Generic placeholder image

Current Traditional Medicine

Editor-in-Chief

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

Review Article

An Overview of Medicinal Importance, Pharmacological Activities and Analytical Aspects of Fraxin from Cortex fraxinus

Author(s): Dinesh Kumar Patel* and Kanika Patel

Volume 9, Issue 5, 2023

Published on: 21 October, 2022

Article ID: e190922208921 Pages: 7

DOI: 10.2174/2215083808666220919114652

Price: $65

Abstract

Background: Medicinal plants and their derived products have been used in the medical field for the treatment of human diseases from a very early age. Phytoproducts, including numerous pure phytoconstituents, have been used for the treatment of human health complications. Coumarin is a pure phytoconstituent of benzopyrone family derivatives. Fraxin is a coumarin glucoside found to be present in the bark of Fraxinus excelsior L. Fraxin has numerous physiological functions in medicine mainly due to its anti-inflammatory, anti-oxidant, hepatoprotective and analgesic activity.

Methods: The present review mainly focuses on the medicinal importance, pharmacological activities and analytical aspects of fraxin. Pharmacological activities of fraxin have been reviewed in the present work through literature data analysis of different scientific research works. Analytical methods developed for the qualitative and quantitative analysis of fraxin in herbal drugs have also been reviewed in the present work through scientific data analysis of different research works. Electronic databases such as PubMed, Science Direct, Google Scholar and Scopus have been searched in the present work in order to collect scientific information on fraxin.

Results: Present paper summarized the biological importance, therapeutic benefit and pharmacological activities of fraxin in medicine. Scientific data analysis of different literature work revealed the biological importance and therapeutic benefit of fraxin in medicine. The biological importance of fraxin in medicine was mainly due to its significant effect on acute respiratory distress syndrome, hepatotoxicity, inflammation, oxidative stress, acute kidney injury, osteoarthritis, liver damage and tumor. Further pharmacokinetic data on fraxin were also collected and discussed in the present work to know the plasma drug concentration profile of fraxin. The analytical data of the present paper will be beneficial for the development of newer and effective analytical tools for the quantitative estimation of fraxin in different biological samples. However, scientific experiments on fraxin for their clinical use in medicine should be developed in order to scientifically validate the medicinal uses of fraxin.

Conclusion: Present paper will be beneficial to all the scientific people in the biological field to know the therapeutic benefit of fraxin against various forms of human disorders.

Keywords: Fraxin, respiratory, hepatotoxicity, inflammation, oxidative stress, kidney injury, osteoarthritis, tumor.

