Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 May 2.
Published in final edited form as: Curr Drug Metab. 2012 Jul;13(6):787–814. doi: 10.2174/138920012800840356

Summary of Information on the Effects of Ionizing and Non-ionizing Radiation on Cytochrome P450 and Other Drug Metabolizing Enzymes and Transporters

Slobodan Rendic 1,*, F Peter Guengerich 2
PMCID: PMC4007673  NIHMSID: NIHMS568180  PMID: 22571481

Abstract

The present paper is an update of data on the effects of ionizing radiation (γ-rays, X-rays, high energy UV, fast neutron) caused by environmental pollution or clinical treatments and the effects of non-ionizing radiation (low energy UV) on the expression and/or activity of drug metabolism (e.g., cytochrome P450,, glutathione transferase), enzymes involved in oxidative stress (e.g., peroxidases, catalase,, aconitase, superoxide dismutase), and transporters. The data are presented in tabular form (Tables 13) and are a continuation of previously published summaries on the effects of drugs and other chemicals on cytochrome P450 enzymes (Rendic, S.; Di Carlo, F. Drug Metab. Rev., 1997, 29 (1–2), 413–580, Rendic, S. Drug Metab. Rev., 2002, 34 (1–2), 83–448) and of the data on the effects of diseases and environmental factors on the expression and/or activity of human cytochrome P450 enzymes and transporters (Guengerich, F.P.; Rendic, S. Curr. Drug Metab., 2010, 11(1), 1–3, Rendic, S.; Guengerich, F.P. Curr. Drug Metab., 2010, 11 (1), 4–84). The collective information is as presented by the cited author(s) in cases where several references are cited the latest published information is included. Remarks and conclusions suggesting clinically important impacts are highlighted, followed by discussion of the major findings. The searchable database is available as an Excel file (for information about file availability contact the corresponding author).

Keywords: Ionizing radiation, UV, γ-rays, X-rays, cytochrome P450, CYP, oxidative stress enzymes, transporters, expression, activity

INTRODUCTION

Preparation of this summary was prompted by nuclear plant disasters (Chernobyl (1986), Fukushima (2011)) and the consequent irradiation of humans and other biological systems caused by this environmental pollution. This summary includes also the influence of clinically applied radiation (γ-rays, X-rays, fast neutron) for treatment of specific diseases, as well as the effects of UVA, UVB, and UVC radiation on the activity and/or expression of drug metabolism enzymes such as cytochrome P450 (CYP), which are of prime importance for new drug development and absorption-distribution-metabolism-elimination (ADME) research, the enzymes involved in regulation of oxidative stress (e.g., peroxidases (PO), catalase (CAT), aconitase (ACO), superoxide dismutase (SOD)), and transporters. Ionizing radiation is defined as radiation composed of particles of different energy that can liberate an electron from an atom or molecule. When interacting with organic compounds or other small molecules in the body, ionizing radiation (γ-rays, α- and β-particles, high-frequency ultraviolet, X-rays, and gamma rays, fast neutron irradiation, and contamination with radioactive particles, e.g. uranium or radioactive pollutant contamination), depending on the energy, produces free radicals that can damage tissue molecules. Most UV light is classified as non-ionizing radiation, but the higher energies of the UV spectrum (e.g., ~ 150 nm, or ‘vacuum’ UV) are ionizing.

Cytochrome P450 enzymes catalyze a number of reactions that have profound effects on the biological activities (therapeutic and/or toxic) of xenobiotics, including drugs, and the significance of the human enzymes in drug metabolism has been reviewed [14]. The roles of the transporters, which (depending on the site of expression) may enhance or limit absorption or excretion of drugs and other xenobiotics from an organ or tissue and have additional effects on the biological activities of drugs/xenobiotics, are reviewed elsewhere [5,6]. In addition to a great number of xenobiotics used as drugs or coming from the environment, and influencing the activity and/or expression of the cytochrome P450 enzymes and transporters [1,2,5], the effects of diseases and environmental factors are also of interest [7]. These factors can have profound effects on enzyme activity and expression and therefore also the final biological activity, efficacy, and safety of drugs and other chemicals. A great number of examples from the literature show that the final effect of a drug or other chemical on an organism, whether pharmacological and/or toxicological, depends on regulation of expression and the activity of cytochrome P450 enzymes and transporters. When these properties are changed (resulting in their overexpression or inhibition) by factors such as irradiation (alone or in combination with specific drugs, e.g. anticancer drugs) a significant change of therapeutic outcome or resistance to a drug might occur.

Damaging effects of ionizing and UV irradiation result from generation of reactive oxygen species (ROS) and subsequent radical formation and from direct damage to cellular macromolecules, including DNA. The most pronounced effect of prolonged exposure to UV (predominately UVB and UVA) in humans is induction and development of skin cancer. UVB radiation (280–320 nm) is readily absorbed by and can cause severe damage to proteins and DNA. Other UV wavelengths of interest include UVA (320–400 nm), which are less detrimental to skin but still can cause harmful effects. Thus high doses of UVB in the epidermis (and UVA in the dermis) may be responsible for the production of reactive oxygen species [8]. Adverse effects of UV radiation are partially reduced by molecular defense mechanisms including glutathione and enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX), which are involved in quenching excessive levels of ROS and other free radicals [9]. Increased activity of the enzymes protecting against oxidative stress by ionizing or UV irradiation might be beneficial, and decreased activity might enhance damaging effects of irradiation. In addition, modification of the expression and activity of enzymes involved in metabolism (e.g., the toxicologically important CYP2E1 and CYP1 Family enzymes or CYP4A11, which is involved in lipid metabolism) or transporters (e.g. the clinically important P-glycoprotein, LRP, or MRP1) can influence the therapeutic outcome or toxicity of other drugs and xenobiotics. In addition, irradiation-induced modification of cytochrome P450 enzyme activity might result in enhanced or diminished formation of important substances, e.g. vitamin D and related compounds [10,11].

The present summary provides a collection of information on the effects of major ionizing and non-ionizing irradiations on the function of cytochrome P450 and other drug metabolizing enzymes and transporters, considering also the effects on therapeutic treatment or human health.

TABULAR PRESENTATION

Data are presented in Tables 13 and formatted in columns 1 – 8: 1. Species used for investigation, 2. Enzyme/Transporter; 3. Category; 4. Effectors; 5. Model; method used; 6. Effect on particular enzyme or transporter; 7. Remarks about effects when stated by the cited authors to additionally characterize the effects. 8. References.

Table 1.

Effects of Ionizing and Non-ionizing Irradiation on Expression and Activity of Transporters

