Yıl: 2023 Cilt: 47 Sayı: 1 Sayfa Aralığı: 171 - 184 Metin Dili: İngilizce DOI: 10.55730/1300-0527.3527 İndeks Tarihi: 13-03-2023

Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies

Öz:
In this study, some novel mono- and di-O-β-D-glycopyranosyl chalcone analogs were designed, synthesized, and characterized. The chalcone derivatives were synthesized with good yields by base-catalyzed Claisen-Schmidt condensation in EtOH solution. Then these chalcones were reacted with TAGBr (2,3,4,6-tetra-O-acetyl-α-D-glucopyranosylbromide) in dry acetone under the anhydrous condition at 0–5 °C. Deacylated was carried out by the Zemplen’s method with $NaOCH_3$ in dry methanol results in substituted chalcone- O-glycosides (mono- and di-O-β-D-glycopyranosyl chalcone analogs). The chemical structures of all synthesized compounds were elucidated based on IR, NMR spectral data, and mass spectrometry. Further, the compounds (7a-c, 8a-c, 12a-c, 16a-c, and 17a-c) were tested for their enzyme inhibition activity against α-glycosidase, tyrosinase, and AChE with in vitro and in silico analysis. Amongst them, compounds 12a-c, 16a-c, and 17a-c displayed moderate or less enzyme inhibition activity against α-glycosidase while other compounds 7a-c and 8a-c) were not active. Remarkably interesting enzyme inhibition effects, with $IC_{50}$ values below 30.59 ± 0.30 μM were recorded with 7c $(IC_{50}=11.07 ± 0.55 μM)$ against tyrosinase.
Anahtar Kelime: Chalcone enzyme inhibition glycoside glycosylation molecular docking

Belge Türü: Makale Makale Türü: Araştırma Makalesi Erişim Türü: Erişime Açık
  • 1. Nguyen HT, Thi TAD, Thi PH, Le-Nhat-Thuy G, Thi QGN et al. A new approach for the synthesis of novel naphthoquinone chalcone hybrid compounds. Tetrahedron Letters 2021; 81: 183337.
  • 2. Asiri AM, Khan SA. Synthesis, characterization, and in vitro antibacterial activities of macromolecules derived from bis chalcone. Journal of Heterocyclic Chemistry 2012; 49: 1434.
  • 3. Hu Y, Hu C, Pan G, Yu C, Ansari MF et al. Novel chalcone-conjugated, multi-flexible end-group coumarin thiazole hybrids as potential antibacterial repressors against methicillin-resistant Staphylococcus aureus. European Journal of Medicinal Chemistry 2021; 222: 113628.
  • 4. Huo K, Chen Y, Sui L, Wang Y, Fu Y et al. Transient expression and enzymatic assay identified uridine-diphosphate glucosyltransferases related to flavonoid glycosylation in Vernonia amygdalina leaves. Industrial Crops and Products 2021; 172: 114005.
  • 5. Fei Y, Shao Y, Wang W, Cheng Y, Yu B et al. Biosynthesis of Three Chalcone β-D-glucosides by Glycosyltransferase from Bacillus subtilis ATCC 6633. Microbiology and Biotechnology Letters 2021; 49 (2): 174–180.
  • 6. Kitamura K, Ando Y, Matsumoto T, Suzuki K. Total Synthesis of Aryl C-Glycoside Natural Products: Strategies and Tactics. Chemical Reviews 2018; 118 (4): 1495–1598.
  • 7. Zhao ZX, Jin J, Lin CZ, Zhu CC, Liu YM et al. Two new chalcone glycosides from the stems of Entada phaseoloides. Fitoterapia 2011; 82 (7): 1102-1105.
  • 8. Ingle VN, Kharche ST, Upadhyay UG. Glucosylation of 4’-hydroxychalcones using glucosyl donor. Indian Journal of Chemistry 2005; 44: 801-805.
