Generic placeholder image

Current Traditional Medicine

Editor-in-Chief

ISSN (Print): 2215-0838
ISSN (Online): 2215-0846

Review Article

Naringenin Scaffold as a Template for Drug Designing

Author(s): Yogesh Murti*, Bhupesh Chander Semwal, Ahsas Goyal and Pradeep Mishra

Volume 7, Issue 1, 2021

Published on: 17 June, 2019

Page: [28 - 44] Pages: 17

DOI: 10.2174/2215083805666190617144652

Price: $65

Abstract

Natural products provide cornucopia of heterocyclic systems. The nucleus of 2- phenyl chromane is one of the important and well-known heterocycles found in the natural products. Naringenin, a plant-derived flavanone (2-phenyl chroman-4-one) belongs to the family of flavanoids. It possesses diverse biologic activities such as antidiabetic, antiatherogenic, antidepressant, antiandrogenic, antiestrogenic, immunomodulatory, antitumor, antimicrobial, anti-inflammatory, antiviral, hypolipidemic, antihypertensive, antioxidant, neuroprotective, anti-obesity, anti-Alzheimer, and memory enhancer activity. It has the potential to be used as an active pharmacophore. There have been reports of a number of molecular mechanisms underlying their beneficial activities. With emerging interest in traditional medicine and exploiting their potential based on a variety of health care systems, naringenin literature was thought to be explored. Further, this review aims to provide a new era of flavonoid-based therapeutic agents with new insights into naringenin and its derivatives as a lead compound in drug design.

Keywords: Naringenin, flavanone, pharmacophore, drug design, flavanoids, lead compound.

