Skip to main content

Transgenic rat models for mutagenesis and carcinogenesis

Abstract

Rats are a standard experimental animal for cancer bioassay and toxicological research for chemicals. Although the genetic analyses were behind mice, rats have been more frequently used for toxicological research than mice. This is partly because they live longer than mice and induce a wider variety of tumors, which are morphologically similar to those in humans. The body mass is larger than mice, which enables to take samples from organs for studies on pharmacokinetics or toxicokinetics. In addition, there are a number of chemicals that exhibit marked species differences in the carcinogenicity. These compounds are carcinogenic in rats but not in mice. Such examples are aflatoxin B1 and tamoxifen, both are carcinogenic to humans. Therefore, negative mutagenic/carcinogenic responses in mice do not guarantee that the chemical is not mutagenic/carcinogenic to rats or perhaps to humans. To facilitate research on in vivo mutagenesis and carcinogenesis, several transgenic rat models have been established. In general, the transgenic rats for mutagenesis are treated with chemicals longer than transgenic mice for more exact examination of the relationship between mutagenesis and carcinogenesis. Transgenic rat models for carcinogenesis are engineered mostly to understand mechanisms underlying chemical carcinogenesis. Here, we review papers dealing with the transgenic rat models for mutagenesis and carcinogenesis, and discuss the future perspective.

Background

In modern industrial society, humans are inevitably exposed to a variety of chemicals. These chemicals are mostly important to sustain the society and improve the quality of life. Antibiotics and other pharmaceuticals are such examples and they significantly prolong longevity and improve peoples’ health conditions. However, there are a number of chemicals that might have adverse effects on humans. Such examples are cigarette smoke, air pollutants and contaminants in water and food. These adverse chemicals are sometimes linked to human cancer. Therefore, international organizations such as Organization for Economic Co-operation and Development (OECD) or World Health Organization (WHO) set up guidelines to evaluate the genotoxic and carcinogenic risk of chemicals [1]. Genotoxicity is regarded as an important biomarker for carcinogenesis because many human carcinogens are reactive to DNA and induce mutations in the target organs of carcinogenesis [2]. In mechanisms, mutations of many oncogenes and suppressor oncogenes are deeply involved in a variety of human cancer [3]. In general, it is believed that DNA reactive carcinogens impose cancer risk on humans even at very low doses [4]. Therefore, regulatory agencies in many countries pay strong attention to identify DNA reactive genotoxic agents to reduce the cancer risk related to exposure to environmental chemicals.

In 1970’s and 1980’s, genotoxicity of chemicals was examined mainly by in vitro short-term assays with bacteria and cultured mammalian cells. Although bacterial mutation assays, i.e., Ames test, is still the gold standard to identify DNA reactive genotoxic chemicals, in vitro genotoxicity assays have some limitations. Bacteria and most of cultured mammalian cells do not possess enough metabolic capacity to activate or inactivate chemical carcinogens [5]. So, rat liver homogenate, i.e., S9, is adopted to mimic the mammalian metabolism. However, some chemical carcinogens such as urethane give negative results in Ames test because of the inefficiency of S9 to activate the chemicals to ultimate mutagens [6]. On the other hand, non-carcinogenic chemicals such as 2,6-diaminotoluene (2,6-DAT) give positive results in Ames test probably because S9 does not have enough detoxication capacities [7, 8]. Recent survey revealed that in vitro mammalian genotoxicity assays such as chromosome aberration assays, gene mutation assays and micronucleus assays give many false positives, i.e., positives in the assays but negatives in rodent cancer bioassays [9]. Thus, in vivo genotoxicity is regarded more important than in vitro results in terms of decision making whether the particular chemical is genotoxic and carcinogenic to humans or not.

Classical in vivo genotoxicity assays are, however, very time consuming and target organs for the assays are quite limited. For example, “mouse spot test” uses developing embryo and detects mutations in the genes controlling the pigmentation of coat color of mice [10]. This test has been adopted into OECD Guidelines for the Testing of Chemicals as Test No. 484. If mutations are induced in the genes that control the pigmentation of coat color, the offspring will have spots of changed color in the coat. The frequency of such spots in the treated mice is compared to that of the spots in untreated mice. Although this assay certainly detects mutations in mice in vivo, the target organ for mutagenesis is only melanoblasts in embryo. Because very few people conduct the assays nowadays, it has been deleted from OECD test guidelines in 2014. Another in vivo genotoxicity assay, that is, “Mouse Dlb-1 mutation assay”, detects mutations at the Dlb-1 locus in colon, which determines the expression of the binding site for the lectin Dolichos biflorus agglutinin [11]. C57BL/6J × SWR F1 mice are exposed to chemicals and the mutants are detected as clones of epithelial cells not stained with a peroxidase conjugated with the agglutinin. The assay is capable of identification of mutagens in colon but is not applicable to other organs such as liver.

To circumvent the above limitations, transgenic mice for mutagenesis have been developed in late 1980’s and 1990’s. Big Blue mice, Muta Mice and gpt delta mice are representative transgenic mice for mutagenesis and they use lambda phage as a vector having reporter genes for mutations [1215]. The phages are recovered from the genomic DNA of mice by in vitro lambda phage packaging reactions and in vivo mutations are detected after introduction of the rescued phage to indicator Escherichia coli (E. coli). Because the vector DNA having the reporter genes is recovered from the mouse genome to bacteria, they are called shuttle vectors. Although the reporter genes are bacteria or phage origin, the assays allow detection of mutations in any organ of mice such as liver, lung, bone marrow or testis. In addition, DNA sequence analysis can reveal mutation spectra associated with chemical exposure. About 10 years later from the development of transgenic mice, transgenic rats were developed because rats are more frequently used for cancer bioassays. Currently Big Blue rats having lambda LIZ and gpt delta rats having lambda EG10 are commercially available and widely used for in vivo mutagenesis [7, 16, 17]. Therefore, we focus on these two in vivo assays and discuss what has been revealed by the assays (Table 1). In the later part of this review, we review several transgenic rat models for chemical carcinogenesis (Table 2) and discuss the future perspective.

Table 1 Summary of experimental data of transgenic rat models for mutagenesis
Table 2 Summary of transgenic rat models for carcinogenesis

Transgenic rats for mutagenesis

Before establishment of transgenic rats for mutagenesis, there was a gap between in vivo genotoxicity assays and rodent cancer bioassays in terms of animal species. In vivo genotoxicity assays such as chromosome aberration test and micronucleus test have been conducted more frequently with mice than with rats because of the ease of handling and clearer genetic background. In contrast, rodent cancer bioassays have been conducted with rats more frequently than mice because of the lower frequency of spontaneous tumors and larger body mass. This species difference leads to discrepancy of test results between mice in mutagenesis and rats in carcinogenesis. Aflatoxin B1 gives negative or weakly positive results in genotoxicity with mice while rats give strong positives in carcinogenicity assays [18]. To fill in the gap, transgenic rats have been engineered. Nowadays, they are used as a standard tool to examine the mutagenicity of chemicals in the target organs of carcinogenesis.

Assay systems

Although both Big Blue rats and gpt delta rats use lambda phage as vectors of reporter genes, the assay systems are different as described below.

Big blue rats

Big Blue rats were generated by microinjection of lambda LIZ phage DNA into fertilized eggs of Fischer 344 (F344) rats [16]. In addition, the embryonic fibroblasts, i.e., Rat 2 cells, were established for an in vitro transgenic assay [19]. Originally, color selection with lacI was adopted for mutant detection but later more convenient cII selection was applied to Big Blue rat assays [20] (Fig. 1a, b). The gene lacI encodes a repressor protein LacI, which suppresses the expression of beta-galactosidase in E. coli. Therefore, inactivation of lacI by mutations results in the expression of beta-galactosidase and production of blue plaque in the presence of X-gal, while wild-type lacI leads to colorless plaques. However, this selection is time consuming and expensive because X-gal is an expensive chromogenic agent. In contrast, the CII protein induces the expression of the cI and the int genes that are required for a phage lysogeny [21]. In the hfl E. coli, phages with active cII gene can’t enter a lytic cycle and form no plaques because of the deficient in Hfl protease. This protease degrades CII protein and lets the phage enter a lytic cycle. The only phages with inactive cII mutants can make plaques with the E. coli hfl cells. Thus, this is a positive selection, and much more convenient and less expensive than the original lacI assay. The coding size of lacI is 1080 bp while that of cII is 294 bp, which makes cII more attractive for determination of mutation spectrum.

Fig. 1
figure 1

Mutant selections for Big Blue rats. a lacI selection. When LacI, the repressor protein of the lac operon, is active, it represses the expression of beta-galactosidase, which leads to colorless plaques. When the lacI gene is inactivated by mutations, beta-galactosidase is expressed, which leads to blue plaques. b cII selection. The cII protein is the critical switch in the lytic/lysogenic cycles of lambda phage. It activates the expression of the lambda cI (repressor) and int (integrase) genes, which are required for the establishment of lysogeny. The cII protein is negatively regulated by host E. coli Hfl protease, which digests the cII protein. In the hfl - background, the cII level is high, and therefore the lambda becomes lysogen. Only cII mutants can enter a lytic cycle and make plaques at 24 °C. The cI - mutants can’t enter the lytic cycle at this temperature. Therefore, the cII selection for Big Blue rats is conducted at 24 °C

Because cII was introduced several years after the original lacI color selection has been established, the level of spontaneous mutations and sensitivity to chemically-induced mutagenesis were compared between the reporter genes. Chen et al. [22] report that spontaneous mutation frequency of cII in liver is markedly higher than that of lacI (80 × 10−6 vs 10 × 10−6). Stuart et al. [23] also report that the mutation frequency of cII in colon mucosa is higher than that of lacI (78 × 10−6 vs 23 × 10−6). The cII gene has six G:C base pairs between nucleotide number 179 and 185, which is one of the hot spots of spontaneous mutagenesis. The high background makes smaller fold increases in mutation frequency after chemical treatments with alpha-hydroxytamoxifen and tamoxifen [22]. However, Gollapudi et al. [20] report that there is no significant difference in spontaneous and dimethyl nitrosamine (DMN)-induced mutation frequencies in liver between cII and lacI of Big Blue rats (99 × 10−6 vs 85 × 10−6 for spontaneous and 415 × 10−6 vs 400 × 10−6 for DMN.)

In both lacI and cII, deamination of 5-methylcytosine (5-MeC), which results in G:C to A:T transitions, is a major source of spontaneous mutations. Full methylation of cII and lacI in Big Blue rats is reported in bone marrow, bladder, liver, spleen and breast [24]. Spontaneous lacI mutation frequencies are lower in bone marrow and bladder compared to liver, which can’t be explained by the status of methylation of 5-MeC [25, 26]. Monroe et al. [24] suggest, therefore, that other mechanisms besides deamination of 5-MeC contribute to spontaneous mutagenesis in Big Blue system.