  • 9. Ninomiya M, Efdi M, Inuzuka T, Koketsu M. Chalcone glycosides from aerial parts of Brassica rapa L. ‘hidabeni’, turnip. Phytochemistry Letters 2010; 3: 96-99.
  • 10. Xue Y, Liu Y, Xie Y, Cong C, Wang G et al. Antioxidant activity and mechanism of dihydrochalcone C-glycosides: Effects of C-glycosylation and hydroxyl groups. Phytochemistry 2020; 179: 112393.
  • 11. Hara H, Nakamura Y, Ninomiya M, Mochizuki R, Kamiya T et al. Inhibitory effects of chalcone glycosides isolated from Brassica rapa L. ‘hidabeni’ and their synthetic derivatives on LPS-induced NO production in microglia. Bioorganic & Medicinal Chemistry 2011; 19: 5559- 5568.
  • 12. Çelik G. New chalcone-3-O-glycoside derivatives: Synthesis and characterization. Journal of Chemical Research 2020; 44: 598-601.
  • 13. Funke I, Melzig MF. Traditionally used plants in diabetes therapy: phytotherapeutics as inhibitors of alpha-amylase activity. Revista Brasileira Farmacognosia 2006; 16 (1): 1–5.
  • 14. Telagari M, Hullatti K. In-vitro α-amylase and α-glucosidase inhibitory activity of Adiantum caudatum Linn. and Celosia argentea Linn. extracts and fractions. Indian Journal of Pharmacology 2015; 47 (4): 425–429.
  • 15. Baltaş N. α-Glucosidase and α-amylase inhibition of some ethanolic propolis samples. Uludag Bee Journal 2021; 21 (1): 1-7.
  • 16. Kumar Y, Goyal RK, Kumar Thakur A. Pharmacotherapeutics of miglitol: an α-glucosidase inhibitor. Journal of Analytical & Pharmaceutical Research 2018; 7 (6): 617-619.
  • 17. Değirmencioğlu İ, Öz Tuncay F, Cakmak U, Kolcuoğlu Y. The synthesis of novel piperazine-benzodioxole substituted phthalocyanines and investigation of their α-amylase and tyrosinase inhibition properties. Journal of Organometallic Chemistry 2021; 951: 122012.
  • 18. Guo C, Shan Y, Yang Z, Zhang L, Ling W et al. Chemical composition, antioxidant, antibacterial, and tyrosinase inhibition activity of extracts from Newhall navel orange (Citrus sinensis Osbeck cv. Newhall) peel. Journal of the Science Food and Agriculture 2020; 100: 2664–2674.
  • 19. Haldys K, Goldeman W, Jewgínski M, Wolínska E, Anger N et al. Inhibitory properties of aromatic thiosemicarbazones on mushroom tyrosinase: Synthesis, kinetic studies, molecular docking and effectiveness in melanogenesis inhibition. Bioorganic Chemistry 2018; 81: 577-586.
  • 20. Chen J, Ye Y, Ran M, Li Q, Ruan Z et al. Inhibition of Tyrosinase by Mercury Chloride: Spectroscopic and Docking Studies. Frontiers in Pharmacology 2020; 11: 81.
  • 21. Raza H, Abbasi MA, Rehman A, Siddiqui SZ, Hassan M et al. Design, synthesis and computational studies of N-(substituted-phenyl)-4- (4-phenyl-1-piperazinyl)butanamides as potent anti-melanogenic and tyrosinase inhibitors. Journal of Molecular Structure 2020; 1210: 127969.
  • 22. Shi F, Xie L, Lin Q, Tong C, Fu Q et al. Profiling of tyrosinase inhibitors in mango leaves for a sustainable agro-industry. Food Chemistry 2020; 312: 126042.
  • 23. Bari A, Ghani U, Syed SA, Riazullah. Thiosemicarbazide binds with the dicopper center in the competitive inhibition of mushroom tyrosinase enzyme: Synthesis and molecular modeling of theophylline analogues. Bioorganic & Medicinal Chemistry Letters 2021; 36: 127826.
