Copyright #ERS Journals Ltd 2001 Eur Respir J 2001; 18: Suppl. 32, 122s–126s European Respiratory Journal Printed in UK – all rights reserved ISSN 0904-1850 ISBN 1-904097-01-4

SELECTED REPORT

Expression of xenobiotic-metabolizing enzymes in human pulmonary tissue: possible role in susceptibility for ILD

J. Hukkanen*, O. Pelkonen*, H. Raunio*,#

Expression of xenobiotic-metabolizing enzymes in human pulmonary tissue: possible *Dept of Pharmacology and Toxico- # logy, University of Oulu, Oulu, and role in susceptibility for ILD. J. Hukkanen, O. Pelkonen, H. Raunio. ERS Journals # Ltd 2001. UniversityofKuopio,Kuopio,Finland. ABSTRACT: The lung is a major target for all inhaled toxicants. Many inhaled Correspondence: O. Pelkonen, Dept of chemicals are not hazardous as such, but are biotransformed to reactive intermediates. Pharmacology and Toxicology, Uni- Therefore, the pathogenesis of interstitial and other lung diseases is intimately linked to versity of Oulu, POB 5000, FIN-90014 exposure to environmental and other chemicals, which may be causative or modifying Oulu, Finland. factors in the cellular pathways and mechanisms mediating oxidative stress and cell Fax: 358 85375247 protection in the pulmonary tissue. Several different xenobiotic-metabolizing (CYP) and phase II Keywords: Cytochrome P450 enzymes (i.e. conjugation enzymes including several transferases) are present in the cytochrome P450 enzymes human lung and lung-derived cell lines, possibly contributing to in situ activation and xenobiotic-metabolizing enzymes inactivation of chemical toxicants. This paper describes the expression and localization of individual CYP-forms in the lung. The studies in the authors9 laboratories Interindividual differences in the expression of these enzymes may contribute to the have been financially supported by the Academy of Finland and the Biomed2 risk of developing interstitial and other lung diseases initiated by agents that require programme (EUROCYP Project). metabolic activation. Eur Respir J 2001; 18: Suppl. 32, 122s–126s.

The lung is a major target for all inhaled toxicants. forms as well as to detoxify them [6]. Finding evidence Many chemicals are not hazardous as such, but are for this in humans is more difficult, but various lines biotransformed to reactive intermediates, often by of research have established that whole lung tissue, as enzymes in the cytochrome P450 (CYP) superfamily well as several cell types in the lung, possess metabolic [1]. The same chemicals are also detoxified by cataly- capacity towards numerous xenobiotics [7]. Although sis via these enzymes. For example, of the several the lung contains several enzymatic pathways capable thousands of compounds present in tobacco smoke, of xenobiotic metabolism, it is generally agreed that several have been found to undergo metabolic acti- the CYP superfamily of enzymes is the main system vation through xenobiotic-metabolizing CYP enzymes catalyzing the oxidative metabolism and metabolic [2, 3]. activation of most toxicants. However, from the Enzymes activating or detoxifying environmental susceptibility point of view, it is equally important chemicals are not the sole factors in the aetiology of to measure variations in phase II detoxifying enzymes chemical-induced lung diseases. Pro-oxidant and anti- (i.e. conjugation enzymes, such as uridine diphos- oxidant enzymes and chemicals (many xenobiotic- phate (UDP)-glucuronyl transferases and glutathione metabolizing enzymes have these same properties), as S-transferases) and pro-oxidant and antioxidant well as various repair systems, also play an impor- systems, although these areas are less well character- tant role in the aetiology or modification of diseases. ized. It is also notable that, like most of the polycyclic Their possible role in interstitial lung disease (ILD) aromatic hydrocarbons (PAH), such as benzo(a)pyr- induced by exogenous agents is outlined by NEMERY ene, the inhaled, highly lipophilic compounds have et al. [4] in this Supplement. longer retention times and higher local doses in pulmonary epithelium than less lipophilic compounds, indicating that at least these lipophilic substances are Metabolic capacity of pulmonary cells primarily site-of-entry toxicants [6].