Because lacI is not an endogenous gene but a bacterial gene, the sensitivity of lacI and an endogenous gene, i.e., Hprt, in spleen was compared in Big Blue rats. Both genes were responded to 7, 12-dimethylbenz[a]anthracene (DMBA) [26, 27], N-hydroxyacetylaminofluorene [28] and thiotepa, an anticancer drug [29], and the mutation frequencies were increased. However, spontaneous mutation frequencies of Hprt were about 10 times lower than those of lacI [27]. Thus, the fold increases were larger in Hprt than in lacI. For example, the mutation frequency of Hprt was increased more than 10 fold by thiotepa treatments (3.5 × 10−6 vs 41.1 × 10−6) while that of lacI was increased about four fold by the same treatment (34.8 × 10−6 vs 140.9 × 10−6) [29]. In addition, the mutation spectra were different where Hprt recovered a fraction of large deletions not found among lacI mutants [29].

In summary, lacI and cII can be regarded as effective surrogate genes for in vivo mutations while spontaneous mutation frequency of cII may be higher than that of lacI. Caution should be payed that deletion mutations may be missed by the surrogate genes.

gpt delta rats

gpt delta rats were generated by microinjection of lambda EG10 DNA into fertilized eggs of Sprague-Dawley (SD) rats [17]. The SD gpt delta rats were later crossed with F344 rats for 15 generations, thereby establishing F344 gpt delta rats [7]. Two distinct selection systems are available for gpt delta mice and rats (Fig. 2a). One is gpt selection for detection of point mutations and the other is Spi- selection for deletions [15, 30]. The gpt gene is a bacterial counterpart of Hprt and encodes guanine phosphoribosyl transferase. When the gpt gene is inactivated by mutations, the E. coli host cells possessing plasmid carrying mutated gpt gene can survive on plates containing 6-thioguanine (6-TG) while those harboring plasmid carrying the wild-type gpt gene die because they phosphoribosylate 6-TG and incorporate 6-TGMP into DNA. Therefore, the gpt selection is a positive selection.

Fig. 2
figure 2

Mutant selection for gpt delta rats. a gpt selection. The E. coli gpt gene encodes guanine phosphoribosyl transferase, which attaches a phosphoribose to 6-TG. The phosphoribosylated 6-TG is further phosphorylated and finally incorporated into DNA. Incorporation of 6-TG is toxic to E. coli and cell death is induced. Therefore, only when the gpt gene is inactivated by mutations, E. coli can make colonies on a plate containing 6-TG. b Spi- selection. The wild-type lambda phages lyse E. coli, thereby making phage plaques. However, if the E. coli chromosome harbors P2 phage DNA, which is called P2 lysogen, the wild-type lambda phage can’t lyse P2 lysogen. Only the defective lambda phage whose red and gam genes are inactivated can lyse P2 lysogen. The resulting plaques are called P2 plaques. Because the red and gam genes are localized in lambda genome side by side, the inactivation of two genes are most likely induced by deletions in the region

Spi- stands for sensitive to P2 interference [31] (Fig. 2b). This selection allows selective detection of deletion mutants of lambda phage. In wild-type E. coli, the wild-type lambda phage lyses the E. coli, thereby forming phage plaques. However, if E. coli chromosome possesses P2 phage DNA, that is called P2 lysogen, the wild-type lambda phage can’t form plaques. This phenomenon is called “P2 interference”. However, when two genes of lambda phage, i.e., the red and gam genes, are simultaneously inactivated, the defective phage can make plaques in P2 lysogen. The plaques are called Spi- plaques. Since the red and gam genes are located side by side in the lambda DNA, the simultaneous inactivation of two genes are most likely induced by deletion of the region containing the two genes. The unique feature of Spi- selection is specific detection of deletion mutations including frameshift mutations.

The transgene lambda EG10 having the gpt gene and the red/gam genes is located in the chromosome four of gpt delta rats. The exact location of the integration site in the rat genome was determined by next generation DNA sequencer (NGS) [32]. About 72 kb genomic sequence was deleted during integration of the transgene and smaller genetic rearrangements were also induced by the integration. Unlike gpt delta mice, which have lambda EG10 in both chromosome 17, gpt delta rats are heterozygous where lambda EG10 is integrated in only one allele of chromosome 4. This is because homozygous gpt delta rats are defective in tooth development and can’t survive after weaning. Specific PCR primers that can be used to amplify the DNA sequence between rat chromosome and the integrated lambda EG10 are available. They can be used to distinguish between wild-type rats and gpt delta rats. The average spontaneous gpt and Spi- mutant frequencies in liver are 4.5 × 10−6 and 2.7 × 10−6, respectively [33]. The frequencies are significantly lower than those of the lacI and cII genes. The low spontaneous mutant frequencies of gpt and Spi- are similar to those of gpt delta mice.

Issues that have been examined by transgenic rat assays

Organ/tissue specificity

An important feature of chemical carcinogens is the organ specificity. They induce cancer in specific organs, which are called target organs for carcinogenesis. Aflatoxin B1, aristolochic acid and o-toluidine are all potent human carcinogens but they induce cancer in different organs, i.e., liver by aflatoxin B1, kidney by aristolochic acid and bladder by o-toluidine [3436]. Thus, an interesting question for transgenic rat assays for mutagenesis is whether mutations can be identified in the target organs for carcinogenesis.

2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) is a heterocyclic amine in cooked food and administration of PhIP in diet causes cancer in the prostate in male rats and in the mammary glands in females [37, 38]. It was examined, therefore, whether PhIP induces mutations in the target organs in a sex specific manner. PhIP-induced mutations were identified in mammary glands of female rats [39, 40] and prostate in males [41, 42]. These results suggest the causal link between mutagenesis and carcinogenesis induced by PhIP in mammary glands and prostates. However, mutations in prostate were identified not only in ventral prostate where cancer is induced but also in dorsolateral and anterior lobe where cancer is sparingly induced [41]. This raised a question as to what factors define the lobe specificity of PhIP-induced carcinogenesis. Interestingly, PhIP acts as a promoter and induces cell proliferation only in the ventral prostate [41]. Thus, PhIP may be an organ- and lobe-specific promoter while it acts as an initiator in all three lobes

PhIP induces colon cancer much more frequently in male rats than in females [43]. Therefore, colon mutations were examined in male and female Big Blue rats. It was revealed that mutations were almost equally induced in both sexes [44, 45]. The mutation spectra induced by PhIP were also similar in both sexes, i.e., one base deletions including the guanine deletion at 5′-GGGA-3′ [44]. These results suggest that factors other than mutagenesis strongly contribute to PhIP-induced carcinogenesis and also that the factors may determine the sex-specific induction of colon cancer by PhIP.

The relationship between mutagenesis and carcinogenesis has been examined even at the sub-organ level as in the case of PhIP in the prostate. Tris(2,3-dibromopropyl)phosphate (TDBP) induces tumors specifically in outer medulla in the kidney of rats [46]. Mutations were examined in the inner medulla, outer medulla and cortex of kidney, and the mutation frequency was in the order of cortex followed by outer medulla (the target site) and inner medulla [47]. The highest mutation induction does not coincide with the localization of tumors. However, cell proliferation is increased specifically in the outer medulla after TDBP treatment [46, 48]. Thus, it was concluded that combined effects of cell proliferation and induction of mutations are responsible for sub-organ-specific tumor formation by TDBP.

Ochratoxin A, a mycotoxin, also induces renal tumors in rats specific in S3 segment of the proximal tubules [49]. Unlike TDBP, mutations are induced only in the outer medulla, which is primarily occupied by the S3 segment of the proximal tubules [50]. No mutations were detected in the cortex. Thus in this case, specific induction of mutations in outer medulla might account for the sub-organ-specific induction of tumors in rats (See more in Genotoxic versus non-genotoxic carcinogens section).

Phenacetin, an analgesic drug, induces tumors in kidney but not in liver [51]. The in vivo mutagenesis in kidney and liver was examined with SD gpt delta rats fed with diet containing phenacetin for 26 and 52 weeks [52]. Mutations were detected in both kidney and liver and the mutation frequency was much higher in liver (non-target organ) than in kidney (target organ). The results suggest the intensity of mutagenicity does not necessarily correlate with the induction of tumor formation.

Carcinogens versus structurally-related non-carcinogens

Chemical carcinogens excert the adverse effects depneding on the chemical structures. Even the structures are similar, their carcinogenicity is sometimes completely different. Transgenic rats for mutagenesis were examined for their ability to distinguish mutagenicity of structural isomers, i.e., one is a carcinogen and the other is a non-carcinogen. 2,4-Diaminotoluene (2,4-DAT) is an intermediate in chemical industry but induces hepatic tumors in male and female rats and mammary and subcutaneous tumors in female rats [53]. The isomer 2,6-DAT is an intermediate of dyes and rubber chemicals and is not carcinogenic in rats and mice despite the structural similarity to 2,4-DAT [54]. Interestingly, both DATs are mutagenic in Ames Salmonella strains [7], suggesing the potential mutagenicity of both chemicals. The in vivo mutagenicity of 2,4-DAT and 2,6-DAT was examined in liver and kidney of male gpt delta rats [7]. The rats were fed 2,4-DAT or 2,6-DAT in diet for 13 weeks and the mutations were examined. Only 2,4-DAT induced gpt and Spi- mutations in liver but not in kidney. 2,6-DAT was negative in gpt and Spi- assays in liver and kideny. The results suggest that in vitro mutagenicity should be carefully examined by in vivo mutagenicty assay. The mutagenicity of 2,4-DAT but not 2,6-DAT in liver of gpt delta rats was also reported by 4 weeks administration of gavage [55].

Tamoxifen is a nonsteroid antiestrogen that is used as adjuvant therapy for breast cancer. However, tamoxifen is carcinogenic in liver in rats [56]. The structural analogue toremifen is not carcinogenic [57]. To examine whether transgenic rats distinguish two compounds in terms of mutagenesis, female F344 gpt delta rats were treated with either tamoxifen or toremifen [58]. Tamoxifen significantly enhanced gpt and Spi- mutation frequencies in the liver. The treatment did not increase the mutation frequencies in the kidney, a non-target organ for carcinogenesis. Toremifen did not increase gpt and Spi- mutation frequencies in liver and kidney. The results clearly indicate that tamoxifen is mutagenic in the target organ for carcinogenesis but the strustural analogue toremifen is not.

