124
Views
0
CrossRef citations to date
0
Altmetric
Review

Natural products as novel scaffolds for the design of glycogen synthase kinase 3β inhibitors

, , , , , , & show all
Pages 377-396 | Received 06 Oct 2021, Accepted 15 Feb 2022, Published online: 09 Mar 2022

References

  • Martinez A, Castro A, Dorronsoro I, et al. Glycogen synthase kinase 3 (GSK-3) inhibitors as new promising drugs for diabetes, neurodegeneration, cancer, and inflammation. Med Res Rev. 2002;22(4):373–384.
  • Martinez A, Castro A, and Medina M. Drug Discov. Dev. In: Wang, Binghe, editor. Glycogen Synthase Kinase 3 (GSK-3) and Its Inhibitors. Hoboken NJ USA: John Wiley & Sons, Inc.; 2006. 978-0-471-77001-5.
  • Woodgett JR. Molecular cloning and expression of glycogen synthase kinase-3/Factor A. EMBO J. 1990;9(8):2431–2438.
  • Ter Haar E, Coll JT, Austen DA, et al. Structure of GSK3β reveals a primed phosphorylation mechanism. Nat Struct Biol. 2001;8(7):593–596.
  • Dajani R, Fraser E, Roe SM, et al., Crystal structure of glycogen synthase kinase 3β: structural basis for phosphate-primed substrate specificity and autoinhibition. Cell. 2001;105(6): 721–732.
  • Johnson LN, Noble MEM, Owen DJ. Active and inactive protein kinases: structural basis for regulation. Cell. 1996;85(2):149–158.
  • Picton C, Aitken A, Bilham T, et al. Multisite phosphorylation of glycogen synthase from rabbit skeletal muscle. Organisation of the seven sites in the polypeptide chain. Eur J Biochem. 1982;124(1):37–45.
  • Hughes K, Nikolakaki E, Plyte SE, et al. Modulation of the glycogen synthase kinase-3 family by tyrosine phosphorylation. EMBO J. 1993;12(2):803–808.
  • Dajani R, Fraser E, Roe SM, et al. Structural basis for recruitment of glycogen synthase kinase 3β to the axin-APC scaffold complex. EMBO J. 2003;22(3):494–501.
  • Bax B, Carter PS, Lewis C, et al. The structure of phosphorylated GSK-3β complexed with a peptide, FRATtide, that inhibits β-catenin phosphorylation. Structure. 2001;9(12):1143–1152.
  • Cross DAE, Alessi DR, Cohen P, et al. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378(6559):785–789.
  • Cohen P. The croonian lecture 1998. Identification of a protein kinase cascade of major importance in insulin signal transduction. Philos Trans R Soc B Biol Sci. 1999;354(1382):485–495.
  • Jope RS, Johnson GVW. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004;29(2):95–102.
  • Fang X, Yu SX, Lu Y, et al. Phosphorylation and inactivation of glycogen synthase kinase 3 by protein kinase A. Proc Natl Acad Sci U S A. 2000;97(22):11960–11965.
  • Li M, Wang X, Meintzer MK, et al. Cyclic AMP promotes neuronal survival by phosphorylation of glycogen synthase kinase 3β. Mol Cell Biol. 2000;20(24):9356–9363.
  • Fang X, Yu S, Tanyi JL, et al. Convergence of multiple signaling cascades at glycogen synthase kinase 3: edg receptor-mediated phosphorylation and inactivation by lysophosphatidic acid through a protein kinase c-dependent intracellular pathway. Mol Cell Biol. 2002;22(7):2099–2110.
  • Sutherland C, Leighton IA, Cohen P. Inactivation of glycogen synthase kinase-3 β by phosphorylation: new kinase connections in insulin and growth-factor signalling. Biochem J. 1993;296(1):15–19.
  • V BR, Shanley J, Correll MP, et al. Regulation and localization of tyrosine216 phosphorylation of glycogen synthase kinase-3beta in cellular and animal models of neuronal degeneration. Proc Natl Acad Sci. 2000;97(20):11074–11079.