Species Transporter Category Effectors Model; method used Effects Remarks Reference
Rat ABCA1, ABC1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), liver, microsomes; quantitative RT-PCR no change of mRNA expression [37]
Mice ABCA1, ABC1 Environmental impact Uranium contamination chronic exposure for 8 months by depleted uranium through drinking water 20 mg/L, cerebral cortex; RT-PCR increase of mRNA level expression by 52% [38]
Rat ABCA1, ABC1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, brain; RT-PCR increase of mRNA level expression by 34% dose corresponds to twice the highest concentration found naturally in Finland [36]
Rat ABCA1, ABC1 Environmental impact Uranium contamination low-level chronic ingestion of depleted uranium in drinking water for 9 months, 40mg/kg, liver; RT-PCR increase of mRNA expression (to 154%) and protein expression (125% but not significant) [39]
Human ABCA1, ABC1 Clinical impact X-rays malignant glioma U87-MG cells, 200KV x-ray-irradiation; immunoblotting increase of protein expression temozolomide co-treatment enhanced expression rate, suggested to be involved in drug resistance following radiation treatment [40,18]
Rat ABCG5 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), liver; quantitative RT-PCR decrease of mRNA expression (by 42%) [37]
Rat ABCG5 Environmental impact Uranium contamination low-level chronic ingestion of depleted uranium in drinking water for 9 months, 40 mg/kg, liver; RT-PCR increase of hepatic gene expression [39]
Rat ABCG8 Environmental impact Uranium contamination low-level chronic ingestion of depleted uranium in drinking water for 9 months, 40 mg/kg, liver; RT-PCR increase of hepatic gene expression [39]
Human EAAT1, GLAST, SLC1A3 Clinical impact γ-radiation, γ-rays NTera2-derived neurons as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy gamma rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometry, immunoblotting, glutamate uptake linear increase of protein expression and appeared to double between times, increase of glutamate uptake no significant differences between doses of radiation [41]
Human EAAT1, GLAST, SLC1A3 Clinical impact γ-radiation, γ-rays NTera2-derived astrocytes as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometer, immunoblotting, glutamate uptake increase of protein expression approximately 1.5-fold between 3h and 2 days and about fivefold between day 2 and day 7 after irradiation, significant decrease in glutamate uptake 2 days after irradiation with all three doses and returned to baseline levels no significant differences between doses of radiation [41]
Human EAAT2, GLT-1, SLC1A2 Clinical impact γ-radiation, γ-rays NTera2-derived neurons as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometry, immunoblotting, glutamate uptake dose-dependent increase in protein expression was seen between doses of 10 and 50 cGy, increase of glutamate uptake no significant differences between doses of radiation [41]
Human EAAT2, GLT-1, SLC1A2 Clinical impact γ-radiation, γ-rays NTera2-derived astrocytes as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometry, immunoblotting, glutamate uptake increase of protein expression approximately 1.5-fold between 3h and 2 days and about fivefold between day 2 and day 7 after irradiation, significant decrease in glutamate uptake 2 days after irradiation with all three doses and returned to baseline levels no significant differences between doses of radiation [41]
Human EAAT3, EAAC1, SLC1A1 Clinical impact γ-radiation, γ-rays NTera2-derived neurons as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays significant increase of protein expression was observed in EAAT3 after 50 cGy 7 days after exposure, increase of glutamate uptake no significant differences between doses of radiation [41]
Human EAAT3, EAAC1, SLC1A1 Clinical impact γ-radiation, γ-rays NTera2-derived astrocytes as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometry, immunoblotting, glutamate uptake increase of expression over time, 16-fold 7 days after 50 cGy irradiation, low level expression, significant decrease in glutamate uptake 2 days after irradiation with all three doses and returned to baseline levels no significant differences between doses of radiation [41]
Human EAAT4, SLC1A6 Clinical impact γ- radiation, γ-rays NTera2-derived neurons as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometry, immunoblotting, glutamate uptake increase of protein expression 3 h after irradiation, increase of glutamate uptake no significant differences between doses of radiation [41]
Human EAAT4, SLC1A6 Clinical impact γ-radiation, γ-rays NTera2-derived astrocytes as pure cultures exposed to doses of 10 cGy, 50 cGy and 2 Gy γ-rays, analyzed at 3 h, 2 days and 7 days after exposure; immunocytochemical labeling, laser scanning cytometry, immunoblotting, glutamate uptake two-fold increase of protein expression between 3 h and day after irradiation, low level expression, significant decrease in glutamate uptake 2 days after irradiation with all three doses and returned to baseline levels no significant differences between doses of radiation [41]
Human LRP (lung resistance-related protein) Clinical impact Ionizing irradiation fractionated irradiation, breast cancer cell lines irradiated with a total dose of 27 Gy, five fractions of 1.8 Gy per week; RT-PCR, flow cytometry increase of mRNA expression and protein level expression in irradiated cells [20]
Human LRP (lung resistance-related protein) Clinical impact Ionizing irradiation fractionated irradiation, colon cancer cell lines irradiated with a total dose of 27 Gy, five fractions of 1.8 Gy per week; RT-PCR, flow cytometry no increase of mRNA expression, high increase of protein level in irradiated cells significant resistance to cisplatin, doxorubicin and bendamustine [20]
Human LRP (lung resistance-related protein) Clinical impact X-rays fractionated irradiation, SW620 colon carcinoma cells exposed to either 27 Gy in 1.8-Gy daily fractions; semiquantitative RT-PCR, flow cytometry gene activated only shortly after radiation irradiated cells less sensitive to cisplatin but not to doxorubicin [22]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact γ-radiation, γ-rays cervical carcinoma HeLa cells exposed to 10 daily fractions of 0.17 Gy γ-rays; immunochemistry increase of protein expression increased resistance to vincristine and vinblastine [15]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact γ-radiation, γ-rays hepatocellular carcinoma HepG2 cells (G cells) were gamma-radiation treated of 2 Gy for 10 days (G2) or 10 Gy for 2 days (G10) and then doxorubicin (Dox) treated as continuous exposure for up to 10 microM; Dox accumulation assay, immunohistochemistry, immunoblotting, Southern blotting, RT-PCR increase of protein and mRNA expression in irradiated cells before Dox treatment radiation treatment may lead to a development of highly resistant phenotype [21]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact γ-radiation, γ-rays fractionated irradiation, MDR KB-VI cancer cells exposed to 1400 and 2800 cGy ionizing radiation administered in 7 and 14 fractions (at 200 cGy per fraction/day, with 137Cs γ cell-40 Exactor); immunoblotting, FISH analysis decrease in MDR1 extrachromosomal gene copy, decrease of protein level low to moderate doses of ionizing radiation reduce multidrug resistance with improved therapeutic response (vinblastine, doxorubicin, colchicine, cisplatin) for some cancers due to loss of extrachromosomally amplified genes from tumor cells [43]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact γ-radiation, γ-rays fractionated irradiation, esophageal cancer biopsy samples; quantitative RT-PCR, immunoblotting increased initial mRNA expression decreased after 7 irradiation Gy/f, 14 fractions differentially modulated MDR1 expression by different doses of fractionated radiation [44]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF and Sk-Mel), single doses (5, 10 and 20 Gy); no significant change of mRNA expression [42]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact Ionizing irradiation radiotherapy, stage I primary breast cancer specimens; immunohistochemistry increase of protein expression [46]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact Ionizing irradiation irradiated nasopharyngeal carcinoma (NPC) CNE1 cells; immunoblotting, RT-PCR, flow cytometry long-term overexpression, reduction in intracellular daunorubicin accumulation irradiation decreased the chemotherapy sensitivity of CNE1 cells [17]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact Ionizing irradiation fractionated irradiation, breast cancer cell lines irradiated with a total dose of 27 Gy, five fractions of 1.8 Gy per week; quantitative RT-PCR, flow cytometry increase of mRNA expression, low increase of protein level [20]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact Ionizing irradiation fractionated irradiation, colon cancer cell lines irradiated with a total dose of 27 Gy, five fractions of 1.8 Gy per week; quantitative RT-PCR, flow cytometry no increase of mRNA expression, high increase of protein level in irradiated cells, significant resistance to cisplatin, doxorubicin and bendamustine [20]
Murine MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact Ionizing irradiation NIH 3T3 cells treated with single doses of 5, 10 and 20 Gy no change of mRNA expression [45]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Environmental impact UV irradiation KB carcinoma cells irradiated with UV light (16 J/m2, 254 nm); luciferase activity, Northern analysis, immunoblotting induction of gene expression, transcriptional activation of the MDR1 gene promoter induction of gene expression through a common stress-induced signal transduction suggested [49,50,51]
Chinese hamster MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, Chinese hamster ovary cell lines (ten fractions of 9 Gy), exposure to multiple lethal doses, or to a single 30-Gy dose of radiation; immunoblotting, mRNA blotting increase of protein level, no gene amplification, no significant alteration in expression and mRNA levels drug cross-resistance to vinca alkaloids (vincristine), epipodophyllotoxins (etoposide), gramicidin D, taxol, puromycin, and navelbine, but sensitive to anthracyclines (daunomycin or mitoxantrone) regulated by post-translational stability [27,28,29,30,31,32]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, lymphoblastic leukemia CEM/MDR cell sublines; susceptibility to anticancer drugs decrease of protein expression drug sensitivity of multidrug-resistant (MDR) cells could be enhanced by fractionated irradiation [33]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, small-cell lung cancer H69 SCLC cells, X- rays to an accumulated dose of 37.5 Gy over 8 months; immunoblotting no change in protein expression [25]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays squamous carcinoma oral cavity T167 cell lines, exposed to either a standard clinical dose of 2 GY and 7GY or low-dose fractionated irradiation therapy (LDFRT) delivered as 0.5 Gy in four fractions; RT-PCR, immunoblotting, activity as efflux of rhodamine 123 increase of expression and activity in response to conventional 2-Gy and high-dose 7-Gy radiation, no increase during LDFRT. the up-regulation of the transport activity may lead to radio-and chemoresistance during the conventional and high-dose ionizing radiation treatments, but not during LDFRT [19]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays radiotherapy, primary oral squamous carcinoma tissues; immunohistochemistry increase of expression in tumor tissue standard radiation might affect the efficacy of subsequent or concurrent chemotherapy [52]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, human epidermoid lung carcinoma xenograft HXL55 exposed to seven irradiation treatments of 10 Gy over period of 9 months; immunofluorescence, Southern blotting increase of protein expression, no gene amplification increased resistance to vincristine [26]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, human ovarian tumor cells exposed to 10 fractions of 5 Gy (the total radiation dose administered was50 Gy); immuno and mRNA blotting or RNase protection assays increase of protein expression, no increase of mRNA expression increased resistance to vincristine and etoposide [24]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, human ovarian tumor cells SK-OV-3 exposed to 2 Gy twice-daily fractions for 5 days on two consecutive weeks; immunocytochemistry increase of protein expression [53]
Human MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays fractionated irradiation, parental drug-sensitive lymphoblastic leukemia CEM cells; susceptibility to anticancer drugs increase of protein expression induced drug resistance in the parental drug-sensitive CEM cells [33]
Rat MDR1, P-glycoprotein, P-gp, ABCB1 Clinical impact X-rays right brain hemispheres irradiated with single doses of 2–25 Gy, or with fractionated irradiation (4×5 Gy), followed by cyclosporine A (CsA) 5 days later, brain samples; immunohistochemistry, immunoblotting, [11C]carvedilol uptake using quantitative autoradiography decrease of protein expression and activity 10 days after start of irradiation, and between day 15 and 20 after single dose irradiation, and increased again thereafter suggested that that brain irradiation could be used to enhance the delivery of P-gp substrates to the brain. [34,35]
Murine MDR1 A, P-glycoprotein, P-gp, ABCB1A Clinical impact Ionizing irradiation fractionated irradiation, Ehrlich ascites tumor cells (EHR2), 60 Gy; immunoblotting, semiquantitative RT-PCR increase of protein expression (threefold), mRNA not detectable increased resistance to etoposide and vincristine [23]
Murine MDR1B, P-glycoprotein, P-gp, ABCB1B Clinical impact Ionizing irradiation fractionated irradiation, Ehrlich ascites tumor cells (EHR2), 60 Gy; immunoblotting, semiquantitative RT-PCR increase of protein expression (threefold), slight reduction of mRNA expression increased resistance to etoposide and vincristine [23]
Rat MRP Clinical impact γ-radiation, Gamma-rays isolated hepatocytes in vitro, dose 8 Gy; real-time PCR, immunoblotting increase of mRNA expression and protein level [47]
Rat MRP Clinical impact γ-radiation, γ-rays liver were irradiated in vivo with 6 MV photons (dose rate 2.4 Gy/min); real-time PCR, immunoblotting increase of mRNA expression and protein level [47]
Human MRP1, MRP, GS-X, ABCC1 Clinical impact γ-radiation, γ-rays fractionated irradiation, CCRF-CEM (CEM) human T-cell leukemia cell line, total 75 Gy (10 cycles of 1.5 Gy daily for 5 days). 6-fold increase in protein level, increase of mRNA expression mRNA increased within 4 hr which, by 24 hr, was greater than 5-fold and suggested to be involved in drug resistance following radiation treatment, buthionine sulphoximine reversed the daunorubicin resistance [16,18]
Human MRP1, MRP, GS-X, ABCC1 Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF and Sk-Mel), single doses (5, 10 and 20 Gy); no significant change of mRNA expression [42]
Human MRP1, MRP, GS-X, ABCC1 Clinical impact Ionizing irradiation fractionated irradiation, breast cancer cell lines irradiated with a total dose of 27 Gy, five fractions of 1.8 Gy per week; quantitative RT-PCR, flow cytometry increase of mRNA expression and protein level in irradiated cells [20]
Human MRP1, MRP, GS-X, ABCC1 Clinical impact Ionizing irradiation fractionated irradiation, colon cancer cell lines irradiated with a total dose of 27 Gy, five fractions of 1.8 Gy per week; quantitative RT-PCR, flow cytometry no increase of mRNA expression, high increase of protein level in irradiated cells significant resistance to cisplatin, doxorubicin and bendamustine [20]
Murine Mrp1, Mrp, GS-X, Abcc1 Clinical impact Ionizing irradiation fractionated irradiation, Ehrlich ascites tumor cells (EHR2), 60 Gy; immunoblotting, semiquantitative RT-PCR increase of protein expression (8-fold), increase of mRNA expression (6-fold) increased resistance to etoposide and vincristine [23]
Human MRP1, MRP, GS-X, ABCC1 Clinical impact X-rays fractionated irradiation, small-cell lung cancer H69 SCLC cells treated to an accumulated dose of 37.5 Gy over 8 months; immunoblotting increase of protein expression may account for the cisplatin resistance [25]
Human MRP1, MRP, GS-X, ABCC1 Clinical impact X-rays fractionated irradiation, human ovarian tumor cells SK-OV-3 exposed to 2 Gy twice-daily fractions for 5 days on two consecutive weeks; immunocytochemistry increase of expression [54]
Human MRP2, cMOAT, ABCC2 Clinical impact X-rays fractionated irradiation, small-cell lung cancer H69 SCLC cells treated to an accumulated dose of 37.5 Gy over 8 months; immunoblotting increase of protein expression may account for the cisplatin resistance [25]
Human MRP2, cMOAT, ABCC2 Clinical impact X-rays fractionated irradiation, SW620 colon carcinoma cells exposed to either 27 Gy in 1.8-Gy daily fractions; semiquantitative RT-PCR, flow cytometry increase of gene expression up to 3 weeks after radiation irradiated cells less sensitive to cisplatin but not to doxorubicin [22]
Mice SGLT1, SLC5A1 Environ mental impact γ-radiation, γ-rays proximal jejunum sections; whole body irradiation with 137Cs γ-rays at doses of 0, 7, 8.5, or 10 Gy, real-time PCR decreased mRNA abundance d-glucose uptake decreased by approximately 10–20% by day 8 post irradiation; vitamin A supplementation had no effect on clinical or transport parameters [48]
Mice VAChT (vesicular acetylcholine transporter) Environmental impact Uranium contamination chronic exposure for 8 months by depleted uranium through drinking water 20 mg/L, cerebral cortex; RT-PCR increase of mRNA level expression by 120% [38]

Table 3.

Effects of Ionizing and Non-ionizing Irradiation on Expression and Activity of Oxidative Stress and Other Enzymes