Graphical Abstract
[1]
Patel K, Kumar V, Verma A, Rahman M, Kumar PD. Health benefits of furanocoumarins ‘Psoralidin’ an active phytochemical of Psoralea corylifolia: The present, past and future scenario. Curr Bioact Compd 2019; 15(4): 369-76.
[http://dx.doi.org/10.2174/1573407214666180511153438]
[2]
Alagesan V, Ramalingam S, Kim M, Venugopal S. Antioxidant activity guided isolation of a coumarin compound from Ipomoea pes-caprea (Convolvulaceae) leaves acetone extract and its biological and molecular docking studies. Eur J Integr Med 2019; 32: 100984.
[http://dx.doi.org/10.1016/j.eujim.2019.100984]
[3]
Patel K, Husain GM, Katiyar DK, Prasad SK, Patel DK. Sophoricoside: Bioactive compounds from Sophora japonica, their role in disease prevention and treatment. Curr Tradit Med 2021; 7(2): 180-8.
[http://dx.doi.org/10.2174/2215083806666200214114106]
[4]
Patel K, Kumar V, Rahman M, Verma A, Patel DK. Rhamnazin: A systematic review on ethnopharmacology, pharmacology and analytical aspects of an important phytomedicine. Curr Tradit Med 2018; 4(2): 120-7.
[http://dx.doi.org/10.2174/2215083804666180416124949]
[5]
Aminah NS, Laili ER, Rafi M, Rochman A, Insanu M, Tun KNW. Secondary metabolite compounds from Sida genus and their bioactivity. Heliyon 2021; 7(4): e06682.
[http://dx.doi.org/10.1016/j.heliyon.2021.e06682] [PMID: 33912700]
[6]
Baba SA, Vishwakarma RA, Ashraf N. Functional characterization of CsBGlu12, a β-glucosidase from Crocus sativus, provides insights into its role in abiotic stress through accumulation of antioxidant flavonols. J Biol Chem 2017; 292(11): 4700-13.
[http://dx.doi.org/10.1074/jbc.M116.762161] [PMID: 28154174]
[7]
Wink M. Evolution of secondary metabolites in legumes (Fabaceae). S Afr J Bot 2013; 89: 164-75.
[http://dx.doi.org/10.1016/j.sajb.2013.06.006]
[8]
Kumar Bachheti R, Fikadu A, Bachheti A, Husen A. Biogenic fabrication of nanomaterials from flower-based chemical compounds, characterization and their various applications: A review. Saudi J Biol Sci 2020; 27(10): 2551-62.
[http://dx.doi.org/10.1016/j.sjbs.2020.05.012] [PMID: 32994711]
[9]
Detering T, Berger RG. Formation of 2,6-DMBQ in anthocyanin-rich foods. Lebensm Wiss Technol 2020; 134: 110178.
[http://dx.doi.org/10.1016/j.lwt.2020.110178]
[10]
Gowd V, Xie L, Sun C, Chen W. Phenolic profile of bayberry followed by simulated gastrointestinal digestion and gut microbiota fermentation and its antioxidant potential in HepG2 cells. J Funct Foods 2020; 70: 103987.
[http://dx.doi.org/10.1016/j.jff.2020.103987]
[11]
Patel K, Singh GK, Patel DK. A review on pharmacological and analytical aspects of naringenin. Chin J Integr Med 2018; 24(7): 551-60.
[http://dx.doi.org/10.1007/s11655-014-1960-x] [PMID: 25501296]
[12]
Yu M, Sun A, Zhang Y, Liu R. Purification of coumarin compounds from Cortex fraxinus by adsorption chromatography. J Chromatogr Sci 2014; 52(9): 1033-7.
[http://dx.doi.org/10.1093/chromsci/bmt153] [PMID: 24114664]
[13]
Sarfraz I, Rasul A, Jabeen F, et al. Fraxinus: A plant with versatile pharmacological and biological activities. Evid Based Complement Alternat Med 2017; 2017: 4269868.
[http://dx.doi.org/10.1155/2017/4269868] [PMID: 29279716]
[14]
Idris S, Mishra A, Khushtar M. Phytochemical, ethanomedicinal and pharmacological applications of escin from Aesculus hippocastanum L. towards future medicine. J Basic Clin Physiol Pharmacol 2020; 31(5): 31.
[http://dx.doi.org/10.1515/jbcpp-2019-0115] [PMID: 32649293]
[15]