  • Hartigan JA, Xiong WC, Johnson GVW. Glycogen synthase kinase 3β is tyrosine phosphorylated by PYK2. Biochem Biophys Res Commun. 2001;284(2):485–489.
  • Ishiguro K, Shiratsuchi A, Sato S, et al. Glycogen synthase kinase 3β is identical to tau protein kinase I generating several epitopes of paired helical filaments. FEBS Lett. 1993;325(3):167–172.
  • Sengupta A, Wu Q, Grundke-Iqbal I, et al. Potentiation of GSK-3-catalyzed Alzheimer-like phosphorylation of human tau by cdk5. Mol Cell Biochem. 1997;167(1–2):99–105.
  • Ciaraldi TP, Oh DK, Christiansen L, et al. Tissue-specific expression and regulation of GSK-3 in human skeletal muscle and adipose tissue. Am J Physiol - Endocrinol Metab. 2006;291(5):E891–8.
  • Summers SA, Kao AW, Kohn AD, et al. The role of glycogen synthase kinase 3β in insulin-stimulated glucose metabolism. J Biol Chem. 1999;274(25):17934–17940.
  • Kaladchibachi SA, Doble B, Anthopoulos N, et al. Glycogen synthase kinase 3, circadian rhythms, and bipolar disorder: a molecular link in the therapeutic action of lithium. J Circadian Rhythms. 2007;5:3.
  • Kozlovsky N, Belmaker RH, Agam G. GSK-3 and the neurodevelopmental hypothesis of schizophrenia. Eur Neuropsychopharmacol. 2002;12(1):13–25.
  • Hooper C, Killick R, Lovestone S. The GSK3 hypothesis of Alzheimer’s disease. J Neurochem. 2008;104(6):1433–1439.
  • Morales-García JA, Susín C, Alonso-Gil S, et al. Glycogen synthase kinase-3 inhibitors as potent therapeutic agents for the treatment of Parkinson's disease. ACS Chem Neurosci. 2013;4(2):350–360.
  • Ougolkov AV, Billadeau DD. Targeting GSK-3: a promising approach for cancer therapy? Futur Oncol. 2006;2(1):91–100.
  • King MK, Pardo M, Cheng Y, et al. Glycogen synthase kinase-3 inhibitors: rescuers of cognitive impairments. Pharmacol Ther. 2014;141(1):1–12.
  • Kramer T, Schmidt B, Lo Monte F. Small-molecule inhibitors of GSK-3: structural insights and their application to Alzheimer’s disease models. Int. J. Alzheimers. Dis. 2012;2012:381029.
  • Xu M, Wang SL, Zhu L, et al. Structure-activity relationship (SAR) studies of synthetic glycogen synthase kinase-3β inhibitors: a critical review. Eur J Med Chem. 2019;164:448–470.
  • Khan I, Tantray MA, Alam MS, et al. Natural and synthetic bioactive inhibitors of glycogen synthase kinase. Eur J Med Chem. 2017;125:464–477.
  • Yu JY, Taylor J, DeRuiter SL, et al. Simultaneous inhibition of GSK3α and GSK3β using hairpin siRNA expression vectors. Mol Ther. 2003;7(2):228–236.
  • Eldar-Finkelman H, Martinez A. GSK-3 inhibitors: preclinical and clinical focus on CNS. Front Mol Neurosci. 2011;4:32.
  • Bertrand JA, Thieffine S, Vulpetti A, et al. Structural characterization of the GSK-3β active site using selective and non-selective ATP-mimetic inhibitors. J Mol Biol. 2003;333(2):393–407.
  • Leost M, Schultz C, Link A, et al. Paullones are potent inhibitors of glycogen synthase kinase-3β and cyclin-dependent kinase 5/p25. Eur J Biochem. 2000;267(19):5983–5994.
