Generic placeholder image

Current Nutraceuticals

Editor-in-Chief

ISSN (Print): 2665-9786
ISSN (Online): 2665-9794

Review Article

Recent Updates on the Pharmacological Potential of Plant-based Rutin

Author(s): Akanksha Pandey, Ramesh Kumar, Amita Mishra, Akhilesh Pandey and Abhay K. Pandey*

Volume 3, Issue 2, 2022

Published on: 03 October, 2022

Article ID: e140622205967 Pages: 10

DOI: 10.2174/2665978603666220614151613

Price: $65

Abstract

Backgrounds: Flavonoids have been found to be beneficial in the treatment of several diseases owing to their intense biological activity, bioavailability, and safety aspects. Rutin, an important polyphenolic flavonoid, is consumed on a daily basis in the diet. Rutin, also known as vitamin P and quercetin-3-O- rutinoside, is a nontoxic and non-oxidizable molecule. It is present in vegetables, food items, and beverages.

Objectives: The present review is aimed at providing a comprehensive overview of the medicinal attributes, metabolism, biological and pharmacological activities, and mechanisms of action of rutin. Limitations and future prospects on rutin related research are also described.

Methods: Various search engines and databases were used for literature search with keyword combinations including rutin, antioxidant, pharmacological efficacy, bioavailability, clinical studies, and molecular targets.

Results: A wide range of in vitro and in vivo studies on rutin suggested its potential biological and pharmacological effects, including antioxidant, anti-inflammatory, antidiabetic, anti-hyperlipidemic, renoprotective, hepatoprotective, cardioprotective, and anticancer activities. Rutin exhibited the drug action by modulating various signaling pathways, including PI3K/AKT pathway, β-catenin signaling, JAK-STAT signaling, and apoptotic pathways.

Conclusion: The available data suggest the potential therapeutic utility of rutin against a diverse number of disorders, mainly derived from its antioxidant activity and the modulation of signaling pathways. Further, it helps in minimizing the side effects of the therapy for the treatment of several chronic diseases. Thus promising activities of rutin make it a potent drug candidate.

Keywords: Rutin, metabolism, antioxidant, antidiabetic, anticancer, cardioprotective, signaling.

