Planta Med 2010; 76(4): 331-338
DOI: 10.1055/s-0029-1186153
Pharmacology
Original Papers
© Georg Thieme Verlag KG Stuttgart · New York

Potent in vitro Inhibition of CYP3A4 and P-Glycoprotein by Rhodiola rosea

Bent H. Hellum1 , Anita Tosse2 , Kathrine Hoybakk1 , Mette Thomsen2 , Jens Rohloff3 , Odd Georg Nilsen1
  • 1Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
  • 2Norwegian Institute for Agricultural and Environmental Research, Bioforsk Arable Crops Division, Apelsvoll, Norway
  • 3Department of Biology, Faculty of Natural Sciences and Technology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
Further Information

Publication History

received June 23, 2009 revised August 18, 2009

accepted August 24, 2009

Publication Date:
29 September 2009 (online)

Abstract

Six clones of Rhodiola rosea, obtained from plants originating from widely different areas in Norway, were investigated for their in vitro inhibitory potential on CYP3A4-mediated metabolism and P‐gp efflux transport activity. Presumed active constituents in the ethanol extracts of the different clones were quantified. C‐DNA baculovirus expressed CYP3A4 and Caco-2 cells were used for inhibitory assays, and as positive control inhibitors ketoconazole and verapamil were applied, respectively. A validated HPLC methodology was used to quantify the formation of 6-β‐OH-testosterone and scintillation counting was used to quantify the transport of 3H-digoxin in Caco-2 cells. All clones showed potent inhibition of CYP3A4 and P‐gp activities, with IC50 values ranging from 1.7 to 3.1 µg/mL and from 16.7 to 51.7 µg/mL, respectively, being below that reported for other herbs and some known classic drug inhibitors, such as St. John's wort and fluoxetine. Rhodiola rosea might thus be a candidate for clinically relevant drug interactions. The concentration of presumed biologically active constituents in the different clones varied considerably, but this variation was not related to the clones' inhibitory potential on CYP3A4 or P‐gp activities. Other constituents might thus be responsible for the observed inhibitory properties. The place of origin seemed to be of minor importance for CYP3A4 or P‐gp inhibition.