Graphical Abstract
[1]
Mbaveng AT, Zhao Q, Kuete V. Harmful and protective effects of phenolic compounds from African medicinal plants Toxicological survey of African medicinal plants. Elsevier 2014.
[http://dx.doi.org/10.1016/B978-0-12-800018-2.00020-0]
[2]
Kawaii S, Tomono Y, Katase E, Ogawa K, Yano M. Quantitation of flavonoid constituents in citrus fruits. J Agric Food Chem 1999; 47(9): 3565-71.
[http://dx.doi.org/10.1021/jf990153+] [PMID: 10552686]
[3]
Brodowska KM. Natural flavonoids: Classification, potential role, and application of flavonoid analogues. Eur J Biol Res 2017; 7(2): 108-23.
[4]
Madrigal-Santillán E, Madrigal-Bujaidar E, Álvarez-González I, et al. Review of natural products with hepatoprotective effects. World J Gastroenterol 2014; 20(40): 14787-804.
[http://dx.doi.org/10.3748/wjg.v20.i40.14787] [PMID: 25356040]
[5]
Rao PV, Kiran SDVS, Rohini P, et al. Flavonoid: A review on naringenin. J Pharmacogn Phytochem 2017; 6(5): 2778-83.
[6]
Mir IA, Tiku AB. Chemopreventive and therapeutic potential of “naringenin,” a flavanone present in citrus fruits. Nutr Cancer 2015; 67(1): 27-42.
[http://dx.doi.org/10.1080/01635581.2015.976320] [PMID: 25514618]
[7]
Mukherjee PK. Quality control of herbal drugs. Bus Horiz 2002; 46-65.
[8]
Sachdeva AK, Kuhad A, Chopra K. Naringin ameliorates memory deficits in experimental paradigm of Alzheimer’s disease by attenuating mitochondrial dysfunction. Pharmacol Biochem Behav 2014; 127: 101-10.
[http://dx.doi.org/10.1016/j.pbb.2014.11.002] [PMID: 25449356]
[9]
Lou H, Jing X, Wei X, Shi H, Ren D, Zhang X. Naringenin protects against 6-OHDA-induced neurotoxicity via activation of the Nrf2/ARE signaling pathway. Neuropharmacology 2014; 79: 380-8.
[http://dx.doi.org/10.1016/j.neuropharm.2013.11.026] [PMID: 24333330]
[10]
Zaki HF, Abd-El-Fattah MA, Attia AS. Naringenin protects against scopolamine-induced dementia in rats. Bull Fac Pharm Cairo Univ 2014; 52(1): 15-25.
[http://dx.doi.org/10.1016/j.bfopcu.2013.11.001]]
[11]
Muthaiah VP, Venkitasamy L, Michael FM, Chandrasekar K, Venkatachalam S. Neuroprotective role of naringenin on carbaryl induced neurotoxicity in mouse neuroblastoma cells. J Pharmacol Pharmacother 2013; 4(3): 192-7.
[http://dx.doi.org/10.4103/0976-500X.114599] [PMID: 23960424]
[12]
Raza SS, Khan MM, Ahmad A, et al. Neuroprotective effect of naringenin is mediated through suppression of NF-κB signaling pathway in experimental stroke. Neuroscience 2013; 230(29): 157-71.
[http://dx.doi.org/10.1016/j.neuroscience.2012.10.041] [PMID: 23103795]
[13]
Yang W, Ma J, Liu Z, Lu Y, Hu B, Yu H. Effect of naringenin on brain insulin signaling and cognitive functions in ICV-STZ induced dementia model of rats. Neurol Sci 2014; 35(5): 741-51.
[http://dx.doi.org/10.1007/s10072-013-1594-3] [PMID: 24337945]
[14]
Kandhare AD, Raygude KS, Ghosh P, Ghule AE, Bodhankar SL. Neuroprotective effect of naringin by modulation of endogenous biomarkers in streptozotocin induced painful diabetic neuropathy. Fitoterapia 2012; 83(4): 650-9.
[http://dx.doi.org/10.1016/j.fitote.2012.01.010] [PMID: 22343014]
[15]
Khan MB, Khan MM, Khan A, et al. Naringenin ameliorates Alzheimer’s disease (AD)-type neurodegeneration with cognitive impairment (AD-TNDCI) caused by the intracerebroventricular-streptozotocin in rat model. Neurochem Int 2012; 61(7): 1081-93.