Graphical Abstract
[1]
Gupta, A.; Singh, A.K.; Kumar, R.; Jamieson, S.; Pandey, A.K.; Bishayee, A. Neuroprotective potential of ellagic acid: A critical review. Adv. Nutr., 2021, 12(4), 1211-1238.
[http://dx.doi.org/10.1093/advances/nmab007] [PMID: 33693510]
[2]
Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. ScientificWorldJournal, 2013, 2013162750.
[http://dx.doi.org/10.1155/2013/162750] [PMID: 24470791]
[3]
Ganeshpurkar, A.; Saluja, A.K. The pharmacological potential of rutin. Saudi Pharm. J., 2017, 25(2), 149-164.
[http://dx.doi.org/10.1016/j.jsps.2016.04.025] [PMID: 28344465]
[4]
Chua, L.S. A review on plant-based rutin extraction methods and its pharmacological activities. J. Ethnopharmacol., 2013, 150(3), 805-817.
[http://dx.doi.org/10.1016/j.jep.2013.10.036] [PMID: 24184193]
[5]
Satari, A.; Ghasemi, S.; Habtemariam, S.; Asgharian, S.; Lorigooini, Z. Rutin: A flavonoid as an effective sensitizer for anticancer therapy; Insights into multifaceted mechanisms and applicability for combination therapy. Evid. Based Complement. Alternat. Med., 2021, 20219913179.
[http://dx.doi.org/10.1155/2021/9913179] [PMID: 34484407]
[6]
Pan, P.H.; Lin, S.Y.; Wang, Y.Y.; Chen, W.Y.; Chuang, Y.H.; Wu, C.C.; Chen, C.J. Protective effects of rutin on liver injury induced by biliary obstruction in rats. Free Radic. Biol. Med., 2014, 73, 106-116.
[http://dx.doi.org/10.1016/j.freeradbiomed.2014.05.001] [PMID: 24815012]
[7]
Sharma, U.K.; Kumar, R.; Ganguly, R.; Gupta, A.; Sharma, A.K.; Pandey, A.K. Cinnamaldehyde, an active component of cinnamon provides protection against food colour induced oxidative stress and hepatotoxicity in albino Wistar rats. Vegetos, 2018, 31(2), 123-129.
[http://dx.doi.org/10.5958/2229-4473.2018.00063.0]
[8]
Dimitrieska-Stojkovic, E.; Zdravkovski, Z. Supercritical fluid extraction of quercetin and rutin from Hyperici herba. J. Liq. Chromatogr. Relat. Technol., 2003, 26(15), 2517-2533.
[http://dx.doi.org/10.1081/JLC-120023798]
[9]
Macikova, P.; Halouzka, V.; Hrbac, J.; Bartak, P.; Skopalova, J. Electrochemical behavior and determination of rutin on modified carbon paste electrodes. ScientificWorldJournal, 2012, 2012, 394756.
[http://dx.doi.org/10.1100/2012/394756] [PMID: 22654602]
[10]
Zhang, Y.; Li, S.F.; Wu, X.W. Pressurized liquid extraction of flavonoids from Houttuynia cordata Thunb. Separ. Purif. Tech., 2008, 58(3), 305-310.
[http://dx.doi.org/10.1016/j.seppur.2007.04.010]
[11]
Michalkiewicz, A.; Biesaga, M.; Pyrzynska, K. Solid-phase extraction procedure for determination of phenolic acids and some flavonols in honey. J. Chromatogr. A, 2008, 1187(1-2), 18-24.
[http://dx.doi.org/10.1016/j.chroma.2008.02.001] [PMID: 18282581]
[12]
Yang, Y.; Zhang, F. Ultrasound-assisted extraction of rutin and quercetin from Euonymus alatus (Thunb.). Sieb. Ultrason. Sonochem., 2008, 15(4), 308-313.
[http://dx.doi.org/10.1016/j.ultsonch.2007.05.001] [PMID: 17606398]
[13]
Zhang, F.; Yang, Y.; Su, P.; Guo, Z. Microwave-assisted extraction of rutin and quercetin from the stalks of Euonymus alatus (Thunb.). Sieb. Phytochem. Anal., 2009, 20(1), 33-37.
[http://dx.doi.org/10.1002/pca.1088] [PMID: 19086093]
[14]
Chahyadi, A. Elfahmi, The influence of extraction methods on rutin yield of cassava leaves (Manihot esculenta Crantz). Saudi Pharm. J., 2020, 28(11), 1466-1473.
[http://dx.doi.org/10.1016/j.jsps.2020.09.012] [PMID: 33250654]
[15]
Liao, J.; Qu, B.; Liu, D.; Zheng, N. New method to enhance the extraction yield of rutin from Sophora japonica using a novel ultrasonic extraction system by determining optimum ultrasonic frequency. Ultrason. Sonochem., 2015, 27, 110-116.
[http://dx.doi.org/10.1016/j.ultsonch.2015.05.005] [PMID: 26186827]
[16]
Andlauer, W.; Stumpf, C.; Fürst, P. Intestinal absorption of rutin in free and conjugated forms. Biochem. Pharmacol., 2001, 62(3), 369-374.
[http://dx.doi.org/10.1016/S0006-2952(01)00638-4] [PMID: 11434911]
[17]