A key question concerning organ-specific chemical toxicity is whether the actual target tissue has the Expression of cytochrome P450 forms in human lung capacity to activate (or efficiently inactivate) chem- icals [5]. Animal models show that in the case of For the most recent update on CYP enzymes, pulmonary toxicity, several target cells in the lung consult [8]. have the capacity to convert chemicals to reactive The detection of individual CYP forms in human XENOBIOTIC-METABOLIZING ENZYMES IN ILD 123s lung by conventional methods, such as and it is inducible by smoking [27]. CYP1B1 is also purification, catalytic activity studies, and Western induced by smoking in alveolar macrophages [28]. immunoblotting, has been difficult due to the low CYP2A6 mRNA was detected in bronchial epithe- abundance of CYPs in lungs [9]. With the advent of lium [17, 29], but two reports on the expression of the the reverse transcriptase-polymerase chain reaction CYP2A6 protein are contradictory [17, 18]. Studies by (RT-PCR) technology it has become possible to detect the author did not demonstrate CYP2A6 mRNA in minute amounts of messenger ribonucleic acid whole lung tissue homogenate, probably due to (mRNA) in tissue samples. RT-PCR is extremely dilution of bronchus-specific mRNA expression with sensitive and results obtained with it cannot be other cell types of the lung [30]. The expression of regarded as a direct indication of the existence of pulmonary CYP2A6 protein would be of utmost corresponding . Rather, RT-PCR is valuable interest because CYP2A6 has a crucial role in the acti- as a screening method, revealing mRNA that can vation of 4-methylnitrosamino-1,3-pyridyl-1-butanone potentially be translated to functional protein in a (NNK), the tobacco-specific procarcinogen [31]. given tissue. Conversely, absence of mRNA in RT- The CYP2B6 is expressed in human lung as a PCR analysis is a strong indication of the lack of a splicing variant called CYP2B7 [21, 27, 32] and the corresponding protein product at biologically mean- corresponding protein is also expressed [17, 33, 34]. A ingful levels. Expression of various CYP enzymes in recent immunohistochemistry study suggests a cell- human lung at mRNA and protein level is summa- specific expression of CYP2C proteins only in the rized in table 1. serous cells of bronchial glands [35]. Out of four CYP1A1 is by far the most studied human previous Western blot studies, two support [19, 36] pulmonary CYP [10]. The first report on the expres- and two oppose the expression of CYP2C proteins in human lung [37, 38]. CYP2C8 and CYP2C18 mRNAs sion of CYP1A1 mRNA in human lung was by have also been detected in the lung [17]. OMIECINSKI et al. [11] in 1990. Soon after, the There has been great interest in studying expression induction of CYP1A1 mRNA by tobacco smoking of pulmonary CYP2D6 due to its alleged role in the [12], the expression of CYP1A1 protein in human lung activation of the tobacco-specific procarcinogen NNK [13] and the localization and induction of CYP1A1 [30]. However, although it is the focus of numerous protein by tobacco smoke [14] were reported. studies [17, 19, 21, 39–41], the data on the expression CYP1A1 protein is only detected in smokers [14] of CYP2D6 in human lung is inconsistent. The and CYP1A1 expression is positively correlated with expression of pulmonary CYP2E1 mRNA and pro- aryl hydrocarbon hydroxylase (AHH) activity in tein have been established in several studies [17, 19, human lung tissue [15, 16]. 21, 29, 37, 42, 43]. CYP2E1 is an interesting CYP Another PAH-metabolizing CYP, CYP1A2, was form because it is the most active CYP enzyme in detected recently by RT-PCR and Western blot in forming oxygen radicals, causing tissue injury [44]. peripheral lung [17], but earlier reports do not corro- The expression of CYP2F1 has been detected at the borate this finding [13, 18–21]. Also, the presence of mRNA level [21, 24, 45], but there are no published CYP1B1 protein in the lung is controversial [22, 23]. results on the expression of CYP2F1 protein. Recom- CYP1B1 mRNA is expressed in human lung [24–26] binant CYP2F1 is capable of activating the pulmo- nary toxicant 3-methylindole [46]. Several studies have demonstrated the expression of Table 1. – Summary of expression of cytochrome P450s in human lung CYP3A protein in human lung [17, 19, 20, 37, 47, 48]. The main pulmonary CYP3A form is CYP3A5 CYP mRNA Protein Comments [17, 21, 48, 49]. There are no published results on the expression of CYP4B1 protein, but mRNA is 1A1 zzzzzz Smokers expressed in human lung [21, 24, 32, 50]. It has been z/- -- Nonsmokers speculated that native human CYP4B1 enzyme is not 1A2 z/- z/- functional due to inability to incorporate haeme [51]. 