Species Enzyme Category Effectors Model; method used Effects Remarks References
Rat ACO (aconitase) Environmental impact γ-radiation, γ-rays whole body irradiation of 3 and 9 gray (G) at a dosage rate of 12.5 cG/min from a 60Co radiation source, liver, mitochondrial post-nuclear fraction; conversion of citrate to isocitrate activity decreased 30–90% by increasing gamma-irradiation [66]
Human ALPL (alkaline phosphatase) Environmental impact γ-radiation, γ-rays workers exposed to short-life radioactive isotopes 131I and 99Tc, blood smears; decreased activity [87]
Human APRT (adenyl phosphoribosyl transferase) Clinical impact γ-radiation, γ-rays single dose of 1-Gy 137Cs-gamma-rays, TK6 lymphoblastoid cells; two-dimensional (2-D) gel electrophoresis, MALDI-TOF, immunoblotting decreased protein level [88]
Guinea pigs CAT (catalase) Environmental impact γ-radiation, γ-rays irradiated with the doses of 8 Gy or 15 Gy, single dose/whole body, 60Co, source axis distance 80 cm, liver; activity activity decreased at 15 Gy [83]
Mice CAT (catalase) Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors irradiated with different doses of γ-rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumor; activity measured as decomposition of H2O2 at 240 nm no activity detected in control or irradiated samples [56]
Mice CAT (catalase) Clinical impact γ-radiation, γ-rays whole body irradiated mice with Ehrlich solid tumor in the thigh pad and non-tumor bearing animals, irradiated with different doses of gamma-radiation (0–9 Gy) at a dose rate of 0.0153 Gy/s, liver; activity measured as decomposition of H2O2 at 240 nm activity decreased with dose in tumor and non-tumor mice, activity was higher in liver of tumor mice than control [81]
Rat CAT (catalase) Environmental impact γ-radiation, γ-rays whole body single dose of γ-radiation (5 Gy); testicular level decreased protein level supplementation with extract of Xylopia aethiopica and vitamin C reversed the effect [82]
Mice CAT (catalase) Environmental impact UV irradiation Skh:HR-1 hairless mice, in vivo, single UVB irradiation; activity assays decrease of activity by 12 h after irradiation [86]
Mice NADPH-quinone reductase Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors irradiated with different doses of γ-rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumor; activity measured as reduction of 2,6-dichlorophenolindophenol activity increased with increase in radiation dose, at 9 Gy it was about 50% higher compared to the unirradiated control [56]
Mice NADPH-quinone reductase Clinical impact γ-radiation, γ-rays whole body irradiated mice with Ehrlich solid tumor in the thigh pad and non-tumor bearing animals, irradiated with different doses of γ-radiation (0–9 Gy) at a dose rate of 0.0153 Gy/s, liver; activity measured as reduction of 2,6-dichlorophenolindophenol activity increased at all doses except 9 Gy, activity higher in liver of tumor compared to non-tumor bearing mice [81]
Mice NADPH-quinone reductase Environmental impact γ-radiation, Gamma-rays whole body irradiation, different doses of gamma-rays at 1.38 Gy/min, liver; activity activity increased up to 5 Gy and decreased thereafter administration of phenothiazines increased the radiation effect at lower doses providing the radioprotective action [55]
Mice GLY I (glyoxalase I) Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors irradiated with different doses of γ-rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumor; activity measured as formation of (S)-D-lactoylglutathione increased activity with increase in radiation dose [56]
Mice GLY I (glyoxalase I) Clinical impact γ-radiation, γ-rays whole body irradiated mice with Ehrlich solid tumor in the thigh pad and non-tumor bearing animals, irradiated with different doses of gamma-radiation (0–9 Gy) at a dose rate of 0.0153 Gy/s, liver; activity measured by formation of (S)-D-lactoylglutathione increased activity in both normal and tumor-bearing animals from 1–4 Gy in a dose dependent manner, declined beyond 4 G, activity higher in liver of tumor-bearing compared to non-tumor bearing mice [81]
Guinea pigs GPX (glutathione peroxidase) Environmental impact γ-radiation, γ-rays irradiated with the doses of 8 Gy or 15 Gy, single dose/whole body, 60Co, source axis distance 80 cm, liver; activity activity increased at 15 Gy [83]
Human GPX (glutathione peroxidase) Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF and Sk-Mel), single doses (5, 10, and 20 Gy); no significant change of mRNA expression [42]
Mice GST (glutathione transferase) Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors irradiated with different doses of gamma rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumors activity measured as formation of GSHCDNB (1-chloro-2,4-dinitrobenzene) conjugate, Western blotting increased activity between 4 an 9 Gy, dose dependent increase of protein level compared to control tissue [56]
Mice GST (glutathione transferase) Clinical impact γ-radiation, γ-rays whole body irradiated mice with Ehrlich solid tumor in the thigh pad and non-tumor bearing animals, irradiated with different doses of gamma-radiation (0–9 Gy) at a dose rate of 0.0153 Gy/s, liver; activity expressed as 1-chloro-2,4-dintrobenzene conjugate formed, Western blotting increased activity depending on the dose of radiation in both normal and tumor-bearing animals [81]
Mice GST (glutathione transferase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of γ-rays at 1.38 Gy/min, liver; activity activity increased up to 5 Gy and decreased thereafter administration of phenothiazines increased the radiation effect at lower doses providing the radio protective action [55]
Mice GST (glutathione transferase) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 and 9 Gy increase of mRNA expression 2-(allylthio)pyrazine pretreatment (100 mg/kg/day, for 2 days) prior to whole body irradiation increased the 30 day survival rate of mice to 91% [89]
Rat GST (glutathione transferase) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry decreased mRNA expression at 3 and 8 hr after irradiation, followed by increase of mRNA level by 2-fold at 15 and 24 hr after irradiation and return to the levels of untreated rats at 48 hr after treatment, paralleled changes of protein levels with those of mRNA oltipraz pretreatment before irradiation resulted in additional increase of mRNA expression and increased the survival rate, protective role of oltipraz suggested [90]
Rat GST (glutathione transferase) Environmental impact γ-radiation, γ-rays whole body single dose of γ-radiation (5 Gy), testicular level; protein level decrease of protein level supplementation with extract of Xylopia aethiopica and vitamin C reversed the effect [82]
Rat GST (glutathione S-transferase) Clinical impact Ionizing irradiation preliminary radiation-exposed, transplanted Guerin’s carcinoma, liver, microsomes decreased activity in the latent and logarithmic phases of oncogenesis, no effect on terminal stages of Guerin’s carcinoma growth [57]
Human GST (glutathione transferase) Environmental impact Radioactivity-contaminated areas placental samples at term, cytosolic fraction; activity measured as GSH conjugation with 1-chloro-2,4 dinitrobenzene down-regulation of activity and mRNA level in samples exposed to highest levels of radioactivity imbalance in detoxification capacity suggested [54]
Rat GST (glutathione transferase) Clinical impact X-rays preliminary radiation-exposed rats with Guerin’s carcinoma, liver, microsomal fraction; activity reduced activity [57]
Rat GSTA2-2 (glutathione transferase A2-2) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry increase of mRNA level (threefold) after 3 Gy g-irradiation dexamethasone prior to 3 Gy irradiation exhibited 80%–93% suppression in the radiation-inducible increases in the mRNA level and reduced the mean survival time [91]
Rat GSTA2-2 (glutathione transferase A2-2) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression 2-to 2.8-fold 2-(allylthio)pyrazine pretreatment mRNA expression at 24 h after 2-AP treatment [89]
Mice GSTA3-2 (glutathione transferase A3-3) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression 2-(allylthio)pyrazine pretreatment increased mRNA expression [89]
Rat GSTA3-3 (glutathione transferase alpha3) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry increase of mRNA level (3-fold) after 3 Gy g-irradiation dexamethasone prior to 3 Gy irradiation exhibited 80%–93% suppression in the radiation-inducible increases in the mRNA level and reduced the mean survival time [91]
Rat GSTA3-3 (glutathione transferase A3-3) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression 2-(Allylthio)pyrazine pretreatment caused smaller increase mRNA expression at 24 h after 2-AP treatment [89]
Rat GSTA5-5 (glutathione transferase A5-5) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry increase of mRNA level (3-fold) after 3 Gy g-irradiation dexamethasone prior to 3 Gy irradiation exhibited 80%–93% suppression in the radiation-inducible increases in the mRNA level and reduced the mean survival time [91]
Rat GSTA5-5 (glutathione transferase A5-5) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression 2-(allylthio)pyrazine pretreatment caused smaller increase mRNA expression at 24 h after treatment [89]
Rat GSTM1-1 (glutathione transferase M1-1) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry increase of mRNA level (3-fold) after 3 Gy g-irradiation dexamethasone prior to 3 Gy irradiation at doses of 1 mg/kg exhibited 68% suppression in the radiation-inducible increases in the mRNA level [91]
Rat GSTM1-1 (glutathione transferase M1-1) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression 2-(allylthio)pyrazine pretreatment caused smaller increase mRNA expression at 24 h after treatment [89]
Rat GSTM2-2 (glutathione transferase M2-2) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry no significant change in mRNA level after 3 Gy g-irradiation dexamethasone prior to 3 Gy irradiation exhibited no significant change in mRNA level [91]
Rat GSTM2-2 (glutathione transferase M2-2) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression 2-(allylthio)pyrazine pretreatment caused smaller increase mRNA expression at 24 h after treatment [89]
Human GSTO1-1 (glutathione transferase O1-1) Clinical impact γ-radiation, γ-rays single dose of 1-Gy 137Cs-gamma-rays, TK6 lymphoblastoid cells; two-dimensional (2D) gel electrophoresis, MALDI-TOF, immunoblotting no change in protein level [88]
Mice GSTP1-1 (glutathione transferase P1-1) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 8 Gy, liver; mRNA blotting increase of mRNA expression no influence of 2-(allylthio)pyrazine pretreatment [89]
Human GSTP1-1 (glutathione transferase P1-1) Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF and Sk-Mel), single doses (5, 10 and 20 Gy); increase of mRNA expression [42]
Human GSTP1-1 (glutathione transferase P1-1) Clinical impact Ionizing irradiation fractionated X-irradiation of AuxB1Chinese hamster ovary cell lines; Western blotting increase of protein expression [30]
Murine GSTP1-1 (glutathione transferase P1-1) Clinical impact Ionizing irradiation NIH 3T3 cells treated with single doses of 5, 10 and 20 Gy increase of mRNA expression and protein level [45]
Murine GSTP1-1 (glutathione transferase P1-) Clinical impact Ionizing irradiation human lung carcinoma cell line LXF 289, single doses of 5, 10, and 20 Gy increase of mRNA expression and protein level [45]
Mice LDH (L-lactate dehydrogenase) Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors irradiated with different doses of gamma rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumor; activity measured as disappearance of NADH (340 nm) increased activity up to 6 Gy, declined beyond 6 Gy [56]
Mice LDH (L-lactate dehydrogenase) Clinical impact γ-radiation, γ-rays whole body irradiated mice with Ehrlich solid tumor in the thigh pad and non-tumor bearing animals, irradiated with different doses of gamma-radiation (0–9 Gy) at a dose rate of 0.0153 Gy/s, liver; activity measured by disappearance of NADH (340 nm) increased activity at lower doses (2 and 4 Gy), declined at higher doses (6–9 Gy) in both normal and tumor-bearing animals [81]
Mice LDH (L-lactate dehydrogenase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma- rays at 1.38 Gy/min, liver; activity progressive increase in activity administration of phenothiazines inhibited activity [55]
Mice LDH (L-lactate dehydrogenase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma radiation (1–9 Gy) at a dose rate of 0.023 Gy/s, liver activity activity increased at doses above 3 Gy phenylmethylsulfonyl fluoride inhibited activity, dithiothreitol inhibited the release of lactate dehydrogenase [92,93]
Mice mEH (microsomal epoxide hydrolase) Environmental impact γ-radiation, γ-rays whole body treatment at the dose of 0.5 and 8 Gy, liver; mRNA blotting increase of mRNA expression 2- to 2.8-fold 2-(allylthio)pyrazine pretreatment mRNA expression at 24 h after 2-AP treatment [89]
Rat mEH (microsomal epoxide hydrolase) Environmental impact γ-radiation, γ-rays whole body treatment with either 3 or 0.5 Gy of radiation/day, dosage of 12.5 cGy/min from a 60Co radiation source, liver; immunoblotting, mRNA blotting, scanning densitometry increase of mRNA level (threefold) after 3 Gy g-irradiation dexamethasone prior to 3 Gy irradiation exhibited 80%–93% suppression in the radiation-inducible increases in the mRNA level and reduced the mean survival time [91]
Rat mEH (microsomal epoxide hydrolase) Environmental impact γ-radiation, γ-rays whole body single dose (3 Gy) treatment, liver; immunoblotting mRNA level transiently decreased at 3 and 8 h after irradiation, increased 3- to 4-fold at 15 to 48 h post-irradiation, returning to the level in untreated animals at 72 h, paralleled changes of protein levels with those of mRNA mRNA level increased by oltipraz treatment [97]
Human MGMT (O6-alkylguanine-DNA alkyltransferase) Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF, and Sk-Mel), single doses (5, 10, and 20 Gy); no significant change of mRNA expression [42]
Human MPO (myeloperoxydase) Environmental impact γ-radiation, γ-rays workers exposed to short-life radioactive isotopes 131I and 99Tc, blood smears; stained for L-ALP and MPO and benzidine method decreased activity [87]
Human PP1α1 (serine/threonine protein phosphatase PP1-α1) Clinical impact γ-radiation, γ-rays single dose of 1-Gy 137Cs- gamma-rays, TK6 lymphoblastoid cells; two-dimensional gel electrophoresis, MALDI-TOF, immunoblotting decreased protein level [88]
Guinea pigs SOD (superoxide dismutase) Environmental impact γ-radiation, γ-rays irradiated with the doses of 8 Gy or 15 Gy, single dose/whole body, 60Co, source axis distance 80 cm, liver; activity activity decreased at 15 Gy [83]
Mice SOD (superoxide dismutase) Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors irradiated with different doses of gamma rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumor; activity measured as inhibition of autoxidation of pyrogallol increased activity with dose up to 4 Gy and then declined beyond 4 Gy at 24 h after irradiation [56]
Mice SOD (superoxide dismutase) Clinical impact γ-radiation, γ-rays whole body irradiated mice with Ehrlich solid tumor in the thigh pad and non-tumor bearing animals, irradiated with different doses of gamma-radiation (0–9 Gy) at a dose rate of 0.0153 Gy/s, liver; activity measured as inhibition of autoxidation of pyrogallol activity increased with radiation dose in the liver of tumor-bearing animals, in the liver of normal animals the activity was increased up to 6 Gy and inhibited thereafter, higher activity in liver of tumor-bearing compared to non-tumor bearing mice [81]
Mice SOD (superoxide dismutase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma- rays at 1.38 Gy/min, liver; activity activity increased up to 5 Gy and decreased thereafter administration of phenothiazines increased the radiation effect at lower doses providing the radioprotective action [55]
Rat SOD (superoxide dismuase) Environmental impact γ-radiation, γ-rays whole body single dose (5 Gy) treatment, testis; testicular level decreased protein level supplementation with extract of Xylopia aethiopica and vitamin C reversed the adverse effects of radiation [82]
Human SOD (superoxide dismutase) Environmental impact Ionizing irradiation medical workers exposed to occupational low-level doses, plasma samples; activity measured spectrophotometrically higher SOD activity in the blood samples of exposed vs. unexposed persons higher activity at occupational doses provide protection against the increased production of reactive oxygen species (ROS) [84]
Human SOD (superoxide dismutase) Environmental impact Ionizing irradiation blood samples irradiated by 2Gy of gamma radiation, dose-rate 0.45 Gy/min, and the distance from the source of 74 cm; SOD activity measured spectrophotometrically decrease of SOD activity after high dose irradiation dysfunction of mitochondrial system suggested at higher doses [84]
Human SOD1 (superoxide dismutase) Environmental impact UV irradiation epidermis, in vivo, chronic UVB irradiation; activity increase of activity [85]
Murine SOD1 (superoxide dismutase) Environmental impact UV irradiation Skh:HR-1 hairless mice, in vivo, single UVB irradiation; activity decrease of activity by 12 h after irradiation [86]
Zebrafish SOD1 (superoxide dismutase) Environmental impact UV irradiation embryos exposed for varying time to UVB on two consecutive days; spectophotometry, RT-PCR increase of mRNA expression [9]
Human TOP2A (topoisomerase (DNA) IIα) Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF, and Sk-Mel), single doses (5, 10, and 20 Gy); increases of mRNA expression [42]
Human TYMS (thymidylate synthetase) Clinical impact Ionizing irradiation tumor cell lines (MCF-7, LXF and Sk-Mel), single doses (5, 10 and 20 Gy); increases of mRNA expression [42]
Human UQCRC1 (ubiquinol-cytochrome c reductase core protein I) Clinical impact γ-radiation, γ-rays single dose of 1-Gy 137Cs-gamma-rays, TK6 lymphoblastoid cells; two-dimensional (2D) gel electrophoresis, MALDI-TOF, Western blotting decreased protein level [88]
Mice XDH (xanthine dehydrogenase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma radiation (1–9 Gy) at a dose rate of 0.023 Gy/s, liver; activity activity decreased at doses above 3 Gy allopurinol and folic acid inhibited activity, phenylmethylsulfonyl fluoride restored activity [92,93,94]
Mice XOR (xanthine oxidoreductase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma-rays (1–9 Gy) at a dose rate of 0.023 Gy/s or 1.38 Gy/min, liver; activity activity progressively increased at doses above 3 Gy phenothiazines, allopurinol, folic acid, and phenylmethylsulfonyl fluoride inhibited activity [55,92,93,94]
Mice XOR (xanthine oxidoreductase) Environmental impact UV irradiation Skh:HR-1 hairless mice, in vivo, single UVB irradiation; activity no effect on activity [86]
Human SDH (succinate dehydrogenase) Environmental impact UV irradiation UV (240–390 nm) irradiation, lymphocytes from human blood donors photoinactivation immediately after UV-irradiation [95]
Human LDH (L-lactate dehydrogenase) Environmental impact UV irradiation UV (240–390 nm) irradiation, lymphocytes from human blood donors photoinactivation immediately after UV-irradiation [95]
Human COX (cytochrome c oxidase) Environmental impact UV irradiation UV (240–390 nm) irradiation, lymphocytes from human blood donors photoinactivation immediately after UV-irradiation [95]
Mice ChAT (choline acetyl transferase) Environmental impact Uranium contamination chronic exposure for 8 months by depleted uranium through drinking water 20 mg/l, cerebral cortex; RT-PCR increase of mRNA level expression by 189% [38]

The data are sorted in Column 2 (Enzymes/Transporters), Column 4 (Effectors), and Column 1 (Species), for an easier approach to the information presented.