Carbonaro, M.; Grant, G. Absorption of quercetin and rutin in rat small intestine. Ann. Nutr. Metab., 2005, 49(3), 178-182.
[http://dx.doi.org/10.1159/000086882] [PMID: 16006787]
[18]
Boyle, S.P.; Dobson, V.L.; Duthie, S.J.; Hinselwood, D.C.; Kyle, J.A.M.; Collins, A.R. Bioavailability and efficiency of rutin as an antioxidant: A human supplementation study. Eur. J. Clin. Nutr., 2000, 54(10), 774-782.
[http://dx.doi.org/10.1038/sj.ejcn.1601090] [PMID: 11083486]
[19]
Manach, C.; Texier, O.; Regerat, F.; Agullo, G.; Demigne, C.; Remesy, C. Dietary quercetin is recovered in rat plasma as conjugated derivatives of isorhamnetin and quercetin. J. Nutr. Biochem., 1996, 7(7), 375-380.
[http://dx.doi.org/10.1016/S0955-2863(96)00058-7]
[20]
Manach, C.; Morand, C.; Demigné, C.; Texier, O.; Régérat, F.; Rémésy, C. Bioavailability of rutin and quercetin in rats. FEBS Lett., 1997, 409(1), 12-16.
[http://dx.doi.org/10.1016/S0014-5793(97)00467-5] [PMID: 9199494]
[21]
Sawai, Y.; Kohsaka, K.; Nishiyama, Y.; Ando, K. Serum concentrations of rutoside metabolites after oral administration of a rutoside formulation to humans. Arzneimittelforschung, 1987, 37(6), 729-732.
[PMID: 3663272]
[22]
Negahdari, R.; Bohlouli, S.; Sharifi, S.; Maleki Dizaj, S.; Rahbar Saadat, Y.; Khezri, K.; Jafari, S.; Ahmadian, E.; Gorbani Jahandizi, N.; Raeesi, S. Therapeutic benefits of rutin and its nanoformulations. Phytother. Res., 2021, 35(4), 1719-1738.
[http://dx.doi.org/10.1002/ptr.6904] [PMID: 33058407]
[23]
Olthof, M.R.; Hollman, P.C.; Buijsman, M.N.; van Amelsvoort, J.M.; Katan, M.B. Chlorogenic acid, quercetin-3-rutinoside and black tea phenols are extensively metabolized in humans. J. Nutr., 2003, 133(6), 1806-1814.
[http://dx.doi.org/10.1093/jn/133.6.1806] [PMID: 12771321]
[24]
Nagasawa, T.; Tabata, N.; Ito, Y.; Aiba, Y.; Nishizawa, N.; Kitts, D.D. Dietary G-rutin suppresses glycation in tissue proteins of streptozotocin-induced diabetic rats. Mol. Cell. Biochem., 2003, 252(1-2), 141-147.
[http://dx.doi.org/10.1023/A:1025563519088] [PMID: 14577587]
[25]
Leong, C.N.A.; Tako, M.; Hanashiro, I.; Tamaki, H. Antioxidant flavonoid glycosides from the leaves of Ficus pumila L. Food Chem., 2008, 109(2), 415-420.
[http://dx.doi.org/10.1016/j.foodchem.2007.12.069] [PMID: 26003366]
[26]
Kumar, S.; Mishra, A.; Pandey, A.K. Antioxidant mediated protective effect of Parthenium hysterophorus against oxidative damage using in vitro models. BMC Complement. Altern. Med., 2013, 13(1), 120.
[http://dx.doi.org/10.1186/1472-6882-13-120] [PMID: 23721571]
[27]
Mishra, A.; Sharma, A.K.; Kumar, S.; Saxena, A.K.; Pandey, A.K. Bauhinia variegata leaf extracts exhibit considerable antibacterial, antioxidant, and anticancer activities. BioMed Res. Int., 2013, 2013915436.
[http://dx.doi.org/10.1155/2013/915436] [PMID: 24093108]
[28]
Kumar, S.; Dwivedi, A.; Kumar, R.; Pandey, A.K. Preliminary evaluation of biological activities and phytochemical analysis of Syngonium podophyllum leaf. Natl. Acad. Sci. Lett., 2015, 38(2), 143-146.
[http://dx.doi.org/10.1007/s40009-014-0318-4]
[29]
Kumar, S.; Pandey, A.K. Phytochemical screening, GC-MS analysis and in-vitro antioxidant potential of Parthenium hysterophorus stem sequentially extracted fractions. Chem. Biol. Lett, 2020, 7(4), 235-241.
[30]
Nguyen, G.T.; Green, E.R.; Mecsas, J. Neutrophils to the ROScue: Mechanisms of NADPH oxidase activation and bacterial resistance. Front. Cell. Infect. Microbiol., 2017, 7, 373.
[http://dx.doi.org/10.3389/fcimb.2017.00373] [PMID: 28890882]
[31]
Gupta, A.; Kumar, R.; Pandey, A.K. Antioxidant and antidiabetic activities of Terminalia bellirica fruit in alloxan induced diabetic rats. S. Afr. J. Bot., 2020, 130, 308-315.
[http://dx.doi.org/10.1016/j.sajb.2019.12.010]
[32]
Aruoma, O.I.; Hayashi, Y.; Marotta, F.; Mantello, P.; Rachmilewitz, E.; Montagnier, L. Applications and bioefficacy of the functional food supplement fermented papaya preparation. Toxicology, 2010, 278(1), 6-16.
