Skip to main content

Advertisement

Log in

Coexistence of Passive and Proton Antiporter-Mediated Processes in Nicotine Transport at the Mouse Blood–Brain Barrier

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

Nicotine, the main tobacco alkaloid leading to smoking dependence, rapidly crosses the blood–brain barrier (BBB) to become concentrated in the brain. Recently, it has been shown that nicotine interacts with some organic cation transporters (OCT), but their influence at the BBB has not yet been assessed in vivo. In this study, we characterized the transport of nicotine at the mouse luminal BBB by in situ brain perfusion. Its influx was saturable and followed the Michaelis–Menten kinetics (K m = 2.60 mM, V max = 37.60 nmol/s/g at pH 7.40). At its usual micromolar concentrations in the plasma, most (79%) of the net transport of nicotine at the BBB was carrier-mediated, while passive diffusion accounted for 21%. Studies on knockout mice showed that the OCT Oct1–3, P-gp, and Bcrp did not alter [3H]-nicotine transport at the BBB. Neither did inhibiting the transporters Mate1, Octn, or Pmat. The in vivo manipulation of intracellular and/or extracellular pH, the chemical inhibition profile, and the trans-stimulation experiments demonstrated that the nicotine transporter at the BBB shared the properties of the clonidine/proton antiporter. The molecular features of this proton-coupled antiporter have not yet been identified, but it also transports diphenhydramine and tramadol and helps nicotine cross the BBB at a faster rate and to a greater extent. The pharmacological inhibition of this nicotine/proton antiporter could represent a new strategy to reduce nicotine uptake by the brain and thus help curb addiction to smoking.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

REFERENCES

  1. Samaha AN, Yau WY, Yang P, Robinson TE. Rapid delivery of nicotine promotes behavioral sensitization and alters its neurobiological impact. Biol Psychiatry. 2005;57(4):351–60.

    Article  PubMed  CAS  Google Scholar 

  2. West R, Hajek P, Foulds J, Nilsson F, May S, Meadows A. A comparison of the abuse liability and dependence potential of nicotine patch, gum, spray and inhaler. Psychopharmacology (Berl). 2000;149(3):198–202.

    Article  CAS  Google Scholar 

  3. Samaha AN, Robinson TE. Why does the rapid delivery of drugs to the brain promote addiction? Trends Pharmacol Sci. 2005;26(2):82–7.

    Article  PubMed  CAS  Google Scholar 

  4. Berridge MS, Apana SM, Nagano KK, Berridge CE, Leisure GP, Boswell MV. Smoking produces rapid rise of [11C]nicotine in human brain. Psychopharmacology (Berl). 2010;209(4):383–94.

    Article  CAS  Google Scholar 

  5. Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37(1):13–25.

    Article  PubMed  CAS  Google Scholar 

  6. Tournier N, Decleves X, Saubamea B, Scherrmann JM, Cisternino S. Opioid transport by ATP-binding cassette transporters at the blood–brain barrier: implications for neuropsychopharmacology. Curr Pharm Des. 2011;17(26):2829–42.

    Article  PubMed  CAS  Google Scholar 

  7. Andre P, Debray M, Scherrmann JM, Cisternino S. Clonidine transport at the mouse blood–brain barrier by a new H+ antiporter that interacts with addictive drugs. J Cereb Blood Flow Metab. 2009;29(7):1293–304.

    Article  PubMed  CAS  Google Scholar 

  8. Fukada A, Saito H, Inui K. Transport mechanisms of nicotine across the human intestinal epithelial cell line Caco-2. J Pharmacol Exp Ther. 2002;302(2):532–8.

    Article  PubMed  CAS  Google Scholar 

  9. Fukada A, Saito H, Urakami Y, Okuda M, Inui K. Involvement of specific transport system of renal basolateral membranes in distribution of nicotine in rats. Drug Metab Pharmacokinet. 2002;17(6):554–60.

    Article  PubMed  Google Scholar 

  10. Takasato Y, Rapoport SI, Smith QR. An in situ brain perfusion technique to study cerebrovascular transport in the rat. Am J Physiol. 1984;247(3 Pt 2):H484–93.

    PubMed  CAS  Google Scholar 

  11. Dagenais C, Rousselle C, Pollack GM, Scherrmann JM. Development of an in situ mouse brain perfusion model and its application to mdr1a P-glycoprotein-deficient mice. J Cereb Blood Flow Metab. 2000;20(2):381–6.

    Article  PubMed  CAS  Google Scholar 

  12. Smith QR. Brain perfusion systems for studies of drug uptake and metabolism in the central nervous system. Pharm Biotechnol. 1996;8:285–307.