1B1 zzz/- Protein in alveolar In conclusion, at least CYPs 1A1 (in smokers), 2B7, macrophages 2E1 and 3A5 proteins are expressed in human lung. 2A6 zzz/- Other CYP forms are also likely to be expressed, but 2B6/7 zzzzzz z z their expression is probably very low or restricted 2C /- /- Protein in serous cells to specific cell types or individuals. Studies on the of bronchial glands 2D6 z/- z/- expression of certain CYP forms (CYP2F1 and 2E1 zzzzzz CYP4B1) are still required. 2F1 zzz 3A4 z/- z Protein in y20% of cases 3A5 zzzzz Localization of individual cytochrome P450 forms in 3A7 z/- the lung 4B1 zzz CYP: cytochrome P450; mRNA: messenger ribonucleic The cell-specific localization of individual CYP acid. --: moderate negative evidence; -: weak negative enzymes in the lung is still largely unknown, since evidence;z/-: conflicting evidence;z: weak positive evidence; there are only a handful of good immunohisto- zz: moderate positive evidence; zzz: strong positive chemical studies concerning CYP forms in human evidence. lung. It would be of great benefit to the understanding 124s J. HUKKANEN ET AL. of cell-specific toxicity to have a comprehensive alveolar macrophages may provide a model for picture of localization of different CYP forms. The investigating the relationship between the capacity to overall distribution of all CYP enzymes can be esti- activate and detoxify chemicals in an easily accessible mated from the immunohistochemical distribution of lung cell type [28]. In these cells, at least CYP1B1, reduced nicotinamide adenine dinucleotide phosphate CYP2E1 and CYP3A5 proteins are expressed, while (NADPH)-cytochrome P450 reductase, which is CYP1A1 is not [15, 28, 42, 49]. detected in bronchial and bronchiolar epithelium, Clara cells, alveolar lining cells and alveolar macro- phages [52]. Inducibility of cytochrome P450 forms in the lung According to immunohistochemistry, CYP1A1 is mainly expressed at the epithelium of the peripheral CYP1A1 is induced by tobacco smoking in human airways. CYP1A1 expression does not extend to the lung, and there is also evidence for the induction of epithelium of bronchi w1 mm in diameter and is CYP1B1 in smokers [14, 27, 28]. Information about expressed only in the lungs of smokers [14]. Alveolar induction of other pulmonary CYP forms in vivo does macrophages do not express CYP1A1 [14, 28]. not exist. However, studies in a continuously growing Induced CYP1A1 may be a precondition for the pulmonary adenocarcinoma cell line (A549) indicate development of peripheral lung cancer in smokers, as that CYP3A5 is inducible by glucocorticoids [56]. not a single case of this disease with noninducible CYP1A1, CYP1B1 and CYP3A5 participate in the CYP1A1 in the lung has been found, and, further- activation of PAHs in the tobacco smoke [10, 57, 58] more, CYP1A1 is localized in the part of the airways and thus, their induction could have practical in which peripheral cancers arise [14, 53]. A study on significance. the localization of CYP1A1 mRNA by in situ hybri- dization corresponded well with the protein data [54]. Immunohistochemical analysis with CYP3A5- Genetic differences in the pulmonary expression of specific antibody shows that CYP3A5 protein is cytochrome P450s present in all lung samples studied and is localized to the bronchial, bronchiolar and alveolar epithelium, The expression of CYP1A1 is regulated by genetic as well as endothelium and alveolar macrophages [49]. polymorphism and it has been claimed that this Compared with CYP1A1 localization, CYP3A5 hereditary variability contributes to individual sus- expression extends up to larger bronchi. The highest ceptibility to environmental chemicals [59]. Although CYP3A5 level is detected in the bronchial lung. Also, the interindividual variability of CYP3A5 expression CYP3A4 protein is found in some cell types in a in the liver and lung is rather large, several 10-fold, minority (y20%) of lung samples with CYP3A4 relative