[http://dx.doi.org/10.1016/j.tox.2010.09.006] [PMID: 20870007]
[33]
Khan, M.M.; Ahmad, A.; Ishrat, T.; Khuwaja, G.; Srivastawa, P.; Khan, M.B.; Raza, S.S.; Javed, H.; Vaibhav, K.; Khan, A.; Islam, F. Rutin protects the neural damage induced by transient focal ischemia in rats. Brain Res., 2009, 1292, 123-135.
[http://dx.doi.org/10.1016/j.brainres.2009.07.026] [PMID: 19631195]
[34]
Kumar, S.; Pandey, A.K. Phenolic content, reducing power and membrane protective activities of Solanum xanthocarpum root extracts. Vegetos, 2013, 26(1), 301-307.
[http://dx.doi.org/10.5958/j.2229-4473.26.1.043]
[35]
Yang, J.; Guo, J.; Yuan, J. In vitro antioxidant properties of rutin. Lebensm. Wiss. Technol., 2008, 41(6), 1060-1066.
[http://dx.doi.org/10.1016/j.lwt.2007.06.010]
[36]
Vlahos, C.J. Phosphatidylinositol 3-kinase inhibitors and their effects on cell signaling pathways. Drugs Future, 1995, 20(2), 165-171.
[http://dx.doi.org/10.1358/dof.1995.020.02.288444]
[37]
You, K.M.; Jong, H.G.; Kim, H.P. Inhibition of cyclooxygenase/lipoxygenase from human platelets by polyhydroxylated/methoxylated flavonoids isolated from medicinal plants. Arch. Pharm. Res., 1999, 22(1), 18-24.
[http://dx.doi.org/10.1007/BF02976430] [PMID: 10071954]
[38]
Farzaei, M.H.; Singh, A.K.; Kumar, R.; Croley, C.R.; Pandey, A.K.; Coy-Barrera, E.; Kumar Patra, J.; Das, G.; Kerry, R.G.; Annunziata, G.; Tenore, G.C.; Khan, H.; Micucci, M.; Budriesi, R.; Momtaz, S.; Nabavi, S.M.; Bishayee, A. Targeting inflammation by flavonoids: Novel therapeutic strategy for metabolic disorders. Int. J. Mol. Sci., 2019, 20(19), 4957.
[http://dx.doi.org/10.3390/ijms20194957] [PMID: 31597283]
[39]
Gupta, A.; Pandey, A.K. Aceclofenac-induced hepatotoxicity: An ameliorative effect of Terminalia bellirica fruit and ellagic acid. World J. Hepatol., 2020, 12(11), 949-964.
[http://dx.doi.org/10.4254/wjh.v12.i11.949] [PMID: 33312421]
[40]
Lindahl, M.; Tagesson, C. Flavonoids as phospholipase A2 inhibitors: Importance of their structure for selective inhibition of group II phospholipase A2. Inflammation, 1997, 21(3), 347-356.
[http://dx.doi.org/10.1023/A:1027306118026] [PMID: 9246576]
[41]
Selloum, L.; Bouriche, H.; Tigrine, C.; Boudoukha, C. Anti-inflammatory effect of rutin on rat paw oedema, and on neutrophils chemotaxis and degranulation. Exp. Toxicol. Pathol., 2003, 54(4), 313-318.
[http://dx.doi.org/10.1078/0940-2993-00260] [PMID: 12710715]
[42]
Lee, W.; Ku, S.K.; Bae, J.S. Barrier protective effects of rutin in LPS-induced inflammation in vitro and in vivo. Food Chem. Toxicol., 2012, 50(9), 3048-3055.
[http://dx.doi.org/10.1016/j.fct.2012.06.013] [PMID: 22721984]
[43]
Sharma, S.; Ali, A.; Ali, J.; Sahni, J.K.; Baboota, S. Rutin: Therapeutic potential and recent advances in drug delivery. Expert Opin. Investig. Drugs, 2013, 22(8), 1063-1079.
[http://dx.doi.org/10.1517/13543784.2013.805744] [PMID: 23795677]
[44]
Jadhav, R.; Puchchakayala, G. Hypoglycemic and antidiabetic activity of flavonoids: Boswellic acid, ellagic acid, quercetin, rutin on streptozotocin-nicotinamide induced type 2 diabetic rats. Int. J. Pharm. Pharm. Sci., 2012, 4, 251-256.
[45]
Ahmed, O.M.; Moneim, A.A.; Yazid, I.A.; Mahmoud, A.M. Antihyperglycemic, anti-hyperlipidemic and antioxidant effects and the probable mechanisms of action of Ruta graveolens infusion and rutin in nicotinamide-streptozotocin-induced diabetic rats. Diabetol. Croat., 2010, 39(1), 15-35.
[46]
Li, Y.Q.; Zhou, F.C.; Gao, F.; Bian, J.S.; Shan, F. Comparative evaluation of quercetin, isoquercetin and rutin as inhibitors of α-glucosidase. J. Agric. Food Chem., 2009, 57(24), 11463-11468.
[http://dx.doi.org/10.1021/jf903083h] [PMID: 19938837]
[47]
Singh, A.K.; Rana, H.K.; Singh, V.; Chand Yadav, T.; Varadwaj, P.; Pandey, A.K. Evaluation of antidiabetic activity of dietary phenolic compound chlorogenic acid in streptozotocin induced diabetic rats: Molecular docking, molecular dynamics, in silico toxicity, in vitro and in vivo studies. Comput. Biol. Med., 2021, 134104462.
