Open Access


Read more
image01

Online Manuscript Submission


Read more
image01

Submitted Manuscript Trail


Read more
image01

Online Payment


Read more
image01

Online Subscription


Read more
image01

Email Alert



Read more
image01

Review Article | OPEN ACCESS

Pharmacological activities and pharmacokinetic study of hyperoside: A short review

Aun Raza, Xiuquan Xu, Huifang Sun, Jian Tang , Zhen Ouyang

School of Pharmacy, Jiangsu University, Zhenjiang 212013, China;

For correspondence:-  Jian Tang   Email: jt.u@hotmail.com   Tel:+8651185038201

Received: 1 March 2016        Accepted: 13 September 2016        Published: 25 February 2017

Citation: Raza A, Xu X, Sun H, Tang J, Ouyang Z. Pharmacological activities and pharmacokinetic study of hyperoside: A short review. Trop J Pharm Res 2017; 16(2):483-489 doi: 10.4314/tjpr.v16i2.30

© 2017 The authors.
This is an Open Access article that uses a funding model which does not charge readers or their institutions for access and distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0) and the Budapest Open Access Initiative (http://www.budapestopenaccessinitiative.org/read), which permit unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited..

Abstract

Hyperoside (quercetin-3-O-D-galactoside) is a flavonol glycoside which has been isolated from different plants. It has different pharmacological actions such as anti-inflammatory, anti-depressant, neuro-protective, cardio-protective, anti-diabetic, anti-cancer, anti-fungal, radio-protective, gastro-protective, and antioxidant activities. Studies on its pharmacokinetic (PK) properties revealed that it is a stable compound with no significant gender variation in its activities. Other significant details on its pharmacological properties and information for future investigations on its components are provided.

Keywords: Hyperoside; Anti-inflammatory, Antidepressant, Neuroprotective, Antidiabetic, Anticancer, Antioxidant, Cytochrome P450

Introduction

Natural products have recently attracted much attention in drug discovery and food supplement research   because they are frequently used due to their chemical diversity and potent bioactivities. Flavonoids are plant polyphenols found in herbs, vegetables and fruits. They are well known for their hepatoprotective, cardioprotective, antipyretic, analgesic, and anti-inflammatory activities [1]. Flavonoids comprise a large group of polyphenolic with a benzo-γ-pyrone structure. They are synthesized by the phenylpropanoid pathway. Available reports show that secondary phenolic metabolites including flavonoids are responsible for a variety of pharmacological activities [2].

Hyperoside (), also known as quercetin-3-O-D-galactoside, is a flavonol glycoside present in a variety of vegetables and fruits [3,4]. It has been isolated from various medicinal plants such as Hypericum perforatum[5], Ligularia fischeri [6], Crataegus davisii [7], Divaricate saposhnikovia [8] and Hypericum mysorense [9]. Hyperoside is associated with several potent pharmacological activities which include anti-inflammatory, anti-thrombotic, anti-diabetic, anti-viral, anti-fungal, hepato-protective, and antioxidant protective effects [10-14] ().

 PHARMACOLOGICAL APPLICATIONS OF HYPEROSIDE

Anti-inflammatory activity

Hyperoside exhibited anti-inflammatory activity in mouse ear edema, with inhibition of arachidonic acid-induced edema by more than 15 %, and crotonoil-induced edema by 19-25 % [15]. When mixed with isoquercitrin it produced anti-inflammatory activity against carrageen-induced hind paw edema in albino mice, with 30 % inhibition for 6 h [16]. This phytochemical  isolated from the leaves of Tripodanthus acutifolius has been reported to produce its anti-inflammatory effect by inhibiting cyclooxygenase-2 (COX-2) and hyaluronidase [17]. Hyperoside suppressed vascular inflammatory processes caused by high glucose level in the human umbilical vein endothelilal cells (HUVECs) and in mice [18]. It  suppresses the production of interleukin-6 (IL-6), tumor necrosis factor (TNF) and nitric oxide (NO) in lipopolysaccharide (LPS) -stimulated mouse peritoneal macrophages [17,19].

Anti-nociceptive activity

Studies have shown that hyperoside exhibits analgesic activity by down regulating calcium (Ca2+) concentrations in afferent nerve endings [18,20]. It has anti-nociceptive activity in p-benzoquinone-induced abdominal constriction in mice with inhibition of 25 % [16].