The tabulated data were obtained either in vivo (clinical experiments and whole animals) or in vitro using various models including clinical tissue samples, cell cultures, and microsomes following irradiation with the effectors.

It is important to emphasize that the experimental results can depend upon the model (human, animal) and/or method of irradiation used for investigation (doses, frequency, duration etc.), and contradictory or insufficient results might have been obtained. Results that might be of clinical importance are designated in the tables (bold-face font). Of the effectors presented in Table 13, a large number result from the environmental impacts (γ-, X-, and/or UV-radiation) and the reminder from clinical application (γ- and X-rays, or just ionizing radiation) on the expression/activity of both transporters and the enzymes. In most of the cases the results are interpreted as “increase” or “decrease” of expression of a particular enzyme/transporter, because this is of the lack of exact limits, diversity of ways of presenting results, and the large numbers of results obtained using immunohistochemistry or immunoblotting which are not always expressed quantitatively and thus making the results difficult to compare with those obtained by other methods [12]. However, standardized detection and quantification techniques and methods (e.g. quantitative real-time PCR (RT-PCR)) can provide more reliable comparisons of experimental results with therapy. In this respect it has been suggested that (of the techniques used to analyze drug transporters) flow cytometry may be preferred to immunoblots, mRNA blots, and immunocytochemical assays, although other report shows immunoblots to be reasonably quantitative [13]. The use of functional flow cytometric tests (assessing modulator-induced changes in fluorophore retention and/or efflux) has been promoted because these allow evaluation of a protein activity, in contrast to immunochemical or molecular tests [14]. As discussed in the previous paper in this series [7], P-gp functional analysis should be preferred when testing effects of radiation in connection with specific clinical treatment (e.g., cancers and other diseases) as a more sensitive predictor of chemoresistance than P-gp expression.

TRANSPORTERS

As presented in Table 1 (Effects and Remarks columns) the experimental results show that although in some cases inconsistent results and conclusions have been obtained for the same effectors using different methods and models, the increases in mRNA and/or protein expression of transporters (P-gp, Mrp1, LRP) by γ-rays or ionizing radiation may link causality with changes of gene expression. For instance, increased P-gp, Mrp1 or Mrp2 mRNA and/or protein expression was associated with increased resistance to anticancer drugs resulting in the development of highly resistant irradiated cancer cell phenotypes following (fractionated) ionizing radiation (total dose up to 75 Gy) using human models [15,16,17,18,19,20,21,22]. Interestingly, by using murine model (total dose of 60 Gy) P-gp (Mdr1a and Mdr1b) mRNA was not detectable or was reduced, but protein expression increased three-fold [23]. In a human model, expression of mRNA and protein was tissue dependent (i.e. in irradiated colon cancer cell lines no increase of MDR1, MRP1, or LRP mRNA expression was observed but a large increase in protein level was observed). On the other hand, in irradiated breast cancer cell lines both mRNA expression and protein levels of the transporters were increased [20]. In both models, murine and human, increased protein levels resulted in significant resistance to anticancer drugs. This result raises the suggestion that functional analysis should be preferred when testing effects of radiation or other effectors as a more sensitive predictor than testing mRNA expression. Following irradiation with X-rays, increased MRP1, MRP2, and/or P-gp protein expression was observed with increased resistance to anticancer drugs [24,25], but no P-gp mRNA increases in human [24, 26] or in animal models [27,28,29,30,31,32]. This result supports the suggestion that standard X-ray radiation might affect clinical efficacy of subsequent or concurrent chemotherapy. When the parental drug-sensitive CEM cells were subjected to fractionated radiation, increased levels of MDR proteins and induced drug resistance was observed. However, fractionated radiation with X-rays resulted in decreased P-gp protein expression, and consequently the drug sensitivity of multidrug-resistant (MDR) cells was enhanced [33]. Using functional analysis, immunohistochemistry, and immunoblotting (following irradiation by either single doses of X-rays up to 25 Gy or fractionated radiation of rat blood-brain barrier), decreased P-gp protein expression and activity were observed [34,35], suggesting that in some cases radiation might be used to enhance the delivery of P-gp substrates to the brain. This result further shows that predictions of the irradiation effects are rather difficult and complex in nature. Chronic contamination of rats by depleted uranium (uranyl nitrate) in their drinking water (dose of 1 mg/rat/day for 9 months) resulted in an increase (34%) in ABCA1 transporter mRNA, which regulates cholesterol transport [36].

CYTOCHROME P450 ENZYMES

Summarized data on the effects of ionizing and non-ionizing radiation on cytochrome P450 enzymes are presented in Table 2. 7-Ethoxycoumarin O-deethylation (ECOD) activity was 7-fold and 2-fold higher in human placental microsomes samples (at term) from radioactivity-contaminated areas compared to a region considered to be “clean.” It was suggested that this effect could be related to the increased formation of reactive metabolites in placenta [54]. Experiments performed using animal models (chronic contamination of rats for 9 months by depleted uranium as uranyl nitrate in drinking water, dose of 1 mg/rat/day) showed that the cholesterol-oxidizing enzyme CYP46A1 displayed a 39% increase in mRNA level, as determined by RT-PCR [36]. In mice it was shown that differential effects of radiation on the components of the cytochrome P450 system were observed after whole body irradiation by γ-rays at different doses. The activities of cytochrome NADPH-P450 reductase, cytochrome b5, and NADH-cytochrome reductase (as well as cytochrome P450 activity) were enhanced by irradiation doses up to ~5 Gy and decreased thereafter. The increase in cytochrome P450 activity was accompanied by the enhanced activity of glutathione transferase (Table 3), and administration of phenothiazines enhanced the radiation effect at lower irradiation doses on components of the cytochrome P450 system (except NADH-cytochrome b5 reductase) by inducing cytochrome P450 enzymes, thus providing radioprotective action [55]. Also, whole body irradiation of mice with Ehrlich solid tumors with different doses of γ-rays (doses of 0–9 Gy, at a dose rate of 0.0153 Gy/s) caused increases in microsomal cytochrome P450 enzymes at doses up to 6 Gy and decreases thereafter [56]. In contrast, cytochrome P450 (Table 2) and glutathione transferase (Table 3) activities in the liver microsomal fractions of radiation-exposed rats were reduced in the latent and logarithmic phases of oncogenesis compared with non-irradiated rats having tumors. In this case, the radiation did not influence the enzyme activity of liver cytochrome P450 and glutathione transferase in the terminal stages of Guerin’s carcinoma growth [57].

Table 2.

Effects of Ionizing and Non-ionizing Irradiation on Expression and Activity of Cytochrome P450 Enzymes