6-p-Dimethylaminophenylazobenzthiazole (6BT) is a potent liver carcinogen in rats [59]. It induces malignant liver tumors after 2-to-3 months of dietary administration in a riboflavin-deficient diet. In contrast, the analogue 5-p-dimethylaminophenylazobenzthiazole (5BT) gives no tumors after 6 month administration. Both chemicals are potent mutagens in Ames Salmonella strains [60]. The mutagenicity of 6BT and 5BT was examined with Big Blue rats and unexpectedly both were mutagenic in liver [61]. Thus, mutagenicity did not account for the marked difference of the carcinogenicity of two closely-related compounds. It is speculated that differential cell proliferation effects on oval cells in liver may explain the difference. 6BT induces the proliferation of ovall cells by either gavage or in diet while 5BT is inactive in this respect. Oval cells may be progenitor cells for hepatocellular carcinoma [62].

Genotoxic versus non-genotoxic carcinogens

A key question for evaluation and regulation of chemical carcinogens is whether mutations are involved in the mechanisms of carcinogenesis. If the chemical induces mutations in the target organ, thereby causing carcinogenesis, the chemical is classified as “a genotoxic carcinogen”, which has no threshold or safety dose for the action [4]. In contrast, when the chemical dose not induce mutations in the target organ despite the carcinogenicity, the chemical is classified as “a non-genotoxic carcinogen”, which has threshold or safety dose and can be used in the society below the safety dose. If the chemical is judged as a genotoxic carcinogen, the chemical is not be considered acceptable for use as food additives, pesticides or veterinary drugs [63, 64].

Several carcinogenic compounds in food were examined for the mutagenicity in the target organs for carcinogenesis with gpt delta rats [63]. It was revealed that citrinin and 3-monochloropropane-1,2-diol (3-MCPD) were negative, and hence they were classified as non-genotoxic carcinogens [65, 66]. Citrinin is a food-contaminated mycotoxin and induces renal tumors in rats [67]. It may induce tumors via cell cycle progression but not genotoxicity [65]. 3-MCPD is regarded as a rat renal and testicular carcinogen [68] and is mutagenic in Salmonella and E. coli strains for mutagenicity assays [69]. The fatty acid esters of 3-MCPD are generated during food processing and exert renal toxicity [70]. The esters are metabolized to 3-MCPD in vivo [71]. Because of the negative mutagenicity in vivo, 3-MCPD and the fatty acid esters are judged as non-genotoxic carcinogens [66]. On the other side, estragole [72], madder color [73] and methyleugenol [74] were positive in the transgenic assay and thus mutagenicity may participate in the carcinogenesis. Estragole is a natural organic compound and frequently used as a flavoring food additive, but is carcinogenic in liver of mice [75]. Despite the in vivo mutagenicity, estragole is not mutagenic in Salmonella and E. coli strains for mutagenicity assays [76]. Madder color is a dye and a potent carcinogen in kidney and liver in rats [77], and thus its use as a food additive has been banned in Japan in 2004. Methyleugenol is a fragrance and flavoring agent but is a hepatocarcinogen in F344 rats [78].

Malachite green is a dye that has been widely used as an antifungal agent in fish industry, and leucomalachite green is a reduction product and a major metabolite of malachite green [79]. Malachite green induces adenoma and/or carcinoma in thyroid gland, liver and mammary gland of female F344 rats and leucomalachite green induces adenoma in the testis of male rats [80]. Female Big Blue rats were fed leucomalachite green for 4, 16 or 32 weeks and mutations were analyzed in Hprt in spleen, micronucleus formation in bone marrow and lacI mutation in liver [81]. No increases were observed in Hprt mutation frequency and micronucleus formation. About three fold increases in lacI mutant frequency were observed in rats treated for 16 weeks [79]. DNA adduct levels increased in liver of rats. However, the following mutation spectrum analysis indicated that the apparent increase in mutation frequency was due to expansion of spontaneous mutations [81]. It is still enigmatic how malachite green and leucomalachite green induce tumors in rats.

Ochratoxin A [49], a mycotoxin, is an interesting agent because it induces Spi- mutations but not gpt [50, 82]. It induces Spi- mutations in the target site of carcinogenesis, i.e., the outer medulla of kidney, when male gpt delta rats were treated with ochratoxin A. Large deletions with the size of more than 1 kb are induced by the treatment. Experiments with p53 deficient gpt delta mice suggest that Spi- mutant frequency, but not gpt, was increased by ochratoxin A treatment [83, 84]. No mutagenicity was observed in p53 proficient mice. It appears that double-strand breaks in DNA are induced in the target site of kidney of rats, which leads to large deletions. It is puzzling, however, why gpt mutations are not induced. When DNA is damaged, gpt mutations are usually more frequently induced compared to Spi- mutations. If ochratoxin A induces DNA adducts, it should induce gpt mutations as well as Spi- mutations. It is tempting to speculate, therefore, that ochratoxin A may interact with proteins involved in DNA replication, repair or chromosome segregation, thereby inducing double-strand breaks in DNA. If so, ochratoxin A may not be a genotoxic carcinogen although it induces mutations in the target organ of carcinogenesis.

Threshold or low dose effects

Although it is supposed that genotoxic carcinogens have no thresholds or safety level, the following experiments exhibit no effective dose levels for in vivo mutations of genotoxic carcinogens. Male Big Blue rats were fed a diet containing 0.001, 0.01, 0.1, 1, 10 or 100 ppm of 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) for 16 weeks and the lacI mutation frequency and glutathione S-transferase placental form (GST-P) positive foci in the liver were examined [85]. MeIQx is a heterocyclic amine formed during cooking and induces liver tumors in rats [86]. The mutation frequencies significantly increased at doses of 10 and 100 ppm, and GST-P positive foci significantly increased at a dose of 100 ppm. No statistical increases in both frequencies were observed, however, at lower doses, indicating the existence of no effective doses for mutagenesis and carcinogenesis.

Similarly, male Big Blue rats were administered with potassium bromate (KBrO3) in drinking water at concentrations of 0, 0.02, 0.2, 2, 8, 30, 125 and 500 ppm for 16 weeks [87]. The lacI mutation in the kidney was induced only at a concentration of 500 ppm. No mutagenicity was detected at 125 ppm or lower doses. Histopathological changes in renal tubular cells were observed at doses of 125 and 500 ppm but not at 30 ppm or lower doses. 8-oxoguanine in DNA was formed only at a dose of 500 ppm. KBrO3 is an oxidizing agent and used as a maturing agent for flour and as a dough conditioner [68]. However, it induces renal cell tumors in male and female rats after oral administration for 2 years in the drinking water [88]. The results suggest that there may be safety dose for the genotoxic carcinogen.

Cyproterone acetate (CPA) is an antiandrogenic drug that is used for women in long term treatments of excel androgen levels. However, it induces liver tumors in rats [89]. Female Big Blue rats were treated with CPA at a single dose of 0, 5, 10, 20, 40, 80 and 100 mg/kg and the lacI mutation frequency was determined in the liver 2 weeks after the last treatment. Significant increase in mutation frequency was observed at a dose of 10 mg/kg or higher, and no mutations were induced at a dose of 5 mg/kg [90]. Because high amounts of DNA adducts were formed at the non-effective dose of 5 mg/kg, it was assumed that the mitotic activity required for conversion of DNA adducts to mutation was not sufficiently strong at the dose.

Collectively, these results suggest the existence of no-effective dose for mutagenesis in the target organs for carcinogenesis even for mutagenic carcinogens. It remains uncertain, however, the sensitivity to detect the mutations is high enough to analyze the subtle increase in mutation frequencies. It is suggested that no-effective levels for mutagenesis vary depending on the in vivo models and also that the lower no-effective levels are detected with lower spontaneous mutation frequencies [91]. To detect the no-effective levels, mathematical models such as Points of Departure (PoD) have been proposed [92].

Multiple exposure or chemoprevention

Genotoxic effects of chemicals are sometimes enhanced or attenuated by dietary supplements. In addition, people are exposed to multiple chemicals in real life. Therefore, they may exert additive or synergistic effects on the genotoxic effects. Transgenic rats for mutagenesis have been utilized to examine the combined genotoxic effects of more than one chemical in vivo.

Ellagic acid, green tea and diallyl sulfide (DAS) were examined for the chemo preventive effects against N-nitrosomethylbenzylamine (NMBA)–induced mutations in the esophagus of Big Blue rats [93]. Addition of ellagic acid in diet, replacing drinking water with green tea or gavage of DAS significantly reduced the mutagenicity of NMBA. In contrast, 5 % ethanol to the drinking water enhanced the mutagenicity.

Endogenous estrogen status and addition of genistein, a phytoestrogen, were examined for the modulating effects on DMBA-induced mutation in liver of Big Blue rats [94]. Ovariectomized female rats exhibited higher mutation frequencies than the intact rats, suggesting the endogenous ovarian hormones may have an inhibitory effect on liver mutagenesis by DMBA. Dietary supplement of genistein in the ovariectomized and the intact rats did not alter the spontaneous and induced mutations in liver. Ovariectomized female Big Blue rats were also used to examine the modulating effects of daidzein, genistein and 17-beta-estradiol on DMBA-induced mutagenesis in the mammary glands [95] and uterus [96]. Daidzein and genistein are major constituents of isoflavones and interact with the alpha and beta estrogen receptors in the mammary glands. Daidzein, genistein and 17-beta-estradiol each did not significantly change DMBA-induced mutagenesis in the mammary glands and uterus.

Conjugated linoleic acid is a mixture of heat-derivatives of linoleic acid, and is shown to be protective against heterocyclic amine-induced carcinogenesis [97]. Antimutagenic effects of conjugated linoleic acid was examined in kidney of male and female Big Blue rats treated with PhIP [98]. Conjugated linoleic acid reduced PhIP-induced mutations of female rats but not those of male rats. Therefore, the protective effects are sex-dependent.

High intake of sucrose is associated with increased risk of colon cancer [99]. Co-mutagenic effects of sucrose were examined in colon of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ)-treated Big Blue rats [100]. Sucrose and IQ increased the mutation frequencies and the combined treatment with sucrose and IQ was additive, indicating that sucrose and IQ induce mutations independently. It is worth noticing that sucrose is mutagenic in vivo [101], which will be discussed more detail below (Sweet diet section). On the other hand, dietary restriction may delay aging and age-related diseases. The effects of dietary restriction on PhIP-induced mutation in the distal colon were examined [102]. However, the restriction did not alter the mutation frequency in male and female Big Blue rats. To examine the interactions between tobacco smoking and asbestos exposure, Big Blue rats were exposed to benzo[a]pyrene (BP) and amosite intratracheally and mutations were analyzed in the lung of Big Blue rats. Combined instillation of amosite and BP exhibited a highly significant synergistic effect [103]. The mutation frequency of BP was enhanced more than two times when combined with amosite, which was not mutagenic in lung.