contributions of environmental and genetic specific antibody [49]. A few studies with nonspecific factors have not been elucidated. "Classical" poly- CYP3A antibody also show the same pattern of morphic enzymes CYP2D6 and CYP2C19 have not expression as CYP3A5 antibody [47, 48]. been demonstrated in the human lung. Among phase CYP2E1 is also localized to human bronchial, II enzymes, at least glutathione S-transferase M1 bronchiolar and alveolar epithelium, and alveolar (GSTM1) has been shown to be polymorphic in macrophages [33, 42, 43]. The localization of the human lungs and may be associated with differential highest expression cannot be evaluated since none of susceptibility to lung cancer [53, 60]. these studies examined both bronchial and peripheral lung. One immunohistochemical report localizes CYP2B7 to human Clara cells [33] and a recent Conclusions study by YOKOSE et al. [35] observed CYP2C protein in serous cells of bronchial glands, but not in any The patterns of constitutive and induced expression other cell type of the lung. of cytochrome P450 forms in several relevant human pulmonary cell types are being unravelled with increasing precision. However, knowledge of the Surrogate tissues functional activities and cell-specific localization of the pulmonary cytochrome P450 forms is still Various surrogate tissues have been used in human incomplete, mostly due to sensitivity problems asso- biomonitoring studies, the most popular being ciated with measurements in tissues with a low peripheral blood lymphocytes [55]. Mitogen-treated abundance of cytochrome P450 enzymes. In addition, lymphocytes display inducible CYP1A1 and forma- very little is known about the consequences of tion of benzo(a) pyrene-deoxyribonucleic acid (DNA) interindividual variations in both basal and induced adducts, and it is suggested that these parameters may pulmonary cytochrome P450 expression. Reactions correspond with those of the lung tissue in the same catalyzed by cytochrome P450 enzymes are often individual, although this issue is controversial and still "leaky", resulting in the transfer of electrons to not definitively settled [5, 9]. Alveolar macrophages, produce oxygen radicals. Cytochrome P450-catalyzed which presumably originate from peripheral blood reactions can thus produce xenobiotic metabolites and monocytes, have an important role as defence cells oxygen species capable of interfering with cell home- against inhaled particles. The expression pattern of ostasis. Thus, the role of xenobiotic metabolism CYP in alveolar macrophages resembles the catalyzed by individual pulmonary cytochrome P450 pattern found in the whole lung tissue [21]. Thus, forms in the aetiology of diseases such as interstitial XENOBIOTIC-METABOLIZING ENZYMES IN ILD 125s lung diseases, lung cancer and chronic obstructive P450IA1 and P450IA2 isozymes in lung cancer pulmonary disease, is still elusive and an intriguing patients. Environ Health Perspect 1992; 98: 179–182. subject for future studies. 16. Bartsch H, Castegnaro M, Rojas M, Camus A-M, Alexandrov K, Lang M. Expression of pulmonary cytochrome P4501A1 and carcinogen DNA adduct References formation in high risk subjects for tobacco-related lung cancer. Toxicol Lett 1992; 64–65: 477–483. 1. Nelson DR, Koymans L, Kamataki T, et al. P450 17. Mace´ K, Bowman ED, Vautravers P, Shields superfamily: update on new sequences, gene mapping, PG, Harris CC, Pfeifer AMA. Characterisation of accession numbers and nomenclature. Pharmaco- xenobiotic-metabolising enzyme expression in human genetics 1996; 6: 1–42. bronchial mucosa and peripheral lung tissue. Eur 2. Guengerich FP. Purification and characterization of J Cancer 1998; 34: 914–920. xenobiotic-metabolizing enzymes from lung tissue. 18. Shimada T, Yun C-H, Yamazaki H, Gautier J-C, Pharmacol Ther 1990; 45: 299–307. Beaune PH, Guengerich FP. Characterization of 3. Gonzalez FJ, Gelboin HV. Role of human cyto- human lung microsomal cytochrome P-4501A1 and chromes P450 in the metabolic activation of chemical its role in the oxidation of chemical carcinogens. Mol carcinogens and toxins. Drug Metab Rev 1994; 26: Pharmacol 1992; 41: 856–864. 165–183. 19. Shimada T, Yamazaki H, Mimura M, et al. Character- 4. Nemery B, Bast A, Behr J, et al. Interstitial lung ization of microsomal cytochrome P450 enzymes disease induced by exogenous agents: factors govern- involved in the oxidation of xenobiotic chemicals in ing susceptibility. Eur Respir J 2001; 18: Suppl. 