Skip to main content

Advertisement

Log in

Natural Xanthones from Garcinia mangostana with Multifunctional Activities for the Therapy of Alzheimer’s Disease

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Natural xanthones have diversity pharmacological activities. Here, a series of xanthones isolated from the pericarps of Garcinia mangostana Linn, named α-Mangostin, 8-Deoxygartanin, Gartanin, Garciniafuran, Garcinone C, Garcinone D, and γ-Mangostin were investigated. Biological screening performed in vitro and in Escherichia coli cells indicated that most of the xanthones exhibited significant inhibition of self-induced β-amyloid (Aβ) aggregation and also β-site amyloid precursor protein-cleaving enzyme 1, acted as potential antioxidants and biometal chelators. Among these compounds, α-Mangostin, Gartanin, Garcinone C and γ-Mangostin showed better antioxidant properties to scavenge Diphenyl-1-(2,4,6-trinitrophenyl) hydrazyl (DPPH) free radical than Trolox, and potent neuroprotective effects against glutamate-induced HT22 cell death partly by up-regulating HO-1 protein level and then scavenging reactive oxygen species. Moreover, Gartanin, Garcinone C and γ-Mangostin could be able to penetrate the blood–brain barrier (BBB) in vitro. These findings suggest that the natural xanthones have multifunctional activities against Alzheimer’s disease (AD) and could be promising compounds for the therapy of AD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Abbreviations

AD:

Alzheimer’s disease

MTDLs:

Multi-target-directed ligands

Aβ:

β-Amyloid

BACE1:

β-Site amyloid precursor protein-cleaving enzyme 1

DPPH:

Diphenyl-1-(2,4,6-trinitrophenyl) hydrazyl

BBB:

Blood–brain barrier

HO-1:

Heme oxygenase-1

References

  1. Rosini M, Simoni E, Minarini A, Melchiorre C (2014) Multi-target design strategies in the context of Alzheimer’s disease: acetylcholinesterase inhibition and NMDA receptor antagonism as the driving forces. Neurochem Res 39(10):1914–1923

    Article  CAS  PubMed  Google Scholar 

  2. Carreiras MC, Mendes E, Perry MJ, Francisco AP, Marco-Contelles J (2013) The multifactorial nature of Alzheimer’s disease for developing potential therapeutics. Curr Top Med Chem 13(15):1745–1770

    Article  CAS  PubMed  Google Scholar 

  3. Kim HG, Oh MS (2012) Herbal medicines for the prevention and treatment of Alzheimer’s disease. Curr Pharm Des 18(1):57–75

    Article  CAS  PubMed  Google Scholar 

  4. Wu TY, Chen CP, Jinn TR (2011) Traditional Chinese medicines and Alzheimer’s disease. Taiwan J Obst Gynecol 50(2):131–135

    Article  Google Scholar 

  5. Kim MH, Kim SH, Yang WM (2014) Mechanisms of action of phytochemicals from medicinal herbs in the treatment of Alzheimer’s disease. Planta Med 80(15):1249–1258

    Article  CAS  PubMed  Google Scholar 

  6. Bajda M, Guzior N, Ignasik M, Malawska B (2011) Multi-target-directed ligands in Alzheimer’s disease treatment. Curr Med Chem 18(32):4949–4975

    Article  CAS  PubMed  Google Scholar 

  7. Guzior N, Wieckowska A, Panek D, Malawska B (2015) Recent development of multifunctional agents as potential drug candidates for the treatment of Alzheimer’s disease. Curr Med Chem 22(3):373–404

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Suttirak W, Manurakchinakorn S (2014) In vitro antioxidant properties of mangosteen peel extract. J Food Sci Technol 51(12):3546–3558

    Article  CAS  PubMed  Google Scholar 

  9. Li G, Thomas S, Johnson JJ (2013) Polyphenols from the mangosteen (Garcinia mangostana) fruit for breast and prostate cancer. Front Pharmacol 4:80

    PubMed  PubMed Central  Google Scholar 

  10. Kosem N, Ichikawa K, Utsumi H, Moongkarndi P (2013) In vivo toxicity and antitumor activity of mangosteen extract. J Nat Med 67(2):255–263

    Article  CAS  PubMed  Google Scholar 

  11. Dharmaratne HR, Sakagami Y, Piyasena KG, Thevanesam V (2013) Antibacterial activity of xanthones from Garcinia mangostana (L.) and their structure-activity relationship studies. Nat Prod Res 27(10):938–941

    Article  CAS  PubMed  Google Scholar 

  12. Wang Y, Xia Z, Xu JR, Wang YX, Hou LN, Qiu Y, Chen HZ (2012) Alpha-mangostin, a polyphenolic xanthone derivative from mangosteen, attenuates beta-amyloid oligomers-induced neurotoxicity by inhibiting amyloid aggregation. Neuropharmacology 62(2):871–881