[http://dx.doi.org/10.1016/j.compbiomed.2021.104462] [PMID: 34148008]
[48]
Joshi, T.; Singh, A.K.; Haratipour, P.; Sah, A.N.; Pandey, A.K.; Naseri, R.; Juyal, V.; Farzaei, M.H. Targeting AMPK signaling pathway by natural products for treatment of diabetes mellitus and its complications. J. Cell. Physiol., 2019, 234(10), 17212-17231.
[http://dx.doi.org/10.1002/jcp.28528] [PMID: 30916407]
[49]
Esmaeili, M.A.; Zohari, F.; Sadeghi, H. Antioxidant and protective effects of major flavonoids from Teucrium polium on β-cell destruction in a model of streptozotocin-induced diabetes. Planta Med., 2009, 75(13), 1418-1420.
[http://dx.doi.org/10.1055/s-0029-1185704] [PMID: 19452438]
[50]
Cai, E.P.; Lin, J.K. Epigallocatechin gallate (EGCG) and rutin suppress the glucotoxicity through activating IRS2 and AMPK signaling in rat pancreatic β cells. J. Agric. Food Chem., 2009, 57(20), 9817-9827.
[http://dx.doi.org/10.1021/jf902618v] [PMID: 19803520]
[51]
Kappel, V.D.; Cazarolli, L.H.; Pereira, D.F.; Postal, B.G.; Zamoner, A.; Reginatto, F.H.; Silva, F.R. Involvement of GLUT-4 in the stimulatory effect of rutin on glucose uptake in rat soleus muscle. J. Pharm. Pharmacol., 2013, 65(8), 1179-1186.
[http://dx.doi.org/10.1111/jphp.12066] [PMID: 23837585]
[52]
Khan, A.H.; Pessin, J.E. Insulin regulation of glucose uptake: A complex interplay of intracellular signalling pathways. Diabetologia, 2002, 45(11), 1475-1483.
[http://dx.doi.org/10.1007/s00125-002-0974-7] [PMID: 12436329]
[53]
Srinivasan, K.; Kaul, C.; Ramarao, R. Partial protective effect of rutin on multiple low dose streptozotocin-induced diabetes in mice. Indian J. Pharmacol., 2005, 37(5), 327.
[http://dx.doi.org/10.4103/0253-7613.16859]
[54]
Ghorbani, A. Best herbs for managing diabetes: A review of clinical studies. Braz. J. Pharm. Sci., 2013, 49(3), 413-422.
[http://dx.doi.org/10.1590/S1984-82502013000300003]
[55]
Pandey, A.K.; Kumar, S.; Pandey, A.K.; Reis, F. Combating redox imbalance-associated complications with natural products. Front. Pharmacol., 2021, 12802750.
[http://dx.doi.org/10.3389/fphar.2021.802750] [PMID: 34899360]
[56]
Kumar, R.; Gupta, A.; Singh, A.K.; Bishayee, A.; Pandey, A.K. The antioxidant and antihyperglycemic activities of bottlebrush plant (Callistemon lanceolatus) stem extracts. Medicines, 2020, 7(3), 11.
[http://dx.doi.org/10.3390/medicines7030011] [PMID: 32143382]
[57]
Niture, N.T.; Ansari, A.A.; Naik, S.R. Anti-hyperglycemic activity of rutin in streptozotocin-induced diabetic rats: An effect mediated through cytokines, antioxidants and lipid biomarkers. Indian J. Exp. Biol., 2014, 52(7), 720-727.
[PMID: 25059040]
[58]
Stanely Mainzen Prince, P.; Kannan, N.K. Protective effect of rutin on lipids, lipoproteins, lipid metabolizing enzymes and glycoproteins in streptozotocin-induced diabetic rats. J. Pharm. Pharmacol., 2006, 58(10), 1373-1383.
[http://dx.doi.org/10.1211/jpp.58.10.0011] [PMID: 17034661]
[59]
Fernandes, A.A.H.; Novelli, E.L.B.; Okoshi, K.; Okoshi, M.P.; Di Muzio, B.P.; Guimarães, J.F.C.; Fernandes, Junior, A. Influence of rutin treatment on biochemical alterations in experimental diabetes. Biomed. Pharmacother., 2010, 64(3), 214-219.
[http://dx.doi.org/10.1016/j.biopha.2009.08.007] [PMID: 19932588]
[60]
Wang, Y.B.; Ge, Z.M.; Kang, W.Q.; Lian, Z.X.; Yao, J.; Zhou, C.Y. Rutin alleviates diabetic cardiomyopathy in a rat model of type 2 diabetes. Exp. Ther. Med., 2015, 9(2), 451-455.
[http://dx.doi.org/10.3892/etm.2014.2090] [PMID: 25574214]
[61]
Guimaraes, J.F.; Muzio, B.P.; Rosa, C.M.; Nascimento, A.F.; Sugizaki, M.M.; Fernandes, A.A.; Cicogna, A.C.; Padovani, C.R.; Okoshi, M.P.; Okoshi, K. Rutin administration attenuates myocardial dysfunction in diabetic rats. Cardiovasc. Diabetol., 2015, 14(1), 90.
