits under the sea: the role of marine compounds in neurodegenerative disorders. Mar Drugs. 2021;19:24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Silva J, Alves C, Pinteus S, Susano P, Simões M, Guedes M, et al. Disclosing the potential of eleganolone for Parkinson’s disease therapeutics: Neuroprotective and anti-inflammatory activities. Pharmacol Res. 2021;168:105589.

    Article  CAS  PubMed  Google Scholar 

  19. Kolesnikova SA, Lyakhova EG, Kalinovsky AI, Popov RS, Yurchenko EA, Stonik VA. Oxysterols from a marine sponge inflatella sp. and their action in 6-hydroxydopamine-induced cell model of Parkinson’s disease. Mar Drugs. 2018;16:458.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Letsiou S, Bakea A, Goff GL, Lopes P, Gardikis K, Weis M, et al. Marine fungus Aspergillus chevalieri TM2-S6 extract protects skin fibroblasts from oxidative stress. Mar Drugs. 2020;18:460.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Stern JH, Rutkowski JM, Scherer PE. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell Metab. 2016;23:770–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Xu J, Huang X. Lipid metabolism at membrane contacts: dynamics and functions beyond lipid homeostasis. Front Cell Dev Biol. 2020;8:615856.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Tong Y, Sun Y, Tian X, Zhou T, Wang H, Zhang T, et al. Phospholipid transfer protein (PLTP) deficiency accelerates memory dysfunction through altering amyloid precursor protein (APP) processing in a mouse model of Alzheimer’s disease. Hum Mol Genet. 2015;24:5388–403.

    Article  CAS  PubMed  Google Scholar 

  24. Galper J, Kim WS, Dzamko N. LRRK2 and lipid pathways: implications for Parkinson’s disease. Biomolecules. 2022;12:1597.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Phillips GR, Hancock SE, Brown SHJ, Jenner AM, Kreilaus F, Newell KA, et al. Cholesteryl ester levels are elevated in the caudate and putamen of Huntington’s disease patients. Sci Rep. 2020;10:20314.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xie J, Duan L, Qian X, Huang X, Ding J, Hu G. K(ATP) channel openers protect mesencephalic neurons against MPP+-induced cytotoxicity via inhibition of ROS production. J Neurosci Res. 2010;88:428–37.

    Article  CAS  PubMed  Google Scholar 

  27. Wei Y, Lu M, Mei M, Wang H, Han Z, Chen M, et al. Pyridoxine induces glutathione synthesis via PKM2-mediated Nrf2 transactivation and confers neuroprotection. Nat Commun. 2020;11:941.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ye H, Robak LA, Yu M, Cykowski M, Shulman JM. Genetics and pathogenesis of Parkinson’s syndrome. Annu Rev Pathol. 2023;18:95–121.

    Article  CAS  PubMed  Google Scholar 

  29. Nicholatos JW, Francisco AB, Bender CA, Yeh T, Lugay FJ, Salazar JE, et al. Nicotine promotes neuron survival and partially protects from Parkinson’s disease by suppressing SIRT6. Acta Neuropathol Commun. 2018;6:120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Davis S, Meltzer PS. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics. 2007;23:1846–7.

    Article  PubMed  Google Scholar 

  31. Lei Y, Xu X, Liu H, Chen L, Zhou H, Jiang J, et al. HBx induces hepatocellular carcinogenesis through ARRB1-mediated autophagy to drive the G1/S cycle. Autophagy. 2021;17:4423–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ma X, McKeen T, Zhang J, Ding WX. Role and mechanisms of mitophagy in liver diseases. Cells. 2020;9:837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Feng Y, Ning X, Wang J, Wen Z, Cao F, You Q, et al. Mace-like plasmonic Au-Pd heterostructures boost near-infrared photoimmunotherapy. Adv Sci. 2023;10:e2204842.

    Article  Google Scholar 

  34. Guo JD, Zhao X, Li Y, Li GR, Liu XL. Damage to dopaminergic neurons by oxidative stress in Parkinson’s disease. Int J Mol Med. 2018;41:1817–25.

    CAS  PubMed  Google Scholar 

  35. Sewastianik T, Straubhaar JR, Zhao JJ, Samur MK, Adler K, Tanton HE, et al. miR-15a/16-1 deletion in activated B cells promotes plasma cell and mature B-cell neoplasms. Blood. 2021;137:1905–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Castillo-Quan JI, Steinbaugh MJ, Fernández-Cárdenas LP, Pohl NK, Wu Z, Zhu F, et al. An antisteatosis response regulated by oleic acid through lipid droplet-mediated ERAD enhancement. Sci Adv. 2023;9:eadc8917.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bezawork-Geleta A, Dimou J, Watt MJ. Lipid droplets and ferroptosis as new players in brain cancer glioblastoma progression and therapeutic resistance. Front Oncol. 2022;12:1085034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Consortium TM. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature. 2018;562:367–72.