[http://dx.doi.org/10.1016/j.neuint.2012.07.025] [PMID: 22898296]
[16]
Olsen HT, Stafford GI, van Staden J, Christensen SB, Jäger AK. Isolation of the MAO-inhibitor naringenin from Mentha aquatica L. J Ethnopharmacol 2008; 117(3): 500-2.
[http://dx.doi.org/10.1016/j.jep.2008.02.015] [PMID: 18372132]
[17]
Baluchnejadmojarad T, Roghani M. Effect of naringenin on intracerebroventricular streptozotocin-induced cognitive deficits in rat: A behavioral analysis. Pharmacology 2006; 78(4): 193-7.
[http://dx.doi.org/10.1159/000096585] [PMID: 17065836]
[18]
Zbarsky V, Datla KP, Parkar S, Rai DK, Aruoma OI, Dexter DT. Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkinson’s disease. Free Radic Res 2005; 39(10): 1119-25.
[http://dx.doi.org/10.1080/10715760500233113]] [PMID: 16298737]
[19]
Heo HJ, Kim MJ, Lee JM, et al. Naringenin from Citrus junos has an inhibitory effect on acetylcholinesterase and a mitigating effect on amnesia. Dement Geriatr Cogn Disord 2004; 17(3): 151-7.
[http://dx.doi.org/10.1159/000076349] [PMID: 14739537]
[20]
Cavia-Saiz M, Busto MD, Pilar-Izquierdo MC, Ortega N, Perez-Mateos M, Muñiz P. Antioxidant properties, radical scavenging activity and biomolecule protection capacity of flavonoid naringenin and its glycoside naringin: A comparative study. J Sci Food Agric 2010; 90(7): 1238-44.
[http://dx.doi.org/10.1002/jsfa.3959] [PMID: 20394007]
[21]
Abaza MSI, Orabi KY, Al-Quattan E, Al-Attiyah RJ. Growth inhibitory and chemo-sensitization effects of naringenin, a natural flavanone purified from Thymus vulgaris, on human breast and colorectal cancer. Cancer Cell Int 2015; 15(46): 46.
[http://dx.doi.org/10.1186/s12935-015-0194-0] [PMID: 26074733]
[22]
Kim JH, Lee JK. Naringenin enhances NK cell lysis activity by increasing the expression of NKG2D ligands on Burkitt’s lymphoma cells. Arch Pharm Res 2015; 38(11): 2042-8.
[http://dx.doi.org/10.1007/s12272-015-0624-5] [PMID: 26100136]
[23]
Kapoor R, Rizvi F, Kakkar P. Naringenin prevents high glucose-induced mitochondria-mediated apoptosis involving AIF, Endo-G and caspases. Apoptosis 2013; 18(1): 9-27.
[http://dx.doi.org/10.1007/s10495-012-0781-7] [PMID: 23192364]
[24]
Bulzomi P, Bolli A, Galluzzo P, Acconcia F, Ascenzi P, Marino M. The naringenin-induced proapoptotic effect in breast cancer cell lines holds out against a high bisphenol a background. IUBMB Life 2012; 64(8): 690-6.
[http://dx.doi.org/10.1002/iub.1049] [PMID: 22692793]
[25]
Lou C, Zhang F, Yang M, et al. Naringenin decreases invasiveness and metastasis by inhibiting TGF-β-induced epithelial to mesenchymal transition in pancreatic cancer cells. PLoS One 2012; 7(12)e50956
[http://dx.doi.org/10.1371/journal.pone.0050956] [PMID: 23300530]
[26]
Anand K, Sarkar A, Kumar A, Ambasta RK, Kumar P. Combinatorial antitumor effect of naringenin and curcumin elicit angioinhibitory activities in vivo. Nutr Cancer 2012; 64(5): 714-24.
[http://dx.doi.org/10.1080/01635581.2012.686648] [PMID: 22642894]
[27]
Subramanian P, Arul D. Attenuation of NDEA-induced hepatocarcinogenesis by naringenin in rats. Cell Biochem Funct 2013; 31(6): 511-7.
[http://dx.doi.org/10.1002/cbf.2929] [PMID: 23172681]
[28]
Leonardi T, Vanamala J, Taddeo SS, et al. Apigenin and naringenin suppress colon carcinogenesis through the aberrant crypt stage in azoxymethane-treated rats. Exp Biol Med (Maywood) 2010; 235(6): 710-7.