[http://dx.doi.org/10.1186/s12933-015-0255-7] [PMID: 26185015]
[62]
Ghorbani, A. Mechanisms of antidiabetic effects of flavonoid rutin. Biomed. Pharmacother., 2017, 96, 305-312.
[http://dx.doi.org/10.1016/j.biopha.2017.10.001] [PMID: 29017142]
[63]
Dronavalli, S.; Duka, I.; Bakris, G.L. The pathogenesis of diabetic nephropathy. Nat. Clin. Pract. Endocrinol. Metab., 2008, 4(8), 444-452.
[http://dx.doi.org/10.1038/ncpendmet0894] [PMID: 18607402]
[64]
Schena, F.P.; Gesualdo, L. Pathogenetic mechanisms of diabetic nephropathy. J. Am. Soc. Nephrol., 2005, 16(3)(Suppl. 1), S30-S33.
[http://dx.doi.org/10.1681/ASN.2004110970] [PMID: 15938030]
[65]
Hao, H.H.; Shao, Z.M.; Tang, D.Q.; Lu, Q.; Chen, X.; Yin, X.X.; Wu, J.; Chen, H. Preventive effects of rutin on the development of experimental diabetic nephropathy in rats. Life Sci., 2012, 91(19-20), 959-967.
[http://dx.doi.org/10.1016/j.lfs.2012.09.003] [PMID: 23000098]
[66]
Hu, Q.H.; Wang, C.; Li, J.M.; Zhang, D.M.; Kong, L.D. Allopurinol, rutin, and quercetin attenuate hyperuricemia and renal dysfunction in rats induced by fructose intake: Renal organic ion transporter involvement. Am. J. Physiol. Renal Physiol., 2009, 297(4), F1080-F1091.
[http://dx.doi.org/10.1152/ajprenal.90767.2008] [PMID: 19605544]
[67]
Alsaif, M.A. Beneficial effects of rutin and vitamin C coadministration in a streptozotocin-induced diabetes rat model of kidney nephrotoxicity. Pak. J. Nutr., 2009, 8(6), 745-754.
[http://dx.doi.org/10.3923/pjn.2009.745.754]
[68]
Kamalakkannan, N.; Prince, P.S. The influence of rutin on the extracellular matrix in streptozotocin-induced diabetic rat kidney. J. Pharm. Pharmacol., 2006, 58(8), 1091-1098.
[http://dx.doi.org/10.1211/jpp.58.8.0010] [PMID: 16872556]
[69]
Sharma, U.K.; Kumar, R.; Gupta, A.; Ganguly, R.; Pandey, A.K. Renoprotective effect of cinnamaldehyde in food color induced toxicity. 3 Biotech, 2018, 8(4), 212.
[http://dx.doi.org/10.1007/s13205-018-1241-z]
[70]
Al-Enazi, M.M. Protective effects of combined therapy of rutin with silymarin on experimentally-induced diabetic neuropathy in rats. Pharmacol. Pharm., 2014, 5(09), 876-889.
[http://dx.doi.org/10.4236/pp.2014.59098]
[71]
Gupta, A.; Atkinson, A.N.; Pandey, A.K.; Bishayee, A. Health-promoting and disease-mitigating potential of Verbascum thapsus L. (common mullein): A review. Phytother. Res., 2022, 36(4), 1507-1522.
[http://dx.doi.org/10.1002/ptr.7393] [PMID: 35088467]
[72]
Harrison, S.A. Liver disease in patients with diabetes mellitus. J. Clin. Gastroenterol., 2006, 40(1), 68-76.
[http://dx.doi.org/10.1097/01.mcg.0000190774.91875.d2] [PMID: 16340637]
[73]
Chu, J.; Li, G.; Han, S. Protective effect of rutin from buckwheat flowers and leaves on hepatic injury at early stage in diabetic rats. Jiangsu Med. J., 2010, 8(026)
[74]
Gupta, A.; Kumar, R.; Ganguly, R.; Singh, A.K.; Rana, H.K.; Pandey, A.K. Antioxidant, anti-inflammatory and hepatoprotective activities of Terminalia bellirica and its bioactive component ellagic acid against diclofenac induced oxidative stress and hepatotoxicity. Toxicol. Rep., 2020, 8, 44-52.
[http://dx.doi.org/10.1016/j.toxrep.2020.12.010] [PMID: 33391996]
[75]
Shah, M.S.; Brownlee, M. Molecular and cellular mechanisms of cardiovascular disorders in diabetes. Circ. Res., 2016, 118(11), 1808-1829.
[http://dx.doi.org/10.1161/CIRCRESAHA.116.306923] [PMID: 27230643]
[76]
Sharma, U.K.; Kumar, R.; Gupta, A.; Ganguly, R.; Singh, A.K.; Ojha, A.K.; Pandey, A.K. Ameliorating efficacy of eugenol against metanil yellow induced toxicity in albino Wistar rats. Food Chem. Toxicol., 2019, 126, 34-40.
[http://dx.doi.org/10.1016/j.fct.2019.01.032] [PMID: 30738991]
[77]
Krishna, K.M.; Annapurna, A.; Gopal, G.S.; Chalam, C.R.V.; Madan, K.; Kumar, V.K.; Prakash, G.J. Partial reversal by rutin and quercetin of impaired cardiac function in streptozotocin-induced diabetic rats. Can. J. Physiol. Pharmacol., 2005, 83(4), 343-355.