Species Enzyme Category Effectors Model; method used Effects Remarks References
Mice CPR (cytochrome P450 reductase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma-rays at 1.38 Gy/min, liver; activity activity increased up to 5 Gy (to 125%) and decreased thereafter (to 105% after 9 Gy) administration of phenothiazines enhanced the radiation effect at lower doses providing the radioprotective action [55]
Mice CYB5 (cytochrome b5) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma-rays at 1.38 Gy/min, liver; activity holoprotein increased up to 5 Gy (to 120%) and decreased thereafter (to 83% with 9 Gy) administration of phenothiazines enhanced the radiation effect at lower doses providing radioprotective action [55]
Mice CYB5R (cytochrome b5 reductase) Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of γ-rays at 1.38 Gy/min, liver; activity activity enhanced up to 3 Gy (to 125%)and decreased thereafter (to 104% after 9 Gy) administration of phenothiazines did not affect radiation effect [55]
Mice CYP Clinical impact γ-radiation, γ-rays whole body irradiation of mice with Ehrlich solid tumors with different doses of gamma rays (0–9 Gy) at a dose rate of 0.0153 Gy/s, solid tumor, microsomal fractions; content measured as CO difference spectrum increase of content with dose up to 6 Gy and decreased thereafter [56]
Mice CYP Environmental impact γ-radiation, γ-rays whole body irradiation, different doses of gamma-rays at 1.38 Gy/min, liver; activity holoprotein enhanced up to 5 Gy (to 119%) and decreased thereafter (to 108% after 9 Gy) administration of phenothiazines enhanced the radiation effect at lower doses providing the radioprotective action [55]
Human CYP Clinical impact Ionizing irradiation fractionated irradiation, ADF human astrocytoma cell line treated with 5 Gy for 4 consecutive days; immunoblotting, microarray analysis upregulation of gene expression suggested role in the reactive oxygen species (ROS) formation [64]
Rat CYP Clinical impact Ionizing irradiation preliminary radiation-exposed, transplanted Guerin’s carcinoma, liver, microsomes; activity decreased activity in the latent and logarithmic phases of oncogenesis, no effect on terminal stages of Guerin’s carcinoma growth [57]
Human CYP Environmental impact Radioactivity-contaminated areas placental samples at term, microsomes; 7-ethoxycoumarin O-deethylase (ECOD) enhanced ECOD activity ECOD activity was 7-fold and 2-fold higher compared to the region considered to be “clean”; increased formation of reactive metabolites suggested [54]
Rat CYP Environmental impact Uranium contamination short-term (3 and 30 days) and long-term (3–24 months) treatment with neutron-activated UO2 particles (9.3 kBq), liver microsomes; testosterone 7α- and 15α-hydroxylase activity decreased activity by 30% (7α-) at 3 days treatment, at 30 days after treatment activity enhanced by 70% (7α-) and 40% (15α-) [58]
Rat CYP Environmental impact Uranium contamination long-term (3–24 months) treatment with neutron-activated UO2 particles (9.3 kBq, cumulated lung dose 0.4–0.66 Gy, 131 and 182 kBq ), lung, liver; testosterone 7β-, 6α-, and 16α-hydroxylase activity at the 1.5-year treatment decreases in lung testosterone 6β-hydroxylase (60%) and testosterone 6α-hydroxylase (30%) activities, hepatic testosterone 16α-hydroxylase activity decreased by 60–75% with both non-activated and neutron-activated particles [58]
Human CYP Environmental impact UV irradiation cultures of fibrobroblasts in a collagen matrix as the dermal component and keratinocytes as the epidermal component, UVB irradiation; calcitriol formation from 7-dehydrocholesterol, HPLC, GC-MS wavelength- and dose dependent ultraviolet-B-triggered conversion of 7-dehydrocholesterol to calcitriol observed [10,11]
Rat CYP Clinical impact X-rays preliminary radiation-exposed rats with Guerin’s carcinoma, liver, microsomal fraction; activity activity reduced [57]
Human CYP19A1 (Aromatase) Environmental impact UV irradiation keratinocytes, combined UVA and UVB irradiation, microsomes; gel electrophoresis, RT-PCR slight induction of mRNA expression [8]
Zebrafish CYP1A Environmental impact UV irradiation embryos exposed for varying time of UVA plus UVB, or UVB alone on two consecutive days; spectophotometry, RT-PCR increase of mRNA expression [9]
Zebrafish CYP1A Environmental impact UV irradiation embryos exposed for varying time of UVA on two consecutive days; spectophotometry, RT-PCR no effect on mRNA expression [9]
Zebrafish CYP1A Environmental impact UV irradiation larvae exposed to single 8-h long UVB exposure; spectophotometry, RT-PCR increase of mRNA expression [9]
Rat CYP1A1 Environmental impact Uranium contamination chronically exposed to depleted uranium (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, brain, liver, lung, kidney, intestine; RT-PCR no change in expression of mRNA [60]
Human CYP1A1 Environmental impact UV irradiation keratinocytes, UVB irradiation; immunohistochemistry, semiquantitative RT-PCR, immunoblotting induction of mRNA and protein [61]
Human CYP1A1 Environmental impact UV irradiation hepatoma cell line HepG2, UVB irradiation; immunohistochemistry, RT-PCR, immunoblotting, 6-formylindolo[3,2-b]carbazole metabolism initial repression (3 hours after treatment) and induction of mRNA following prolonged treatment (9 hours after treatment), inhibition of activity [63]
Human CYP1A1 Environmental impact UV irradiation keratinocytes, UVB irradiation; RT-PCR induction of mRNA and protein induction was higher in the presence of tryptophan [62]
Human CYP1A1 Environmental impact UV irradiation primary human blood lymphocytes, UVB irradiation; immunohistochemistry, RT-PCR, immunoblotting induction of mRNA and protein induction was higher in the presence of tryptophan [62]
Mice CYP1A1 Environmental impact UV irradiation Hepa-1 cells, UVB irradiation; immunohistochemistry, RT-PCR, immunoblotting induction of mRNA and protein [62]
Rat CYP1A1 Environmental impact UV irradiation liver, UVB irradiation; EROD activity induction of activity [65]
Rat CYP1A2 Environmental impact Gamma radiation, Gamma-rays whole body irradiation of 3 gray (G) at a dosage rate of 12.5 cG/min from a 60Co radiation source, liver, microsomes; immunoblotting, mRNA blotting no change in the mRNA expression at 24 h [66]
Rat CYP1A2 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), plasma, liver, microsomes; chlorzoxazone pharmacokinetics i.v., microsomal metabolism, immunoblotting, mRNA blotting no change of protein or mRNA expression induced acute renal failure observed [67,68,69,70,71]
Human CYP1B1 Environmental impact UV irradiation keratinocytes, UVB irradiation; immunohistochemistry, semiquantitative RT-PCR, immunoblotting induction of mRNA and protein connected with enhanced bioactivation of polycyclic aromatic hydrocarbons and other environmental pollutants [61]
Zebrafish CYP1B1 Environmental impact UV irradiation embryos exposed for varying time of UVB on two consecutive days; spectophotometry, RT-PCR increase of mRNA expression [9]
Zebrafish CYP1B1 Environmental impact UV irradiation larvae exposed to single 8-h long UVB exposure; spectophotometry, RT-PCR increase of mRNA expression [9]
Zebrafish CYP1C1 Environmental impact UV irradiation larvae exposed to single 8-h long UVB exposure; spectophotometry, RT-PCR no effect on mRNA expression [9]
Zebrafish CYP1C2 Environmental impact UV irradiation larvae exposed to single 8-h long UVB exposure; spectophotometry, RT-PCR no effect on mRNA expression [9]
Zebrafish CYP1D1 Environmental impact UV irradiation larvae exposed to single 8-h long UVB exposure; spectophotometry, RT-PCR no effect on mRNA expression [9]
Rat CYP24A1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), kidney; RT-PCR, activity ([4-14C]cholesterol as substrate) no significant change of mRNA expression [72]
Rat CYP24A1 Environmental impact Uranium contamination single acute depleted uranium (as uranyl nitrate) intragastric administration (204 mg/kg body weight dissolved in 1.5 ml), kidney; RT-PCR no change in expression of mRNA contamination by short-term exposure to depleted uranium [73]
Rat CYP24A1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, brain, mitochondria; RT-PCR mRNA not detected dose corresponds to the double of highest concentration found naturally in Finland [74]
Rat CYP24A1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, kidney, mitochondria; RT-PCR decreased mRNA expression by 38%, dose corresponds to the double of highest concentration found naturally in Finland [74]
Rat CYP24R1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, liver, mitochondria; RT-PCR no change of mRNA level expression dose corresponds to the double of highest concentration found naturally in Finland [74]
Rat CYP27A1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), liver, mitochondria; quantitative RT-PCR, [4-14C]Cholesterol 27-hydroxyation activity no significant change of mRNA expression, increase of activity by 34% [37,72,96]
Rat CYP27A1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), brain; RT-PCR no significant change of mRNA expression [72]
Rat CYP27A1 Environmental impact Uranium contamination single acute depleted uranium (as uranyl nitrate) intragastric administration (204 mg/kg body weight dissolved in 1.5 ml), liver, mitochondria; RT-PCR, [4-14C]Cholesterol 27-hydroxyation activity no gross modifications in the expression, activity decreased at day 1 days and increased (threefold) at day 3 after treatment [73]
Rat CYP27A1 Environmental impact Uranium contamination single depleted uranium (as uranyl nitrate) subcutaneous administration, sublethal toxic dose of 11.5 mg/kg, liver, mitochondria; [4-14C]cholesterol 27-hydroxylation activity activity quintupled at day 1 after treatment and then returned to levels similar to controls at day 3 [59]
Rat CYP27A1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, brain, mitochondria; RT-PCR decreased mRNA level expression by 32%, activity decreased at day 1 days and increased (threefold) at day 3 after treatment dose corresponds to the double of highest concentration found naturally in Finland [74]
Rat CYP27A1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, liver, mitochondria; RT-PCR, [4-14C] cholesterol 27-hydroxyastion activity no change of mRNA level expression or activity dose corresponds to the double of highest concentration found naturally in Finland [74]
Rat CYP27B1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), brain; RT-PCR increase of mRNA expression by 35% [72]
Rat CYP27B1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), kidney; RT-PCR no significant change of mRNA expression [72]
Rat CYP27B1 Environmental impact Cesium contamination newborn rats chronically exposed with post-accidental doses of 137Cs in drinking water durin lactation period, at a dose of 6500 Bq/l (150 Bq/rat/day), liver, kidney; RT-PCR decrease of mRNA expression (by 39%) [75]
Rat CYP27B1 Environmental impact Uranium contamination single acute depleted uranium (as uranyl nitrate) intragastric administration (204 mg/kg body weight dissolved in 1.5 ml), kidney; RT-PCR increase of mRNA expression at days 1 and 3 after treatment (11- and 4-fold respectively) [73]
Rat CYP27B1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, kidney, mitochondria; RT-PCR no change of mRNA level expression dose corresponds to the double of highest concentration found naturally in Finland [74]
Rat CYP2A Environmental impact Uranium contamination chronically exposed to depleted uranium (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, liver microsomes; testosterone 7α-hydroxylase activity no change in hepatic activity [60]
Rat CYP2B Environmental impact Uranium contamination chronically exposed to depleted uranium (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, liver microsomes; testosterone 16α-hydroxylase activity no change in hepatic activity [60]
Rat CYP2B Environmental impact UV irradiation liver, UVB irradiation; ADM activity no change of activity [65]
Rat CYP2B1 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), liver, microsomes; Western blotting, Northern blotting no change of protein or mRNA expression induced acute renal failure observed [67,68,69,70,71]
Rat CYP2B1 Environmental impact Uranium contamination chronically exposed to depleted uranium (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, brain, liver, lung, kidney, intestine; RT-PCR increase of mRNA expression in kidney [60]
Rat CYP2B2 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), liver, microsomes; immunoblotting, mRNA blotting no change of protein or mRNA expression induced acute renal failure observed [67,68,69,70,71]
Rat CYP2C Environmental impact Uranium contamination chronically exposed to depleted uranium (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, liver microsomes; testosterone 2α-hydroxylase activity no change in hepatic activity [60]
Rat CYP2C11 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), liver, microsomes; immunoblotting, mRNA blotting decrease of protein level to 20% and mRNA expression to 25% of control induced acute renal failure observed [67,68,69,70,71]
Mice CYP2E1 Environmental impact Fast neutron irradiation whole body fast neutron irradiation of 0, 0.25, 1, 2, 4 and 8 Gy, liver samples, hepatocytes; histopathology, immunohistochemistry dose-dependent increase of protein expression [78]
Mice CYP2E1 Environmental impact γ-radiation, γ-rays low doses of continuous γ-radiation, liver; decreased mRNA expression and protein levels [79]
Mice CYP2E1 Environmental impact γ-radiation, γ-rays low doses of acute γ-radiation, liver; increased protein level, decreased mRNA expression [79]
Mice CYP2E1 Environmental impact γ-radiation, γ-rays high doses of acute γ-radiation, liver; decreased protein level, decreased mRNA expression [79]
Mice CYP2E1 Environmental impact γ-radiation, γ-rays low intensity gamma-radiation and ethanol combined administration, liver; protein level increased in the first week, back to normal on second week changes of CYP2E1 protein amount at the end of the fifth week accompanied by a decrease of CYP2E1 mRNA level [80]
Rat CYP2E1 Environmental impact γ-radiation, γ-rays whole body irradiation of 3 gray (G) at a dosage rate of 12.5 cG:min from a 60Co radiation source, liver; immunoblotting, mRNA blotting increased mRNA (3.6-fold) and protein (2.5-fold) expression at 24 h [66]
Rat CYP2E1 Environmental impact γ-radiation, γ-rays whole body irradiation of 3 gray (G) at a dosage rate of 12.5 cG/min from a 60Co radiation source; chlorzoxazone pharmacokinetics i.v. significantly greater plasma concentration-time curve and the amount of 6-hydroxychlorzoxazone excreted in 8 h urine [66]
Rat CYP2E1 Environmental impact γ-radiation, γ-rays whole body irradiation of 0.5–1 gray (G) at a dosage rate of 12.5 cG/min from a 60Co radiation source, liver, microsomes; immunoblotting, mRNA blotting no change of mRNA expression [66]
Rat CYP2E1 Environmental impact γ-radiation, γ-rays whole body irradiation of 3–9 gray (G) at a dosage rate of 12.5 cG/min from a 60Co radiation source, liver microsomes small but significant increase mRNA expression at 24 h than those irradiated at a single dose of 3 G g-rays liver injury observed [66]
Rat CYP2E1 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), plasma, liver, microsomes; chlorzoxazone CZX pharmacokinetics i.v., microsomal metabolism, immunoblotting, mRNA blotting increase of protein level 2.3 times and mRNA expression 3 times, increase of activity induced acute renal failure, subcutaneous injection of rHGH for one day on the fourth day after uranyl nitrate or glucose (dissolved drinking water for 5 days) reduced the expression of CYP2E1 [67,68,69,70,71]
Rat CYP2R1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), liver; RT-PCR increase of mRNA expression (by 40%) [72]
Rat CYP2R1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), brain; RT-PCR decrease of mRNA expression (by 20%) [72]
Rat CYP2R1 Environmental impact Cesium contamination newborn rats chronically exposed with post-accidental doses of 137Cs in drinking water during lactation period, at a dose of 6500 Bq/l (150 Bq/rat/day), liver, kidney; RT-PCR decrease of mRNA expression (by 26%) [75]
Rat CYP2R1 Environmental impact Uranium contamination single acute depleted uranium (as uranyl nitrate) intragastric administration (204 mg/kg body weight dissolved in 1.5 ml), liver, mitochondria; RT-PCR, activity ([4-14C]cholesterol as substrate) no gross modifications in the expression, slight increase of mRNA expression at day 3 after treatment [73]
Rat CYP3A Environmental impact γ-radiation, γ-rays whole body irradiation of 3 gray (G) at a dosage rate of 12.5 cG/min from a 60Co radiation source, liver, microsomes; immunoblotting, mRNA blotting no change in the mRNA expression at 24 h [66]
Rat CYP3A Environmental impact Uranium contamination chronically exposed to depleted (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, liver microsomes; testosterone 6β-hydroxylation activity no change in hepatic activity [60]
Rat CYP3A Environmental impact Uranium contamination single subcutaneous administration of depleted uranium, sublethal toxic dose of 11.5 mg/kg, liver, microsomes; testosterone 6β-hydroxylation activity decrease of activity at day 1 but returned to levels similar to controls at day 3 [59]
Rat CYP3A Environmental impact UV irradiation liver, UVB irradiation; EMDM activity no change of activity [65]
Rat CYP3A Environmental impact UV irradiation skin, AVA and UVB irradiation; EROD activity induction of activity [65]
Rat CYP3A1 Environmental impact Uranium contamination chronically exposed to depleted (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, brain, liver, lung, kidney, intestine; RT-PCR increase of mRNA expression in brain, liver, and kidney stimulatory effect might lead to hepatic or extrahepatic toxicity (or both) during drug treatment [60]
Rat CYP3A1 Environmental impact Uranium contamination single subcutaneous administration of depleted uranium (as uranyl nitrate) sublethal toxic dose of 11.5 mg/kg, liver, microsomes; RT-PCR increase in expression of mRNA 3 days after treatment, no change at day1 [59]
Rat CYP3A2 Environmental impact Uranium contamination chronically exposed to depleted (as uranyl nitrate) in drinking water, 1 mg/(rat day) for 9 months, brain, liver, lung, kidney, intestine; RT-PCR increase of mRNA expression in lungs and liver stimulatory effect might lead to hepatic or extrahepatic toxicity (or both) during drug treatment [60]
Rat CYP3A2 Environmental impact Uranium contamination single subcutaneous administration of depleted uranium (as uranyl nitrate) sublethal toxic dose of 11.5 mg/kg, liver, microsomes; RT-PCR no change in expression of mRNA [59]
Rat CYP3A2 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), liver, microsomes; immunoblotting, mRNA blotting no change of protein or mRNA expression induced acute renal failure observed [68,71]
Rat CYP3A23 Environmental impact Uranium contamination uranyl nitrate solution injected once via the tail vein (5 mg/kg), liver, microsomes; immunoblotting, mRNA blotting increase of protein level 4 times, no change of mRNA expression induced acute renal failure observed [67,68,69,70]
Rat CYP46A1 Environmental impact Uranium contamination chronic contamination for 9 months by depleted uranium (uranyl nitrate) through drinking water of dose 1 mg/rat/day, brain; RT-PCR increase of mRNA expression by 39% dose corresponds to the double of highest concentration found naturally in Finland [36]
Human CYP4A11 Environmental impact UV irradiation keratinocytes, UVA irradiation, microsomes; gel electrophoresis, RT-PCR, immunoblotting, thin-layer chromatography mRNA expression detected mRNA was not detected in any keratinocyte preparations under control conditions, proposed that CYP4A11 may participate in the defense mechanism against UVA-induced oxidative damage [8]
Human CYP4A11 Environmental impact UV irradiation keratinocytes, combined UVA and UVB or UVA irradiation, microsomes; gel electrophoresis, RT-PCR, immunoblotting, thin layer chromatography induction of mRNA and protein mRNA was not detected in any keratinocyte preparations under control conditions, proposed that CYP4A11 may participate in the defense mechanism against UVA-induced oxidative damage [8]
Human CYP4A11 Environmental impact UV irradiation keratinocytes, UVB irradiation, microsomes; gel electrophoresis, RT-PCR mRNA not detected [8]
Rat CYP7A1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 3 months, at a dose of 6500 Bq/l (150 Bq/rat/day), liver, microsomes; quantitative RT-PCR, activity with [4-14C]Cholesterol as substrate no change in expression of mRNA and activity [37,96]
Rat CYP7A1 Environmental impact Uranium contamination single subcutaneous administration of depleted uranium (as uranyl nitrate) sublethal toxic dose of 11.5 mg/kg, liver, microsomes; [4-14C]cholesterol 7α-hydroxyase activity no significant change in the activity [59]
Rice CYP84A Environmental impact UV irradiation UVB or UVC irradiation on 1- and 2-week-old plants for 6 hours; semiquantitative RT-PCR increase of gene expression under UVB and UVC irradiation protection against damage due to UV irradiation proposed [76]
Rat CYP27A1 Environmental impact Cesium contamination chronic exposure with post-accidental doses of 137Cs in drinking water for 9 months, at a dose of 6500 Bq/l (150 Bq/rat/day), brain; RT-PCR decrease of mRNA expression plasma profile, and brain and liver cholesterol concentrations were unchanged [77]
Rat CYP7A1 Environmental impact Uranium contamination low-level chronic ingestion of depleted uranium in drinking water for 9 months, 40 mg/kg, liver; RT-PCR, specific activity decrease of the activity [39]
Rat CYP7B1 Environmental impact Uranium contamination low-level chronic ingestion of depleted uranium in drinking water for 9 months, 40 mg/kg, liver; RT-PCR, specific activity decrease of expression [39]