[http://dx.doi.org/10.1258/ebm.2010.009359] [PMID: 20511675]
[29]
Qin L, Jin L, Lu L, et al. Naringenin reduces lung metastasis in a breast cancer resection model. Protein Cell 2011; 2(6): 507-16.
[http://dx.doi.org/10.1007/s13238-011-1056-8] [PMID: 21748601]
[30]
Park JH, Jin CY, Lee BK, Kim GY, Choi YH, Jeong YK. Naringenin induces apoptosis through downregulation of Akt and caspase-3 activation in human leukemia THP-1 cells. Food Chem Toxicol 2008; 46(12): 3684-90.
[http://dx.doi.org/10.1016/j.fct.2008.09.056] [PMID: 18930780]
[31]
Ekambaram G, Rajendran P, Magesh V, Sakthisekaran D. Naringenin reduces tumor size and weight lost in N-methyl-N'-nitro-N-nitrosoguanidine-induced gastric carcinogenesis in rats. Nutr Res 2008; 28(2): 106-12.
[http://dx.doi.org/10.1016/j.nutres.2007.12.002] [PMID: 19083396]
[32]
Gao K, Henning SM, Niu Y, et al. The citrus flavonoid naringenin stimulates DNA repair in prostate cancer cells. J Nutr Biochem 2006; 17(2): 89-95.
[http://dx.doi.org/10.1016/j.jnutbio.2005.05.009] [PMID: 16111881]
[33]
Yilma AN, Singh SR, Morici L, Dennis VA. Flavonoid naringenin: A potential immunomodulator for Chlamydia trachomatis inflammation. Mediators Inflamm 2013.2013102457
[http://dx.doi.org/10.1155/2013/102457] [PMID: 23766556]
[34]
Du G, Jin L, Han X, Song Z, Zhang H, Liang W. Naringenin: a potential immunomodulator for inhibiting lung fibrosis and metastasis. Cancer Res 2009; 69(7): 3205-12.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-3393] [PMID: 19318568]
[35]
Annadurai T, Muralidharan AR, Joseph T, Hsu MJ, Thomas PA, Geraldine P. Antihyperglycemic and antioxidant effects of a flavanone, naringenin, in streptozotocin-nicotinamide-induced experimental diabetic rats. J Physiol Biochem 2012; 68(3): 307-18.
[http://dx.doi.org/10.1007/s13105-011-0142-y] [PMID: 22234849]
[36]
Kannappan S, Anuradha CV. Naringenin enhances insulin-stimulated tyrosine phosphorylation and improves the cellular actions of insulin in a dietary model of metabolic syndrome. Eur J Nutr 2010; 49(2): 101-9.
[http://dx.doi.org/10.1007/s00394-009-0054-6] [PMID: 19727895]
[37]
Mulvihill EE, Allister EM, Sutherland BG, et al. Naringenin prevents dyslipidemia, apolipoprotein B overproduction, and hyperinsulinemia in LDL receptor-null mice with diet-induced insulin resistance. Diabetes 2009; 58(10): 2198-210.
[http://dx.doi.org/10.2337/db09-0634] [PMID: 19592617]
[38]
Ortiz-Andrade RR, Sánchez-Salgado JC, Navarrete-Vázquez G, et al. Antidiabetic and toxicological evaluations of naringenin in normoglycaemic and NIDDM rat models and its implications on extra-pancreatic glucose regulation. Diabetes Obes Metab 2008; 10(11): 1097-104.
[http://dx.doi.org/10.1111/j.1463-1326.2008.00869.x] [PMID: 18355329]
[39]
Allister EM, Borradaile NM, Edwards JY, Huff MW. Inhibition of microsomal triglyceride transfer protein expression and apolipoprotein B100 secretion by the citrus flavonoid naringenin and by insulin involves activation of the mitogen-activated protein kinase pathway in hepatocytes. Diabetes 2005; 54(6): 1676-83.
[http://dx.doi.org/10.2337/diabetes.54.6.1676]] [PMID: 15919788]
[40]
Teng J, Li Y, Yu W, et al. Naringenin, a common flavanone, inhibits the formation of AGEs in bread and attenuates AGEs-induced oxidative stress and inflammation in RAW264.7 cells. Food Chem 2018; 269: 35-42.