The compound 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is an environmental contaminant and a potent carcinogen in laboratory rodents [104]. Modulating effects of TCDD on mutagenesis was examined with male and female Big Blue rats [105]. The rats were pre-exposed to TCDD for 6 weeks (2 μg twice per week) and then they were given aflatoxin B1 at a dose of 0.5 mg/kg by gavage. After 2 weeks, the lacI mutation frequency was measured. TCDD pre-treatments did not significantly modulate the mutation frequency in male. However, the female mutation frequency was reduced to the control level. DNA sequence analysis confirmed the absence of aflatoxin B1-induced transversion mutations in female rats. It is speculated that sex-specific factors such as estrogens or estrogen receptors may play a role in the sex-dependent chemopreventive effects of TCDD against aflatoxin B1-induced mutagenesis.

Tamoxifen

As described in Carcinogens versus structurally-related non-carcinogens, tamoxifen is widely used for adjuvant therapy in the breast cancer patient for many years. However, tamoxifen induces endometrial cancer in women, and liver and endometrial tumors in rats [106]. There is no evidence, however, that tamoxifen induces liver tumors in humans. Tamoxifen is metabolically activated to alpha-hydroxytamoxifen, which is further acitvated by sulfotransferase and finally induces DNA adducts. Rat sulfotransferase activates alpha-hydroxytamoxifen but human enzyme does not [107]. This may be the reason for the species difference between human and rat for liver tumorigenesis by tamoxifen. Because tamoxifen is inactive in a battery of short-term tests for mutagenesis [108], the in vivo mutagenicity was examined with Big Blue rats and gpt delta rats. Tamoxifen induced lacI, cII, gpt and Spi- mutations in the liver, mainly G:C to T:A transversions and −1 frameshift [58, 108110]. Alpha-hydroxytamoxifen also induces mutations in the liver with the spectrum of mutation of G:C to T:A [22]. Thus, it appears that tamoxifen induces liver tumors in rats via alpha-hydroxytamoxifen-induced mutagenesis.

Naturally occurring carcinogens

Several plant constituents often used for herbal treatments were examined for the mutagenicity in vivo because of the carcinogenicity in experimental animals and in humans. Aristolochic acid is a nephrotoxin and carcinogenic in kidney and forestomach in rodents [111]. It has been associated with the development of urothelial cancer in humans. Male Big Blue rats were gavaged with aristolochic acid for 3 months, and the DNA adduct levels and mutations were examined in liver (a non-target organ) and kidney (a target organ) [112, 113]. Kidney exhibited at least two fold higher levels of DNA adducts and mutations than liver. A:T to T:A transversions were the predominant mutation in both organs. In this case, higher DNA damage and mutation frequencies were observed in the target organ than in the non-target organ.

Riddelliine is a naturally occurring pyrrolizidine alkaloid that induces liver hemangiosarcomas in rats and mice [114]. Female Big Blue rats were gavaged with riddelliine for 12 weeks and the mutations were analyzed in liver [115]. Mutations were induced in a dose-dependent manner and the major mutation was G:C to T:A. Later, liver was dissected into parenchymal and endothelial cells and riddelliine-induced mutations were analyzed in the cells [116]. Mutation was specifically induced in the endothelial cells but not in the parenchymal cells. Because hemangiosarcomas are derived from endothelial cells, the results indicate a good correlation between mutagenesis and carcinogenesis at a cell-type level.

Oxidative damage

Oxidative stress is an important factor for in vivo mutagenesis and carcinogenesis. Although KBrO3 induces 8-oxoguanine in DNA, which leads to G:C to T:A mutations, in vitro genotoxicity assays suggest that KBrO3 induces deletions rather than G:C to T:A transversions [117, 118]. Male SD gpt delta rats were given KBrO3 in drinking water for 13 weeks and the level of 8-oxoguanine in DNA and mutations were analyzed in the kidney [119]. Increases of 8-oxoguanine in DNA occurred after 1 week treatment at 500 ppm. Spi- mutations were increased after 9 weeks administration at 500 ppm but no significant increases in mutation frequency were observed at 500 ppm earlier than 9 weeks. No gpt mutations were observed even at week 13. The results suggest that deletions but not G:C to T:A are induced by KBrO3 in kideny of rats and also that 9 weeks may be necessary to convert the induced 8-oxoguanine in DNA to mutations. It is worth noticing, however, that male Big Blue rats (F344) exhibited mainly G:C to T:A transversions in kideny when they were treated with KBrO3 in drinking water at 500 ppm for 16 weeks [87] (see Threshold or low dose effects). Different genetic background of rats (SD versus F344) might affect the spectrum of mutations. When female F344 gpt delta rats were given KBrO3 in drinking water at 500 ppm for 9 weeks, gpt mutation frequency was significantly increased along with slight increase of Spi- mutations [120]. However, the spectrum of induced gpt mutations was not predominated by G:C to T:A but various types of mutations including −1 frameshift were observed. Thus, it remains to be clarified what types of mutations are induced by KBrO3 in vivo.

DNA non-reactive carcinogens (metals, asbestos and TCDD)

Several nickel compounds are carcinogenic in humans and animals [121]. Nickel subsulfide (Ni3S2) is one of them and induces lung tumors in F344 rats following inhalation exposure [122]. Although Ni3S2 increased lacI mutation frequency in in vitro Rat2 cells, it did not enhance lacI mutation in the lung and nasal mucosa of male Big Blue rats when the rats were treated by inhalation through the nose [123]. Male F344 gpt delta rats were also treated with Ni3S2 by intratracheal instillation, but no increases in gpt and Spi- mutant frequencies were observed in the lung [124].

Asbestos is a well-known human carcinogen that induces mesothelioma and lung cancer in exposed persons [125]. Male Big Blue rats were given amosite asbestos by intratracheal instillation with single doses of 1 or 2 mg/animal, or 4 weekly doses of 2 mg [126]. The in vivo mutations were analyzed at 4 weeks or 16 weeks after the last treatment. The average length of amosite was more than 20 μm and the average thickness of the fiber was 0.7 micron, leading to persistent presence in the lung. About two fold induction of lacI mutations was observed in the lung after 16 weeks exposure possibly because of the persistent inflammation induced by the treatment. Similarly, two asbestos substituent mineral fibers, i.e., rock (stone) wool RW1 and glass wool MMVF10, were examined for the in vivo mutagenicity with male Big Blue rats [127]. The man-made fibers were given to the rats by intratracheal instillation with single doses of 1 or 2 mg/animal, or 4 weekly doses of 2 mg. Exposure of RW1 for 16 weeks increased lacI mutant frequency about two-fold in the lung but MMVF10 did not. Because RW1 induces mild inflammation in the lung, the mutagenicity may be due to DNA damage induced by the inflammation.

TCDD induces various tumors in rats [104]. Male and female Big Blue rats were exposed to 2 μg TCDD/kg by gavage for 6 weeks but no increase in lacI mutation frequency was observed in the liver of both sexes [128].

Mechanical irritation by uracil-induced urolithiasis was examined for the in vivo mutagenicity with male Big Blue rats [25]. The rats were fed 3 % uracil in the diet for 50 weeks and the lacI mutation frequency was determined in the bladder. About three to five fold increases in the mutation frequency were observed at weeks 10, 20 and 51. The mutation spectra were similar to those of the spontaneous mutations, i.e., G:C to A:T transitions at CpG sites. Therefore, it is suggested that the elevation of spontaneous mutations may be due to cell proliferations induced by the uracil treatment.

Polluted air

Diesel exhaust (DE) is a factor of air pollution and a suspected cause of lung cancer and other respiratory diseases [129]. Male Big Blue rats were exposed to 1 or 6 mg/m3 of DE for 4 weeks [130]. The mutant frequency in lung was increased about five times over the control level by exposure to six DE mg/m3 but no increases were observed with 1 mg DE/m3. The results clearly indicate that DE is mutagenic in rat lung. When male Big Blue rats were treated with a diet containing DE from 0 to 80 mg/kg for 3 weeks, no mutation induction was observed in the lung although DNA adducts and DNA strand breaks were observed [131]. The results suggest that inhalation exposure, but not dietary exposure, is needed to evaluate the mutagenic potential of DE in lung. Road paving workers are exposed to bitumen fumes, a complex mixture of various polycyclic aromatic amines. Big Blue rats were exposed to bitumen fumes through nose, and DNA adduct levels and mutation frequencies were examined in the lung [132]. Although DNA adducts were increased by the exposure, the mutation frequencies were not enhanced. Perhaps, cell proliferation is not fully induced by the treatment.

4-Monochlorobiphenyl (PCB3) is found in indoor and outdoor air and in food [133]. Unlike polychlorinated biphenyls, PCB3 is more readily metabolized to monohydroxy-PCBs by CYP drug metabolizing enzymes and further dihydroxy-metabolites, which can be oxidized to quinones [134]. The mutagenicity of PCB3 and the metabolite, i.e., 4-hydroxy-PCB3, were examined with male Big Blue rats [133, 135]. The rats were given PCB3 or 4-hydroxy-PCB3 by intraperitoneal injection once per week for 4 weeks. In liver and lung, the mutant frequency in PCB-3-treated rats was significantly elevated and 4-hydroxy-PCB3 induced a non-significant increase in the mutant frequency.

Sweet diet

Cancer incidence in colon and other organs is strongly affected by diet and life style. Intake of sucrose-rich diet was examined for the in vivo mutagenicity with Big Blue rats [136]. Male Big Blue rats were fed diet with sucrose of 3.4 % (control), 6.9, 13.8 and 34.5 % for 3 weeks without affecting the overall energy and carbohydrate intake. The cII mutation frequency was increased about two fold in a dose-dependent manner in the colonic mucosa but no increases in the liver. No oxidative DNA damage was increased. Later, male Big Blue rats were fed diet containing 30 % sucrose or the composed sugar, i.e., either 30 % glucose or 30 % fructose for 35 days [101]. In these experiments, however, any sugar did not significantly increase the cII mutations in the colon and the liver, although DNA adduct levels were increased by the diet in both organs. It is suggested that indirect effects such as alterations of chemical environment in colon may account for the apparent genotoxicity.

Transgenic rats for carcinogenesis

In carcinogenesis study field, transgenic rats provide good models too. Rats rather than mice are more frequently used in chemical carcinogenesis studies for various reasons. For example, in the liver, GST-P has been utilized as a reliable marker for early detection of preneoplastic lesions [137]. So far, more than 30 different transgenic rats have been reported and utilized in neurosciences, endocrinology and carcinogenesis fields. Transgenic rats that are highly susceptible to carcinogens or exhibit high incidence of spontaneous neoplasm are good models for screening of chemopreventive agents and mechanism studies of carcinogenesis process.