    Article  Google Scholar 

  39. Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases. Nat Neurosci. 2010;13:805–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Raudenska M, Balvan J, Masarik M. Crosstalk between autophagy inhibitors and endosome-related secretory pathways: a challenge for autophagy-based treatment of solid cancers. Mol Cancer. 2021;20:140.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Nakamura S, Yoshimori T. New insights into autophagosome-lysosome fusion. J Cell Sci. 2017;130:1209–16.

    CAS  PubMed  Google Scholar 

  42. Wang X, Zhang X, Chu ESH, Chen X, Kang W, Wu F, et al. Defective lysosomal clearance of autophagosomes and its clinical implications in nonalcoholic steatohepatitis. FASEB J. 2018;32:37–51.

    Article  CAS  PubMed  Google Scholar 

  43. Tian X, Zhang G, Shao Y, Yang Z. Towards enhanced metabolomic data analysis of mass spectrometry image: multivariate curve resolution and machine learning. Anal Chim Acta. 2018;1037:211–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Castellanos DB, Martín-Jiménez CA, Rojas-Rodríguez F, Barreto GE, González J. Brain lipidomics as a rising field in neurodegenerative contexts: perspectives with machine learning approaches. Front Neuroendocrinol. 2021;61:100899.

    Article  CAS  PubMed  Google Scholar 

  45. Fahy E, Subramaniam S, Brown HA, Glass CK, Merrill AH, Murphy RC, et al. A comprehensive classification system for lipids. J Lipid Res. 2005;46:839–61.

    Article  CAS  PubMed  Google Scholar 

  46. Farooqui AA. Lipid mediators in the neural cell nucleus: their metabolism, signaling, and association with neurological disorders. Neuroscientist. 2009;15:392–407.

    Article  CAS  PubMed  Google Scholar 

  47. Shamim A, Mahmood T, Ahsan F, Kumar A, Bagga P. Lipids: an insight into the neurodegenerative disorders. Clin Nutr Exp. 2018;20:1–19.

    Article  Google Scholar 

  48. Farese RV, Walther TC. Lipid droplets finally get a little R-E-S-P-E-C-T. Cell. 2009;139:855–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Liu L, Zhang K, Sandoval H, Yamamoto S, Jaiswal M, Sanz E, et al. Glial lipid droplets and ROS induced by mitochondrial defects promote neurodegeneration. Cell. 2015;160:177–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Marschallinger J, Iram T, Zardeneta M, Lee SE, Lehallier B, Haney MS, et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat Neurosci. 2020;23:194–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Guo Y, Walther TC, Rao M, Stuurman N, Goshima G, Terayama K, et al. Functional genomic screen reveals genes involved in lipid-droplet formation and utilization. Nature. 2008;453:657–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Tanaka S, Hikita H, Tatsumi T, Sakamori R, Nozaki Y, Sakane S, et al. Rubicon inhibits autophagy and accelerates hepatocyte apoptosis and lipid accumulation in nonalcoholic fatty liver disease in mice. Hepatology. 2016;64:1994–2014.

    Article  CAS  PubMed  Google Scholar 

  53. Nakamura S, Oba M, Suzuki M, Takahashi A, Yamamuro T, Fujiwara M, et al. Suppression of autophagic activity by Rubicon is a signature of aging. Nat Commun. 2019;10:847.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Wong SW, Sil P, Martinez J. Rubicon: LC3-associated phagocytosis and beyond. FEBS J. 2018;285:1379–88.

    Article  CAS  PubMed  Google Scholar 

  55. Parzych KR, Klionsky DJ. An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal. 2014;20:460–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, et al. Autophagy regulates lipid metabolism. Nature. 2009;458:1131–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bae EJ, Lee HJ, Jang YH, Michael S, Masliah E, Min DS, et al. Phospholipase D1 regulates autophagic flux and clearance of α-synuclein aggregates. Cell Death Differ. 2014;21:1132–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (No. 81991523, 82003725) and the National Key R&D Programme of China (No. 2021ZD0202901). We thank Prof. RXT from Nanjing University for supplying the marine-derived natural compounds.

Author information

Authors and Affiliations

Authors

Contributions

GH and RXT conceived and designed the study. The natural compounds were provided by ZWT and RXT. PY, YL, QHH, XHX and SYM performed the experiments and analysed the data. PY and YL wrote the manuscript. JHD provided technical support. GH and ML revised the manuscript. All the authors have read and approved the final version of the manuscript.

Corresponding authors

Correspondence to Ming Lu, Ren-xiang Tan or Gang Hu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, P., Liu, Y., Tong, Zw. et al. The marine-derived compound TAG alleviates Parkinson’s disease by restoring RUBCN-mediated lipid metabolism homeostasis. Acta Pharmacol Sin (2024). https://doi.org/10.1038/s41401-024-01259-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41401-024-01259-y

Keywords

Search

Quick links