  • Martinez A, Alonso M, Castro A, et al. First Non-ATP competitive glycogen synthase kinase 3 β (gsk-3β) inhibitors: thiadiazolidinones (tdzd) as potential drugs for the treatment of Alzheimer’s disease. J Med Chem. 2002;45(6):1292–1299.
  • Bidon-Chanal A, Fuertes A, Alonso D, et al. Evidence for a new binding mode to GSK-3: allosteric regulation by the marine compound palinurin. Eur J Med Chem. 2013;60:479–489.
  • Palomo V, Soteras I, Perez DI, et al. Exploring the binding sites of glycogen synthase kinase 3. identification and characterization of allosteric modulation cavities. J Med Chem. 2011;54(24):8461–8470.
  • Duda P, Akula SM, Abrams SL, et al. Targeting GSK3 and associated signaling pathways involved in cancer. Cells. 2020;9(5):1110.
  • Li, Wang, Zhang, et al., A systematic review of recently reported marine derived natural product kinase inhibitors. Mar Drugs. 2019;17(9): 493.
  • Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J Nat Prod. 2020;83(3):770–803.
  • de Pedro N, Cantizani J, Ortiz-López FJ, et al. Protective effects of isolecanoric acid on neurodegenerative in vitro models. Neuropharmacology. 2016;101:538–548.
  • Leclerc S, Garnier M, Hoessel R, et al. Indirubins inhibit glycogen synthase kinase-3β and CDK5/P25, two protein kinases involved in abnormal tau phosphorylation in Alzheimer’s disease. A property common to most cyclin-dependent kinase inhibitors? J Biol Chem. 2001;276(1):251–260.
  • Meijer L, Skaltsounis AL, Magiatis P, et al. GSK-3-selective inhibitors derived from tyrian purple indirubins. Chem Biol. 2003;10(12):1255–1266.
  • Polychronopoulos P, Magiatis P, Skaltsounis AL, et al. Structural basis for the synthesis of indirubins as potent and selective inhibitors of glycogen synthase kinase-3 and cyclin-dependent kinases. J Med Chem. 2004;47(4):935–946.
  • Vougogiannopoulou K, Ferandin Y, Bettayeb K, et al. Soluble 3′,6-substituted indirubins with enhanced selectivity toward glycogen synthase kinase −3 alter circadian period. J Med Chem. 2008;51(20):6421–6431.
  • Libnow S, Methling K, Hein M, et al. Synthesis of indirubin-N′-glycosides and their anti-proliferative activity against human cancer cell lines. Bioorg Med Chem. 2008;16(10):5570–5583.
  • Sakai R, Higa T, Jefford CW, et al. Manzamine A, a novel antitumor alkaloid from a sponge. J Am Chem Soc. 1986;108(20):6404–6405.
  • V RK, Donia MS, Peng J, et al. Manzamine B and E and Ircinal A related alkaloids from an Indonesian acanthostrongylophora sponge and their activity against infectious, tropical parasitic, and Alzheimer’s diseases. J Nat Prod. 2006;69(7):1034–1040.
  • Hamann M, Alonso D, Martín-Aparicio E, et al. Glycogen synthase kinase-3 (gsk-3) inhibitory activity and structure–activity relationship (sar) studies of the manzamine alkaloids. Potential for Alzheimer’s Disease. J Nat Prod. 2007;70:1397–1405. Internet.
  • Peng J, Kudrimoti S, Prasanna S, et al. Structure - activity relationship and mechanism of action studies of manzamine analogues for the control of neuroinflammation and cerebral infections. J Med Chem. 2010;53(1):61–76.
  • Skropeta D, Pastro N, Zivanovic A. Kinase inhibitors from marine sponges. Mar Drugs. 2011;9(10):2131–2154.
  • Van Epps S, Fiamengo B, Edmunds J, et al. Design and synthesis of tricyclic cores for kinase inhibition. Bioorganic Med Chem Lett. 2013;23(3):693–698.