The complexity of effects of radiation on cytochrome P450 activity are exemplified in the results obtained after short-term (3 and 30 day) and long-term (3–24 month) treatment of rats with neutron-activated UO2 particles (9.3 kBq) [58]. After 3 days of treatment a decreased activity of 30% (testosterone 7α-hydroxylase) was observed, and after 30 days after treatment activity was enhanced by 70% (testosterone 7α-hydroxylase) and 40% (testosterone 15α-hydroxylase). After 1.5-year treatment, decreases in activity in lung testosterone 6β- (60%) and 6α-hydroxylase (30%) occurred; hepatic testosterone 16α-hydroxylase activity was decreased by 60–75% with both non-activated and neutron-activated particles [58].

A single subcutaneous administration of depleted uranium (as uranyl nitrate) to rats (at a sublethal toxic dose) affected bile acid cytochrome P450 activity in the following way: 7α-hydroxycholesterol plasma levels decreased by 52% at day 3, whereas microsomal CYP7A1 activity in the liver did not change significantly and mitochondrial CYP27A1 activity quintupled after day 1 [59]. Chronic exposure of rats with post-accidental doses of 137Cs in drinking water for 3 months showed no significant change of CYP27A1 mRNA expression but increased the activity by 34% in rat liver [37,72,96]. However, experiments with rats chronically exposed to depleted uranium in drinking water (1 mg/rat day) for 9 months showed that CYP3A1 mRNA expression was significantly higher in the brain (200%), liver (300%), and kidneys (900%) of exposed rats compared with control rats, and CYP3A2 mRNA levels were higher in the lungs (300%) and liver (200%), and CYP2B1 mRNA expression was elevated in the kidneys (300%). Expression of CYP1A1 mRNA did not change significantly during this study. It was suggested that the stimulating effect of uranium on cytochrome P450 enzymes might lead to hepatic or extrahepatic toxicity (or both) during drug treatment [60]. For further effects of ionizing irradiation on cytochrome P450 enzymes see Table 2.

The effects of UV irradiation were examined using human keratinocytes, and it was reported that combined UVA and UVB—UVA irradiation alone—caused induction of CYP4A11 mRNA expression and protein expression levels [8]. CYP1B1 mRNA and protein expression were induced by UVB irradiation of keratinocytes [51]. CYP1A1 was induced in keratinocytes, primary human blood lymphocytes, and a hepatoma cell line [61,62,63] slight induction of CYP19A1 (aromatase) mRNA was induced by combined UVA and UVB irradiation in keratinocytes [8]. Increased mRNA expression of toxicologically important CYP1A was observed in zebrafish larvae and embryos (exposed for varying time to UVA plus UVB, or UVB alone on two consecutive days), while embryos exposed to UVA alone had no effect on mRNA expression and UVB irradiation increased mRNA expression of CYP1B1 [9]. Induction of CYP1A1 and CYP1B1 mRNA and protein expression levels was suggested to be related to enhanced bioactivation of polycyclic aromatic hydrocarbons and other environmental pollutants in skin [61].

OXIDATIVE STRESS AND OTHER ENZYMES

Different effects (Table 3) on the oxidative stress enzymes (catalase (CAT), NADPH-quinone reductase (NOR), glyoxylase I (GLO1), superoxide dismutase (SOD)) were obtained after whole body irradiation of mice with Ehrlich solid tumor and non-tumor bearing animals with different doses of γ-radiation (0–9 Gy). While the activity of most of the enzymes increased with the radiation dose in the both tumor-bearing and non-tumor bearing mice up to 6 Gy and decreased thereafter (NADPH-quinone reductase, glyoxalase I, superoxide dismutase, xanthine oxidoreductase) [55,56,81], catalase was either not detected or decreased with the irradiation dose in both tumor and non-tumor animals [56,81]. At the same time a progressive increase was noticed in peroxidative damage with increasing irradiation dose [56]. As with the effects on cytochrome P450 enzymes (Table 2), administration of phenothiazines enhanced the effect of radiation at lower doses on the activities of superoxide dismutase and NADPH-quinone reductase but inhibited the activity of L-lactate dehydrogenase (LDH). The effect of phenothiazines was also connected to radioprotective action at lower irradiation doses in this case. In addition, phenothiazines inhibited lipid peroxidation and xanthine oxidoreductase (XOR) [55]. Taking superoxide dismutase as an example, the effects of the ionizing irradiation were the following: the lower doses of γ-irradiation or ionizing radiation (up to 6 Gy) enhanced the enzyme activity, but decreases of activity and protein levels were observed at higher doses. This result was confirmed using different models and methods, including human models [55,82,83]. It was reported that the decrease of the activity and protein levels at higher radiation doses could be reversed by supplementation with extracts of Xylopia aethiopica and vitamin C [82]. At lower occupational doses (e.g., exposed medical workers), enhanced activity of superoxide dismutase might provide protection against the increased production of reactive oxygen species but that dysfunction of enzyme system can occur following higher doses of irradiation [84]. Similarly, chronic UV irradiation also caused increased superoxide dismutase activity and mRNA expression in both human and in animal models [9,85] but decreased activity was observed after single UVB radiation of mice skin [86].

CONCLUSION

As reported before [7], increases in expression levels of cytochrome P450 enzymes (for instance CYP1B1 and CYP2J2) and transporters (MRP1) in tumor cells and tissues have been suggested as tumor markers in the diagnosis and prognosis of malignancies and/or other diseases. However, results reported before [1,2] and in this summary show that when using such markers and before complex considerations one has to take into account the effects of drug and diseases, as well as natural (UV) or “artificial” radiation coming from the environment (e. g., nuclear plant disasters, radon exposure) or applied clinically (γ- and X-rays). This summary presents the complex effects of different types of radiation on the activity and expression of both the enzymes (e.g., cytochrome P450, oxidative stress enzymes) and transporters, and shows that these effects might impact ADME properties of drugs.

Acknowledgments

This work is dedicated to the family of the corresponding author (R.S.), including his wife Vjekoslava, son Borut, and daughter Petra.

Footnotes

DECLARATION OF INTEREST:

The authors report no financial conflicts of interest.