Human c-Ha-ras proto-oncogene transgenic rats (Hras128)

Hras128 carries a human c-Ha-ras proto-oncogene including its own promoter region. Female Hras128 is highly susceptible to breast carcinogens such as N-methyl-N-nitrosourea (MNU) and PhIP [138, 139]. These chemicals induced estrogen-independent breast tumors because they did not respond to ovariectomy [140]. Esophagus and bladder tumors were highly inducible in carcinogen–treated male Hras128 [141, 142]. This Hras128 is deposited to National BioResource Project (NBRP Rat No.0376), and available from it [143]. In addition, cell lines (RMC-1, RMC-2, RMC-3, RMC-6, RMC-11, RMC-17) derived from Hras128 mammary adenocarcinoma are also available from RIKEN cell bank [144].

Probasin-SV40 T antigen transgenic rats (TRAP)

TRAP expresses the simian virus 40 (SV40) large T antigen under probasin promoter control. This animal was established to obtain sufficient size of samples of prostate cancer. In the male TRAP, prostate carcinomas are developed at 100 % incidence in all lobes (ventral, dorsolateral and anterior) before 15 weeks of age [145]. Since these tumors are androgen dependent, it is expected to utilize TRAP as a model for understanding the mechanisms of relapsing of tumors that are androgen independent. Chemopreventive studies and mechanism studies utilizing TRAP have been also reported [146148].

Connexin 32 dominant-negative transgenic rats (Cx32Δ Tg)

Employment of the dominant negative mutants is one of the alternatives to gene targeting in rat. Cx32Δ Tg expresses a dominant negative mutant of connexin 32 (Cx32). Cx32 is a major gap junction protein in the liver. They formed transmembrane channels between adjacent cells. In the liver of this animal, localization of normal connexins is disrupted and gap junction capacities are markedly decreased [149]. Chemical-induced carcinogenesis studies using Cx32Δtransgenic revealed that disruption of gap junctional intercellular communications in vivo resulted in hepatocarcinogenesis and its progression [150, 151]. In addition, this transgenic rat can be utilized to mechanism studies of the onset of toxicity which are related to cell-cell communications [149].

Transgenic rats carrying a mutated H- or K-ras gene controlled by Cre/loxP activation (Hras250 and Kras327)

These transgenic rats express a human activated RAS oncogene regulated by the Cre/lox system. Targeted pancreatic activation of the transgene was accomplished by injection of adenovirus carrying Cre into the pancreatic ducts and acini [152, 153]. Tumors in the model exhibit similarities to the human pancreatic ductal adenocarcinoma. Hras250 is deposited to National BioResource Project (NBRP Rat No.0568), and available [143].

Transgenic rats as carcinogenic models promise our understanding of the behavior of cancer in vivo, and will be useful to explore new therapeutic approaches. For carcinogenicity studies, rasH2 mice and p53+/− mice are utilized because of their high susceptibility for carcinogens [154]. Several transgenic rats in Table 2 exhibit high sensitivity to carcinogens and oncogenic events are easily initiated. However, their background data are still not enough and amassed research evidence may be needed for applying them to short-term carcinogenicity tests. In this decade, gene-targeting technology using rats might be about to enter a new period. Gene-targeting technology using zinc-finger nucleases (ZFNs) allowed generation of the first knock-out rat in 2009 [155, 156]. And, generation of knock-out rats was achieved using rat ES cell-based technology in 2010 [157]. More recently, transcription activator-like effector nucleases (TALEN) and CRISPR/Cas9 systems were introduced to generate knock-out and knock-in rats [158]. The study utilizing gene-modified animals might be stepped up by advent of knock-out rats. p53 knock out rats are expected to be highly susceptible to chemical carcinogens. They will be applied to short-term carcinogenicity assays even though the p53 knock out rats and p53 knock out mice reveal differing phenotypes [159]. Recently, the data with transgenic rats for evaluation of carcinogenic potency of chemicals have been remarkably accumulated. Transgenic rats for mutagenesis and carcinogenesis will be principal models in future carcinogenesis studies and drug developments.

Perspective

Development of transgenic rats for mutagenesis opened a possibility to use them in repeat dose toxicity assays, thereby enabling general toxicity and genotoxicity assays in same rats [7, 160]. This approach is consistent with the principle of 3Rs (Replacement, Refinement and Reduction) of animal use in laboratory experiments. For this purpose, SD and F344 gpt delta rats were compared with non-transgenic SD and F344 rats for their toxic and genotoxic responses to diethylnitrosamine (DEN) and di(2-ethylhexyl)phthalate (DEHP) [161]. DEN induced similar levels of GST-P foci in the liver of both transgenic and non-transgenic rats. DEN but not DEHP increased gpt and Spi mutation frequency in the liver of transgenic rats. It was concluded that SD and F344 gpt delta rats exhibited comparable toxic and genotoxic responses to DEHP and DEN to those with non-transgenic SD and F344 rats. Therefore, introduction of transgenic rats to repeat dose toxicity assays seems a promising future of toxicology and genotoxicology studies. However, standardization of assay procedures still needs more experimental results and discussion. For example, 4 weeks treatment of chemicals is recommended for gene mutation assays with transgenic rats by OECD TG488. However, KBrO3 at 500 ppm in drinking water needs 9 weeks to detect Spi mutations in the kidney of rats although 8-oxoguanien in DNA is formed by 1 week treatment [119]. Amosite at 2 mg by intratracheal instillation induced lacI mutations in the lung after treatment period of 16 weeks but not after 1 week administration [126]. Administration periods longer than 4 weeks may be required to detect mutations induced by weak mutagens or oxidative stress such as inflammation.

Epigenetic influence of environmental chemicals is an important research area in a field of chemical carcinogenesis. It is well documented that methylation of cytosine and demethylation of 5-MC in DNA, and methylation, acetylation and phosphorylation of histone strongly affect the expression of genes and the phenotypes [162, 163]. Perhaps epigenetic changes may underlie the mechanisms of some of non-genotoxic carcinogens. In fact, one of the mechanisms of nickel-induced carcinogenesis is epigenetic alterations [164]. Although there is no literature where Big Blue rats or gpt delta rats are used for epigenetic studies as far as we searched, one paper reported mechanical irritation increased mutation frequency in bladder without alteration of mutation spectrum [25]. It may be interesting to investigate the epigenetic alterations associated with chemical treatments when the mutation frequency increases without changing the mutation spectrum. Perhaps methylation status of cytosine in DNA may be altered by the treatments.

Recent advance in genome editing technology such as CRISPR/Cas9 has an impact on biomedical research including mutagenesis and carcinogenesis. In the near future, knock-out and knock-in rats will be generated more extensively. Aflatoxin B1 and tamoxifen induce tumors in rats more frequently compared with mice [18, 58]. Thus, genetic factors that affect the carcinogenesis may be investigated with knock-out or knock-in rats. In addition to the genome editing technology, DNA sequence analysis with NGS is greatly evolved recent years. NGS has been employed to characterize lacZ mutations in transgenic mice for mutagenesis [165] and for exome analysis of ENU-induced germ line mutation in gpt delta mice [166]. DNA adducts and mutation signature in human cancer may reflect the history of exposure of the patients to environmental chemicals. Since sensitivity of mass spectrometer has been increased substantially, relationships among DNA adducts, mutations and human cancer will be more extensively studied.

Conclusions

Although mutation is an underlying mechanism of carcinogenesis, the literature reviewed here exhibits complex relationships between in vivo mutagenesis and carcinogenesis even for genotoxic carcinogens. The simplest relationship between mutagenesis and carcinogenesis is that mutations are induced only in the target organs or sub-organs for carcinogenesis. However, mutations are induced by PhIP not only in target lobe of prostate but also in non-target lobes [41]. PhIP induces mutations in the colon of male and female rats while it induces tumors predominantly in male rats. Phenacetin induced mutations in the liver (a non-target organ) much more strongly than in the kidney (the target organ) [52]. Similarly, TDBP induces mutations in the cortex of kidney (a non-target site) more extensively than outer medulla of kidney (the target site) [47]. These results suggest that the highest mutation induction does not coincide with the localization of tumors. The relationship between DNA adduct and mutation is not simple too. Leucomalachite green induces DNA adducts in the liver of rats but no mutations are induced [79]. Bitumen fumes induces DNA adducts in the lung without induction of detectable mutations [132]. Obviously, factors other than mutation such as cell proliferation strongly affect the carcinogenesis. Nevertheless, transgenic rat models for mutagenesis and carcinogenesis are useful tools for various purposes such as regulation of chemicals, chemoprevention studies and mechanistic investigations. Mutation spectra induced by chemical exposure with transgenic rats may be useful to interpret the mutation signatures of human cancer. Advanced sequencing technology coupled with transgenic rat models may contribute significantly to further development of research on chemical mutagenesis and carcinogenesis.

Abbreviations

2,4-DAT:

2,4-diaminotoluene

2,6-DAT:

2,6-diaminotoluene

3-MCPD:

3-monochloropropane-1,2-diol

3Rs:

Replacement, Refinement and Reduction

4-OH-PCB3:

4-hydroxy-PCB3

5-BT:

5-p-dimethylaminophenylazaobenzthiazole

5-MC:

5-methylcytosine

6-BT:

6-p-dimethylaminophenylazaobenzthiazole

BP:

Benzo[a]pyrene

CPA:

Cyproterone acetate

Cx32:

Connexin 32

Cx32Δ transgenic:

Connexin 32 dominant-negative transgenic rats

DAS:

Diallyl sulfide

DE:

Diesel exhaust

DEHP:

Di(2-ethylhexyl)phthalate

DEN:

Diethylnitrosamine

DMBA:

7, 12-dimethylbenz[a]anthracene

DMH:

Dimethyl hydrazine

DMN:

Dimethyl nitrosamine

E. coli :

Escherichia coli

ENU:

N-ethyl-N-nitrosourea

F344:

Fischer 344

GST-P:

Glutathione S-transferase placental form

Hras128:

Human c-Ha-ras proto-oncogene transgenic rats

IQ:

2-amino-3-methylimidazo[4,5-f]quinoline

KBrO3 :

Potassium bromate

MeIQx:

2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline

MNU:

N-methyl-N-nitrosourea

NGS:

Next-generation DNA sequencer

Ni3S2 :

Nickel subsulfide

NMBA:

N-nitrosomethylbenzylamine

OECD:

Organization for Economic Co-operation and Development

PCB3:

4-monochlorobiphenyl

PhIP:

2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine

PoD:

Points of Departure

SD:

Sprague—Dawley

SV40:

Simian virus 40

TALEN:

Transcription activator-like effector nucleases

TCDD:

2,3,7,8-tetrachlorodizenzo-p-dioxin

TDBP:

Tris(2,3-dibromopropyl)phosphate

TRAP:

Probasin-SV40 T antigen transgenic rats

WHO:

World Health Organization

ZFNz:

Zinc-finger nucleases

References

  1. Eastmond DA, Hartwig A, Anderson D, Anwar WA, Cimino MC, Dobrev I, et al. Mutagenicity testing for chemical risk assessment: update of the WHO/IPCS harmonized scheme. Mutagenesis. 2009;24:341–9.