Ulrich-Merzenich G, Hartbrod F, Kelber O, Müller J, Koptina A, Zeitler H. Salicylate-based phytopharmaceuticals induce adaptive cytokine and chemokine network responses in human fibroblast cultures. Phytomedicine 2017; 34: 202-11.
[http://dx.doi.org/10.1016/j.phymed.2017.08.002] [PMID: 28899503]
[16]
Wang Z, Zhu W, Liu H, et al. Simultaneous determination of aesculin, aesculetin, fraxetin, fraxin and polydatin in beagle dog plasma by UPLC-ESI-MS/MS and its application in a pharmacokinetic study after oral administration extracts of Ledum palustre L. Molecules 2018; 23(9): 2285.
[http://dx.doi.org/10.3390/molecules23092285] [PMID: 30205426]
[17]
Akdemir FNE, Tanyeli A. The antioxidant effect of fraxin against acute organ damage in polymicrobial sepsis model induced by cecal ligation and puncture. Turkish J Sci 2019; 4: 229.
[18]
Ma X, Liu X, Feng J, et al. Fraxin alleviates LPS-Induced ARDS by downregulating inflammatory responses and oxidative damages and reducing pulmonary vascular permeability. Inflammation 2019; 42(5): 1901-12.
[http://dx.doi.org/10.1007/s10753-019-01052-8] [PMID: 31273573]
[19]
Whang WK, Park HS, Ham I, et al. Natural compounds,fraxin and chemicals structurally related to fraxin protect cells from oxidative stress. Exp Mol Med 2005; 37(5): 436-46.
[http://dx.doi.org/10.1038/emm.2005.54] [PMID: 16264268]
[20]
Hassanein EHM, Sayed AM, Hussein OE, Mahmoud AM. Coumarins as modulators of the Keap1/Nrf2/ARE signaling pathway. Oxid Med Cell Longev 2020; 2020: 1675957.
[http://dx.doi.org/10.1155/2020/1675957] [PMID: 32377290]
[21]
Li W, Li W, Zang L, et al. Fraxin ameliorates lipopolysaccharide-induced acute lung injury in mice by inhibiting the NF-κB and NLRP3 signalling pathways. Int Immunopharmacol 2019; 67: 1-12.
[http://dx.doi.org/10.1016/j.intimp.2018.12.003] [PMID: 30530164]
[22]
Chang BY, Jung YS, Yoon C-S, et al. Fraxin prevents chemically induced hepatotoxicity by reducing oxidative stress. Molecules 2017; 22(4): 587.
[http://dx.doi.org/10.3390/molecules22040587] [PMID: 28383514]
[23]
Niu X, Liu F, Li W, et al. Hepatoprotective effect of fraxin against carbon tetrachloride-induced hepatotoxicity in vitro and in vivo through regulating hepatic antioxidant, inflammation response and the MAPK-NF-κB signaling pathway. Biomed Pharmacother 2017; 95: 1091-102.
[http://dx.doi.org/10.1016/j.biopha.2017.09.029] [PMID: 28922728]
[24]
Ekinci-Akdemir FN, Bingöl Ç, Yıldırım S, Kandemir FM, Küçükler S, Sağlam YS. The investigation of the effect of fraxin on hepatotoxicity induced by cisplatin in rats. Iraqi J Sci 2020; 23: 13827.
[25]
Li W, Li W, Yu J, et al. Fraxin inhibits lipopolysaccharide-induced inflammatory cytokines and protects against endotoxic shock in mice. Fundam Clin Pharmacol 2020; 34(1): 91-101.
[http://dx.doi.org/10.1111/fcp.12500] [PMID: 31325387]
[26]
Topdağı Ö, Tanyeli A, Akdemir FNE, Eraslan E, Güler MC, Çomaklı S. Preventive effects of fraxin on ischemia/reperfusion-induced acute kidney injury in rats. Life Sci 2020; 242: 117217.
[http://dx.doi.org/10.1016/j.lfs.2019.117217] [PMID: 31884094]
[27]
Li J-M, Zhang X, Wang X, Xie Y-C, Kong L-D. Protective effects of cortex fraxini coumarines against oxonate-induced hyperuricemia and renal dysfunction in mice. Eur J Pharmacol 2011; 666(1-3): 196-204.
[http://dx.doi.org/10.1016/j.ejphar.2011.05.021] [PMID: 21620826]
[28]
Qian Z, Ru X, Liu C, Huang X, Sun Q. Fraxin prevents knee osteoarthritis through inhibiting chondrocyte apoptosis in an experimental rat osteoarthritis model. Protein Pept Lett 2021; 28(11): 1298-302.
