Species Differences on Intestinal CYP Expression
Published: 2023-02-28
Page: 19-29
Issue: 2023 - Volume 6 [Issue 1]
Rupali Ghosh
Department of Pharmacy, East West University, Dhaka, Bangladesh.
Tirtha Nandi *
Clinical Pharmacology and AMP, Pharmacometrics (CPPM), Alnylam Pharmaceuticals, USA.
*Author to whom correspondence should be addressed.
Abstract
The CYP450 enzyme family is most responsible for the biotransformation of endobiotic and exobiotic. Eventually, this family will consist of the most abundant drug-metabolizing enzymes. Though the liver is the major metabolizing organ for drugs and xenobiotics, orally administered drugs can be pre-metabolized before reaching systemic circulation. This metabolism takes place in the small intestine, which is the second-largest metabolizing organ after the liver. Because intestinal cells from various laboratory animals can be examined to predict the pre-systemic metabolism of orally administered drugs, this study hopes to shed light on species differences in intestinal gene expression. The most abundant human CYP subfamilies were CYP3A, and the most highly expressed isoform was CYP3A4, which was completely mimicked by monkeys’ intestinal CYP expression. CYP1A1 was expressed in all species' intestines except dogs. Other commonly expressed isoforms for all species were CYP1B1, CYP2E1, CYP2J2, and CYP3A5. Humans, mice, and monkeys have shown the most similarities. The homologies of CYP1A1 and CYP1A2 were found to be identical in monkeys, but substrate specificity varied. Humans have interindividual variation in the expression of enzymes, which is not an attributing problem for other species. In all the species studied, the distribution of enzymes varies with the different segments of the intestine, and the level of protein expression also varies while reaching distal regions. Further summarization of CYP expression in different segments of the small intestines of humans will help in pharmacokinetic studies.
Keywords: CYP450, small intestine, liver, human, monkey, mouse, pharmacokinetics, CYP3A4
How to Cite
Downloads
References
Ghosh R, Nayan MIH, Mitu MM, Nandi T. Common Approaches of Cytochrome P450 (CYP) Induction Assays. International Blood Research & Reviews. 2023;14(1): 6-14.
US-FDA. In Vitro Drug Interaction Studies — Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions
Guidance for Industry. In: Services USDoHaH, editor. Center for Drug Evaluation and Research (CDER)2020.
Burkina V, Rasmussen MK, Pilipenko N, Zamaratskaia G. Comparison of xenobiotic-metabolising human, porcine, rodent, and piscine cytochrome P450. Toxicology. 2017;375:10-27.
Epub 2016/11/26. DOI: 10.1016/j.tox.2016.11.014. PubMed PMID: 27884721
Xie F, Ding X, Zhang QY. An update on the role of intestinal cytochrome P450 enzymes in drug disposition. Acta Pharm Sin B. 2016;6(5):374-83. Epub 2016/10/07. DOI: 10.1016/j.apsb.2016.07.012. PubMed PMID: 27709006; PubMed Central PMCID: PMCPMC5045550.
Chovan JP, Ring SC, Yu E, Baldino JP. Cytochrome P450 probe substrate metabolism kinetics in Sprague Dawley rats. Xenobiotica. 2007;37(5):459-73.
Nandi T. Importance of Sufficient Time-Points For Efficient Pharmacokinetic (Pk) Compartmental Modeling. International Journal of Applied Pharmaceutics. 2023; 15(1):87-92.
Nandi T. Time-Dependent Inactivation Of Intestinal Cytochrome P450s And Its Impact On Systemic Bioavailability. 2022.
Nandi T, Korzekwa K, Nagar S, editors. In-vitro enzyme kinetics of NCD metabolism to DNCD in RLM and RIM2022.
Nandi T, Korzekwa K, Nagar S, editors. In vitro midazolam metabolism and CYP atypical kinetics in SD rat microsomes; 2021.
Nandi T, Korzekwa K, Nagar S, editors. Inclusion of intestinal metabolism in a continuous intestinal absorption model to predict clinical pharmacokinetics of oral midazolam2022.