  • Wan Y, Hur W, Cho CY, et al. Synthesis and target identification of hymenialdisine analogs. Chem Biol. 2004;11(2):247–259.
  • Meijer L, Thunnissen AMWH, White AW, et al. Inhibition of cyclin-dependent kinases, GSK-3β and CK1 by hymenialdisine, a marine sponge constituent. Chem Biol. 2000;7(1):51–63.
  • Masi M, Di Lecce R, Cimmino A, et al. Advances in the chemical and biological characterization of amaryllidaceae alkaloids and natural analogues isolated in the last decade. Molecules. 2020;25(23):5621.
  • Hulcová D, Maříková J, Korábečný J, et al. Amaryllidaceae alkaloids from narcissus pseudonarcissus l. cv. Dutch master as potential drugs in treatment of Alzheimer’s disease. Phytochemistry. 2019;162:165.
  • Kohelová E, Peřinová R, Maafi N, et al. Derivatives of the β-crinane amaryllidaceae alkaloid haemanthamine as multi-target directed ligands for Alzheimer’s disease. Molecules. 2019;24(7):1307.
  • Omura S, Iwai Y, Hirano A, et al. A new alkaloid am-2282 of streptomyces origin taxonomy, fermentation, isolation and preliminary characterization. J Antibiot (Tokyo). 1977;30(4):275–282.
  • Bharate SB, Sawant SD, Singh PP, et al. Kinase inhibitors of marine origin. Chem Rev. 2013;113(8):6761–6815.
  • Ōmura S, Asami Y, Crump A. Staurosporine: new lease of life for parent compound of today’s novel and highly successful anti-cancer drugs. J. Antibiot. (Tokyo). Nature Publishing Group. 2018;71(8):688–701.
  • Kozikowski AP, Gaisina IN, Petukhov PA, et al. Highly potent and specific GSK-3β inhibitors that block tau phosphorylation and decrease α-synuclein protein expression in a cellular model of Parkinson’s disease. ChemMedChem. 2006;1(2):256–266.
  • Liu J-G, Zhao D, Gong Q, et al. Development of bisindole-substituted aminopyrazoles as novel GSK-3β inhibitors with suppressive effects against microglial inflammation and oxidative neurotoxicity. ACS Chem Neurosci. 2020;11(20):3398–3408.
  • BH H. The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 2002;96(2–3):67–202.
  • Ravishankar D, Watson KA, Greco F, et al. Novel synthesised flavone derivatives provide significant insight into the structural features required for enhanced anti-proliferative activity. RSC Adv. 2016;6(69):64544–64556.
  • Hole KL, Williams RJ. Flavonoids as an Intervention for alzheimer’s disease: progress and hurdles towards defining a mechanism of action. Brain Plast. 2020;6:167–192.
  • Ragab FA, Yahya TAA, El-Naa MM, et al. Design, synthesis and structure–activity relationship of novel semi-synthetic flavonoids as antiproliferative agents. Eur J Med Chem. 2014;82:506–520.
  • Wang H-K. The therapeutic potential of flavonoids. Expert Opin Investig Drugs. 2000;9:2103–2119.
  • Kumara PM, Soujanya KN, Ravikanth G, et al. Rohitukine, a chromone alkaloid and a precursor of flavopiridol, is produced by endophytic fungi isolated from dysoxylum binectariferum Hook.f and amoora rohituka (Roxb).Wight & Arn. Phytomedicine. 2014;21(4):541–546.
  • Ibrahim N, Bonnet P, Brion J-D, et al. Identification of a new series of flavopiridol-like structures as kinase inhibitors with high cytotoxic potency. Eur J Med Chem. 2020;199:112355.
  • Liang Z, Zhang B, Su WW, et al. C Glycosylflavones alleviate tau phosphorylation and amyloid neurotoxicity through GSK3β inhibition. ACS Chem Neurosci. 2016;7(7):912–923.