[http://dx.doi.org/10.1139/y05-009] [PMID: 15877109]
[78]
Rauter, A.P.; Martins, A.; Borges, C.; Mota-Filipe, H.; Pinto, R.; Sepodes, B.; Justino, J. Antihyperglycaemic and protective effects of flavonoids on streptozotocin-induced diabetic rats. Phytother. Res., 2010, 24(S2)(Suppl. 2), S133-S138.
[http://dx.doi.org/10.1002/ptr.3017] [PMID: 20309949]
[79]
Sprague, A.H.; Khalil, R.A. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem. Pharmacol., 2009, 78(6), 539-552.
[http://dx.doi.org/10.1016/j.bcp.2009.04.029] [PMID: 19413999]
[80]
Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as anticancer agents. Nutrients, 2020, 12(2), 457.
[http://dx.doi.org/10.3390/nu12020457] [PMID: 32059369]
[81]
Araújo, J.R.; Gonçalves, P.; Martel, F. Chemopreventive effect of dietary polyphenols in colorectal cancer cell lines. Nutr. Res., 2011, 31(2), 77-87.
[http://dx.doi.org/10.1016/j.nutres.2011.01.006] [PMID: 21419311]
[82]
Gupta, A.; Singh, A.K.; Loka, M.; Pandey, A.K.; Bishayee, A. Ferulic acid-mediated modulation of apoptotic signaling pathways in cancer. Adv. Protein Chem. Struct. Biol., 2021, 125, 215-257.
[http://dx.doi.org/10.1016/bs.apcsb.2020.12.005] [PMID: 33931140]
[83]
Kushwaha, P.P.; Kumar, R.; Neog, P.R.; Behara, M.R.; Singh, P.; Kumar, A.; Prajapati, K.S.; Singh, A.K.; Shuaib, M.; Sharma, A.K.; Pandey, A.K.; Kumar, S. Characterization of phytochemicals and validation of antioxidant and anticancer activity in some Indian polyherbal ayurvedic products. Vegetos, 2021, 34(2), 286-299.
[http://dx.doi.org/10.1007/s42535-021-00205-1]
[84]
Lin, J.P.; Yang, J.S.; Lin, J.J.; Lai, K.C.; Lu, H.F.; Ma, C.Y.; Sai-Chuen Wu, R.; Wu, K.C.; Chueh, F.S.; Gibson Wood, W.; Chung, J.G. Rutin inhibits human leukemia tumor growth in a murine xenograft model in vivo. Environ. Toxicol., 2012, 27(8), 480-484.
[http://dx.doi.org/10.1002/tox.20662] [PMID: 21254320]
[85]
Chen, H.; Miao, Q.; Geng, M.; Liu, J.; Hu, Y.; Tian, L.; Pan, J.; Yang, Y. Anti-tumor effect of rutin on human neuroblastoma cell lines through inducing G2/M cell cycle arrest and promoting apoptosis. ScientificWorldJournal, 2013, 2013269165.
[http://dx.doi.org/10.1155/2013/269165] [PMID: 24459422]
[86]
Muraoka, K.; Shimizu, K.; Sun, X.; Tani, T.; Izumi, R.; Miwa, K.; Yamamoto, K. Flavonoids exert diverse inhibitory effects on the activation of NF-kappaB. Transplant. Proc., 2002, 34(4), 1335-1340.
[http://dx.doi.org/10.1016/S0041-1345(02)02795-1] [PMID: 12072354]
[87]
Rabišková, M.; Bautzová, T.; Gajdziok, J. Dvořáčková, K.; Lamprecht, A.; Pellequer, Y.; Spilková, J. Coated chitosan pellets containing rutin intended for the treatment of inflammatory bowel disease: In vitro characteristics and in vivo evaluation. Int. J. Pharm., 2012, 422(1-2), 151-159.
[http://dx.doi.org/10.1016/j.ijpharm.2011.10.045] [PMID: 22079717]
[88]
Ragheb, S.R.; El Wakeel, L.M.; Nasr, M.S.; Sabri, N.A. Impact of Rutin and Vitamin C combination on oxidative stress and glycemic control in patients with type 2 diabetes. Clin. Nutr. ESPEN, 2020, 35, 128-135.
[http://dx.doi.org/10.1016/j.clnesp.2019.10.015] [PMID: 31987106]
[89]
Corsale, I.; Carrieri, P.; Martellucci, J.; Piccolomini, A.; Verre, L.; Rigutini, M.; Panicucci, S. Flavonoid mixture (diosmin, troxerutin, rutin, hesperidin, quercetin) in the treatment of I-III degree hemorroidal disease: A double-blind multicenter prospective comparative study. Int. J. Colorectal Dis., 2018, 33(11), 1595-1600.
[http://dx.doi.org/10.1007/s00384-018-3102-y] [PMID: 29934701]
[90]
Suzuki, T.; Morishita, T.; Noda, T.; Ishiguro, K. Acute and subacute toxicity studies on Rutin-rich tartary buckwheat dough in experimental animals. J. Nutr. Sci. Vitaminol. (Tokyo), 2015, 61(2), 175-181.
[http://dx.doi.org/10.3177/jnsv.61.175] [PMID: 26052149]
[91]
Tiwari, R.; Siddiqui, M.H.; Mahmood, T.; Farooqui, A.; Bagga, P.; Ahsan, F.; Shamim, A. An exploratory analysis on the toxicity & safety profile of polyherbal combination of curcumin, quercetin and rutin. Clin. Phytosci., 2020, 6(1), 82.