    Article  CAS  PubMed  Google Scholar 

  13. Bumrungpert A, Kalpravidh RW, Chuang CC, Overman A, Martinez K, Kennedy A, McIntosh M (2010) Xanthones from mangosteen inhibit inflammation in human macrophages and in human adipocytes exposed to macrophage-conditioned media. J Nutr 140(4):842–847

    Article  CAS  PubMed  Google Scholar 

  14. Huang HJ, Chen WL, Hsieh RH, Hsieh-Li HM (2014) Multifunctional effects of mangosteen pericarp on cognition in C57BL/6J and triple transgenic Alzheimer’s mice. Evid-Based Complement Altern Med eCAM 2014:813672

    Google Scholar 

  15. Xu Z, Huang L, Chen XH, Zhu XF, Qian XJ, Feng GK, Lan WJ, Li HJ (2014) Cytotoxic prenylated xanthones from the pericarps of Garcinia mangostana. Molecules 19(2):1820–1827 (Basel, Switzerland)

    Article  PubMed  Google Scholar 

  16. Di L, Kerns EH, Fan K, McConnell OJ, Carter GT (2003) High throughput artificial membrane permeability assay for blood-brain barrier. Eur J Med Chem 38(3):223–232

    Article  CAS  PubMed  Google Scholar 

  17. Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297(5580):353–356

    Article  CAS  PubMed  Google Scholar 

  18. Biancalana M, Makabe K, Koide A, Koide S (2009) Molecular mechanism of thioflavin-T binding to the surface of beta-rich peptide self-assemblies. J Mol Biol 385(4):1052–1063

    Article  CAS  PubMed  Google Scholar 

  19. Fodera V, Groenning M, Vetri V, Librizzi F, Spagnolo S, Cornett C, Olsen L, van de Weert M, Leone M (2008) Thioflavin T hydroxylation at basic pH and its effect on amyloid fibril detection. J Phys Chem B 112(47):15174–15181

    Article  CAS  PubMed  Google Scholar 

  20. Pouplana S, Espargaro A, Galdeano C, Viayna E, Sola I, Ventura S, Munoz-Torrero D, Sabate R (2014) Thioflavin-S staining of bacterial inclusion bodies for the fast, simple, and inexpensive screening of amyloid aggregation inhibitors. Curr Med Chem 21(9):1152–1159

    Article  CAS  PubMed  Google Scholar 

  21. Luo Y, Bolon B, Kahn S, Bennett BD, Babu-Khan S, Denis P, Fan W, Kha H, Zhang J, Gong Y, Martin L, Louis JC, Yan Q, Richards WG, Citron M, Vassar R (2001) Mice deficient in BACE1, the Alzheimer’s beta-secretase, have normal phenotype and abolished beta-amyloid generation. Nat Neurosci 4(3):231–232

    Article  CAS  PubMed  Google Scholar 

  22. Yan R, Vassar R (2014) Targeting the beta secretase BACE1 for Alzheimer’s disease therapy. Lancet Neurol 13(3):319–329

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Padurariu M, Ciobica A, Lefter R, Serban IL, Stefanescu C, Chirita R (2013) The oxidative stress hypothesis in Alzheimer’s disease. Psychiatr Danub 25(4):401–409

    CAS  PubMed  Google Scholar 

  24. Azizi G, Navabi SS, Al-Shukaili A, Seyedzadeh MH, Yazdani R, Mirshafiey A (2015) The Role of Inflammatory Mediators in the Pathogenesis of Alzheimer’s Disease. Sultan Qaboos Univ Med J 15(3):e305–e316

    Article  PubMed  PubMed Central  Google Scholar 

  25. Buendia I, Michalska P, Navarro E, Gameiro I, Egea J, Leon R (2016) Nrf2-ARE pathway: an emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases. Pharmacol Ther 157:84–104

    Article  CAS  PubMed  Google Scholar 

  26. Biswas C, Shah N, Muthu M, La P, Fernando AP, Sengupta S, Yang G, Dennery PA (2014) Nuclear heme oxygenase-1 (HO-1) modulates subcellular distribution and activation of Nrf2, impacting metabolic and anti-oxidant defenses. J Biol Chem 289(39):26882–26894

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lim JL, Wilhelmus MM, de Vries HE, Drukarch B, Hoozemans JJ, van Horssen J (2014) Antioxidative defense mechanisms controlled by Nrf2: state-of-the-art and clinical perspectives in neurodegenerative diseases. Arch Toxicol 88(10):1773–1786

    Article  CAS  PubMed  Google Scholar 

  28. Schipper HM (2007) Biomarker potential of heme oxygenase-1 in Alzheimer’s disease and mild cognitive impairment. Biomark Med 1(3):375–385

    Article  CAS  PubMed  Google Scholar 

  29. Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ, Mattson MP (2010) Cellular stress responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid Redox Signal 13(11):1763–1811