Ramsden D, Wu JT, Zerler B, Iqbal S, Jiang J, Clausen V, et al. In Vitro Drug-Drug Interaction Evaluation of GalNAc Conjugated siRNAs Against CYP450 Enzymes and Transporters. Drug Metab Dispos. 2019;47(10):1183-94.
Epub 2019/07/05. DOI: 10.1124/dmd.119.087098. PubMed PMID: 31270142.
Nakanishi T, Tamai I. Interaction of Drug or Food with Drug Transporters in Intestine and Liver. Curr Drug Metab. 2015;16(9):753-64. Epub 2015/12/04. DOI:10.2174/138920021609151201113537. PubMed PMID: 26630906.
Galetin A, Gertz M, Houston JB. Contribution of intestinal cytochrome p450-mediated metabolism to drug-drug inhibition and induction interactions. Drug Metab Pharmacokinet. 2010;25(1):28-47. Epub 2010/03/09. DOI: 10.2133/dmpk.25.28. PubMed PMID: 20208387.
Galetin A, Gertz M, Houston JB. Potential role of intestinal first-pass metabolism in the prediction of drug-drug interactions. Expert Opin Drug Metab Toxicol. 2008;4(7):909-22. Epub 2008/07/16. DOI: 10.1517/17425255.4.7.909. PubMed PMID: 18624679.
Nelson DR. Cytochrome P450 Nomenclature, 2004. Cytochrome P450 protocols: Springer; 2006. p. 1-10.
Nelson DR, Zeldin DC, Hoffman SMG, Maltais LJ, Wain HM, Nebert DW. Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics and Genomics. 2004;14(1):1-18.
Zhou Z-W, Zhou S-F. Application of mechanism-based CYP inhibition for predicting drug–drug interactions. Expert opinion on drug metabolism & toxicology. 2009;5(6):579-605.
Zhou SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Curr Drug Metab. 2008;9(4):310-22. Epub 2008/05/14. doi: 10.2174/138920008784220664. PubMed PMID: 18473749.
Venkatakrishnan K, von Moltke LL, Harmatz JS, Crespi CL, Greenblatt DJ. Comparison between cytochrome P450 (CYP) content and relative activity approaches to scaling from cDNA-expressed CYPs to human liver microsomes: ratios of accessory proteins as sources of discrepancies between the approaches. Drug metabolism and disposition. 2000;28(12):1493-504.
Martignoni M, Groothuis GMM, de Kanter R. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert opinion on drug metabolism & toxicology. 2006;2(6): 875-94.
Nandi T. Importance of sufficient time-points for efficient pharmacokinetic (PK) compartmental modeling. International Journal of Applied Pharmaceutics. 2023;15(1).
Fedi A, Vitale C, Ponschin G, Ayehunie S, Fato M, Scaglione S. In vitro models replicating the human intestinal epithelium for absorption and metabolism studies: A systematic review. Journal of Controlled Release. 2021;335:247-68.
Horspool AM, Wang T, Scaringella Y-S, Taub ME, Chan TS. Human liver microsomes immobilized on magnetizable beads: a novel approach to study in vitro drug metabolism. Drug Metabolism and Disposition. 2020;48(8):645-54.
Humphreys SC, Thayer MB, Lade JM, Wu B, Sham K, Basiri B, et al. Plasma and Liver Protein Binding of N-Acetylgalactosamine-Conjugated Small Interfering RNA. Drug Metab Dispos. 2019;47(10):1174-82.
Epub 2019/05/18. DOI: 10.1124/dmd.119.086967. PubMed PMID: 31097425.
Kang W, Podtelezhnikov AA, Tanis KQ, Pacchione S, Su M, Bleicher KB, et al. Development and application of a transcriptomic signature of bioactivation in an advanced in vitro liver model to reduce drug-induced liver injury risk early in the pharmaceutical pipeline. Toxicological Sciences. 2020;177(1):121-39.
Kratochwil NA, Meille C, Fowler S, Klammers F, Ekiciler A, Molitor B, et al. Metabolic profiling of human long-term liver models and hepatic clearance predictions from in vitro data using nonlinear mixed-effects modeling. The AAPS journal. 2017;19(2):534-50.
Martignoni M. Species and strain differences in drug metabolism in liver and intestine: University Library Groningen][Host]; 2006.