    PubMed  CAS  Google Scholar 

  13. Cattelotte J, Andre P, Ouellet M, Bourasset F, Scherrmann JM, Cisternino S. In situ mouse carotid perfusion model: glucose and cholesterol transport in the eye and brain. J Cereb Blood Flow Metab. 2008;28(8):1449–59.

    Article  PubMed  Google Scholar 

  14. Taylor CJ, Nicola PA, Wang S, Barrand MA, Hladky SB. Transporters involved in regulation of intracellular pH in primary cultured rat brain endothelial cells. J Physiol. 2006;576(Pt 3):769–85.

    Article  PubMed  CAS  Google Scholar 

  15. ChemSpider. The free chemical database. http://www.chemspider.com/Chemical-Structure.917.html. Accessed 16 June 2012.

  16. Oldendorf W, Braun L, Cornford E. pH dependence of blood–brain barrier permeability to lactate and nicotine. Stroke. 1979;10(5):577–81.

    Article  PubMed  CAS  Google Scholar 

  17. Putney Jr JW, Borzelleca JF. On the mechanisms of 14C-nicotine distribution in rat submaxillary gland in vitro. J Pharmacol Exp Ther. 1971;178(1):180–91.

    PubMed  CAS  Google Scholar 

  18. Seelig A, Gottschlich R, Devant RM. A method to determine the ability of drugs to diffuse through the blood–brain barrier. Proc Natl Acad Sci USA. 1994;91(1):68–72.

    Article  PubMed  CAS  Google Scholar 

  19. Beschiaschvili G, Seelig J. Peptide binding to lipid bilayers. Nonclassical hydrophobic effect and membrane-induced pK shifts. Biochemistry (Mosc). 1992;31(41):10044–53.

    Article  CAS  Google Scholar 

  20. Busto U, Bendayan R, Sellers EM. Clinical pharmacokinetics of non-opiate abused drugs. Clin Pharmacokinet. 1989;16(1):1–26.

    Article  PubMed  CAS  Google Scholar 

  21. Aceto MD, Awaya H, Martin BR, May EL. Antinociceptive action of nicotine and its methiodide derivatives in mice and rats. Br J Pharmacol. 1983;79(4):869–76.

    Article  PubMed  CAS  Google Scholar 

  22. Ghosheh OA, Dwoskin LP, Miller DK, Crooks PA. Accumulation of nicotine and its metabolites in rat brain after intermittent or continuous peripheral administration of [2′-(14)C]nicotine. Drug Metab Dispos. 2001;29(5):645–51.

    PubMed  CAS  Google Scholar 

  23. Doran A, Obach RS, Smith BJ, Hosea NA, Becker S, Callegari E, et al. The impact of P-glycoprotein on the disposition of drugs targeted for indications of the central nervous system: evaluation using the MDR1A/1B knockout mouse model. Drug Metab Dispos. 2005;33(1):165–74.

    Article  PubMed  CAS  Google Scholar 

  24. Linnet K, Ejsing TB. A review on the impact of P-glycoprotein on the penetration of drugs into the brain. Focus on psychotropic drugs. Eur Neuropsychopharmacol. 2008;18(3):157–69.

    Article  PubMed  CAS  Google Scholar 

  25. Manda VK, Mittapalli RK, Bohn KA, Adkins CE, Lockman PR. Nicotine and cotinine increases the brain penetration of saquinavir in rat. J Neurochem. 2010;115(6):1495–507.

    Article  PubMed  CAS  Google Scholar 

  26. Koepsell H, Lips K, Volk C. Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm Res. 2007;24(7):1227–51.

    Article  PubMed  CAS  Google Scholar 

  27. Itagaki S, Ganapathy V, Ho HT, Zhou M, Babu E, Wang J. Electrophysiological characterization of the polyspecific organic cation transporter plasma membrane monoamine transporter. Drug Metab Dispos. 2012;40(6):1138–43.

    Article  PubMed  CAS  Google Scholar 

  28. Tsuda M, Terada T, Asaka J, Ueba M, Katsura T, Inui K. Oppositely directed H+ gradient functions as a driving force of rat H+/organic cation antiporter MATE1. Am J Physiol Renal Physiol. 2007;292(2):F593–8.

    Article  PubMed  CAS  Google Scholar 

  29. Liou HH, Hsu HJ, Tsai YF, Shih CY, Chang YC, Lin CJ. Interaction between nicotine and MPTP/MPP+ in rat brain endothelial cells. Life Sci. 2007;81(8):664–72.