[http://dx.doi.org/10.1016/j.foodchem.2018.06.126] [PMID: 30100446]
[41]
Annadurai T, Thomas PA, Geraldine P. Ameliorative effect of naringenin on hyperglycemia-mediated inflammation in hepatic and pancreatic tissues of Wistar rats with streptozotocin- nicotinamide-induced experimental diabetes mellitus. Free Radic Res 2013; 47(10): 793-803.
[http://dx.doi.org/10.3109/10715762.2013.823643] [PMID: 23841752]
[42]
Tsai SJ, Huang CS, Mong MC, Kam WY, Huang HY, Yin MC. Anti-inflammatory and antifibrotic effects of naringenin in diabetic mice. J Agric Food Chem 2012; 60(1): 514-21.
[http://dx.doi.org/10.1021/jf203259h] [PMID: 22117528]
[43]
Jayaraman J, Jesudoss VA, Menon VP, Namasivayam N. Anti-inflammatory role of naringenin in rats with ethanol induced liver injury. Toxicol Mech Methods 2012; 22(7): 568-76.
[http://dx.doi.org/10.3109/15376516.2012.707255] [PMID: 22900548]
[44]
Vafeiadou K, Vauzour D, Lee HY, Rodriguez-Mateos A, Williams RJ, Spencer JP. The citrus flavanone naringenin inhibits inflammatory signalling in glial cells and protects against neuroinflammatory injury. Arch Biochem Biophys 2009; 484(1): 100-9.
[http://dx.doi.org/10.1016/j.abb.2009.01.016] [PMID: 19467635]
[45]
Shi Y, Dai J, Liu H, et al. Naringenin inhibits allergen-induced airway inflammation and airway responsiveness and inhibits NF-kappaB activity in a murine model of asthma. Can J Physiol Pharmacol 2009; 87(9): 729-35.
[http://dx.doi.org/10.1139/Y09-065] [PMID: 19794524]
[46]
Amaro MI, Rocha J, Vila-Real H, et al. Anti-inflammatory activity of naringin and the biosynthesised naringenin by naringinase immobilized in microstructured materials in a model of DSS-induced colitis in mice. Food Res Int 2009; 42(8): 1010-7.
[http://dx.doi.org/10.1016/j.foodres.2009.04.016]]
[47]
Bodet C, La VD, Epifano F, Grenier D. Naringenin has anti-inflammatory properties in macrophage and ex vivo human whole-blood models. J Periodontal Res 2008; 43(4): 400-7.
[http://dx.doi.org/10.1111/j.1600-0765.2007.01055.x] [PMID: 18503517]
[48]
Yen FL, Wu TH, Lin LT, Cham TM, Lin CC. Naringenin-loaded nanoparticles improve the physicochemical properties and the hepatoprotective effects of naringenin in orally-administered rats with CCl(4)-induced acute liver failure. Pharm Res 2009; 26(4): 893-902.
[http://dx.doi.org/10.1007/s11095-008-9791-0] [PMID: 19034626]
[49]
Lee CH, Jeong TS, Choi YK, et al. Anti-atherogenic effect of citrus flavonoids, naringin and naringenin, associated with hepatic ACAT and aortic VCAM-1 and MCP-1 in high cholesterol-fed rabbits. Biochem Biophys Res Commun 2001; 284(3): 681-8.
[http://dx.doi.org/10.1006/bbrc.2001.5001] [PMID: 11396955]
[50]
Galluzzo P, Ascenzi P, Bulzomi P, Marino M. The nutritional flavanone naringenin triggers antiestrogenic effects by regulating estrogen receptor α-palmitoylation. Endocrinology 2008; 149(5): 2567-75.
[http://dx.doi.org/10.1210/en.2007-1173]] [PMID: 18239068]
[51]
Yi LT, Li CF, Zhan X, et al. Involvement of monoaminergic system in the antidepressant-like effect of the flavonoid naringenin in mice. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34(7): 1223-8.
[http://dx.doi.org/10.1016/j.pnpbp.2010.06.024] [PMID: 20603175]
[52]
Olsen HT, Stafford GI, van Staden J, Christensen SB, Jäger AK. Isolation of the MAO-inhibitor naringenin from Mentha aquatica L. J Ethnopharmacol 2008; 117(3): 500-2.