Anti-depressant effect

Hyperoside isolated from Apocynum venetum leaves has been shown to exhibit excellent anti-depressant activity. In vitro study confirmed that 2.5-10 μg/ml of the compound shielded PC12 cells from the lesions and high influx of intracellular Ca2+ induced by corticosteroid (10 μM). Its ability to boost brain-derived neurotrophic factor (BDNF) and cAMP response element binding protein (CREB) has also been reported [21]. In mice orally administered hyperoside at doses of 10-30 mg/kg for 10 days,  antidepressant-like activity was noticed in suspension test (TST) and forced swimming test (FST) that may be affected in their interaction with serotonergic (5-HT2A and 5-HT2) systems [20,22]. Antidepressant-like effects of hyperoside have also been observed in forced swimming tests in rats. The antidepressant effect in rats was prevented by the administration of sulpiride (D2-antagonist). Evidence indicates that the antidepressant effect of hyperoside is mediated through the dopaminergic system [21,23].

Neuroprotective activity

In vitro studies have shown that pretreatment with hyperoside (0.01-1 µmol/L) for 4 h decreased apoptosis in PC12 cells induced by sodium azide (20 µM).This neuro-protective action has been attributed to inhibition of ROS production [22,24]. At 5-20 µM, hyperoside protected primary rat cortical neuron from neurotoxcity of β-amyloid protein (Aβ) induced by sodium azide at 20 µM for 24 h. Thus, hyperosdie could be a useful therapeutic remedy for Alzheimer’s disease (AD) and other neurodegenerative diseases [23,25]. Furthermore, in in vitro ischemic models of oxygen–glucose deprivation followed by reperfusion (OGD-R), it was observed that hyperoside offered protection from ischemic neuron damage by nitric oxide signaling process [26]. It has also been reported that hyperoside attenuates neuronal apoptosis triggered by L-glutamate through inhibition of NR2B-containing N-methyl-D-aspartate (NMDA) subtype of glutamate receptors [27]. At 16 µM it has been shown to effectively reduce elevated levels of NO and Ca2+ from 34.4 µM and 640 nM to 25.0 µM and 331 nM in brain cells, respectively. Hyperoside also reduces the level of neuronal damage caused by anoxia as a result of increased concentration of NO and Ca2+ in brain cells [28].

Protective activity on brain

Hyperoside at 1~10 µM reduced the degree of cerebral injury induced by oxygen-glucose deprivation followed by reperfusion [29].This provided protective effect on focal brain ischemia-reperfusion injury and restrained brain edema in rats [30].Moreover, it was observed that hyperoside has  protective effect on focal brain ischemia and reperfusion injury induced by oxidative damage in  rats [31]. Furthermore, it was observed that brain tissues treated with this compound  had lower water content [32]. Another study revealed that it shields rats from brain ischemia-reperfusion injury and protects them from cognitive disability [33].

Cardiovascular protective activity

Hyperoside at 25 and 50 mg/kg protected rats from cardiomyocyte apoptosis caused by ischemia and perfusion, and at 0.50 μmol/L protected cultured rat neonatal cardiomyocyte from apoptosis [34]. It was also reported that hyperoside suppressed levels of malonodialdehyde (MDA) and NO in the myocardium, but increased the level of superoxide dismutase(SOD) [35]. It has also been reported that hyperoside protects cardiomyocytes from oxidative stress caused by ischemia and perfusion through significant up-regulation of extracellular signal-regulated protein kinase (ERK) [36]. Hyperoside inhibited vascular lesions produced in a carotid artery ligation rat model [37]. It also possesses anticoagulant effects, as demonstrated through its capacity to restrain the production of thrombin and activated factor X (FXa), and extension of activated partial thromboplastin time (aPTT) and prothrombin time (PT) as well as inhibition of thrombin and FXa in HUVECs [13].

Antidiabetic activity

Hyperoside is well regarded for its anti-diabetic effects. It has been reported that hyperoside from Rhododendron arboretum and Crataegus pinnatifida exhibited anti-diabetic activity in streptozotocin-induced diabetic rats [38,39]. Methanolic extract of Artemisia capillaris which contains hyperoside has been reported to show excellent α-glucosidase inhibitory action [40]. In a molecular docking studies, it was shown that hyperoside could be an effective docking agent with targets of Type Ⅱdiabetes mellitus [12,41].