  • Tan X, Liang Z, Li Y, et al. Isoorientin, a GSK-3β inhibitor, rescues synaptic dysfunction, spatial memory deficits and attenuates pathological progression in APP/PS1 model mice. Behav Brain Res. 2021;398:112968.
  • L Z, L QX. Discovery of selective, substrate-competitive, and passive membrane permeable glycogen synthase kinase-3β Inhibitors: synthesis, biological evaluation, and molecular modeling of new c glycosylflavones. ACS Chem Neurosci. 2018;9(5):1166–1183.
  • Sufi SA, Adigopula LN, Syed SB, et al. In-silico and in-vitro anti-cancer potential of a curcumin analogue (1E, 6E)-1,7-di (1H-indol-3-yl) hepta-1, 6-diene-3, 5-dione. Biomed Pharmacother. 2017;85:389–398.
  • Duda P, Wiśniewski J, Wójtowicz T, et al. Targeting GSK3 signaling as a potential therapy of neurodegenerative diseases and aging. Expert Opin Ther Targets. 2018;22(10):833–848.
  • Di MRMC, Pruccoli L, Bisi A, et al. Novel curcumin-diethyl fumarate hybrid as a dualistic gsk-3β inhibitor/Nrf2 Inducer for the treatment of parkinson’s disease. ACS Chem Neurosci. 2020;11(17):2728–2740.
  • Di Martino RMC, De Simone A, Andrisano V, et al. Versatility of the curcumin scaffold: discovery of potent and balanced dual bace-1 and gsk-3β inhibitors. J Med Chem. 2016;59(2):531–544.
  • Leri M, Scuto M, Ontario ML, et al. Healthy effects of plant polyphenols: molecular mechanisms. Int J Mol Sci. 2020;21(4):1250.
  • Karim N, Khan H, Khan I, et al. An increasing role of polyphenols as novel therapeutics for Alzheimer’s: a review. Med Chem (Los Angeles). 2019;16:1007–1021.
  • Jurcau A. The role of natural antioxidants in the prevention of dementia—where do we stand and future perspectives. Nutrients. 2021;13(2):282.
  • Singh YP, Rai H, Singh G, et al. A review on ferulic acid and analogs based scaffolds for the management of Alzheimer’s disease. Eur J Med Chem. 2021;215:113278.
  • Paudel P, Seong SH, Zhou Y, et al. Rosmarinic acid derivatives’ inhibition of glycogen synthase kinase-3β is the pharmacological basis of kangen-karyu in Alzheimer’s disease. Molecules. 2018;23(11):2919.
  • De Simone A, Bartolini M, Baschieri A, et al. Hydroxy-substituted trans-cinnamoyl derivatives as multifunctional tools in the context of Alzheimer’s disease. Eur J Med Chem. 2017;139:378–389.
  • Yao H, Liu J, Xu S, et al. The structural modification of natural products for novel drug discovery. Expert Opin Drug Discov. 2017;12:121–140. **This review highlights the importance of structural modification of NPs in drug development.
  • De Simone A, Tumiatti V, Andrisano V, et al. Glycogen synthase kinase 3β: a new gold rush in Anti-Alzheimer’s disease multitarget drug discovery? J Med Chem. 2021;64(1):26–41.
  • AG A, W B, P-w EM, et al. Discovery and resupply of pharmacologically active plant-derived natural products: a review. Biotechnol Adv. 2015;33(8):1582–1614.
  • Harvey AL, Edrada-Ebel R, Quinn RJ. The re-emergence of natural products for drug discovery in the genomics era. Nat Rev Drug Discov. 2015;14(2):111–129.
  • Wilkinson B, Micklefield J. Mining and engineering natural-product biosynthetic pathways. Nat Chem Biol. 2007;3(7):379–386.
  • Wilkinson B, Moss SJ. Biosynthetic engineering of natural products for lead optimization and development. Curr Opin Drug Discov Dev. 2005;8:748–756.
  • Atanasov AG, Zotchev SB, Dirsch VM, et al., Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discov. 2021;20(3): 200–216.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.