[http://dx.doi.org/10.2174/0929866528666211022152556] [PMID: 34719360]
[29]
Kim YR, Park B-K, Seo C-S, Kim NS, Lee MY. Antidepressant and anxiolytic-like effects of the stem bark extract of Fraxinus rhynchophylla hance and its components in a mouse model of depressive-like disorder induced by reserpine administration. Front Behav Neurosci 2021; 15: 650833.
[http://dx.doi.org/10.3389/fnbeh.2021.650833] [PMID: 34220460]
[30]
Wu S, Tong L, Liu B, et al. Bioactive ingredients obtained from Cortex Fraxini impair interactions between FAS and GPI. Free Radic Biol Med 2020; 152: 504-15.
[http://dx.doi.org/10.1016/j.freeradbiomed.2019.11.022] [PMID: 31784059]
[31]
Wang H, Xiao B, Hao Z, Sun Z. Simultaneous determination of fraxin and its metabolite, fraxetin, in rat plasma by liquid chromatography-tandem mass spectrometry and its application in a pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1017-1018: 70-4.
[http://dx.doi.org/10.1016/j.jchromb.2016.02.030] [PMID: 26945887]
[32]
Yasuda T, Fukui M, Nakazawa T, Hoshikawa A, Ohsawa K. Metabolic fate of fraxin administered orally to rats. J Nat Prod 2006; 69(5): 755-7.
[http://dx.doi.org/10.1021/np0580412] [PMID: 16724835]
[33]
Wang X, Sun H, Fan Y, Li L, Makino T, Kano Y. Analysis and bioactive evaluation of the compounds absorbed into blood after oral administration of the extracts of Vaccinium vitis-idaea in rat. Biol Pharm Bull 2005; 28(6): 1106-8.
[http://dx.doi.org/10.1248/bpb.28.1106] [PMID: 15930756]
[34]
Zhou Y, Zhang X, Li C, et al. Research on the pharmacodynamics and mechanism of Fraxini Cortex on hyperuricemia based on the regulation of URAT1 and GLUT9. Biomed Pharmacother 2018; 106: 434-42.
[http://dx.doi.org/10.1016/j.biopha.2018.06.163] [PMID: 29990831]
[35]
Owczarek A, Kłys A, Olszewska MA. A validated 1H qNMR method for direct and simultaneous quantification of esculin, fraxin and (-)-epicatechin in Hippocastani cortex. Talanta 2019; 192: 263-9.
[http://dx.doi.org/10.1016/j.talanta.2018.09.036] [PMID: 30348388]
[36]
Li C, Chen A, Chen X, Ma X, Chen X, Hu Z. Non-aqueous capillary electrophoresis for separation and simultaneous determination of fraxin, esculin and esculetin in Cortex fraxini and its medicinal preparations. Biomed Chromatogr 2005; 19(9): 696-702.
[http://dx.doi.org/10.1002/bmc.497] [PMID: 15828063]
[37]
Wang Y, Xu Q, Shen Z, et al. Simultaneous determination of eight active compounds in baitouweng decotion and its single herbs. J Chromatogr Sci 2019; 57(6): 502-10.
[http://dx.doi.org/10.1093/chromsci/bmz022] [PMID: 30929002]
[38]
Qazi SS, Lombardo DA, Abou-Zaid MM. A metabolomic and HPLC-MS/MS analysis of the foliar phenolics, flavonoids and coumarins of the Fraxinus species resistant and susceptible to emerald ash borer. Molecules 2018; 23(11): 2734.
[http://dx.doi.org/10.3390/molecules23112734] [PMID: 30360500]
[39]
Szűcs Z, Cziáky Z, Kiss-Szikszai A, Sinka L, Vasas G, Gonda S. Comparative metabolomics of Tilia platyphyllos Scop. bracts during phenological development. Phytochemistry 2019; 167: 112084.
[http://dx.doi.org/10.1016/j.phytochem.2019.112084] [PMID: 31415913]
[40]
Devkota HP, Basnet P, Yahara S. A new phenolic compound, 4-dehydrochebulic acid-1,6-dimethyl ester from Sapium insigne leaves. J Nat Med 2010; 64(2): 191-3.
[http://dx.doi.org/10.1007/s11418-009-0378-9] [PMID: 20037803]
[41]
Lin S, Liu MT, Wang SJ, Li S, Yang YC, Shi JG. Coumarins from branch of Fraxinus sieboldiana and their antioxidative activity. Zhongguo Zhongyao Zazhi 2008; 33(14): 1708-10.
[PMID: 18841773]
[42]