Anticancer activity

In an in vitro study on osteosarcoma cells, hyperoside was reported to restrain the growth of U2OS osteosarcoma cell lines (IC50 value 223.5 ± 9.5 µg/ml for 72 h) and MG63 ( IC50 value 239 ± 22.4 µg/ml for 72 h) by revitalizing the G0/G1 arrest in cell cycle [42]. In addition, hyperoside in combination with Quercetin produced anti-cancer effects in 786-O renal cancer cells (IC50 value 18.2, 18.7,11.8µg/ml at 48, 72 and 96 h) [43].

Antiviral activity

Hyperoside from Abelmoschus manihot (L) medik inhibited the production of HBeAg and HBsAg by hepatitis B virus (HBV)-producing 2.2.15 cells, and inhibited the duck hepatitis B virus (DHBV)-DNA levels in HBV- infected duck models [2]. Its antiviral activity was investigated in covalently-closed circular DNA (cccDNA) of duck hepatitis B virus [44]. Yinqiaosan powder which contains hyperoside is known to possess antiviral action against influenza virus [45].

Hepatoprotective activity

Hyperoside isolated from Artemisia Capillaris has been reported to protect mice from liver damage induced by carbon tetrachloride (CCl4). Furthermore, it increased inducible nitric oxide synthase (iNOS), hemeoxygenase-1 (HO-1) and mRNA expression of tumor necrosis factor-R (TNF-R) [46].

Antioxidant activity

Hyperoside protected PC12 cells against cytotoxicity induced by hydrogen peroxide (H2O2) and tert-butylhydroperoxide (t-BuOOH), and also protected lung fibroblast cells from oxidative stress induced by H2O2  [47]. 

Moreover, hawthorn fruit extract containing hyperoside has been reported to attenuate oxidative damage by protecting mouse bone marrow cells from genotoxicity caused by cyclophosphamide [3]. It also protected hepatic L02 cells and HUVEC cells from oxidative damage induced by H2O2 [48,49]. Recently hyperoside isolated from lotus seed epicarp showed significant DPPH and ABTS+ radical scavenging activities [50].

Immunomodulatory activity

Hyperoside has been shown to exhibit immuno-modulatory activity in mice in non-specific, cellular and humoral immunity tests. In carbon clearance test (non-specific immunity), clearance index at doses of 12.5, 25 and  50 mg/kg was enhanced 34.6 %, 7.7 % and 7.7 %, respectively, and phagocytic index was increased by 18.3 %, 12.9 % and 2.1 % respectively when compared to normal group. In lymphocyte proliferation test, lymphocyte proliferation was elevated by hyperoside action [51]. It also raised the concentration of effector T helper cells secreting cytokines in mouse spleen lymphocytes [44].

Inhibitory activity against cytochrome P450(CYP) isoform

It has been reported that hyperoside inhibits different isoforms of CYP, especially CYP2D6 [52].

Other activities of hyperoside

Hyperoside has remarkable antifungal effect against Pestalotia guepinii, Drechslera sp. and Fusarium avenaceum [11]. It protected mice from gastric mucosal damage induced by ethanol via its anti-oxidant properties, as well as reduction of NO level in gastric mucosal tissues [53]. It decreased the formation of calcium oxalate stones in rat kidney [54] and diminished renal fibrosis in vivo [55]. Hyperoside also shielded hamster lung fibroblast cells from gamma ray radiation-stimulated apoptosis [56] and inhibited ultraviolet B (UVB) -stimulated transactivation of activator protein-1(AP-1), as well as mitogen-activated protein kinase signaling pathway(ERK1/2) in JB6 P+ mouse epidermal cells line [57].

Pharmacokinetics of hyperoside

In vitro studies show that hyperoside is a stable compound. In an oral absorption study of this compound and isoquercetrin in rats, it was demonstrated that isoquercetrin was more easily hydrolyzed to its aglycone moiety in the gastrointestinal tract than hyperoside, and that in vitro, it was more stable than isoquercetrin [58]. In another study, binding interaction between hyperoside and bovine serum albumin (BSA) was investigated by fluorescence spectroscopy [59]. Fluorescence spectroscopy data was examined with the aid of Tachiya model and Stern–Volmer equation. Tachiya model equation revealed that the binding sites and binding constants were increased with increase in temperature. On the other hand, Stern-Volmer equation showed that binding constants reduced with temperature elevation, although the binding sites were independent of temperature. The binding of hyperoside to bovine serum albumin  was enhanced in the presence of Cu2+, Ca2+, Zn2+ and K+ but  decreased when Mg2+ was present [59]. When hyperoside and 3'-O-methyl-hyperoside were administered through gastric probe and intraperitoneal (i.p.) route to rats, it was observed that hyperoside and 3'-O-methyl-hyperoside were present in rat brain after i.p. administration, but were not detected when administered by gastric probe [60]. In another pharmacokinetic profile study of  hyperoside in male and female dogs, it was shown that sex has no remarkable impact  on its pharmacokinetic profile [61]. 