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Trovato Salinaro A, Cornelius C, Koverech G, Koverech A, Scuto M, Lodato F, Fronte V, Muccilli V, Reibaldi M, Longo A, Uva MG, Calabrese V (2014) Cellular stress response, redox status, and vitagenes in glaucoma: a systemic oxidant disorder linked to Alzheimer’s disease. Front Pharmacol 5:129

    Article  PubMed  PubMed Central  Google Scholar 

  31. Calabrese V, Cornelius C, Mancuso C, Pennisi G, Calafato S, Bellia F, Bates TE, Giuffrida Stella AM, Schapira T, Dinkova Kostova AT, Rizzarelli E (2008) Cellular stress response: a novel target for chemoprevention and nutritional neuroprotection in aging, neurodegenerative disorders and longevity. Neurochem Res 33(12):2444–2471

    Article  CAS  PubMed  Google Scholar 

  32. Calabrese V, Cornelius C, Dinkova-Kostova AT, Iavicoli I, Di Paola R, Koverech A, Cuzzocrea S, Rizzarelli E (1822) Calabrese EJ (2012) cellular stress responses, hormetic phytochemicals and vitagenes in aging and longevity. Biochim Biophys Acta 5:753–783

    Google Scholar 

  33. Viayna E, Sola I, Bartolini M, De Simone A, Tapia-Rojas C, Serrano FG, Sabate R, Juarez-Jimenez J, Perez B, Luque FJ, Andrisano V, Clos MV, Inestrosa NC, Munoz-Torrero D (2014) Synthesis and multitarget biological profiling of a novel family of rhein derivatives as disease-modifying anti-Alzheimer agents. J Med Chem 57(6):2549–2567

    Article  CAS  PubMed  Google Scholar 

  34. Khaw KY, Choi SB, Tan SC, Wahab HA, Chan KL, Murugaiyah V (2014) Prenylated xanthones from mangosteen as promising cholinesterase inhibitors and their molecular docking studies. Phytomed Int J Phytother Phytopharmacol 21(11):1303–1309

    Article  CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to thank Dr. Ling Huang for their technical expertise. This study was supported by Guangdong Provincial International Cooperation Project of Science & Technology (No. 2013B051000038), National Natural Science Foundation of China (No. 31371070) and the Fundamental Research Funds for the Central Universities (No. 15ykjc08b) to R. Pi.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Wen-jian Lan, Xiao-bo Yang or Jing-kao Chen.

Additional information

Sheng-nan Wang, Qian Li and Ming-hua Jing have equally contributed to this work.

Electronic supplementary material

Below is the link to the electronic supplementary material.

S1 Fig. 

1H NMR spectrum of α-Mangostin in CDCl3 (300 MHz) (TIFF 548 kb)

S2 Fig. 

13C NMR spectrum of α-Mangostin in CDCl3 (75 MHz) (TIFF 837 kb)

S3 Fig. 

1H NMR spectrum of 8-Deoxygartanin in DMSO-d 6 (300 MHz) (TIFF 697 kb)

S4 Fig. 

13C NMR spectrum of 8-Deoxygartanin in DMSO-d 6 (75 MHz) (TIFF 936 kb)

S5 Fig. 

1H NMR spectrum of Gartanin in DMSO-d 6 (300 MHz) (TIFF 612 kb)

S6 Fig. 

13C NMR spectrum of Gartanin in DMSO-d 6 (75 MHz) (TIFF 1051 kb)

S7 Fig. 

1H NMR spectrum of Garciniafuran in CDCl3 (300 MHz) (TIFF 551 kb)

S8 Fig. 

13C NMR spectrum of Garciniafuran in CDCl3 (75 MHz) (TIFF 1047 kb)

S9 Fig. 

1H NMR spectrum of Garcinone in DMSO-d 6 (300 MHz) (TIFF 616 kb)

S10 Fig. 

13C NMR spectrum of Garcinone in DMSO-d 6 (75 MHz) (TIFF 791 kb)

S11 Fig. 

1H NMR spectrum of Garcinone D in DMSO-d6 (300 MHz) (TIFF 637 kb)

S12 Fig. 

13C NMR spectrum of Garcinone D in DMSO-d6 (75 MHz) (TIFF 985 kb)

S13 Fig.

S13 Fig. 1H NMR spectrum of γ-Mangostin in DMSO-d6 (300 MHz) (TIFF 611 kb)

S14 Fig. 

13C NMR spectrum of γ-Mangostin in DMSO-d6 (75 MHz) (TIFF 906 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Sn., Li, Q., Jing, Mh. et al. Natural Xanthones from Garcinia mangostana with Multifunctional Activities for the Therapy of Alzheimer’s Disease. Neurochem Res 41, 1806–1817 (2016). https://doi.org/10.1007/s11064-016-1896-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-016-1896-y

Keywords

Navigation