[http://dx.doi.org/10.1186/s40816-020-00228-2]
[92]
Jung, Y.S.; Park, J.I. Wnt signaling in cancer: Therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp. Mol. Med., 2020, 52(2), 183-191.
[http://dx.doi.org/10.1038/s12276-020-0380-6] [PMID: 32037398]
[93]
Dijksterhuis, J.P.; Petersen, J.; Schulte, G. WNT/Frizzled signalling: Receptor-ligand selectivity with focus on FZD-G protein signalling and its physiological relevance: IUPHAR Review 3. Br. J. Pharmacol., 2014, 171(5), 1195-1209.
[http://dx.doi.org/10.1111/bph.12364] [PMID: 24032637]
[94]
Reya, T.; Clevers, H. Wnt signalling in stem cells and cancer. Nature, 2005, 434(7035), 843-850.
[http://dx.doi.org/10.1038/nature03319] [PMID: 15829953]
[95]
Bourogaa, E.; Bertrand, J.; Despeaux, M.; Jarraya, R.; Fabre, N.; Payrastre, L.; Demur, C.; Fournié, J.J.; Damak, M.; Feki, A.E.; Racaud-Sultan, C. Hammada scoparia flavonoids and rutin kill adherent and chemoresistant leukemic cells. Leuk. Res., 2011, 35(8), 1093-1101.
[http://dx.doi.org/10.1016/j.leukres.2010.12.011] [PMID: 21216465]
[96]
Jiang, Y.; Zhang, Y.; Wark, L.; Ortiz, E.; Lim, S.; He, H.; Wang, W.; Medeiros, D.; Lin, D. Wolfberry water soluble phytochemicals down-regulate ER Stress biomarkers and modulate multiple signaling pathways leading to inhibition of proliferation and induction of apoptosis in Jurkat cells. J. Nutr. Food Sci., 2011, S2, S2-S001.
[PMID: 22685690]
[97]
Amado, N.G.; Fonseca, B.F.; Cerqueira, D.M.; Reis, A.H.; Simas, A.B.; Kuster, R.M.; Mendes, F.A.; Abreu, J.G. Effects of natural compounds on Xenopus embryogenesis: A potential read out for functional drug discovery targeting Wnt/β-catenin signaling. Curr. Top. Med. Chem., 2012, 12(19), 2103-2113.
[http://dx.doi.org/10.2174/156802612804910241] [PMID: 23167799]
[98]
Gupta, A.; Singh, A.K.; Kumar, R.; Ganguly, R.; Rana, H.K.; Pandey, P.K.; Sethi, G.; Bishayee, A.; Pandey, A.K. Corilagin in cancer: A critical evaluation of anticancer activities and molecular mechanisms. Molecules, 2019, 24(18), 3399.
[http://dx.doi.org/10.3390/molecules24183399] [PMID: 31546767]
[99]
Choi, K.S.; Kundu, J.K.; Chun, K.S.; Na, H.K.; Surh, Y.J. Rutin inhibits UVB radiation-induced expression of COX-2 and iNOS in hairless mouse skin: p38 MAP kinase and JNK as potential targets. Arch. Biochem. Biophys., 2014, 559, 38-45.
[http://dx.doi.org/10.1016/j.abb.2014.05.016] [PMID: 24875145]
[100]
Choi, S.; Lim, T.G.; Hwang, M.K.; Kim, Y.A.; Kim, J.; Kang, N.J.; Jang, T.S.; Park, J.S.; Yeom, M.H.; Lee, K.W. Rutin inhibits B[a]PDE-induced cyclooxygenase-2 expression by targeting EGFR kinase activity. Biochem. Pharmacol., 2013, 86(10), 1468-1475.
[http://dx.doi.org/10.1016/j.bcp.2013.08.066] [PMID: 24021351]
[101]
Singh, A.K.; Kumar, R.; Pandey, A.K. Hepatocellular carcinoma: Causes, mechanism of progression and biomarkers. Curr. Chem. Genomics Transl. Med., 2018, 12(1), 9-26.
[http://dx.doi.org/10.2174/2213988501812010009] [PMID: 30069430]
[102]
Sharma, A.K.; Kumar, S.; Chashoo, G.; Saxena, A.K.; Pandey, A.K. Cell cycle inhibitory activity of Piper longum against A549 cell line and its protective effect against metal-induced toxicity in rats. Indian J. Biochem. Biophys., 2014, 51(5), 358-364.
[PMID: 25630105]
[103]
Ravi, A.; Alvala, M.; Sama, V.; Kalle, A.M.; Irlapati, V.K.; Reddy, B.M. Anticancer activity of Pupalia lappacea on chronic myeloid leukemia K562 cells. Daru, 2012, 20(1), 86.
[http://dx.doi.org/10.1186/2008-2231-20-86] [PMID: 23351440]
[104]
Okonogi, S.; Khonkarn, R.; Mankhetkorn, S.; Unger, F.M.; Viernstein, H. Antioxidant activity and cytotoxicity of Cyrtosperma johnstonii extracts on drug sensitive and resistant leukemia and small cell lung carcinoma cells. Pharm. Biol., 2013, 51(3), 329-338.
[http://dx.doi.org/10.3109/13880209.2012.729064] [PMID: 23153173]

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