  • 24. Yu Q, Fan L. Understanding the combined effect and inhibition mechanism of 4-hydroxycinnamic acid and ferulic acid as tyrosinase inhibitors. Food Chemistry 2021; 352: 129369.
  • 25. Akhtar MN, Sakeh NM, Zareen S, Gul S, Lo KM et al. Design and synthesis of chalcone derivatives as potent inhibitors and their structural activity relationship. Journal of Molecular Structure 2015; 1085: 97-103.
  • 26. Kim BH, Park KC, Park JH, Lee CG, Ye SK et al. Inhibition of tyrosinase activity and melanin production by the chalcone derivative 1-(2-cyclohexylmethoxy-6-hydroxy-phenyl)-3-(4-hydroxymethyl-phenyl)-propenone. Biochemical and Biophysical Research Communications 2016; 480: 648-654.
  • 27. Jamir K, Ganguly R, Seshagirirao K. ZCPG, a cysteine protease from Zingiber montanum rhizome exhibits enhanced anti-inflammatory and acetylcholinesterase inhibition potential. International Journal of Biological Macromolecules 2020; 163: 2429-2438.
  • 28. Alout H, Djogbénou L, Berticat C, Chandre F, Weill M. Comparison of Anopheles gambiae and Culex pipiens acetycholinesterase 1 biochemical properties. Comparative Biochemistry and Physiology, Part B 2008; 150: 271-277.
  • 29. Grutzendler J, Morris JC. Cholinesterase inhibitors for Alzheimer’s disease. Drugs 2001; 61: 41-52.
  • 30. Schneider LS. New therapeutic approaches to Alzheimer’s disease. The journal of Clinical Psychiatry 1996; 57 (14): 30-36.
  • 31. Kızıltas H, Bingöl Z, Gören AC, Polat Kose L, Durmaz L et al. LC-HRMS Profiling and Antidiabetic, Anticholinergic, and Antioxidant Activities of Aerial Parts of Kınkor (Ferulago stellata). Molecules 2021; 26: 2469-2486.
  • 32. Liu HR, Liu XJ, Fan HQ, Tang JJ, Gao XH et al. Design, synthesis and pharmacological evaluation of chalcone derivatives as acetylcholinesterase inhibitors. Bioorganic & Medicinal Chemistry 2014; 22: 6124-6133.
  • 33. Zhang X, Rakesh KP, Bukhari SNA, Balakrishna M, Manukumar HM et al. Multi-targetable chalcone analogs to treat deadly Alzheimer’s disease: Current view and upcoming advice. Bioorganic Chemistry 2018; 80: 86-93.
  • 34. Sapavat M, Reddymasu S, Mohammad YA, Mohamed JA, Rudraraju RR. Synthesis, Biological Evaluation and Molecular Docking Studies of Pyridine Incorporated Chalcone Derivatives as Anticancer Agents. Lettres in Organic Chemistry 2016; 13: 682-692.
  • 35. Antus S, Farkas L, Gott segen A, Nogradi M, Strelisky J et al. The Synthesis of Some Dihydrochalcones. Acta Chimica Academiae Scientiarum Hungaricae 1978; 98: 231-240.
  • 36. Zhao ZX, Jin J, Lin CZ, Zhu CC, Liu Y et al. Two new chalcone glycosides from the stems of Entada phaseoloides. Fitoterapia 2011; 82: 1102-1105.
  • 37. Baiming AT, Ren FX, Yang Y, Zhang Y, Qu AT et al. Chemical constituents of Bauhinia glauca (Wall. ex Benth.) Benth. subsp. hupehana (Craib) T. Chen Journal of Pharmaceutical Sciences 2012; 47: 1796-1798.
  • 38. Shai LJ, Magano SR, Lebelo SL, Mogale AM. Inhibitory effects of five medicinal plants on rat alpha-glucosidase: Comparison with their effects on yeast alpha-glucosidase. Journal of Medicinal Plant Research 2011; 5: 2863–2867.