Conclusion

For this review, a number of scientific databases (Science Direct, ISI Web of Knowledge, Google Scholar and China Knowledge Resource Integrated Database) were employed to access information. Research articles published in both English and Chinese languages were included. According to available information, hyperoside from different fruits and vegetables has become important due to its numerous pharmacological activities such as anti-inflammatory, anti-analgesic, neuro-protective, anti-coagulant, anti-diabetic, anti-cancer, anti-viral, anti-fungal, and anti-oxidant activities. The information presented in this review will assist in future studies on hyperoside.

Declarations

Acknowledgement

The authors are thankful to Chinese Government Scholarship (CSC No: 2014DFH792), China Postdoctoral Foundation (2012M521023), Natural Science Foundation of Jiangsu Province (BK20130526) and Zhenjiang Social Develop-ment Project (SH2014062).

References

  1. Romagnolo DF, Selmin OI. Flavonoids and cancer prevention: a review of the evidence.  J Nutr Gerontol Geriatr 2012; 31(3): 206-238.
  2. Mahomoodally MF, A. Gurib-Fakim, Subratty AH. Antimicrobial activities and phytochemical profiles of endemic medicinal plants of Mauritius, PharmalBiol 2005; 43(1): 237-242.
  3. Hosseinimehr SJ, Azadbakht M, Abadi AJ. Protective effect of hawthorn extract against genotoxicity induced by cyclophosphamide in mouse bone marrow cells. Environ Toxicol Pharmacol 2008; 25(1): 51-56.
  4. Wu LL, Yang XB, Huang ZM, Liu HZ, Wu GX. In vivo and in vitro antiviral activity of hyperoside extracted from Abelmoschus manihot (L) medik. Acta Pharmacol Sin 2007; 28(3): 404-409.
  5. Zhou T, Chen B, Fan G, Chai Y, Wu Y. Application of high-speed counter-current chromatography coupled with high-performance liquid chromatography–diode array detection for the preparative isolation and purification of hyperoside from Hypericum perforatum with online purity monitoring. J Chromatography A 2006; 1116(1-2): 97-101.
  6. Piao XL, Mi XY, Jang YP, Jang MH, Cho EJ. Determination of hyperoside and 2"-acetylhyperoside from Ligularia fischeri by high performance liquid chromatography. Food Chem 2011; 129(1): 213-216.
  7. Sözer U, Donmez AA, Meriçli AH. Constituents from the leaves of Crataegus davisii Browicz. Sci Pharm 2006; 74: 203-208.
  8. Li L, Gui Y, Wang J, Zhang H, Zong X, Liu CM. Preparative separation of hyperoside of seeds extract of Saposhnikovia divaricata by high performance counter-current chromatography. J Med Plants Res 2012; 6(5): 884-887.
  9. Hariharapura RC, Mahal HS, Srinivasan R, Jagani H, Vijayan P. A pulse radiolysis study of hyperoside isolated from Hypericum mysorense. Radiat Phys Chem 2015; 107: 149-159.
  10. Huang K, Yang XB, Huang ZM. Research progress in pharmacological activities of hyperoside. Herald Med2008; 28: 1046-1048.
  11. Li S, Zhang Z, Cain A, Wang B,Long M, Taylor J. Antifungal activity of camptothecin, trifolin, and hyperoside isolated from Camptotheca acuminate. J Agric Food Chem 2005; 53: 32-37.
  12. Makheswari MU, Sudarsanam DD. Interaction of hyperoside and β-sitosterol with alanine transaminase using molecular docking. Int j Pharma Res Rev 2013; 2: 17-24.
  13. Ku SK, Kim TH, Lee S, Kim SM, Bae JS. Antithrombotic and profibrinolytic activities of isorhamnetin-3-O-galactoside and hyperoside. Food Chem Toxicol 2013; 53: 197-204.