Kim B, Kim YS, Hwang Y-H, et al. Quercus acuta Thunb. (Fagaceae) and its component, isoquercitrin, inhibit HSV-1 replication by suppressing virus-induced ROS production and NF-κB activation. Antioxidants 2021; 10(10): 1638.
[http://dx.doi.org/10.3390/antiox10101638] [PMID: 34679772]
[43]
Zakharenko AM, Razgonova MP, Pikula KS, Golokhvast KS. Simultaneous determination of 78 compounds of Rhodiola rosea extract by supercritical CO2-extraction and HPLC-ESI-MS/MS spectrometry. Biochem Res Int 2021; 2021: 9957490.
[http://dx.doi.org/10.1155/2021/9957490] [PMID: 34306755]
[44]
Owczarek A, Kolodziejczyk-Czepas J, Woźniak-Serwata J, et al. Potential activity mechanisms of Aesculus hippocastanum Bark: Antioxidant effects in chemical and biological in vitro models. Antioxidants 2021; 10(7): 995.
[http://dx.doi.org/10.3390/antiox10070995] [PMID: 34206691]
[45]
Wang W-H, Yan P-Z, Yang B. Study on fragmentation patterns of coumarins in Notopterygium inchum with ultrahigh performance liquid chromatography combined with quadrupole time-of-flight mass spectrometry. Zhongguo Zhongyao Zazhi 2021; 46(5): 1179-90.
[PMID: 33787113]
[46]
Chen Y, Cai X, Li G, et al. Chemical constituents of radix Actinidia chinensis planch by UPLC-QTOF-MS. Biomed Chromatogr 2021; 35(7): e5103.
[http://dx.doi.org/10.1002/bmc.5103] [PMID: 33629744]
[47]
Patel DK, Patel K, Dhanabal SP. Development of bioanalytical parameters for standardization of Terminalia arjuna. J Acute Dis 2013; 2(4): 287-91.
[http://dx.doi.org/10.1016/S2221-6189(13)60145-6]
[48]
Patel DK, Patel K, Duraiswamy B, Dhanabal SP. Phytochemical analysis and standardization of Strychnos nux-vomica extract through HPTLC techniques. Asian Pac J Trop Dis 2012; 2: S56-60.
[http://dx.doi.org/10.1016/S2222-1808(12)60124-8]
[49]
Patel K, Kumar V, Verma A, Rahman M, Patel DK. β-sitosterol: Bioactive compounds in foods, their role in health promotion and disease prevention “A concise report of its phytopharmaceutical importance”. Curr Tradit Med 2017; 3(3): 168-77.
[http://dx.doi.org/10.2174/2215083803666170615111759]
[50]
Patel DK, Kumar R, Laloo D, Hemalatha S. Natural medicines from plant source used for therapy of diabetes mellitus: An overview of its pharmacological aspects. Asian Pac J Trop Dis 2012; 2(3): 239-50.
[http://dx.doi.org/10.1016/S2222-1808(12)60054-1]
[51]
Patel K, Jain A, Patel DK. Medicinal significance, pharmacological activities, and analytical aspects of anthocyanidins ‘delphinidin’: A concise report. J Acute Dis 2013; 2(3): 169-78.
[http://dx.doi.org/10.1016/S2221-6189(13)60123-7]
[52]
Patel K, Gadewar M, Tahilyani V, Patel DK. A review on pharmacological and analytical aspects of diosmetin: A concise report. Chin J Integr Med 2013; 19(10): 792-800.
[http://dx.doi.org/10.1007/s11655-013-1595-3] [PMID: 24092244]
[53]
Patel DK, Kumar R, Sairam K, Hemalatha S. Pharmacologically tested aldose reductase inhibitors isolated from plant sources - A concise report. Chin J Nat Med 2012; 10(5): 388-400.
[http://dx.doi.org/10.1016/S1875-5364(12)60078-8]
[54]
Patel DK, Patel K, Gadewar M, Tahilyani V. Pharmacological and bioanalytical aspects of galangin- A concise report. Asian Pac J Trop Biomed 2012; 2(1): S449-55.
[http://dx.doi.org/10.1016/S2221-1691(12)60205-6]
[55]
Turgut-Kara N, Cakir O, Hasancebi S, Karabey F, Ari S. Identification and production of phenolic nicotiflorin in Astragalus chrysochlorus callus. Farmacia 2018; 66(3): 558-62.
[http://dx.doi.org/10.31925/farmacia.2018.3.25]

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