References

  • 1.Rendic S, Di Carlo F. Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers and inhibitors. Drug Metab Rev. 1997;29(1–2):413–580. doi: 10.3109/03602539709037591. [DOI] [PubMed] [Google Scholar]
  • 2.Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34(1–2):83–448. doi: 10.1081/dmr-120001392. [DOI] [PubMed] [Google Scholar]
  • 3.Guengerich FP. Update information on human P450s. Drug Metab Rev. 2002;34(1–2):7–15. doi: 10.1081/dmr-120001386. [DOI] [PubMed] [Google Scholar]
  • 4.Guengerich FP, Rendic S. Update information on drug metabolism systems - 2009; Part I. Curr Drug Metab. 2010;11(1):1–3. doi: 10.2174/138920010791110908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kim RB. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34(1–2):47–54. doi: 10.1081/dmr-120001389. [DOI] [PubMed] [Google Scholar]
  • 6.Gradhand U, Kim RB. Pharmacogenomics of MRP transporters (ABCC1-5) and BCRP (ABCG2) Drug Metab Rev. 1998;40(2):317–354. doi: 10.1080/03602530801952617. [DOI] [PubMed] [Google Scholar]
  • 7.Rendic S, Guengerich FP. Update information on drug metabolism systems - 2009; Part II. Curr Drug Metab. 2010;11(1):4–84. doi: 10.2174/138920010791110908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gonzalez MC, Marteau C, Franchi J, Migliore-Samour D. Cytochrome P450 4A11 expression in human keratinocytes: effects of ultraviolet irradiation. Br J Dermatol. 2001;145(5):749–757. doi: 10.1046/j.1365-2133.2001.04490.x. [DOI] [PubMed] [Google Scholar]
  • 9.Behrendt L, Jönsson ME, Goldstone JV, Stegeman JJ. Induction of cytochrome P450 1 genes and stress response genes in developing zebrafish exposed to ultraviolet radiation. Aquat Toxicol. 2010;98(1):74–82. doi: 10.1016/j.aquatox.2010.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lehmann B, Knuschke P, Meurer M. UVB-induced conversion of 7-dehydrocholesterol to 1α,25-dihydroxyvitamin D3 (calcitriol) in the human keratinocyte line HaCaT. Photochem Photobiol. 2000;72(6):803–809. doi: 10.1562/0031-8655(2000)072<0803:uicodt>2.0.co;2. [DOI] [PubMed] [Google Scholar]
  • 11.Lehmann B, Genehr T, Knuschke P, Pietzsch J, Meurer M. UVB-induced conversion of 7-dehydrocholesterol to 1α,25-dihydroxyvitamin D3 in an in vitro human skin equivalent model. J Invest Dermatol. 2001;117(5):1179–1185. doi: 10.1046/j.0022-202x.2001.01538.x. [DOI] [PubMed] [Google Scholar]
  • 12.Faneyte IF, Kristel PM, van de Vijver MJ. Determining MDR1/P-glycoprotein expression in breast cancer. Int J Cancer. 2001;93(1):114–122. doi: 10.1002/1097-0215(20010701)93:1<114::aid-ijc1309>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
  • 13.Guengerich FP, Wang P, Davidson NK. Estimation of isozymes of microsomal cytochrome P-450 in rats, rabbits, and humans using immunochemical staining coupled with sodium dodecyl sulfate-polyacrylamide gel electrophoresis. 1982;21(7):1698–1706. doi: 10.1021/bi00536a035. [DOI] [PubMed] [Google Scholar]
  • 14.Swerts K, De Moerloose B, Dhooge C, Laureys G, Benoit Y, Philippé J. Prognostic significance of multidrug resistance-related proteins in childhood acute lymphoblastic leukaemia. Eur J Cancer. 2006;42(3):295–309. doi: 10.1016/j.ejca.2005.09.017. [DOI] [PubMed] [Google Scholar]
  • 15.Osmak M, Miljanić S, Kapitanović S. Low doses of gamma-rays can induce the expression of mdr gene. Mutat Res. 1994;324(1–2):35–41. doi: 10.1016/0165-7992(94)90065-5. [DOI] [PubMed] [Google Scholar]
  • 16.Harvie RM, Davey MW, Davey RA. Increased MRP expression is associated with resistance to radiation, anthracyclines and etoposide in cells treated with fractionated gamma-radiation. Int J Cancer. 1997;73(1):164–167. doi: 10.1002/(sici)1097-0215(19970926)73:1<164::aid-ijc25>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 17.Bu JG, Yuan YW, Chen J. Expression of multidrug resistance gene in nasopharyngeal carcinoma cells after irradiation exposure. Di Yi Jun Yi Da Xue Xue Bao. 2004;24(6):646–649. [PubMed] [Google Scholar]
  • 18.Korystov YN, Shaposhnikova VV, Korystova AF, Emel’yanov MO, Kublik LN. Modification of multidrug resistance of tumor cells by ionizing radiation. Cancer Chemother Pharmacol. 2008;61(1):15–21. doi: 10.1007/s00280-007-0439-2. [DOI] [PubMed] [Google Scholar]
  • 19.Shareef MM, Brown B, Shajahan S, Sathishkumar S, Arnold SM, Mohiuddin M, Ahmed MM, Spring PM. Lack of P-glycoprotein expression by low-dose fractionated radiation results from loss of nuclear factor-kappaB and NF-Y activation in oral carcinoma cells. Mol Cancer Res. 2008;6(1):89–98. doi: 10.1158/1541-7786.MCR-07-0221. [DOI] [PubMed] [Google Scholar]
  • 20.Bottke D, Koychev D, Busse A, Heufelder K, Wiegel T, Thiel E, Hinkelbein W, Keilholz U. Fractionated irradiation can induce functionally relevant multidrug resistance gene and protein expression in human tumor cell lines. Radiat Res. 2008;170(1):41–48. doi: 10.1667/RR0986.1. [DOI] [PubMed] [Google Scholar]
  • 21.Tsang TY, Tsang SW, Lai KP, Tsang WP, Co NN, Kwok TT. Facilitation of drug resistance development by gamma-irradiation in human cancer cells. Oncol Rep. 2009;22(4):921–926. doi: 10.3892/or_00000518. [DOI] [PubMed] [Google Scholar]
  • 22.Bartkowiak D, Stempfhuber M, Wiegel T, Bottke D. Radiation- and chemoinduced multidrug resistance in colon carcinoma cells. Strahlenther Onkol. 2009;185(12):815–820. doi: 10.1007/s00066-009-1993-9. [DOI] [PubMed] [Google Scholar]
  • 23.Nielsen D, Maare C, Eriksen J, Litman T, Skovsgaard T. Expression of P-glycoprotein and multidrug resistance associated protein in Ehrlich ascites tumor cells after fractionated irradiation. Int J Radiat Oncol Biol Phys. 2001;51(4):1050–1057. doi: 10.1016/s0360-3016(01)01719-9. [DOI] [PubMed] [Google Scholar]
  • 24.Hill BT, Whelan RD, Hurst HC, McClean S. Identification of a distinctive P-glycoprotein-mediated resistance phenotype in human ovarian carcinoma cells after their in vitro exposure to fractionated X-irradiation. Cancer. 1994;73(12):2990–2999. doi: 10.1002/1097-0142(19940615)73:12<2990::aid-cncr2820731217>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
  • 25.Henness S, Davey MW, Harvie RM, Davey RA. Fractionated irradiation of H69 small-cell lung cancer cells causes stable radiation and drug resistance with increased MRP1, MRP2, and topoisomerase IIalpha expression. In J Radiat Oncol Biol Phys. 2002;54(3):895–902. doi: 10.1016/s0360-3016(02)03037-7. [DOI] [PubMed] [Google Scholar]
  • 26.Mattern J, Efferth T, Volm M. Overexpression of P-glycoprotein in human lung carcinoma xenografts after fractionated irradiation in vivo. Radiat Res. 1991;127(3):335–338. [PubMed] [Google Scholar]
  • 27.Hill BT. Differing patterns of cross-resistance resulting from exposures to specific antitumour drugs or to radiation in vitro. Cytotechnology. 1993;12(1–3):265–288. doi: 10.1007/BF00744668. [DOI] [PubMed] [Google Scholar]
  • 28.McClean S, Hill BT. Modified multiple drug resistance phenotype of Chinese hamster ovary cells selected with X-rays and vincristine versus X-rays only. Br J Cancer. 1994;69(4):711–716. doi: 10.1038/bjc.1994.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.McClean S, Hosking LK, Hill BT. Dominant expression of multiple drug resistance after in vitro X-irradiation exposure in intraspecific Chinese hamster ovary hybrid cells. J Natl Cancer Inst. 1993;85(1):48–53. doi: 10.1093/jnci/85.1.48. [DOI] [PubMed] [Google Scholar]
  • 30.McClean S, Whelan RD, Hosking LK, Hodges GM, Thompson FH, Meyers MB, Schuurhuis GJ, Hill BT. Characterization of the P-glycoprotein over-expressing drug resistance phenotype exhibited by Chinese hamster ovary cells following their in-vitro exposure to fractionated X-irradiation. Biochim Biophys Acta. 1993;1177(2):117–126. doi: 10.1016/0167-4889(93)90030-s. [DOI] [PubMed] [Google Scholar]
  • 31.McClean S, Hosking LK, Hill BT. Expression of P-glycoprotein-mediated drug resistance in CHO cells surviving a single X-ray dose of 30 Gy. Int J Radiat Biol. 1993;63(6):765–773. doi: 10.1080/09553009314552171. [DOI] [PubMed] [Google Scholar]
  • 32.Hill BT, Deuchars K, Hosking LK, Ling V, Whelan RD. Overexpression of P-glycoprotein in mammalian tumor cell lines after fractionated X irradiation in vitro. J Natl Cancer Inst. 1990;82(7):607–612. doi: 10.1093/jnci/82.7.607. [DOI] [PubMed] [Google Scholar]
  • 33.Ryu JS, Um JH, Kang CD, Bae JH, Kim DU, Lee YJ, Kim DW, Chung BS, Kim SH. Fractionated irradiation leads to restoration of drug sensitivity in MDR cells that correlates with down-regulation of P-gp and DNA-dependent protein kinase activity. Radiat Res. 2004;162(5):527–535. doi: 10.1667/rr3260. [DOI] [PubMed] [Google Scholar]
  • 34.Mima T, Toyonaga S, Mori K, Taniguchi T, Ogawa Y. Early decrease of P-glycoprotein in the endothelium of the rat brain capillaries after moderate dose of irradiation. Neurol Res. 1999;21(2):209–215. doi: 10.1080/01616412.1999.11740920. [DOI] [PubMed] [Google Scholar]
  • 35.Bart J, Nagengast WB, Coppes RP, Wegman TD, van der Graaf WT, Groen HJ, Vaalburg W, de Vries EG, Hendrikse NH. Irradiation of rat brain reduces P-glycoprotein expression and function. Br J Cancer. 2007;97(3):322–326. doi: 10.1038/sj.bjc.6603864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Racine R, Gueguen Y, Gourmelon P, Veyssiere G, Souidi M. Modifications of the expression of genes involved in cerebral cholesterol metabolism in the rat following chronic ingestion of depleted uranium. J Mol Neurosci. 2009;38(2):159–165. doi: 10.1007/s12031-008-9145-8. [DOI] [PubMed] [Google Scholar]
  • 37.Souidi M, Tissandie E, Grandcolas L, Grison S, Paquet F, Voisin P, Aigueperse J, Gourmelon P, Guéguen Y. Chronic contamination with 137cesium in rat: effect on liver cholesterol metabolism. Int J Toxicol. 2006;25(6):493–497. doi: 10.1080/10915810600961317. [DOI] [PubMed] [Google Scholar]
  • 38.Lestaevel P, Bensoussan H, Racine R, Airault F, Gourmelon P, Souidi M. Transcriptomic effects of depleted uranium on acetylcholine and cholesterol metabolisms in Alzheimer’s disease model. C R Biol. 2011;334(2):85–90. doi: 10.1016/j.crvi.2010.12.004. [DOI] [PubMed] [Google Scholar]
  • 39.Racine R, Grandcolas L, Grison S, Stefani J, Delissen O, Gourmelon P, Veyssière G, Souidi M. Cholesterol 7alpha-hydroxylase (CYP7A1) activity is modified after chronic ingestion of depleted uranium in the rat. J Steroid Biochem Mol Biol. 2010;120(1):60–66. doi: 10.1016/j.jsbmb.2010.03.066. [DOI] [PubMed] [Google Scholar]
  • 40.Trog D, Moenkemann H, Haertel N, Schüller H, Golubnitschaja O. Expression of ABC-1 transporter is elevated in human glioma cells under irradiation and temozolomide treatment. Amino Acids. 2005;28(2):213–219. doi: 10.1007/s00726-005-0171-3. [DOI] [PubMed] [Google Scholar]
  • 41.Sanchez MC, Benitez A, Ortloff L, Green LM. Alterations in glutamate uptake in NT2-derived neurons and astrocytes after exposure to gamma radiation. Radiat Res. 2009;171(1):41–52. doi: 10.1667/RR1361.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Stammler G, Pommerenke EW, Masanek U, Mattern J, Volm MJ. Messenger RNA expression of resistance factors in human tumor cell lines after single exposure to radiation. Exp Ther Oncol. 1996;1(1):39–48. [PubMed] [Google Scholar]
  • 43.Sanchez AM, Barrett JT, Schoenlein PV. Fractionated ionizing radiation accelerates loss of amplified MDR1 genes harbored by extrachromosomal DNA in tumor cells. Cancer Res. 1998;58(17):3845–3854. [PubMed] [Google Scholar]
  • 44.Zhang GJ, Gao R, Wang JS, Fu JK, Zhang MX, Jin X. Various doses of fractioned irradiation modulates multidrug resistance 1 expression differently through hypoxia-inducible factor 1α in esophageal cancer cells. Dis Esophagus. 2011:1442–2050. doi: 10.1111/j.1442-2050.2010.01168.x. [DOI] [PubMed] [Google Scholar]
  • 45.Stammler G, Pommerenke EW, Mattern J, Volm M. Effects of single doses of irradiation on the expression of resistance-related proteins in murine NIH 3T3 and human lung carcinoma cells. Carcinogenesis. 1995;16(9):2051–2055. doi: 10.1093/carcin/16.9.2051. [DOI] [PubMed] [Google Scholar]
  • 46.Akashi-Tanaka S, Fukutomi T, Tsuda H. Higher Level of P-glycoprotein expression in human breast cancer cells after radiation therapy. Breast Cancer. 1995;2(2):99–103. doi: 10.1007/BF02966947. [DOI] [PubMed] [Google Scholar]
  • 47.Christiansen H, Batusic D, Saile B, Hermann RM, Dudas J, Rave-Frank M, Hess CF, Schmidberger H, Ramadori G. Identification of genes responsive to gamma radiation in rat hepatocytes and rat liver by cDNA array gene expression analysis. Radiat Res. 2006;165(3):318–325. doi: 10.1667/rr3503.1. [DOI] [PubMed] [Google Scholar]
  • 48.Roche M, Neti PV, Kemp FW, Agrawal A, Attanasio A, Douard V, Muduli A, Azzam EI, Norkus E, Brimacombe M, Howell RW, Ferraris RP. Radiation-induced reductions in transporter mRNA levels parallel reductions in intestinal sugar transport. Am J Physiol Regul Integr Comp Physiol. 2010;298(1):R173–182. doi: 10.1152/ajpregu.00612.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hu Z, Jin S, Scotto KW. Transcriptional activation of the MDR1 gene by UV irradiation. Role of NF-Y and Sp1. J Biol Chem. 2000;275(4):2979–2985. doi: 10.1074/jbc.275.4.2979. [DOI] [PubMed] [Google Scholar]
  • 50.Uchiumi T, Kohno K, Tanimura H, Matsuo K, Sato S, Uchida Y, Kuwano M. Enhanced expression of the human multidrug resistance 1 gene in response to UV light irradiation. Cell Growth Differ. 1993;4(3):147–157. [PubMed] [Google Scholar]