References

  • 1 Ming D S, Hillhouse B J, Guns E S, Eberding A, Xie S, Vimalanathan S, Towers G H. Bioactive compounds from Rhodiola rosea (Crassulaceae).  Phytother Res. 2005;  19 740-743
  • 2 Spasov A A, Wikman G K, Mandrikov V B, Mironova I A, Neumoin V V. A double-blind, placebo-controlled pilot study of the stimulating and adaptogenic effect of Rhodiola rosea SHR-5 extract on the fatigue of students caused by stress during an examination period with a repeated low-dose regimen.  Phytomedicine. 2000;  7 85-89
  • 3 Mattioli L, Perfumi M. Rhodiola rosea L. extract reduces stress- and CRF-induced anorexia in rats.  J Psychopharmacol. 2007;  21 742-750
  • 4 Brown R P, Gerbarg P L, Ramazanov Z. Rhodiola rosea. A phytochemical overview.  Herbalgram. 2002;  56 40-52
  • 5 Bystritsky A, Kerwin L, Feusner J D. A pilot study of Rhodiola rosea (Rhodax) for generalized anxiety disorder (GAD).  J Altern Complement Med. 2008;  14 175-180
  • 6 Roland P D, Amundstuen L. [Roseroot – statements and documentation].  Tidsskr Nor Laegeforen. 2007;  127 882-883
  • 7 Scott I M, Leduc R I, Burt A J, Marles R J, Arnason J T, Foster B C. The inhibition of human cytochrome P450 by ethanol extracts of North American botanicals.  Pharm Biol. 2006;  44 315-327
  • 8 Kucinskaite A, Poblocka-Olech L, Krauze-Baranowska M, Sznitowska M, Savickas A, Briedis V. Evaluation of biologically active compounds in roots and rhizomes of Rhodiola rosea L. cultivated in Lithuania.  Medicina (Kaunas). 2007;  43 487-494
  • 9 Southwell I A, Bourke C A. Seasonal variation in hypericin content of Hypericum perforatum L. (St. John's wort).  Phytochemistry. 2001;  56 437-441
  • 10 de los Reyes G C, Koda R T. Determining hyperforin and hypericin content in eight brands of St. John's wort.  Am J Health Syst Pharm. 2002;  59 545-547
  • 11 Ortiz de Montellano P R. Cytochrome P450. Structure, mechanism and biochemistry. New York; Kluwer Academic/Plenum Publishers 2005
  • 12 Hellum B H, Hu Z, Nilsen O G. The induction of CYP1A2, CYP2D6 and CYP3A4 by six trade herbal products in cultured primary human hepatocytes.  Basic Clin Pharmacol Toxicol. 2007;  100 23-30
  • 13 Gaudineau C, Beckerman R, Welbourn S, Auclair K. Inhibition of human P450 enzymes by multiple constituents of the Ginkgo biloba extract.  Biochem Biophys Res Commun. 2004;  318 1072-1078
  • 14 Ameer B, Weintraub R A. Drug interactions with grapefruit juice.  Clin Pharmacokinet. 1997;  33 103-121
  • 15 Gurley B J, Gardner S F, Hubbard M A, Williams D K, Gentry W B, Khan I A, Shah A. In vivo effects of goldenseal, kava kava, black cohosh, and valerian on human cytochrome P450 1A2, 2D6, 2E1, and 3A4/5 phenotypes.  Clin Pharmacol Ther. 2005;  77 415-426
  • 16 Al-Shawi M K, Omote H. The remarkable transport mechanism of P-glycoprotein: a multidrug transporter.  J Bioenerg Biomembr. 2005;  37 489-496
  • 17 Flanagan D. Understanding the grapefruit-drug interaction.  Gen Dent. 2005;  53 282-285
  • 18 Elameen A, Klemsdal S S, Dragland S, Fjellheim S, Rognli O A. Genetic diversity in a germplasm collection of roseroot (Rhodiola rosea) in Norway studied by AFLP.  Biochem Syst Ecol. 2008;  36 706-715
  • 19 Hellum B H, Nilsen O G. In vitro inhibition of CYP3A4 metabolism and P-glycoprotein-mediated transport by trade herbal products.  Basic Clin Pharmacol Toxicol. 2008;  102 466-475
  • 20 Engdal S, Nilsen O G. In vitro inhibition of CYP3A4 by herbal remedies frequently used by cancer patients.  Phytother Res. 2009;  23 906-912
  • 21 Brodie R, Hill H. Validation issues arising from the new FDA guidance for industry on bioanalytical method validation.  Chromatographia. 2002;  55 S91-S94
  • 22 Engdal S, Nilsen O G. Inhibition of P-glycoprotein in Caco-2 cells: effects of herbal remedies frequently used by cancer patients.  Xenobiotica. 2008;  38 559-573
  • 23 Oppiah-Opong R, Commandeur J N, Axson C, Vermeulen N P. Interactions between cytochromes P450, glutathione S-transferases and Ghanaian medicinal plants.  Food Chem Toxicol. 2008;  46 3598-3603