Ito K, Houston JB. Prediction of human drug clearance from in vitro and preclinical data using physiologically based and empirical approaches. Pharm Res. 2005;22(1):103-12. Epub 2005/03/18. doi: 10.1007/s11095-004-9015-1. PubMed PMID: 15771236.
Busse D, Leandersson S, Amberntsson S, Darnell M, Hilgendorf C. Industrial approach to determine the relative contribution of seven major UGT isoforms to hepatic glucuronidation. Journal of pharmaceutical sciences. 2020;109(7):2309-20.
Camenisch G, Umehara Ki. Predicting human hepatic clearance from in vitro drug metabolism and transport data: a scientific and pharmaceutical perspective for assessing drug–drug interactions. Biopharmaceutics & drug disposition. 2012;33(4):179-94.
Lušin TT, Mrhar A, Stieger B, Kullak-Ublick GA, Marc J, Ostanek B, et al. Influence of hepatic and intestinal efflux transporters and their genetic variants on the pharmacokinetics and pharmacodynamics of raloxifene in osteoporosis treatment. Translational research. 2012;160(4): 298-308.
Hallifax D, Houston JB. Evaluation of hepatic clearance prediction using in vitro data: emphasis on fraction unbound in plasma and drug ionisation using a database of 107 drugs. Journal of pharmaceutical sciences. 2012; 101(8):2645-52.
Chiba M, Ishii Y, Sugiyama Y. Prediction of hepatic clearance in human from in vitro data for successful drug development. The AAPS journal. 2009;11(2):262-76.
Galetin A, Houston JB. Intestinal and hepatic metabolic activity of five cytochrome P450 enzymes: impact on prediction of first-pass metabolism. Journal of Pharmacology and Experimental Therapeutics. 2006;318(3):1220-9.
Naritomi Y, Terashita S, Kimura S, Suzuki A, Kagayama A, Sugiyama Y. Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans. Drug metabolism and disposition. 2001;29(10):1316-24.
Yoshida K, Maeda K, Sugiyama Y. Hepatic and intestinal drug transporters: prediction of pharmacokinetic effects caused by drug-drug interactions and genetic polymorphisms. Annual review of pharmacology and toxicology. 2013;53: 581-612.
Gertz M, Harrison A, Houston JB, Galetin A. Prediction of human intestinal first-pass metabolism of 25 CYP3A substrates from in vitro clearance and permeability data. Drug Metabolism and Disposition. 2010;38(7):1147-58.
Galetin A, Hinton LK, Burt H, Obach RS, Houston JB. Maximal inhibition of intestinal first-pass metabolism as a pragmatic indicator of intestinal contribution to the drug-drug interactions for CYP3A4 cleared drugs. Current drug metabolism. 2007;8(7):685-93.
Bruyere A, Decleves X, Bouzom F, Ball K, Marques C, Treton X, et al. Effect of variations in the amounts of P-glycoprotein (ABCB1), BCRP (ABCG2) and CYP3A4 along the human small intestine on PBPK models for predicting intestinal first pass. Molecular pharmaceutics. 2010;7(5):1596-607.
Kato M, Chiba K, Hisaka A, Ishigami M, Kayama M, Mizuno N, et al. The intestinal first-pass metabolism of substrates of CYP3A4 and P-glycoprotein—quantitative analysis based on information from the literature. Drug metabolism and pharmacokinetics. 2003;18(6):365-72.
Paine MF, Shen DD, Kunze KL, Perkins JD, Marsh CL, McVicar JP, et al. First‐pass metabolism of midazolam by the human intestine. Clinical Pharmacology & Therapeutics. 1996;60(1):14-24.
Porat D, Dahan A. Active intestinal drug absorption and the solubility-permeability interplay. International journal of pharmaceutics. 2018;537(1-2):84-93.
Pang KS. Modeling of intestinal drug absorption: roles of transporters and metabolic enzymes (for the Gillette Review Series). Drug metabolism and disposition. 2003;31(12):1507-19.
Kaminsky LS, Zhang Q-Y. The small intestine as a xenobiotic-metabolizing organ. Drug metabolism and disposition. 2003;31(12):1520-5.