  • 51.Ohga T, Uchiumi T, Makino Y, Koike K, Wada M, Kuwano M, Kohno K. Direct involvement of the Y-box binding protein YB-1 in genotoxic stress–induced activation of the human multidrug resistance 1 gene. J Biol Chem. 1998;273(11):5997–6000. doi: 10.1074/jbc.273.11.5997. [DOI] [PubMed] [Google Scholar]
  • 52.Ng IO, Lam KY, Ng M, Kwong DL, Sham JS. Expression of P-glycoprotein, a multidrug-resistance gene product, is induced by radiotherapy in patients with oral squamous cell carcinoma. Cancer. 1998;83(5):851–857. doi: 10.1002/(sici)1097-0142(19980901)83:5<851::aid-cncr8>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
  • 53.Hill BT, Moran E, Etiévant C, Perrin D, Masterson A, Larkin A, Whelan RD. Low-dose twice-daily fractionated X-irradiation of ovarian tumor cells in vitro generates drug-resistant cells overexpressing two multidrug resistance-associated proteins, P-glycoprotein and MRP1. Anticancer Drugs. 2000;11(3):193–200. doi: 10.1097/00001813-200003000-00007. [DOI] [PubMed] [Google Scholar]
  • 54.Obolenskaya MY, Teplyuk NM, Divi RL, Poirier MC, Filimonova NB, Zadrozna M, Pasanen MJ. Human placental glutathione S-transferase activity and polycyclic aromatic hydrocarbon DNA adducts as biomarkers for environmental oxidative stress in placentas from pregnant women living in radioactivity- and chemically-polluted regions. Toxicol Lett. 2010;196(2):80–86. doi: 10.1016/j.toxlet.2010.03.1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chandra D, Kale RK. Influence of gamma-rays on the mouse liver cytochrome P450 system and its modulation by phenothiazine drugs. Int J Radiat Biol. 1999;75(3):335–349. doi: 10.1080/095530099140519. [DOI] [PubMed] [Google Scholar]
  • 56.Agrawal A, Choudhary D, Upreti M, Rath PC, Kale RK. Radiation induced oxidative stress: I. Studies in Ehrlich solid tumor in mice. Mol Cell Biochem. 2001;223(1–2):71–80. doi: 10.1023/a:1017900810837. [DOI] [PubMed] [Google Scholar]
  • 57.Marchenko MM, Kopyl’chuk GP, Ketsa OV. Effect of low doses of x-ray irradiation on the liver detoxication system in rats with transplanted Guerin’s carcinoma. Biomed Khim. 2010;56(2):266–273. doi: 10.18097/pbmc20105602266. [DOI] [PubMed] [Google Scholar]
  • 58.Pasanen M, Lang S, Kojo A, Kosma VM. Effects of simulated nuclear fuel particles on the histopathology and CYP enzymes in the rat lung and liver. Environ Res. 1995;70(2):126–133. doi: 10.1006/enrs.1995.1057. [DOI] [PubMed] [Google Scholar]
  • 59.Guéguen Y, Souidi M, Baudelin C, Dudoignon N, Grison S, Dublineau I, Marquette C, Voisin P, Gourmelon P, Aigueperse J. Short-term hepatic effects of depleted uranium on xenobiotic and bile acid metabolizing cytochrome P450 enzymes in the rat. Arc Toxicol. 2006;80(4):187–95. doi: 10.1007/s00204-005-0027-3. [DOI] [PubMed] [Google Scholar]
  • 60.Souidi M, Gueguen Y, Linard C, Dudoignon N, Grison S, Baudelin C, Marquette C, Gourmelon P, Aigueperse J, Dublineau I. In vivo effects of chronic contamination with depleted uranium on CYP3A and associated nuclear receptors PXR and CAR in the rat. Toxicology. 2005;214(1–2):113–122. doi: 10.1016/j.tox.2005.06.006. [DOI] [PubMed] [Google Scholar]
  • 61.Katiyar SK, Matsui MS, Mukhtar H. Ultraviolet-B exposure of human skin induces cytochromes P450 1A1 and 1B1. J Invest Dermatol. 2000;114(2):328–333. doi: 10.1046/j.1523-1747.2000.00876.x. [DOI] [PubMed] [Google Scholar]
  • 62.Wei YD, Rannug U, Rannug A. UV-induced CYP1A1 gene expression in human cells is mediated by tryptophan. Chem-Biol Interact. 1999;118(2):127–140. doi: 10.1016/s0009-2797(98)00118-5. [DOI] [PubMed] [Google Scholar]
  • 63.Luecke S, Wincent E, Backlund M, Rannug U, Rannug A. Cytochrome P450 1A1 gene regulation by UVB involves crosstalk between the aryl hydrocarbon receptor and nuclear factor kappaB. Chem-Biol Interact. 2010;184(3):466–473. doi: 10.1016/j.cbi.2010.01.038. [DOI] [PubMed] [Google Scholar]
  • 64.Bucci B, Misiti S, Cannizzaro A, Marchese R, Raza GH, Miceli R, Stigliano A, Amendola D, Monti O, Biancolella M, Amati F, Novelli G, Vecchione A, Brunetti E, De Paula U. Fractionated ionizing radiation exposure induces apoptosis through caspase-3 activation and reactive oxygen species generation. Anticancer Res. 2006;26(6B):4549–4557. [PubMed] [Google Scholar]
  • 65.Goerz G, Barnstorf W, Winnekendonk G, Bolsen K, Fritsch C, Kalka K, Tsambaos D. Influence of UVA and UVB irradiation on hepatic and cutaneous P450 isoenzymes. Arch Dermatol Res. 1996;289(1):46–51. doi: 10.1007/s004030050151. [DOI] [PubMed] [Google Scholar]
  • 66.Chung HC, Kim SH, Lee MG, Cho CK, Kim TH, Lee DH, Kim SG. Mitochondrial dysfunction by gamma-irradiation accompanies the induction of cytochrome P450 2E1 (CYP2E1) in rat liver. Toxicology. 2001;161(1–2):79–91. doi: 10.1016/s0300-483x(01)00332-8. [DOI] [PubMed] [Google Scholar]
  • 67.Chung HC, Kim SH, Lee MG, Kim SG. Increase in urea in conjunction with L-arginine metabolism in the liver leads to induction of cytochrome P450 2E1 (CYP2E1), the role of urea in CYP2E1 induction by acute renal failure. Drug Metab Dispos. 2002;30(6):739–746. doi: 10.1124/dmd.30.6.739. [DOI] [PubMed] [Google Scholar]
  • 68.Moon YJ, Lee AK, Chung HC, Kim EJ, Kim SH, Lee DC, Lee I, Kim SG, Lee MG. Effects of acute renal failure on the pharmacokinetics of chlorzoxazone in rats. Drug Metab Dispos. 2003;31(6):776–784. doi: 10.1124/dmd.31.6.776. [DOI] [PubMed] [Google Scholar]
  • 69.Chung W, Kim EJ, Lee I, Kim SG, Lee MG, Kim SH. Effects of recombinant human growth hormone on the pharmacokinetics of intravenous chlorzoxazone in rats with acute renal failure induced by uranyl nitrate. Life Sci. 2003;73(3):253–263. doi: 10.1016/s0024-3205(03)00268-6. [DOI] [PubMed] [Google Scholar]
  • 70.Ahn CY, Kim EJ, Lee I, Kwon JW, Kim WB, Kim SG, Lee MG. Effects of glucose on the pharmacokinetics of intravenous chlorzoxazone in rats with acute renal failure induced by uranyl nitrate. J Pharm Sci. 2003;92(8):1604–1613. doi: 10.1002/jps.10426. [DOI] [PubMed] [Google Scholar]
  • 71.Lee MG, Lee JH, Oh JM. Pharmacokinetic changes of drugs in rat model of acute renal failure induced by uranyl nitrate: correlation between drug metabolism and hepatic microsomal cytochrome P450 isozymes. Curr Clin Pharmacol. 2006;1(2):193–205. doi: 10.2174/157488406776872569. [DOI] [PubMed] [Google Scholar]
  • 72.Tissandie E, Guéguen Y, Lobaccaro JM, Aigueperse J, Gourmelon P, Paquet F, Souidi M. Chronic contamination with 137cesium affects vitamin D3 metabolism in rats. Toxicology. 2006;225(1):75–80. doi: 10.1016/j.tox.2006.05.006. [DOI] [PubMed] [Google Scholar]
  • 73.Tissandie E, Guéguen Y, Lobaccaro JM, Paquet F, Aigueperse J, Souidi M. Effects of depleted uranium after short-term exposure on vitamin D metabolism in rat. Arch Toxicol. 2006;80(8):473–480. doi: 10.1007/s00204-006-0068-2. [DOI] [PubMed] [Google Scholar]
  • 74.Tissandié E, Guéguen Y, Lobaccaro JM, Grandcolas L, Voisin P, Aigueperse J, Gourmelon P, Souidi M. In vivo effects of chronic contamination with depleted uranium on vitamin D3 metabolism in rat. Biochim Biophys Acta. 2007;1770(2):266–272. doi: 10.1016/j.bbagen.2006.10.006. [DOI] [PubMed] [Google Scholar]
  • 75.Tissandie E, Guéguen Y, Lobaccaro JM, Grandcolas L, Grison S, Aigueperse J, Souidi M. Vitamin D metabolism impairment in the rat’s offspring following maternal exposure to 137cesium. Arch Toxicol. 2009;83(4):357–362. doi: 10.1007/s00204-008-0351-5. [DOI] [PubMed] [Google Scholar]
  • 76.Sasaki T, Akutsu H, Shimada H, Miura S. A rice cytochrome P450 OsCYP84A that may interact with the UV tolerance pathway. Biosci Biotechnol Biochem. 2010;74(5):1045–1049. doi: 10.1271/bbb.90942. [DOI] [PubMed] [Google Scholar]
  • 77.Racine R, Grandcolas L, Grison S, Gourmelon P, Guéguen Y, Veyssière G, Souidi M. Molecular modifications of cholesterol metabolism in the liver and the brain after chronic contamination with cesium 137. Food Chem Toxicol. 2009;47(7):1642–1647. doi: 10.1016/j.fct.2009.04.022. [DOI] [PubMed] [Google Scholar]
  • 78.Jeong WI, DOSH, Kim TH, Jeong DH, Hong IH, Ki MR, Kwak DM, Lee SS, Jee YH, Kim SB, Jeong KS. Acute effects of fast neutron irradiation on mouse liver. J Radiat Res (Tokyo) 2007;48(3):233–240. doi: 10.1269/jrr.0629. [DOI] [PubMed] [Google Scholar]
  • 79.Maksymchuk OV, Bezdrobna LK, Sydoryk LL, Kysel’ova OK, Chashchyn MO. Cytochrome P450 2E1 expression in mice liver after exposure to continuous and acute gamma-radiation. Ukr Biokhim Zh. 2008;80(4):59–65. [PubMed] [Google Scholar]
  • 80.Maksymchuk OV, Bobyk VI, Sydoryk LL, Chashchyn MO. Influence of long-term combined gamma-radiation and ethanol on cytochrome P450 2E1 expression in the mice liver. Ukr Biokhim Zh. 2008;80(5):105–111. [PubMed] [Google Scholar]
  • 81.Agrawal A, Chandra D, Kale RK. Radiation induced oxidative stress: II studies in liver as a distant organ of tumor bearing mice. Mol Cell Biochem. 2001;224(1–2):9–17. doi: 10.1023/a:1011990704575. [DOI] [PubMed] [Google Scholar]
  • 82.Adaramoye OA, Adedara IA, Popoola B, Farombi EO. Extract of Xylopia aethiopica (Annonaceae) protects against gamma-radiation induced testicular damage in Wistar rats. J Basic Clin Physiol Pharmacol. 2010;21(4):295–313. doi: 10.1515/jbcpp.2010.21.4.295. [DOI] [PubMed] [Google Scholar]
  • 83.Guney Y, Bukan N, Dizman A, Hicsonmez A, Bilgihan A. Effects of two different high doses of irradiation on antioxidant system in the liver of guinea pigs. Eksp Onkol. 2004;26(1):71–74. [PubMed] [Google Scholar]
  • 84.Durović B, Spasić-Jokić V, Durović B. Influence of occupational exposure to low-dose ionizing radiation on the plasma activity of superoxide dismutase and glutathione level. Vojnosanit Pregl. 2008;65(8):613–618. doi: 10.2298/vsp0808613d. [DOI] [PubMed] [Google Scholar]
  • 85.Punnonen K, Lehtola K, Autio P, Kiistala U, Ahotupa M. Chronic UVB irradiation induces superoxide dismutase activity in human epidermis in vivo. J Photochem Photobiol B. 1995;30(1):43–8. doi: 10.1016/1011-1344(95)07131-k. [DOI] [PubMed] [Google Scholar]
  • 86.Pence BC, Naylor MF. Effects of single-dose ultraviolet radiation on skin superoxide dismutase, catalase, and xanthine oxidase in hairless mice. J Invest Dermatol. 1990;95(2):213–216. doi: 10.1111/1523-1747.ep12478037. [DOI] [PubMed] [Google Scholar]
  • 87.Obolenskaya MY, Teplyuk NM, Divi RL, Poirier MC, Filimonova NB, Zadrozna M, Pasanen MJ. Human placental glutathione S-transferase activity and polycyclic aromatic hydrocarbon DNA adducts as biomarkers for environmental oxidative stress in placentas from pregnant women living in radioactivity- and chemically-polluted regions. Toxicol Lett. 2010;196(2):80–86. doi: 10.1016/j.toxlet.2010.03.1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tapio S, Danescu-Mayer J, Asmuss M, Posch A, Gomolka M, Hornhardt S. Combined effects of gamma radiation and arsenite on the proteome of human TK6 lymphoblastoid cells. Mutat Res. 2005;581(1–2):141–152. doi: 10.1016/j.mrgentox.2004.11.016. [DOI] [PubMed] [Google Scholar]
  • 89.Kim SG, Nam SY, Kim CW, Cho CK, Kim ND. Radioprotective effects of 2-(allylthio)pyrazine an experimental chemopreventive agent: effects on detoxifying enzyme induction. Res Commun Mol Pathol Pharmacol. 1998;101(3):275–288. [PubMed] [Google Scholar]
  • 90.Kim SG, Nam SY, Kim CW, Kim JH, Cho CK, Yoo SY. Enhancement of radiation-inducible hepatic glutathione-S-transferases Ya, Yb1, Yb2, Yc1, and Yc2 gene expression by oltipraz: possible role in radioprotection. Mol Pharmacol. 1997;51(2):225–233. doi: 10.1124/mol.51.2.225. [DOI] [PubMed] [Google Scholar]
  • 91.Nam SY, Cho CK, Kim SG. Correlation of increased mortality with the suppression of radiation-inducible microsomal epoxide hydrolase and glutathione S-transferase gene expression by dexamethasone: effects on vitamin C and E-induced radioprotection. Biochem Pharmacol. 1998;56(10):1295–1304. doi: 10.1016/s0006-2952(98)00203-2. [DOI] [PubMed] [Google Scholar]
  • 92.Srivastava M, Chandra D, Kale RK. Modulation of radiation-induced changes in the xanthine oxidoreductase system in the livers of mice by its inhibitors. Radiat Res. 2002;157(3):290–297. doi: 10.1667/0033-7587(2002)157[0290:morici]2.0.co;2. [DOI] [PubMed] [Google Scholar]
  • 93.Srivastava M, Kale RK. Radiomodfication of xanthine oxidoreductase system in the liver of mice by phenylmethylsulfonyl fluoride and dithiothreitol. Radiat Res. 2000;154(1):94–103. doi: 10.1667/0033-7587(2000)154[0094:roxosi]2.0.co;2. [DOI] [PubMed] [Google Scholar]
  • 94.Srivastava M, Kale RK. Effect of radiation on the xanthine oxidoreductase system in the liver of mice. Radiat Res. 1999;152(3):257–264. [PubMed] [Google Scholar]
  • 95.Artiukhov VG, Zemchenkova OV, Basharina OV, Kim IV. Peculiarities of metabolism of UV-irradiated lymphocytes. Radiat Biol Radioecol. 2011;51(2):252–257. [PubMed] [Google Scholar]
  • 96.Racine R, Grandcolas L, Blanchardon E, Gourmelon P, Veyssiere G, Souidi M. Hepatic cholesterol metabolism following a chronic ingestion of cesium-137 starting at fetal stage in rats. J Radiat Res (Tokyo) 2010;51(1):37–45. doi: 10.1269/jrr.09064. [DOI] [PubMed] [Google Scholar]
  • 97.Nam SY, Kim JH, Cho CK, Yoo SY, Kim SG. Enhancement of radiation-induced hepatic microsomal epoxide hydrolase gene expression by oltipraz in rats. Radiat Res. 1997;147(5):613–620. [PubMed] [Google Scholar]

RESOURCES