  14. Piao MJ, Kang KA, Zhang R, Ko DO, Wang ZH, You HJ, Kim HS, Kim JS, Kang SS, Hyun JW. Hyperoside prevents oxidative damage induced by hydrogen peroxide in lung fibroblast cells via an antioxidant effect. Biochim Biophys Acta 2008; 1780(12): 1448-1457.
  15. Lee SJ,  Son KH,  Chang HW , Do LC , Jung YK , Kang SS ,Kim HP. Antiinflammatory activity of naturally occurring flavone and fiavonol glycosides. Arch Pharm Res 1993; 16(1): 25-28.
  16. Erdemoglua N, Akkol EK, Yesiladab E, Calıs I. Bioassay-guided isolation of anti-inflammatory and antinociceptive principles from a folk remedy, Rhododendron ponticum L. leaves. J Ethnopharmacol 2008; 119: 172-178.
  17. Soberón JR, Sgariglia MA, Sampietro DA, Quiroga EN, Vattuone MA. Study of antiinflammatory activity of metabolites isolated from Tripodanthus acutifolius. Mol Med Chem 2010; 21 88-90.
  18. Ku SK, Kwak S, Kwon OJ, Bae JS. Hyperoside inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflammation 2014; 37: 1389-1400.
  19. Kim SJ, Um JY, Hong SH, Lee JY. Anti-inflammatory activity of hyperoside through the suppression of nuclear factor-κb activation in mouse peritoneal macrophages. Am J Chin Med 2011; 39(1): 171.
  20. Chen ZW, Ma CG, Xu SY. Mechanism of analgesic action of hyperin. Acta Pharm Sin 1989; 24: 326-330.
  21. Zheng M, Liu C, Pan F, Shi D, Zhang Y. Antidepressant-like effect of hyperoside isolated from Apocynum venetum leaves: possible cellular mechanisms. Phytomedicine 2012; 19(2): 145-149.
  22. Zheng M, Liu C, Fan Y, Shi D. Involvement of serotonergic system in the antidepressant-like effect of hyperoside from Apocynum venetum leaves. Lat Am J Pharm 2012; 31: 982-989.
  23. Haas JS, Stolz ED, Betti AH, Stein AC, Schripsema J, Poser GL, Rates SM. The anti-immobility effect of hyperoside on the forced swimming test in rats is mediated by the D2-like receptors activation. Planta Med 2011; 77(4): 334-339.
  24. Zhang L, Cheng XR, Hu JJ, Sun L, Du GH. Neuroprotective effects of hyperoside on sodium azide-induced apoptosis in pc12 cells. Chin J Natural Med 2011; 9: 450-455.
  25. Zeng KW, Wang XM, Ko H, Kwon HC, Cha JW, Yang HO. Hyperoside protects primary rat cortical neurons from neurotoxicity induced by amyloid beta-protein via the PI3K/Akt/Bad/Bcl(XL)-regulated mitochondrial apoptotic pathway. Eur J Pharmacol 2011; 672(1-3): 45-55.
  26. Liu RL, Xiong QJ, Shu Q, Wu WN, Cheng J, Fu H, Wang F, Chen JG, Hu ZL. Hyperoside protects cortical neurons from oxygen-glucose deprivation-reperfusion induced injury via nitric oxide signal pathway. Brain Res 2012; 1469: 164-173.
  27. Zhang XN, Li JM, Yang Q, Feng B, Liu SB, Xu ZH, Guo YY, Zhao MG. Anti-apoptotic effects of hyperoside via inhibition of NR2B-containing NMDA receptors. Pharmacol Rep 2010; 62(5): 949-955.
  28. Zhou WL, Chen ZW. Action mechanism of hyperin on neonatal rats neuron with anoxia-reoxygenation. Chin Pharmacol Bulletin 2010; 26: 83-86.
  29. Peng GP, Wei EQ, Ge QF, Li XX. Effects of hyperin on brain injury induced by oxygen-glucose deprivation/reperfusion invitro. Chin Pharm J 2005; 40: 434-437.
  30. Chen HY, Wang JH, Ren ZX, Yang XB. Protective effect of hyperin on focal cerebral ischemia reperfusion injury in rats. J Chin Integr Med 2006; 4: 526-529.
  31. Chen HY, Wang JH, Yang XB, Geng M, Huang ZM , Cao WB. Effects of hyperin preconditioning on focal cerebral ischemia reperfusion injury via ameliorating oxidative stress in rats. Pharm J Chin PLA 2007; 23: 88-91.
  32. Fu Q, Qi Z, Yang R. Inflammatory mechanism of hyperin on focal cerebral ischemia reperfusion injury in rats. Chin J Prac Nerv Dis 2008; 11: 20-22.