Granvil CP, Yu A-M, Elizondo G, Akiyama TE, Cheung C, Feigenbaum L, et al. Expression of the human CYP3A4 gene in the small intestine of transgenic mice: in vitro metabolism and pharmacokinetics of midazolam. Drug metabolism and disposition. 2003;31(5):548-58.
Lindell M, Lang M, Lennernäs H. Expression of genes encoding for drug metabolising cytochrome P450 enzymes and P-glycoprotein in the rat small intestine; comparison to the liver. European journal of drug metabolism and pharmacokinetics. 2003;28(1):41-8.
Stoll BR, Batycky RP, Leipold HR, Milstein S, Edwards DA. A theory of molecular absorption from the small intestine. Chemical Engineering Science. 2000;55(3):473-89.
Ramdhani A, Ramdhani MA, Amin AS. Writing a Literature Review Research Paper: A step-by-step approach. International Journal of Basic and Applied Science. 2014;3(1):47-56.
Zhang Q-Y, Dunbar D, Kaminsky LS. Characterization of mouse small intestinal cytochrome P450 expression. Drug Metabolism and Disposition. 2003;31(11):1346-51.
Zhang Q-Y, Wikoff J, Dunbar D, Kaminsky L. Characterization of rat small intestinal cytochrome P450 composition and inducibility. Drug Metabolism and Disposition. 1996;24(3):322-8.
Hansen T, Borlak J, Bader A. Cytochrome P450 enzyme activity and protein expression in primary porcine enterocyte and hepatocyte cultures. Xenobiotica. 2000;30(1):27-46.
Vaessen SF, van Lipzig MM, Pieters RH, Krul CA, Wortelboer HM, van de Steeg E. Regional expression levels of drug transporters and metabolizing enzymes along the pig and human intestinal tract and comparison with Caco-2 cells. Drug metabolism and disposition. 2017;45(4):353-60.
Heikkinen AT, Friedlein A, Matondo M, Hatley OJ, Petsalo A, Juvonen R, et al. Quantitative ADME proteomics–CYP and UGT enzymes in the beagle dog liver and intestine. Pharmaceutical research. 2015;32:74-90.
Heikkinen AT, Friedlein A, Lamerz J, Jakob P, Cutler P, Fowler S, et al. Mass spectrometry-based quantification of CYP enzymes to establish in vitro/in vivo scaling factors for intestinal and hepatic metabolism in beagle dog. Pharmaceutical research. 2012;29:1832-42.
Nakanishi Y, Matsushita A, Matsuno K, Iwasaki K, Utoh M, Nakamura C, et al. Regional distribution of cytochrome p450 mRNA expression in the liver and small intestine of cynomolgus monkeys. Drug metabolism and pharmacokinetics. 2010;25(3):290-7.
Nakanishi Y, Matsushita A, Matsuno K, Iwasaki K, Utoh M, Nakamura C, et al. Regional distribution of drug-metabolizing enzyme activities in the liver and small intestine of cynomolgus monkeys. Drug metabolism and pharmacokinetics. 2011;26(3):288-94.
Emoto C, Yoda N, Uno Y, Iwasaki K, Umehara K, Kashiyama E, et al. Comparison of p450 enzymes between cynomolgus monkeys and humans: p450 identities, protein contents, kinetic parameters, and potential for inhibitory profiles. Current drug metabolism. 2013;14(2):239-52.
Thelen K, Dressman JB. Cytochrome P450-mediated metabolism in the human gut wall. Journal of pharmacy and pharmacology. 2009;61(5):541-58.
Zhang Q-Y, Dunbar D, Ostrowska A, Zeisloft S, Yang J, Kaminsky LS. Characterization of human small intestinal cytochromes P-450. Drug Metabolism and Disposition. 1999;27(7):804-9.
Obach RS, Zhang Q-Y, Dunbar D, Kaminsky LS. Metabolic characterization of the major human small intestinal cytochrome p450s. Drug Metabolism and Disposition. 2001;29(3):347-52.
Grangeon A, Clermont V, Barama A, Gaudette F, Turgeon J, Michaud V. Determination of CYP450 expression levels in the human small intestine by mass spectrometry- based targeted proteomics. International Journal of Molecular Sciences. 2021;22(23): 12791.