    Article  PubMed  CAS  Google Scholar 

  30. Dickens D, Owen A, Alfirevic A, Giannoudis A, Davies A, Weksler B, et al. Lamotrigine is a substrate for OCT1 in brain endothelial cells. Biochem Pharmacol. 2012;83(6):805–14.

    Article  PubMed  CAS  Google Scholar 

  31. André P, Saubaméa B, Cochois-Guégan V, Marie-Claire C, Cattelotte J, Smirnova M, et al. Transport of biogenic amine neurotransmitters at the mouse blood–retina and blood–brain barriers by uptake1 and uptake2. J Cereb Blood Flow Metab. 2012;1989–2001.

  32. Smith QR. Transport of glutamate and other amino acids at the blood–brain barrier. J Nutr. 2000;130(4S Suppl):1016S–22S.

    PubMed  CAS  Google Scholar 

  33. Okura T, Hattori A, Takano Y, Sato T, Hammarlund-Udenaes M, Terasaki T, et al. Involvement of the pyrilamine transporter, a putative organic cation transporter, in blood–brain barrier transport of oxycodone. Drug Metab Dispos. 2008;36(10):2005–13.

    Article  PubMed  CAS  Google Scholar 

  34. Fischer W, Bernhagen J, Neubert RH, Brandsch M. Uptake of codeine into intestinal epithelial (Caco-2) and brain endothelial (RBE4) cells. Eur J Pharm Sci. 2010;41(1):31–42.

    Article  PubMed  CAS  Google Scholar 

  35. Sadiq MW, Borgs A, Okura T, Shimomura K, Kato S, Deguchi Y, et al. Diphenhydramine active uptake at the blood–brain barrier and its interaction with oxycodone in vitro and in vivo. J Pharm Sci. 2011;100(9):3912–23.

    Article  PubMed  CAS  Google Scholar 

  36. Takami K, Saito H, Okuda M, Takano M, Inui KI. Distinct characteristics of transcellular transport between nicotine and tetraethylammonium in LLC-PK1 cells. J Pharmacol Exp Ther. 1998;286(2):676–80.

    PubMed  CAS  Google Scholar 

  37. Zevin S, Schaner ME, Giacomini KM. Nicotine transport in a human choriocarcinoma cell line (JAR). J Pharm Sci. 1998;87(6):702–6.

    Article  PubMed  CAS  Google Scholar 

  38. Fischer W, Metzner L, Hoffmann K, Neubert RH, Brandsch M. Substrate specificity and mechanism of the intestinal clonidine uptake by Caco-2 cells. Pharm Res. 2006;23(1):131–7.

    Article  PubMed  CAS  Google Scholar 

  39. Kuwayama K, Inoue H, Kanamori T, Tsujikawa K, Miyaguchi H, Iwata Y, et al. Uptake of 3,4-methylenedioxymethamphetamine and its related compounds by a proton-coupled transport system in Caco-2 cells. Biochim Biophys Acta. 2008;1778(1):42–50.

    Article  PubMed  CAS  Google Scholar 

  40. Mizuuchi H, Katsura T, Ashida K, Hashimoto Y, Inui K. Diphenhydramine transport by pH-dependent tertiary amine transport system in Caco-2 cells. Am J Physiol Gastrointest Liver Physiol. 2000;278(4):G563–9.

    PubMed  CAS  Google Scholar 

  41. Kanaan M, Daali Y, Dayer P, Desmeules J. Uptake/efflux transport of tramadol enantiomers and O-desmethyl-tramadol: focus on P-glycoprotein. Basic Clin Pharmacol Toxicol. 2009;105(3):199–206.

    Article  PubMed  CAS  Google Scholar 

  42. Suzuki T, Ohmuro A, Miyata M, Furuishi T, Hidaka S, Kugawa F, et al. Involvement of an influx transporter in the blood–brain barrier transport of naloxone. Biopharm Drug Dispos. 2010;31(4):243–52.

    PubMed  CAS  Google Scholar 

Download references

ACKNOWLEDGMENTS

We thank Dr. Alfred H. Schinkel for supplying the knockout mice, GSK for generously supplying GF120918, and Novartis for generously supplying PSC833. We thank Dr. S. Rasika for editing the English text.

Conflict of Interest

No conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Salvatore Cisternino.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cisternino, S., Chapy, H., André, P. et al. Coexistence of Passive and Proton Antiporter-Mediated Processes in Nicotine Transport at the Mouse Blood–Brain Barrier. AAPS J 15, 299–307 (2013). https://doi.org/10.1208/s12248-012-9434-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12248-012-9434-6

KEY WORDS

Navigation