  33. Zou Y, Nie H, Zhang F, Wei B, Zeng S, Deng X. Effect of hyperin preconditioning on neuroethology after global cerebral ischemia reperfusion in rats. Modern J Integr Trad Chin West Med 2009; 18: 1340-1342.
  34. Li QL, Chou GX, Chen Z, MA CG. Inhibitory mechanism of hyperin on the apoptosis in myocardial ischemia reperfusion in rats. Acta Pharma Sin 2002; 37: 849-852.
  35. Li QL, Chou GX, Chen ZW, Ma CG. Protective effects of hyperin against ischemia/reperfusion injury in rats. Chin Pharm J 2002; 37: 829-832.
  36. Li ZL, Hu J, Li YL, Xue F, Zhang L, Xie JQ, Liu ZH, Li H, Yi DH, Liu JC. The effect of hyperoside on the functional recovery of the ischemic/reperfused isolated rat heart: potential involvement of the extracellular signal-regulated kinase 1/2 signaling pathway. Free Radical Bio Med 2013; 57: 132-140.
  37. Huo Y, Yi B, Chen M, Wang N, Chen P, Guo C, Sun J. Induction of Nur77 by hyperoside inhibits vascular smooth muscle cell proliferation and neointimal formation. Biochem Pharmacol 2014; 92(4): 590-598.
  38. Verma N, Amresh G, Sahu PK, Mishra N, Rao ChV, Singh AP. Pharmacological evaluation of hyperin for antihyperglycemic activity and effect on lipid profile in diabetic rats. Indian J Exp Biol 2013; 51: 65-72.
  39. Chowdhury SS, Islam MM, Jung HA, Choi JS. In vitro antidiabetic potential of the fruits of Crataegus pinnatifida. Res Pharm Sci 2014; 9: 11-22.
  40. Islam MM, Jung HA, Sohn HS, Kim HM, Choi JS. Potent α-glucosidase and protein tyrosine phosphatase 1B inhibitors from Artemisia capillaris. Arch Pharm Res 2013; 36: 542-552.
  41. Makheswari MU, Sudarsanam DD. Molecular docking studies of hyperoside and beta sitosterol with haemoglobin as a target for type 2 diabetes. Int J Pharm Bio Sci 2013; 4: 1366-1372.
  42. Zhang N, Ying MD, Wu YP, Zhou ZH, Ye ZM, Li H, Lin DS. Hyperoside, a flavonoid compound, inhibits proliferation and stimulates osteogenic differentiation of human osteosarcoma cells. PloS one 2014; 9(7): e98973.
  43. Li W, Liu M, Xu YF. Feng Y, Che JP, Wang GC, Zheng JH. Combination of quercetin and hyperoside has anticancer effects on renal cancer cells through inhibition of oncogenic microRNA-27a. Oncol Rep 2014; 31(1): 117-124.
  44. Geng M, Wang JH, Chen HY, Yang XB, Huang ZM. Effects of hyperin on the cccDNA of duck hepatitis B virus and its immunological regulation. Acta Pharm Sin 2009; 44:1440-1444.
  45. SHI Y, Shi RB, LIU B, LU YR. Studies on antiviral flavonoids in yinqiaosan powder. Chin J Chin Mater Med 2001; 26: 320-322.
  46. Choi JH, Kim DW, Yun N, Choi JS, Islam MN, Kim YS, Lee SM. Protective effects of hyperoside against carbon tetrachloride-induced liver damage in mice. J Nat Prod 2011; 74(5): 1055-1060.
  47. Zhao W, Chen ZW, Song BW, Wang Yl, Wang Q, Tang M, Jiang Q. The protective effect of hyperin on gastric mucosal injury in mice and its mechanism. Acta Univ Med Anhui 1999; 34: 178-180.
  48. Zhu W, Xu YF, Feng Y, Peng B, Che JP, Liu M, Zheng JH. Prophylactic effects of quercetin and hyperoside in a calcium oxalate stone forming rat model. Urolithiasis 2014; 42(6): 519-526.
  49. Yan Y, Feng Y, Li W, Che JP, Wang GC, Liu M, Zheng JH. Protective effects of quercetin and hyperoside on renal fibrosis in rats with unilateral ureteral obstruction. Exp Ther Med 2014; 8(3): 727-730.
  50. Mei JP, Ki CK, Suk JC, Sungwook C, Sam SK, Jin WH. Hyperoside protects cells against gamma ray radiation-induced apoptosis in hamster lung fibroblast. Nat Prod Sci 2013; 19: 127-136.