    Article  CAS  PubMed  Google Scholar 

  2. Shelby MD. The genetic toxicity of human carcinogens and its implications. Mutat Res. 1988;204:3–15.

    Article  CAS  PubMed  Google Scholar 

  3. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Madle S, von der HW, Broschinski L, Janig G. Threshold effects in genetic toxicity: perspective of chemicals regulation in Germany. Mutat Res. 2000;464:117–21.

  5. Ames BN, Durston WE, Yamasaki E, Lee FD. Carcinogens are mutagens: a simple test system combining liver homogenates for activation and bacteria for detection. Proc Natl Acad Sci U S A. 1973;70:2281–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. McCann J, Ames BN. Detection of carcinogens as mutagens in the Salmonella/microsome test: assay of 300 chemicals: discussion. Proc Natl Acad Sci U S A. 1976;73:950–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Toyoda-Hokaiwado N, Inoue T, Masumura K, Hayashi H, Kawamura Y, Kurata Y, et al. Integration of in vivo genotoxicity and short-term carcinogenicity assays using F344 gpt delta transgenic rats: in vivo mutagenicity of 2,4-diaminotoluene and 2,6-diaminotoluene structural isomers. Toxicol Sci. 2010;114:71–8.

    Article  CAS  PubMed  Google Scholar 

  8. Cunningham ML, Burka LT, Matthews HB. Metabolism, disposition, and mutagenicity of 2,6-diaminotoluene, a mutagenic noncarcinogen. Drug Metab Dispos. 1989;17:612–7.

    CAS  PubMed  Google Scholar 

  9. Kirkland D, Aardema M, Henderson L, Muller L. Evaluation of the ability of a battery of three in vitro genotoxicity tests to discriminate rodent carcinogens and non-carcinogens I. Sensitivity, specificity and relative predictivity. Mutat Res. 2005;584:1–256.

    Article  CAS  PubMed  Google Scholar 

  10. Fahrig R. A mammalian spot test: induction of genetic alterations in pigment cells of mouse embryos with x-rays and chemical mutagens. Mol Gen Genet. 1975;138:309–14.

    Article  CAS  PubMed  Google Scholar 

  11. Winton DJ, Peacock JH, Ponder BA. Effect of gamma radiation at high- and low-dose rate on a novel in vivo mutation assay in mouse intestine. Mutagenesis. 1989;4:404–6.

    Article  CAS  PubMed  Google Scholar 

  12. Gossen JA, de Leeuw WJ, Tan CH, Zwarthoff EC, Berends F, Lohman PH, et al. Efficient rescue of integrated shuttle vectors from transgenic mice: a model for studying mutations in vivo. Proc Natl Acad Sci U S A. 1989;86:7971–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kohler SW, Provost GS, Fieck A, Kretz PL, Bullock WO, Sorge JA, et al. Spectra of spontaneous and mutagen-induced mutations in the lacI gene in transgenic mice. Proc Natl Acad Sci U S A. 1991;88:7958–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Nohmi T, Katoh M, Suzuki H, Matsui M, Yamada M, Watanabe M, et al. A new transgenic mouse mutagenesis test system using Spi and 6-thioguanine selections. Environ Mol Mutagen. 1996;28:465–70.

    Article  CAS  PubMed  Google Scholar 

  15. Nohmi T, Suzuki T, Masumura K. Recent advances in the protocols of transgenic mouse mutation assays. Mutat Res. 2000;455:191–215.

    Article  CAS  PubMed  Google Scholar 

  16. Dycaico MJ, Provost GS, Kretz PL, Ransom SL, Moores JC, Short JM. The use of shuttle vectors for mutation analysis in transgenic mice and rats. Mutat Res. 1994;307:461–78.

    Article  CAS  PubMed  Google Scholar 

  17. Hayashi H, Kondo H, Masumura K, Shindo Y, Nohmi T. Novel transgenic rat for in vivo genotoxicity assays using 6-thioguanine and Spi selection. Environ Mol Mutagen. 2003;41:253–9.

    Article  CAS  PubMed  Google Scholar 

  18. Dycaico MJ, Stuart GR, Tobal GM, de Boer JG, Glickman BW, Provost GS. Species-specific differences in hepatic mutant frequency and mutational spectrum among lambda/lacI transgenic rats and mice following exposure to aflatoxin B1. Carcinogenesis. 1996;17:2347–56.

    Article  CAS  PubMed  Google Scholar 

  19. Wyborski DL, Malkhosyan S, Moores J, Perucho M, Short JM. Development of a rat cell line containing stably integrated copies of a lambda/lacI shuttle vector. Mutat Res. 1995;334:161–5.

    Article  CAS  PubMed  Google Scholar 

  20. Gollapudi BB, Jackson KM, Stott WT. Hepatic lacI and cII mutation in transgenic (lambdaLIZ) rats treated with dimethylnitrosamine. Mutat Res. 1998;419:131–5.

    Article  CAS  PubMed  Google Scholar 

  21. Jakubczak JL, Merlino G, French JE, Muller WJ, Paul B, Adhya S, et al. Analysis of genetic instability during mammary tumor progression using a novel selection-based assay for in vivo mutations in a bacteriophage lambda transgene target. Proc Natl Acad Sci U S A. 1996;93:9073–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen T, da Gamboa CG, Marques MM, Shelton SD, Beland FA, Manjanatha MG. Mutations induced by alpha-hydroxytamoxifen in the lacI and cII genes of Big blue transgenic rats. Carcinogenesis. 2002;23:1751–7.

    Article  CAS  PubMed  Google Scholar 

  23. Stuart GR, Thorleifson E, Okochi E, de Boer JG, Ushijima T, Nagao M, et al. Interpretation of mutational spectra from different genes: analyses of PhIP-induced mutational specificity in the lacI and cII transgenes from colon of Big Blue rats. Mutat Res. 2000;452:101–21.

  24. Monroe JJ, Manjanatha MG, Skopek TR. Extent of CpG methylation is not proportional to the in vivo spontaneous mutation frequency at transgenic loci in Big Blue rodents. Mutat Res. 2001;476:1–11.

    Article  CAS  PubMed  Google Scholar 

  25. Takahashi S, Ikeda Y, Kimoto N, Okochi E, Cui L, Nagao M, et al. Mutation induction by mechanical irritation caused by uracil-induced urolithiasis in Big Blue rats. Mutat Res. 2000;447:275–80.

    Article  CAS  PubMed  Google Scholar 

  26. Shelton SD, Cherry V, Manjanatha MG. Mutant frequency and molecular analysis of in vivo lacI mutations in the bone marrow of Big Blue rats treated with 7, 12-dimethylbenz[a]anthracene. Environ Mol Mutagen. 2000;36:235–42.

    Article  CAS  PubMed  Google Scholar 

  27. Manjanatha MG, Shelton SD, Aidoo A, Lyn-Cook LE, Casciano DA. Comparison of in vivo mutagenesis in the endogenous Hprt gene and the lacI transgene of Big BlueR rats treated with 7,12-dimethylbenz[a]anthracene. Mutat Res. 1998;401:165–78.

    Article  CAS  PubMed  Google Scholar 

  28. Chen T, Mittelstaedt RA, Aidoo A, Hamilton LP, Beland FA, Casciano DA, et al. Comparison of hprt and lacI mutant frequency with DNA adduct formation in N-hydroxy-2-acetylaminofluorene-treated Big Blue rats. Environ Mol Mutagen. 2001;37:195–202.

    Article  CAS  PubMed  Google Scholar 

  29. Chen T, Aidoo A, Manjanatha MG, Mittelstaedt RA, Shelton SD, Lyn-Cook LE, et al. Comparison of mutant frequencies and types of mutations induced by thiotepa in the endogenous hprt gene and transgenic lacI gene of Big Blue rats. Mutat Res. 1998;403:199–214.

    Article  CAS  PubMed  Google Scholar 

  30. Nohmi T. Novel DNA, polymerases and novel genotoxicity assays. Genes Environ. 2007;29:75–88.

    Article  CAS  Google Scholar 

  31. Nohmi T, Suzuki M, Masumura K, Yamada M, Matsui K, Ueda O, et al. Spi selection: an efficient method to detect gamma-ray-induced deletions in transgenic mice. Environ Mol Mutagen. 1999;34:9–15.

    Article  CAS  PubMed  Google Scholar 

  32. Masumura K, Sakamoto Y, Kumita W, Honma M, Nishikawa A, Nohmi T. Genomic integration of lambda EG10 transgene in gpt delta transgenic rodents. Genes Environ. 2015;37:24. doi:10.1186/s41021-015-0024-6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Masumura K. Spontaneous and induced gpt and Spi mutant frequencies in gpt delta rodents. Genes Environ. 2009;31:105–18.

    Article  CAS  Google Scholar 

  34. IARC. Aflatoxins. IARC Monogr Eval Carcinog Risks Hum. 2002;82:171–300.

    Google Scholar 

  35. IARC. Aristolochic species and aristolochic acid. IARC Monogr Eval Carcinog Risks Hum. 2002;82:69–128.

    Google Scholar 

  36. IARC. ortho-toluidine. IARC Monogr Eval Carcinog Risks Hum. 2000;77:267–322.

    Google Scholar 

  37. Shirai T, Kato K, Futakuchi M, Takahashi S, Suzuki S, Imaida K, et al. Organ differences in the enhancing potential of 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine on carcinogenicity in the prostate, colon and pancreas. Mutat Res. 2002;506–507:129–36.

    Article  PubMed  Google Scholar 

  38. Nagao M, Ushijima T, Watanabe N, Okochi E, Ochiai M, Nakagama H, et al. Studies on mammary carcinogenesis induced by a heterocyclic amine, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine, in mice and rats. Environ Mol Mutagen. 2002;39:158–64.

    Article  CAS  PubMed  Google Scholar 

  39. Okochi E, Watanabe N, Shimada Y, Takahashi S, Wakazono K, Shirai T, et al. Preferential induction of guanine deletion at 5’-GGGA-3’ in rat mammary glands by 2-amino- 1-methyl-6-phenylimidazo[4,5-b]pyridine. Carcinogenesis. 1999;20:1933–8.

    Article  CAS  PubMed  Google Scholar 

  40. Shan L, Yu M, Schut HA, Snyderwine EG. Susceptibility of rats to mammary gland carcinogenesis by the food-derived carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) varies with age and is associated with the induction of differential gene expression. Am J Pathol. 2004;165:191–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nakai Y, Nelson WG, De Marzo AM. The dietary charred meat carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine acts as both a tumor initiator and promoter in the rat ventral prostate. Cancer Res. 2007;67:1378–84.