  • 39. Şöhretoğlu D, Sari S, Barut B, Özel A. Tyrosinase inhibition by a rare neolignan: Inhibition kinetics and mechanistic insights through in vitro and in silico studies. Computional Biology and Chemistry 2018; 76: 61-66.
  • 40. Barut B, Sari S, Sabuncuoğlu S, Özel A. Azole antifungal compounds could have dual cholinesterase inhibitory potential according to virtual screening, enzyme kinetics, and toxicity studies of an inhouse library. Journal of Molecular Structure 2021;1235: 130268.
  • 41. Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK et al. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry 2009; 30: 2785-2791.
  • 42. Jurrus E, Engel D, Star K, Monson K, Brandi J et al. Improvements to the APBS biomolecular solvation software suite, Protein Science 2018; 27; 112-128.
  • 43. Ravindranath PA, Forli S, Goodsell DS, Olson AJ, Sanner MF. AutoDockFR: Advances in protein-ligand docking with explicitly specified binding site flexibility, PLOS Computational Biology 2015; 11: e1004586.
  • 44. Osman MS, Awad TA, Shantier SW, Garelnabi EA, Osman W et al. Identification of some chalcone analogues as potential antileishmanial agents: An integrated in vitro and in silico evaluation. Arabian Journal of Chemistry 2022; 15: 103717.
  • 45. Ingle VN, Kharche ST, Upadhyay UG. Glucosylation of 4′-hydroxychalcones using glucosyl donor. Indian Journal of Chemistry Section B 2005; 44: 801.
  • 46. Itoh T, Ninomiya M, Nozawa Y, Koketsu M. Chalcone glycosides isolated from aerial parts of Brassica rapa L. ‘hidabeni’ suppress antigen- stimulated degranulation in rat basophilic leukemia RBL-2H3 cells. Bioorganic & Medicinal Chemistry 2010; 18: 7052-7057.
  • 47. Antus S, Farkas L, Gottsegen A, Nogradi M, Strelisky J et al. Dihydrochalcone-type sweetening agents. II.The synthesis of some dihydrochalcones. Acta Chimica Academiae Scientiarum Hungaricae 1978; 98 (2): 231-40.
  • 48. Ammaji S, Masthanamma S, Bhandare RR, Annadurai S, Shaik AB. Antitubercular and antioxidant activities of hydroxy and chloro substituted chalcone analogues: Synthesis, biological and computational studies. Arabian Journal of Chemistry 2022; 15: 103581.
  • 49. Ngameni B, Cedric K, Mbaveng AT, Erdoğan M, Simo I et al. Design, synthesis, characterization, and anticancer activity of a novel series of O-substituted chalcone derivatives. Bioorganic & Medicinal Chemistry Letters 2021; 35: 127827.
  • 50. Yaylı N, Kılıç G, Kahriman N, Kanbolat Ş, Bozdeveci A et al. Synthesis, biological evaluation (antioxidant, antimicrobial, enzyme inhibition, and cytotoxic) and molecular docking study of hydroxy methoxy benzoin/benzil analogous. Bioorganic Chemistry 2021; 115: 105183.
  • 51. Nishina A, Kimura H, Tsukagoshi H, Kozawa K, Koketsu M et al. Neurite Outgrowth of PC12 Cells by 4′-O-β-D-Glucopyranosyl-3′,4- Dimethoxychalcone from Brassica rapa L. ‘hidabeni’ was Enhanced by Pretreatment with p38MAPK Inhibitor. Neorochemical Research 2013; 38: 2397-2407.
  • 52. Zhang Z, Yu B, Schmidt RR. Synthesis of Mono and Di O -β- d -glucopyranoside Conjugates of ( E )Resveratrol. Synthesis 2006; 8: 1301- 1306.
  • 53. Evranos Aksöz B, Ertan R. Chemical and Structural Properties of Chalcones I. FABAD Journal of Pharmaceutical Science 2011; 36: 223- 242.
  • 54. Fandaklı S, Doğan İS, Sellitepe HE, Yaşar A, Yaylı N. Synthesis, theoretical calculation and α-glucosidase inhibition of new chalcone oximes. Organic Communications 2018; 11(1): 23-34.