[http://dx.doi.org/10.1016/j.jep.2008.02.015] [PMID: 18372132]
[53]
Hasanein P, Fazeli F. Role of naringenin in protection against diabetic hyperalgesia and tactile allodynia in male Wistar rats. J Physiol Biochem 2014; 70(4): 997-1006.
[http://dx.doi.org/10.1007/s13105-014-0369-5] [PMID: 25407136]
[54]
Al-Rejaie SS, Abuohashish HM, Al-Enazi MM, Al-Assaf AH, Parmar MY, Ahmed MM. Protective effect of naringenin on acetic acid-induced ulcerative colitis in rats. World J Gastroenterol 2013; 19(34): 5633-44.
[http://dx.doi.org/10.3748/wjg.v19.i34.5633] [PMID: 24039355]
[55]
Dou W, Zhang J, Sun A, et al. Protective effect of naringenin against experimental colitis via suppression of Toll-like receptor 4/NF-κB signalling. Br J Nutr 2013; 110(4): 599-608.
[http://dx.doi.org/10.1017/S0007114512005594] [PMID: 23506745]
[56]
Oršolić N, Gajski G, Garaj-Vrhovac V, Dikić D, Prskalo ZŠ, Sirovina D. DNA-protective effects of quercetin or naringenin in alloxan-induced diabetic mice. Eur J Pharmacol 2011; 656(1-3): 110-8.
[http://dx.doi.org/10.1016/j.ejphar.2011.01.021] [PMID: 21277296]
[57]
Pérez-Pastén R, Martínez-Galero E, Chamorro-Cevallos G. Quercetin and naringenin reduce abnormal development of mouse embryos produced by hydroxyurea. J Pharm Pharmacol 2010; 62(8): 1003-9.
[http://dx.doi.org/10.1111/j.2042-7158.2010.01118.x] [PMID: 20663034]
[58]
Rajadurai M, Prince PSM. Naringin ameliorates mitochondrial lipid peroxides, antioxidants and lipids in isoproterenol-induced myocardial infarction in Wistar rats. Phytother Res 2009; 23(3): 358-62.
[http://dx.doi.org/10.1002/ptr.2632] [PMID: 18844325]
[59]
Arafa HM, Abd-Ellah MF, Hafez HF. Abatement by naringenin of doxorubicin-induced cardiac toxicity in rats. J Egypt Natl Canc Inst 2005; 17(4): 291-300.
[PMID: 17102822]
[60]
Zandi K, Teoh BT, Sing-Sin S, et al. In vitro antiviral activity of fisetin, rutin and naringenin against dengue virus type-2. J Med Plants Res 2011; 8(6): 5534-9.
[61]
Kumar S, Pandey AK. Chemistry and biological activities of flavonoids: an overview. ScientificWorldJournal 2013.2013162750
[http://dx.doi.org/10.1155/2013/162750] [PMID: 24470791]
[62]
Brodowska KM. Natural flavonoids: Classification, potential role, and application of flavonoid analogues. Eur J Biol Res 2017; 7(2): 108-23.
[63]
Kozłowska J, Potaniec B, Żarowska B, Anioł M. Synthesis and biological activity of novel O-alkyl derivatives of naringenin and their oximes. Molecules 2017; 22(9): 1485-99.
[http://dx.doi.org/10.3390/molecules22091485] [PMID: 28878189]
[64]
Brodowska K, Sykuła A, Garribba E, et al. Naringenin Schiff base: Antioxidant activity, acid-base profile, and interactions with DNA. Transition Met Chem 2016; 41(2): 179-89.
[http://dx.doi.org/10.1007/s11243-015-0010-7]
[65]
Murti Y, Mishra P. Synthesis, characterization, and biological evaluation of novel naringenin derivatives as anticancer agents. Curr Bioact Compd 2019; 15: 1-6.
[66]
Kocyigit A, Koyuncu I, Dikilitas M, et al. Cytotoxic, genotoxic and apoptotic effects of naringenin-oxime relative to naringenin on normal and cancer cell lines. Asian Pac J Trop Biomed 2016; 6(10): 872-80.
[http://dx.doi.org/10.1016/j.apjtb.2016.08.004]
[67]
Li H, Zhu F, Chen H, et al. 6-C-(E-phenylethenyl)-naringenin suppresses colorectal cancer growth by inhibiting cyclooxygenase-1. Cancer Res 2014; 74(1): 243-52.