    Article  CAS  PubMed  Google Scholar 

  42. Stuart GR, Holcroft J, de Boer JG, Glickman BW. Prostate mutations in rats induced by the suspected human carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine. Cancer Res. 2000;60:266–8.

    CAS  PubMed  Google Scholar 

  43. Ito N, Hasegawa R, Sano M, Tamano S, Esumi H, Takayama S, et al. A new colon and mammary carcinogen in cooked food, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). Carcinogenesis. 1991;12:1503–6.

    Article  CAS  PubMed  Google Scholar 

  44. Okonogi H, Stuart GR, Okochi E, Ushijima T, Sugimura T, Glickman BW, et al. Effects of gender and species on spectra of mutation induced by 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine in the lacI transgene. Mutat Res. 1997;395:93–9.

    Article  CAS  PubMed  Google Scholar 

  45. Stuart GR, de Boer JG, Haesevoets R, Holcroft J, Kangas J, Sojonky K, et al. Mutations induced by 2-amino-1-methyl-6-phenylimidazo [4,5-b]pyridine (PhIP) in cecum and proximal and distal colon of lacI transgenic rats. Mutagenesis. 2001;16:431–7.

    Article  CAS  PubMed  Google Scholar 

  46. Cunningham ML, Elwell MR, Matthews HB. Site-specific cell proliferation in renal tubular cells by the renal tubular carcinogen tris(2,3-dibromopropyl)phosphate. Environ Health Perspect. 1993;101 Suppl 5:253–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. de Boer JG, Holcroft J, Cunningham ML, Glickman BW. Tris(2,3-dibromopropyl)phosphate causes a gradient of mutations in the cortex and outer and inner medullas of the kidney of lacI transgenic rats. Environ Mol Mutagen. 2000;36:1–4.

    Article  PubMed  Google Scholar 

  48. Cunningham ML, Elwell MR, Matthews HB. Relationship of carcinogenicity and cellular proliferation induced by mutagenic noncarcinogens vs carcinogens. III. Organophosphate pesticides vs tris(2,3-dibromopropyl)phosphate. Fundam Appl Toxicol. 1994;23:363–9.

    Article  CAS  PubMed  Google Scholar 

  49. National Toxicology Program (NTP). Toxicology and carcinogenesis studies of ochratoxin A (CAS No.303-47-9) in F344/N rats (gavage studies). Natl Toxicol Program Tech Rep Ser. 1989;1–142.

  50. Hibi D, Suzuki Y, Ishii Y, Jin M, Watanabe M, Sugita-Konishi Y, et al. Site-specific in vivo mutagenicity in the kidney of gpt delta rats given a carcinogenic dose of ochratoxin A. Toxicol Sci. 2011;122:406–14.

    Article  CAS  PubMed  Google Scholar 

  51. Isaka H, Yoshii H, Otsuji A, Koike M, Nagai Y, Koura M, et al. Tumors of Sprague-Dawley rats induced by long-term feeding of phenacetin. Gann. 1979;70:29–36.

    CAS  PubMed  Google Scholar 

  52. Kawamura Y, Hayashi H, Masumura K, Numazawa S, Nohmi T. Genotoxicity of phenacetin in the kidney and liver of Sprague-Dawley gpt delta transgenic rats in 26-week and 52-week repeated-dose studies. Toxicology. 2014;324:10–7.

    Article  CAS  PubMed  Google Scholar 

  53. National Toxicology Program (NTP). Bioassay of 2,4-diaminotoluene for possible carcinogenicity. Natl Cancer Inst. Carcinog. Tech. Res. Ser. 1979: 1–139.

  54. National Toxicology Program (NTP). Bioassay of 2,6-diaminotoluene dihydrochloride for possible carcinogenicity (CAS No. 15481-70-6). Natl. Toxicol Program Tech Rep Ser. 1980;1–123.

  55. Sui H, Ohta R, Shiragiku T, Akahori A, Suzuki K, Nakajima M, et al. Evaluation of in vivo mutagenicity by 2,4-diaminotoluene and 2,6-diaminotoluene in liver of F344 gpt delta transgenic rat dosed for 28 days: a collaborative study of the gpt delta transgenic rat mutation assay. Genes Environ. 2012;34:25–33.

    Article  CAS  Google Scholar 

  56. Jordan VC. Overview from the international conference on long-term tamoxifen therapy for breast cancer. J Natl Cancer Inst. 1992;84:231–4.

    Article  CAS  PubMed  Google Scholar 

  57. Li D, Dragan Y, Jordan VC, Wang M, Pitot HC. Effects of chronic administration of tamoxifen and toremifene on DNA adducts in rat liver, kidney, and uterus. Cancer Res. 1997;57:1438–41.

    CAS  PubMed  Google Scholar 

  58. Kawamura Y, Hayashi H, Kurata Y, Hiratsuka K, Masumura K, Nohmi T. Evaluation of the genotoxicity of tamoxifen in the liver and kidney of F344 gpt delta transgenic rat in 3-week and 13-week repeated dose studies. Toxicology. 2013;312:56–62.

    Article  CAS  PubMed  Google Scholar 

  59. Brown EV, Sanchorawala CJ. Carcinogenic activity of analogs of p-dimethylaminoazobenzene. VI. Activity of the benzimidazole and benzthiazole analogs. J Med Chem. 1968;11:1074–5.

    Article  CAS  PubMed  Google Scholar 

  60. Ashby J, Lefevre PA, Styles JA, Charlesworth J, Paton D. Comparisons between carcinogenic potency and mutagenic potency to Salmonella in a series of derivatives of 4-dimethylaminoazobenzene (DAB). Mutat Res. 1982;93:67–81.

    Article  CAS  PubMed  Google Scholar 

  61. Fletcher K, Soames AR, Tinwell H, Lefevre PA, Ashby J. Hepatic gene mutations induced in Big Blue rats by both the potent rat liver azo-carcinogen 6BT and its reported noncarcinogenic analogue 5BT. Environ Mol Mutagen. 1999;34:148–53.

    Article  CAS  PubMed  Google Scholar 

  62. Lemire JM, Shiojiri N, Fausto N. Oval cell proliferation and the origin of small hepatocytes in liver injury induced by D-galactosamine. Am J Pathol. 1991;139:535–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Nohmi T. Past, present and future directions of gpt delta rodent gene mutation assays. Food Saf. 2016. doi:10.14252/foodsafetyfscj.2015024.

  64. FAO, WHO. Risk Characterization. Principles and methods for the risk assessment of chemicals in food. 2009. p. 7–1–7–18.

  65. Kuroda K, Ishii Y, Takasu S, Kijima A, Matsushita K, Watanabe M, et al. Cell cycle progression, but not genotoxic activity, mainly contributes to citrinin-induced renal carcinogenesis. Toxicology. 2013;311:216–24.

    Article  CAS  PubMed  Google Scholar 

  66. Onami S, Cho YM, Toyoda T, Horibata K, Ishii Y, Umemura T, et al. Absence of in vivo genotoxicity of 3-monochloropropane-1,2-diol and associated fatty acid esters in a 4-week comprehensive toxicity study using F344 gpt delta rats. Mutagenesis. 2014;29:295–302.

    Article  CAS  PubMed  Google Scholar 

  67. Arai M, Hibino T. Tumorigenicity of citrinin in male F344 rats. Cancer Lett. 1983;17:281–7.

    Article  CAS  PubMed  Google Scholar 

  68. IARC. 3-Monochloro-1,2-propanediol. IARC Monogr Eval Carcinog Risks Hum. 2012;101:349–74.

    Google Scholar 

  69. Zeiger E, Anderson B, Haworth S, Lawlor T, Mortelmans K. Salmonella mutagenicity tests: IV. Results from the testing of 300 chemicals. Environ Mol Mutagen. 1988;11 Suppl 12:1–157.

    Article  CAS  PubMed  Google Scholar 

  70. Onami S, Cho YM, Toyoda T, Mizuta Y, Yoshida M, Nishikawa A, et al. A 13-week repeated dose study of three 3-monochloropropane-1,2-diol fatty acid esters in F344 rats. Arch Toxicol. 2014;88:871–80.

    Article  CAS  PubMed  Google Scholar 

  71. Abraham K, Appel KE, Berger-Preiss E, Apel E, Gerling S, Mielke H, et al. Relative oral bioavailability of 3-MCPD from 3-MCPD fatty acid esters in rats. Arch Toxicol. 2013;87:649–59.

    Article  CAS  PubMed  Google Scholar 

  72. Suzuki Y, Umemura T, Hibi D, Inoue T, Jin M, Ishii Y, et al. Possible involvement of genotoxic mechanisms in estragole-induced hepatocarcinogenesis in rats. Arch Toxicol. 2012;86:1593–601.

    Article  CAS  PubMed  Google Scholar 

  73. Ishii Y, Takasu S, Kuroda K, Matsushita K, Kijima A, Nohmi T, et al. Combined application of comprehensive analysis for DNA modification and reporter gene mutation assay to evaluate kidneys of gpt delta rats given madder color or its constituents. Anal Bioanal Chem. 2014;406:2467–75.

    Article  CAS  PubMed  Google Scholar 

  74. Jin M, Kijima A, Hibi D, Ishii Y, Takasu S, Matsushita K, et al. In vivo genotoxicity of methyleugenol in gpt delta transgenic rats following medium-term exposure. Toxicol Sci. 2013;131:387–94.

    Article  CAS  PubMed  Google Scholar 

  75. European Commission Scientific Committee on Food. Estragole (1-Allyl-4-methoxybenzene). Opinion of the Scientific Committee on Food. 2001;http://ec.europa.eu/food/fs/sc/scf/out104_en.pdf.

  76. Zeiger E, Anderson B, Haworth S, Lawlor T, Mortelmans K, Speck W. Salmonella mutagenicity tests: III. Results from the testing of 255 chemicals. Environ Mutagen. 1987;9 Suppl 9:1–109.

    Article  CAS  PubMed  Google Scholar 

  77. Inoue K, Yoshida M, Takahashi M, Shibutani M, Takagi H, Hirose M, et al. Induction of kidney and liver cancers by the natural food additive madder color in a two-year rat carcinogenicity study. Food Chem Toxicol. 2009;47:184–91.

    Article  CAS  PubMed  Google Scholar 

  78. National Toxicology Program (NTP). Toxicology and carcinogenesis studies of Methyleugenol (CAS No. 93-15-2) in F344/N and B6C3F1 mice (Gavage studies). Natl Toxicol Program Tech Rep Ser. 2000;1–412.