  • 24 Liu K H, Kim M J, Jeon B H, Shon J H, Cha I J, Cho K H, Lee S S, Shin J G. Inhibition of human cytochrome P450 isoforms and NADPH-CYP reductase in vitro by 15 herbal medicines, including Epimedii herba.  J Clin Pharm Ther. 2006;  31 83-91
  • 25 Zou L, Harkey M R, Henderson G L. Effects of herbal components on cDNA-expressed cytochrome P450 enzyme catalytic activity.  Life Sci. 2002;  71 1579-1589
  • 26 Eagling V A, Wiltshire H, Whitcombe I W, Back D J. CYP3A4-mediated hepatic metabolism of the HIV-1 protease inhibitor saquinavir in vitro.  Xenobiotica. 2002;  32 1-17
  • 27 McConn D J, Lin Y S, Allen K, Kunze K L, Thummel K E. Differences in the inhibition of cytochromes P450 3A4 and 3A5 by metabolite-inhibitor complex-forming drugs.  Drug Metab Dispos. 2004;  32 1083-1091
  • 28 Ridtitid W, Wongnawa M, Mahatthanatrakul W, Raungsri N, Sunbhanich M. Ketoconazole increases plasma concentrations of antimalarial mefloquine in healthy human volunteers.  J Clin Pharm Ther. 2005;  30 285-290
  • 29 Rengelshausen J, Banfield M, Riedel K D, Burhenne J, Weiss J, Thomsen T, Walter-Sack I, Haefeli W E, Mikus G. Opposite effects of short-term and long-term St. John's wort intake on voriconazole pharmacokinetics.  Clin Pharmacol Ther. 2005;  78 25-33
  • 30 Yoshida N, Takagi A, Kitazawa H, Kawakami J, Adachi I. Inhibition of P-glycoprotein-mediated transport by extracts of and monoterpenoids contained in Zanthoxyli fructus.  Toxicol Appl Pharmacol. 2005;  209 167-173
  • 31 Keogh J P, Kunta J R. Development, validation and utility of an in vitro technique for assessment of potential clinical drug-drug interactions involving P-glycoprotein.  Eur J Pharm Sci. 2006;  27 543-554
  • 32 Pauli-Magnus C, Rekersbrink S, Klotz U, Fromm M F. Interaction of omeprazole, lansoprazole and pantoprazole with P-glycoprotein.  Naunyn Schmiedebergs Arch Pharmacol. 2001;  364 551-557
  • 33 Igel S, Drescher S, Murdter T, Hofmann U, Heinkele G, Tegude H, Glaeser H, Brenner S S, Somogyi A A, Omari T, Schafer C, Eichelbaum M, Fromm M F. Increased absorption of digoxin from the human jejunum due to inhibition of intestinal transporter-mediated efflux.  Clin Pharmacokinet. 2007;  46 777-785
  • 34 Kiley C A, Cragin D J, Roth B J. Omeprazole-associated digoxin toxicity.  South Med J. 2007;  100 400-402
  • 35 Rautio J, Humphreys J E, Webster L O, Balakrishnan A, Keogh J P, Kunta J R, Serabjit-Singh C J, Polli J W. In vitro P-glycoprotein inhibition assays for assessment of clinical drug interaction potential of new drug candidates: a recommendation for probe substrates.  Drug Metab Dispos. 2006;  34 786-792
  • 36 Fromm M F, Kim R B, Stein C M, Wilkinson G R, Roden D M. Inhibition of P-glycoprotein-mediated drug transport: a unifying mechanism to explain the interaction between digoxin and quinidine.  Circulation. 1999;  99 552-557
  • 37 Artursson P, Palm K, Luthman K. Caco-2 monolayers in experimental and theoretical predictions of drug transport.  Adv Drug Deliv Rev. 2001;  46 27-43
  • 38 Collett A, Tanianis-Hughes J, Carlson G L, Harwood M D, Warhurst G. Comparison of P-glycoprotein-mediated drug-digoxin interactions in Caco-2 with human and rodent intestine: relevance to in vivo prediction.  Eur J Pharm Sci. 2005;  26 386-393
  • 39 Lowes S, Cavet M E, Simmons N L. Evidence for a non-MDR1 component in digoxin secretion by human intestinal Caco-2 epithelial layers.  Eur J Pharmacol. 2003;  458 49-56
  • 40 Hillhouse B J, Ming D S, French C J, Towers N G H. Acetylcholine esterase inhibitors in Rhodiola rosea.  Pharm Biol. 2004;  42 68-72
  • 41 Majewska A, Hoser G, Furmanowa M, Urbanska N, Pietrosiuk A, Zobel A, Kuras M. Antiproliferative and antimitotic effect, S phase accumulation and induction of apoptosis and necrosis after treatment of extract from Rhodiola rosea rhizomes on HL-60 cells.  J Ethnopharmacol. 2006;  103 43-52

Dr. Bent H. Hellum

Department of Cancer Research and Molecular Medicine
Medical Technical Research Centre

Olav Kyrresgt. 9

7489 Trondheim

Norway

Phone: + 47 73 59 89 89

Fax: + 47 73 59 86 55

Email: bent.h.hellum@ntnu.no

>