  51. Jung SK, Lim TG, Kim JE, Byun S, Kim G-W, Choi JN, Lee CH, Kim BY, Lee KW, Lee HJ. Inhibitory effect of ERK1/2 and AP-1 by hyperoside isolated from Acanthopanax sessiliflorus. Food Chem 2012; 130(4): 915-920.
  52. Liu Z, Tao X, Zhang C, Lu Y, Wei D. Protective effects of hyperoside (quercetin-3-o-galactoside) to PC12 cells against cytotoxicity induced by hydrogen peroxide and tert-butyl hydroperoxide. Biomed Pharmacotherapy 2005; 59(9): 481-490.
  53. Xing HY, Liu Y, Chen JH, Sun FJ, Shi HQ, Xia PY. Hyperoside attenuates hydrogen peroxide-induced L02 cell damage via MAPK-dependent Keap (1)-Nrf(2)-ARE signaling pathway. Biochem Biophy Res Commun 2011; 410(4): 759-765.
  54. Li ZL, Liu JC, Hu J, Li XQ, Wang SW, Yi DH, Zhao MG. Protective effects of hyperoside against human umbilical vein endothelial cell damage induced by hydrogen peroxide. J Ethnopharmacol 2012; 139(2): 388-394.
  55. Liu Y, Ma SS, Ibrahim SA, Li EH, Yang H, Huang W. Identification and antioxidant properties of polyphenols in lotus seed epicarp at different ripening stages. Food Chem 2015; 185: 159-164.
  56. Huang K, Yang XB, Huang ZM , Geng M , Chen HY , Wang JH, Yang JR. The effect of hyperin on immune function in normal mice. Pharm J Chin  PLA 2009;  25: 133-135
  57. Song M, Hong M, Lee MY, Jee JG, Lee YM, Bae JS, Jeong TC, Lee S. Selective inhibition of the cytochrome P450 isoform by hyperoside and its potent inhibition of CYP2D6. Food Chem Toxicol 2013; 59: 549-553.
  58. Chang Q, Zuo Z, Chow MS, Ho WK. Difference in absorption of the two structurally similar flavonoid glycosides, hyperoside and isoquercitrin, in rats. Eur J Pharm Biopharm 2005; 59(3): 549-555.
  59. Qin Y, Zhang Y, Yan S, Ye L. A comparison study on the interaction of hyperoside and bovine serum albumin with Tachiya model and Stern-Volmer equation. Spectrochimica acta Part A, Mol Biomol Spect 2010; 75(5): 1506-1510.
  60. Guo JM, Lin P, Duan JA, Shang EX, Qian DW, Tang YP. Application of microdialysis for elucidating the existing form of hyperoside in rat brain: comparison between intragastric and intraperitoneal administration. J Ethnopharmacol 2012; 144(3): 664-670.
  61. AI G, Huang ZM, Liu CX. Pharmacokinetic study on hyperoside in beagle’s dogs. Chin Herb Med 2012; 4: 213-217.
Impact Factor
Thompson Reuters (ISI): 0.523 (2021)
H-5 index (Google Scholar): 39 (2021)

Article Tools

Share this article with



Article status: Free
Fulltext in PDF
Similar articles in Google
Similar article in this Journal:

Archives

2024; 23: 
1,   2,   3,   4
2023; 22: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2022; 21: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2021; 20: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2020; 19: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2019; 18: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2018; 17: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2017; 16: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2016; 15: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2015; 14: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2014; 13: 
1,   2,   3,   4,   5,   6,   7,   8,   9,   10,   11,   12
2013; 12: 
1,   2,   3,   4,   5,   6
2012; 11: 
1,   2,   3,   4,   5,   6
2011; 10: 
1,   2,   3,   4,   5,   6
2010; 9: 
1,   2,   3,   4,   5,   6
2009; 8: 
1,   2,   3,   4,   5,   6
2008; 7: 
1,   2,   3,   4
2007; 6: 
1,   2,   3,   4
2006; 5: 
1,   2
2005; 4: 
1,   2
2004; 3: 
1
2003; 2: 
1,   2
2002; 1: 
1,   2

News Updates