32, human fetal livers and adult lungs. Drug Metab Dispos 30s–42s. 1996; 24: 515–522. 5. Raunio H, Hakkola J, Hukkanen J, et al. Expression 20. Kelly JD, Eaton DL, Guengerich FP, Coulombe RA. of xenobiotic-metabolizing CYPs in human pulmo- Aflatoxin B1 activation in human lung. Toxicol Appl nary tissue. Exp Toxic Pathol 1999; 51: 412–417. Pharmacol 1997; 144: 88–95. 6. Gerde P, Muggenburg BA, Scott GG, Lewis JL, Pyon 21. Hukkanen J, Hakkola J, Anttila S, et al. Detection of KH, Dahl AR. Local metabolism in lung airways mRNA encoding xenobiotic-metabolizing cytochrome increases the uncertainty of pyrene as a biomarker of P450s in human bronchoalveolar macrophages and polycyclic aromatic hydrocarbon exposure. Carcino- peripheral blood lymphocytes. Mol Carcinog 1997; 20: genesis 1998; 19: 493–500. 224–230. 7. Hecht SS. Tobacco smoke carcinogens and lung 22. Murray GI, Taylor MC, McFadyen MCE, et al. cancer. J Natl Cancer Inst 1999; 91: 1194–1210. Tumor-specific expression of cytochrome P450 8. Nelson D. Cytochrome P450. drnelson.utmem.edu/ CYP1B1. Cancer Res 1997; 57: 3026–3031. nelsonhomepage.html. (Last updated July 3, 2001). 23. Tang YM, Chen G-F, Thompson PA, Lin D-X, Lang 9. Raunio H, Pasanen M, Ma¨enpa¨a¨ J, Hakkola J, NP, Kadlubar FF. Development of an antipeptide Pelkonen O. Expression of extrahepatic cytochrome antibody that binds to the C-terminal region of human P450 in humans. In: Pacifici GM, Fracchia GN, eds. CYP1B1. Drug Metab Dispos 1999; 27: 274–280. Advances in Drug Metabolism in Man. Luxembourg, 24. Willey JC, Coy E, Brolly C, et al. Xenobiotic European Commission, Office for Official Publica- metabolism gene expression in human bronchial tions of the European Communities, 1995; pp. 234– epithelial and alveolar macrophage cells. Am J Respir 287. Cell Mol Biol 1996; 14: 262–271. 10. Shou M, Korzekwa KR, Crespi CL, Gonzalez FJ, 25. Shimada T, Hayes CL, Yamazaki H, et al. Activation Gelboin HV. The role of 12 cDNA-expressed human, of chemically diverse procarcinogens by human cyto- rodent and rabbit cytochromes P450 in the metabo- chrome P-450 1B1. Cancer Res 1996; 56: 2979–2984. lism of benzo[a]pyrene and benzo[a]pyrene trans-7,8- 26. Sutter TR, Tang YM, Hayes CL, et al. Complete diol. Mol Carcinog 1994; 10: 159–168. cDNA sequence of a human dioxin-inducible mRNA 11. Omiecinski CJ, Redlich CA, Costa P. Induction and identifies a new gene subfamily of a cytochrome that developmental expression of cytochrome P450IA1 maps to 2. J Biol Chem 1994; 269: messenger RNA in rat and human tissues. Cancer 13092–13099. Res 1990; 50: 4315–4321. 27. Willey JC, Coy EL, Frampton MW, et al. Quantita- 12. McLemore TL, Adelberg S, Liu MC, et al. Expression tive RT-PCR measurement of cytochromes p450 1A1, of CYP1A1 gene in patients with lung cancer: evidence 1B1, and 2B7, microsomal epoxide hydrolase, and for cigarette smoke-induced gene expression in normal NADPH oxidoreductase expression in lung cells of lung tissue and for altered gene regulation in primary smokers and nonsmokers. Am J Respir Cell Mol Biol pulmonary carcinomas. J Natl Cancer Inst 1990; 83: 1997; 17: 114–124. 1333–1339. 28. Piipari R, Savela K, Nurminen T, et al. Expression 13. Wheeler CW, Park S-S, Guenthner TM. Immuno- of CYP1A1, CYP1B1 and CYP3A, and polycyclic chemical analysis of a cytochrome P-450IA1 homo- aromatic hydrocarbon-DNA adduct formation in logue in human lung microsomes. Mol Pharmacol bronchoalveolar macrophages of smokers and non- 1990; 38: 643. smokers. Int J Cancer 2000; 86: 610–616. 14. Anttila S, Hietanen E, Vainio H, et al. Smoking and 29. Crawford EL, Weaver DA, DeMuth JP, et al. peripheral type of cancer are related to high levels of Measurement of cytochrome P450 2A6 and 2E1 gene pulmonary cytochrome P450IA in lung cancer expression in primary human bronchial epithelial cells. patients. Int J Cancer 1991; 47: 681–685. Carcinogenesis 1998; 19: 1867–1871. 15. Anttila S, Vainio H, Hietanen E, et al. Immuno- 30. Koskela S, Hakkola J, Hukkanen J, et al. Expression histochemical detection of pulmonary cytochrome of CYP2A genes in human liver and extrahepatic P450IA and metabolic activities associated with tissues. Biochem Pharmacol 1999; 57: 1407–1413. 126s J. HUKKANEN ET AL.