[http://dx.doi.org/10.1158/0008-5472.CAN-13-2245] [PMID: 24220240]
[68]
Yoon H, Kim TW, Shin SY, et al. Design, synthesis and inhibitory activities of naringenin derivatives on human colon cancer cells. Bioorg Med Chem Lett 2013; 23(1): 232-8.
[http://dx.doi.org/10.1016/j.bmcl.2012.10.130] [PMID: 23177257]
[69]
Kim JH, Kim H, Bak Y, et al. Naringenin derivative diethyl (5,4′-dihydroxy flavanone-7-yl) phosphate inhibits cell growth and induces apoptosis in A549 human lung cancer cells. J Korean Soc Appl Biol Chem 2012; 55(1): 75-82.
[http://dx.doi.org/10.1007/s13765-012-0013-4]
[70]
Bak Y, Kim H, Kang JW, et al. A synthetic naringenin derivative, 5-hydroxy-7,4′-diacetyloxyflavanone-N-phenyl hydrazone (N101-43), induces apoptosis through up-regulation of Fas/FasL expression and inhibition of PI3K/Akt signaling pathways in non-small-cell lung cancer cells. J Agric Food Chem 2011; 59(18): 10286-97.
[http://dx.doi.org/10.1021/jf2017594] [PMID: 21877710]
[71]
Park JH, Lee JW, Paik HD, et al. Cytotoxic effects of 7-O-butyl naringenin on human breast cancer MCF-7 cells. Food Sci Biotechnol 2010; 19(3): 717-24.
[http://dx.doi.org/10.1007/s10068-010-0101-3]
[72]
Lee ER, Kang YJ, Choi HY, et al. Induction of apoptotic cell death by synthetic naringenin derivatives in human lung epithelial carcinoma A549 cells. Biol Pharm Bull 2007; 30(12): 2394-8.
[http://dx.doi.org/10.1248/bpb.30.2394] [PMID: 18057732]
[73]
Kim JY, Lee YS, Kim HJ, et al. Synthesis of naringenin amino acid esters as potential CDK2 inhibitors. Bull Korean Chem Soc 2005; 26(12): 2065-8.
[http://dx.doi.org/10.5012/bkcs.2005.26.12.2065]]
[74]
Tokalov SV, Henker Y, Schwab P, Metz P, Gutzeit HO. Toxicity and cell cycle effects of synthetic 8-prenylnaringenin and derivatives in human cells. Pharmacology 2004; 71(1): 46-56.
[http://dx.doi.org/10.1159/000076261] [PMID: 15051922]
[75]
Swarnkar G, Sharan K, Siddiqui JA, et al. A naturally occurring naringenin derivative exerts potent bone anabolic effects by mimicking oestrogen action on osteoblasts. Br J Pharmacol 2012; 165(5): 1526-42.
[http://dx.doi.org/10.1111/j.1476-5381.2011.01637.x] [PMID: 21864313]
[76]
Han X, Ren D, Fan P, Shen T, Lou H. Protective effects of naringenin-7-O-glucoside on doxorubicin-induced apoptosis in H9C2 cells. Eur J Pharmacol 2008; 581(1-2): 47-53.
[http://dx.doi.org/10.1016/j.ejphar.2007.11.048] [PMID: 18154951]
[77]
Lee S, Lee CH, Moon SS, et al. Naringenin derivatives as anti-atherogenic agents. Bioorg Med Chem Lett 2003; 13(22): 3901-3.
[http://dx.doi.org/10.1016/j.bmcl.2003.09.009] [PMID: 14592471]
[78]
Roelens F, Heldring N, Dhooge W, et al. Subtle side-chain modifications of the hop phytoestrogen 8-prenylnaringenin result in distinct agonist/antagonist activity profiles for estrogen receptors alpha and beta. J Med Chem 2006; 49(25): 7357-65.
[http://dx.doi.org/10.1021/jm060692n] [PMID: 17149865]
[79]
Zierau O, Hamann J, Tischer S, et al. Naringenin-type flavonoids show different estrogenic effects in mammalian and teleost test systems. Biochem Biophys Res Commun 2005; 326(4): 909-16.
[http://dx.doi.org/10.1016/j.bbrc.2004.11.124] [PMID: 15607756]
[80]
Moon SH, Kim KT, Lee NK, et al. Inhibitory effects of naringenin and its novel derivatives on hyaluronidase. Food Sci Biotechnol 2009; 18(1): 267-70.

Rights & Permissions Print Export Cite as
© 2024 Bentham Science Publishers | Privacy Policy