  • 55. Mukhtar A, Shah S, Kanwal, Khan KM, Khan SU et al. Synthesis of Chalcones as Potential α-Glucosidase Inhibitors, In-Vitro and In-Silico Studies. Chemistry Select 2021; 6 (37): 9933-9940.
  • 56. Barak D, Kronmang C, Ordentlichg A, Ariel N, Bromberg A et al. Acetylcholinesterase peripheral anionic site degeneracy conferred by amino acid arrays sharinga common core. Journal of Biological Chemistry 1994; 269: 6296-6305.
  • 57. Quinn DM. Acetylcholinesterase: enzyme structure, reaction dynamics, and virtual transition states. Chemical Reviews 1987; 87: 955-979.
  • 58. Zou C, Huang W, Zhao G. Determination of the bridging ligand in the active site of tyrosinase. Molecule 2017; 22: 1836.
APA ÇELİK G, Tatar Yılmaz G, Sahin H, Barut B, YAYLI N (2023). Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. , 171 - 184. 10.55730/1300-0527.3527
Chicago ÇELİK Gonca,Tatar Yılmaz Gizem,Sahin Huseyin,Barut Burak,YAYLI Nurettin Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. (2023): 171 - 184. 10.55730/1300-0527.3527
MLA ÇELİK Gonca,Tatar Yılmaz Gizem,Sahin Huseyin,Barut Burak,YAYLI Nurettin Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. , 2023, ss.171 - 184. 10.55730/1300-0527.3527
AMA ÇELİK G,Tatar Yılmaz G,Sahin H,Barut B,YAYLI N Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. . 2023; 171 - 184. 10.55730/1300-0527.3527
Vancouver ÇELİK G,Tatar Yılmaz G,Sahin H,Barut B,YAYLI N Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. . 2023; 171 - 184. 10.55730/1300-0527.3527
IEEE ÇELİK G,Tatar Yılmaz G,Sahin H,Barut B,YAYLI N "Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies." , ss.171 - 184, 2023. 10.55730/1300-0527.3527
ISNAD ÇELİK, Gonca vd. "Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies". (2023), 171-184. https://doi.org/10.55730/1300-0527.3527
APA ÇELİK G, Tatar Yılmaz G, Sahin H, Barut B, YAYLI N (2023). Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. Turkish Journal of Chemistry, 47(1), 171 - 184. 10.55730/1300-0527.3527
Chicago ÇELİK Gonca,Tatar Yılmaz Gizem,Sahin Huseyin,Barut Burak,YAYLI Nurettin Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. Turkish Journal of Chemistry 47, no.1 (2023): 171 - 184. 10.55730/1300-0527.3527
MLA ÇELİK Gonca,Tatar Yılmaz Gizem,Sahin Huseyin,Barut Burak,YAYLI Nurettin Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. Turkish Journal of Chemistry, vol.47, no.1, 2023, ss.171 - 184. 10.55730/1300-0527.3527
AMA ÇELİK G,Tatar Yılmaz G,Sahin H,Barut B,YAYLI N Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. Turkish Journal of Chemistry. 2023; 47(1): 171 - 184. 10.55730/1300-0527.3527
Vancouver ÇELİK G,Tatar Yılmaz G,Sahin H,Barut B,YAYLI N Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies. Turkish Journal of Chemistry. 2023; 47(1): 171 - 184. 10.55730/1300-0527.3527
IEEE ÇELİK G,Tatar Yılmaz G,Sahin H,Barut B,YAYLI N "Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies." Turkish Journal of Chemistry, 47, ss.171 - 184, 2023. 10.55730/1300-0527.3527
ISNAD ÇELİK, Gonca vd. "Design, synthesis, and enzyme inhibition evaluation of some novel Mono- and Di-O-β-D-Glycopyranosyl Chalcone analogues with molecular docking studies". Turkish Journal of Chemistry 47/1 (2023), 171-184. https://doi.org/10.55730/1300-0527.3527