31. Hecht SS. Biochemistry, biology, and carcinogenicity epoxidation of naphthalene by recombinant human of tobacco-specific N-nitrosamines. Chem Res Toxicol CYP2F1 expressed in lymphoblastoid cells. Drug 1998; 11: 559–603. Metab Dispos 1999; 27: 798–803. 32. Czerwinski M, McLemore TL, Gelboin HV, Gonzalez 47. Kivisto¨ KT, Fritz P, Linder A, Friedel G, Beaune P, FJ. Quantitation of CYP2B7, CYP4B1, and CYPOR Kroemer HK. Immunohistochemical localization of messenger RNAs in normal human lung and lung cytochrome P450 3A in human pulmonary carcinomas tumors. Cancer Res 1994; 54: 1085–1091. and normal bronchial tissue. Histochemistry 1995; 33. Mori M, Tezuka F, Chiba R, et al. Atypical 103: 25–29. adenomatous hyperplasia and adenocarcinoma of 48. Kivisto¨ KT, Griese E-U, Fritz P, et al. Expression the human lung. Their heterology in form and analogy of cytochrome P4503A enzymes in human lung: a in immunohistochemical characteristics. Cancer 1996; combined RT-PCR and immunohistochemical analy- 77: 665–674. sis of normal tissue and lung tumors. Naunyn- 34. Gervot L, Rochat B, Gautier JC, et al. Human Schmiedeberg Arch Pharmacol 1996; 353: 207–212. CYP2B6: expression, inducibility and catalytic activi- 49. Anttila S, Hukkanen J, Hakkola J, et al. Expression ties. Pharmacogenetics 1999; 9: 295–306. and localization of CYP3A4 and CYP3A5 in human 35. Yokose T, Doy M, Taniguchi T, et al. Immuno- lung. Am J Respir Cell Mol Biol 1997; 16: 242–249. histochemical study of cytochrome P450 2C and 3A 50. Nhamburo PT, Gonzalez FJ, McBride OW, Gelboin in human non-neoplastic and neoplastic tissues. HV, Kimura S. Identification of a new P450 expressed Virchows Arch 1999; 434: 401–411. in human lung: complete cDNA sequence, cDNA- 36. Nakajima T, Elovaara E, Gonzalez FJ, et al. Styrene directed expression, and chromosome mapping. metabolism by cDNA-expressed human hepatic and Biochemistry 1989; 28: 8060–8066. pulmonary cytochromes P450. Chem Res Toxicol 51. Zheng Y-M, Fisher MB, Yokotani N, Fujii-Kuriyama 1994; 7: 891–896. Y, Rettie AE. Identification of a meander region 37. Wheeler CW, Wrighton SA, Guenthner TM. Detec- proline residue critical for heme binding to cyto- tion of human lung cytochrome P450 that are chrome P450: implications for the catalytic function of immunochemically related to cytochrome P450IIE1 human CYP4B1. Biochemistry 1998; 37: 12847–12851. and cytochrome P450IIIA. Biochem Pharmacol 1992; 52. Hall PM, Stupans I, Burgess W, Birkett DJ, 44: 911–917. McManus ME. Immunohistochemical localization 38. Toussaint C, Albin N, Massaad L, et al. Main drug- of NADPH-cytochrome P450 reductase in human and carcinogen-metabolizing enzyme systems in tissues. Carcinogenesis 1989; 10: 521–530. human non-small cell lung cancer and peritumoral 53. Anttila S, Hirvonen A, Husgafvel-Pursiainen K, tissues. Cancer Res 1993; 53: 4608–4612. Karjalainen A, Nurminen T, Vainio H. Combined 39. Kivisto¨ KT, Griese E-U, Stuven T, et al. Analysis of effect of CYP1A1 inducibility and GSTM1 poly- CYP2D6 expression in human lung: implications for morphism on histological type of lung cancer. the association between CYP2D6 activity and sus- Carcinogenesis 1994; 15: 1133–1135. ceptibility to lung cancer. Pharmacogenetics 1997; 7: 54. Saarikoski ST, Husgafvel-Pursiainen K, Hirvonen A, 295–302. Vainio H, Gonzalez FJ, Anttila S. Localization of 40. Huang Z, Fasco MJ, Spivack S, Kaminsky LS. CYP1A1 mRNA in human lung by in situ hybridiza- Comparisons of CYP2D messenger RNA splice tion: comparison with immunohistochemical findings. variant profiles in human lung tumors and normal Int J Cancer 1998; 77: 33–39. tissues. Cancer Res 1997; 57: 2589–2592. 55. Law MR. Genetic predisposition to cancer.Br 41. Guidice J-ML, Marez D, Sabbagh N, et al. Evidence J Cancer 1990; 61: 195–206. for CYP2D6 expression in human lung. Biochem 56. Hukkanen J, Lassila A, Pa¨iva¨rinta K, et al. Induction Biophys Res Commun 1997; 241: 79–85. and regulation of xenobiotic-metabolizing cyto- 42. Botto F, Seree E, Khyari SE, et al. Tissue-specific chrome P450s in the human A549 lung adenocarci- expression and methylation of the human CYP2E1 noma cell line. Am J Respir Cell Mol Biol 2000; 22: gene. Biochem Pharmacol 1994; 48: 1095–1103. 360–366. 43. Kivisto¨ KT, Linder A, Friedel G, et al. Immuno- 57. Roberts-Thomson SJ, McManus ME, Tukey RH, histochemical localization of cytochrome P450 2E1 in Gonzalez FJ, Holder GM. The catalytic activity of human pulmonary carcinoma and normal bronchial four expressed human cytochromes P450s towards tissue. Virchows Arch 1995; 426: 243–247. benzo(a)pyrene and the isomers of its proximate 44. Ronis MJJ, Lindros KO, Ingelman-Sundberg M, carcinogen. Biochem Biophys Res Commun 1993; 3: Ioannides C, Parke DV, eds. Cytochromes P450: 1373–1379. Metabolic and Toxicological Aspects. 9, The CYP2E 58. Kim JH, Stansbury KH, Walker NJ, Trush MA, subfamily. Boca Raton, CRC Press, 1996; pp. 211– Strickland PT, Sutter TR. Metabolism of benzo(a)- 239. pyrene and benzo(a)pyrene-7,8-diol by human cyto- 45. Nhamburo PT, Kimura S, McBride OW, Kozak CA, chrome P450 1B1. Carcinogenesis 1998; 19: 1847–1853. Gelboin HV, Gonzalez FJ. The human CYP2F gene 59. Hirvonen A. Polymorphisms of xenobiotic-metabo- family: identification of a cDNA encoding a new lizing enzymes and susceptibility to cancer. Environ cytochrome P450, cDNA-directed expression, and Health Perspect 1999; 107: 37–47. chromosome mapping. Biochemistry 1990; 29: 5491– 60. Anttila S, Hirvonen A, Vainio H, Husgafvel-Pursiainen 5499. K, Hayes JD, Ketterer B. Immunohistochemical 46. Lanza DL, Code E, Crespi CL, Gonzalez FJ, Yost localization of glutathione S-transferase in human GS. Specific dehydrogenation of 3-methylindole and lung. Cancer Res 1993; 53: 5643–5648.