  79. Culp SJ, Beland FA, Heflich RH, Benson RW, Blankenship LR, Webb PJ, et al. Mutagenicity and carcinogenicity in relation to DNA adduct formation in rats fed leucomalachite green. Mutat Res. 2002;506–507:55–63.

    Article  PubMed  Google Scholar 

  80. Culp SJ, Mellick PW, Trotter RW, Greenlees KJ, Kodell RL, Beland FA. Carcinogenicity of malachite green chloride and leucomalachite green in B6C3F1 mice and F344 rats. Food Chem Toxicol. 2006;44:1204–12.

    Article  CAS  PubMed  Google Scholar 

  81. Manjanatha MG, Shelton SD, Bishop M, Shaddock JG, Dobrovolsky VN, Heflich RH, et al. Analysis of mutations and bone marrow micronuclei in Big Blue rats fed leucomalachite green. Mutat Res. 2004;547:5–18.

    Article  CAS  PubMed  Google Scholar 

  82. Kuroda K, Hibi D, Ishii Y, Takasu S, Kijima A, Matsushita K, et al. Ochratoxin A induces DNA double-strand breaks and large deletion mutations in the carcinogenic target site of gpt delta rats. Mutagenesis. 2014;29:27–36.

  83. Hibi D, Kijima A, Suzuki Y, Ishii Y, Jin M, Sugita-Konishi Y, et al. Effects of p53 knockout on ochratoxin A-induced genotoxicity in p53-deficient gpt delta mice. Toxicology. 2013;304:92–9.

  84. Kuroda K, Hibi D, Ishii Y, Yokoo Y, Takasu S, Kijima A, et al. Role of p53 in the progression from ochratoxin A-induced DNA damage to gene mutations in the kidneys of mice. Toxicol Sci. 2015;144:65–76.

  85. Hoshi M, Morimura K, Wanibuchi H, Wei M, Okochi E, Ushijima T, et al. No-observed effect levels for carcinogenicity and for in vivo mutagenicity of a genotoxic carcinogen. Toxicol Sci. 2004;81:273–9.

    Article  CAS  PubMed  Google Scholar 

  86. Kato T, Ohgaki H, Hasegawa H, Sato S, Takayama S, Sugimura T. Carcinogenicity in rats of a mutagenic compound, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline. Carcinogenesis. 1988;9:71–3.

    Article  CAS  PubMed  Google Scholar 

  87. Yamaguchi T, Wei M, Hagihara N, Omori M, Wanibuchi H, Fukushima S. Lack of mutagenic and toxic effects of low dose potassium bromate on kidneys in the Big Blue rat. Mutat Res. 2008;652:1–11.

    Article  CAS  PubMed  Google Scholar 

  88. Kurokawa Y, Hayashi Y, Maekawa A, Takahashi M, Kokubo T. Induction of renal cell tumors in F-344 rats by oral administration of potassium bromate, a food additive. Gan. 1982;73:335–8.

    CAS  PubMed  Google Scholar 

  89. Schuppler J, Gunzel P. Liver tumors and steroid hormones in rats and mice. Arch Toxicol Suppl. 1979;(2):181–95.

  90. Wolff T, Topinka J, Deml E, Oesterle D, Schwarz LR. Dose dependent induction of DNA adducts, gene mutations, and cell proliferation by the antiandrogenic drug cyproterone acetate in rat liver. Adv Exp Med Biol. 2001;500:687–96.

    Article  CAS  PubMed  Google Scholar 

  91. Cao X, Mittelstaedt RA, Pearce MG, Allen BC, Soeteman-Hernandez LG, Johnson GE, et al. Quantitative dose-response analysis of ethyl methanesulfonate genotoxicity in adult gpt-delta transgenic mice. Environ Mol Mutagen. 2014;55:385–99.

  92. Gollapudi BB, Johnson GE, Hernandez LG, Pottenger LH, Dearfield KL, Jeffrey AM, et al. Quantitative approaches for assessing dose-response relationships in genetic toxicology studies. Environ Mol Mutagen. 2013;54:8–18.

  93. de Boer JG, Yang H, Holcroft J, Skov K. Chemoprotection against N-nitrosomethylbenzylamine-induced mutation in the rat esophagus. Nutr Cancer. 2004;50:168–73.

    Article  PubMed  Google Scholar 

  94. Chen T, Hutts RC, Mei N, Liu X, Bishop ME, Shelton S, et al. Endogenous estrogen status, but not genistein supplementation, modulates 7,12-dimethylbenz[a]anthracene-induced mutation in the liver cII gene of transgenic big blue rats. Environ Mol Mutagen. 2005;45:409–18.

    Article  CAS  PubMed  Google Scholar 

  95. Manjanatha MG, Shelton S, Bishop ME, Lyn-Cook LE, Aidoo A. Dietary effects of soy isoflavones daidzein and genistein on 7,12-dimethylbenz[a]anthracene-induced mammary mutagenesis and carcinogenesis in ovariectomized Big Blue transgenic rats. Carcinogenesis. 2006;27:2555–64.

    Article  CAS  PubMed  Google Scholar 

  96. Aidoo A, Bishop ME, Shelton SD, Lyn-Cook LE, Chen T, Manjanatha MG. Effects of daidzein, genistein, and 17beta-estradiol on 7,12-dimethylbenz[a]anthracene-induced mutagenicity and uterine dysplasia in ovariectomized rats. Nutr Cancer. 2005;53:82–90.

    Article  CAS  PubMed  Google Scholar 

  97. Josyula S, Schut HA. Effects of dietary conjugated linoleic acid on DNA adduct formation of PhIP and IQ after bolus administration to female F344 rats. Nutr Cancer. 1998;32:139–45.

    Article  CAS  PubMed  Google Scholar 

  98. Yang H, Glickman BW, de Boer JG. Sex-specific induction of mutations by PhIP in the kidney of male and female rats and its modulation by conjugated linoleic acid. Environ Mol Mutagen. 2002;40:116–21.

    Article  CAS  PubMed  Google Scholar 

  99. Giovannucci E. Insulin, insulin-like growth factors and colon cancer: a review of the evidence. J Nutr. 2001;131:3109S–20S.

    CAS  PubMed  Google Scholar 

  100. Hansen M, Hald MT, Autrup H, Vogel U, Bornholdt J, Moller P, et al. Sucrose and IQ induced mutations in rat colon by independent mechanism. Mutat Res. 2004;554:279–86.

    Article  CAS  PubMed  Google Scholar 

  101. Hansen M, Baunsgaard D, Autrup H, Vogel UB, Moller P, Lindecrona R, et al. Sucrose, glucose and fructose have similar genotoxicity in the rat colon and affect the metabolome. Food Chem Toxicol. 2008;46:752–60.

    Article  CAS  PubMed  Google Scholar 

  102. Cooney GT, Holcroft J, de Boer JG. The effect of dietary restriction on PhIP-induced mutation in the distal colon and B[a]P- and ENU-induced mutation in the liver of the rat. Nutr Cancer. 2004;50:63–70.

    Article  CAS  PubMed  Google Scholar 

  103. Loli P, Topinka J, Georgiadis P, Dusinska M, Hurbankova M, Kovacikova Z, et al. Benzo[a]pyrene-enhanced mutagenesis by asbestos in the lung of lambda-lacI transgenic rats. Mutat Res. 2004;553:79–90.

    Article  CAS  PubMed  Google Scholar 

  104. Knerr S, Schrenk D. Carcinogenicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in experimental models. Mol Nutr Food Res. 2006;50:897–907.

    Article  CAS  PubMed  Google Scholar 

  105. Thornton AS, Oda Y, Stuart GR, Holcroft J, de Boer JG. The dioxin TCDD protects against aflatoxin-induced mutation in female rats, but not in male rats. Mutat Res. 2004;561:147–52.

    Article  CAS  PubMed  Google Scholar 

  106. Stearns V, Gelmann EP. Does tamoxifen cause cancer in humans? J Clin Oncol. 1998;16:779–92.

    CAS  PubMed  Google Scholar 

  107. Glatt H, Davis W, Meinl W, Hermersdorfer H, Venitt S, Phillips DH. Rat, but not human, sulfotransferase activates a tamoxifen metabolite to produce DNA adducts and gene mutations in bacteria and mammalian cells in culture. Carcinogenesis. 1998;19:1709–13.

    Article  CAS  PubMed  Google Scholar 

  108. Davies R, Oreffo VI, Bayliss S, Dinh PA, Lilley KS, White IN, et al. Mutational spectra of tamoxifen-induced mutations in the livers of lacI transgenic rats. Environ Mol Mutagen. 1996;28:430–3.

    Article  CAS  PubMed  Google Scholar 

  109. Davies R, Gant TW, Smith LL, Styles JA. Tamoxifen induces G:C-- > T:A mutations in the cII gene in the liver of lambda/lacI transgenic rats but not at 5’-CpG-3’ dinucleotide sequences as found in the lacI transgene. Carcinogenesis. 1999;20:1351–6.

  110. Styles JA, Davies R, Fenwick S, Walker J, White IN, Smith LL. Tamoxifen mutagenesis and carcinogenesis in livers of lambda/lacI transgenic rats: selective influence of phenobarbital promotion. Cancer Lett. 2001;162:117–22.

    Article  CAS  PubMed  Google Scholar 

  111. IARC. Some traditional herbal medicines, some mycotoxins, naphthalene and stylene. IARC Monogr Eval Carcinog Risk Chem Hum. 2002;82:69–128.

  112. Mei N, Arlt VM, Phillips DH, Heflich RH, Chen T. DNA adduct formation and mutation induction by aristolochic acid in rat kidney and liver. Mutat Res. 2006;602:83–91.

    Article  CAS  PubMed  Google Scholar 

  113. Chen L, Mei N, Yao L, Chen T. Mutations induced by carcinogenic doses of aristolochic acid in kidney of Big Blue transgenic rats. Toxicol Lett. 2006;165:250–6.

  114. IARC. Senecio species and riddelline. IARC Monogr Eval Carcinog Risks Hum. 2002;82:153–68.

    Google Scholar 

  115. Mei N, Heflich RH, Chou MW, Chen T. Mutations induced by the carcinogenic pyrrolizidine alkaloid riddelliine in the liver cII gene of transgenic big blue rats. Chem Res Toxicol. 2004;17:814–8.

    Article  CAS  PubMed  Google Scholar 

  116. Mei N, Chou MW, Fu PP, Heflich RH, Chen T. Differential mutagenicity of riddelliine in liver endothelial and parenchymal cells of transgenic big blue rats. Cancer Lett. 2004;215:151–8.

    Article  CAS  PubMed  Google Scholar 

  117. Luan Y, Suzuki T, Palanisamy R, Takashima Y, Sakamoto H, Sakuraba M, et al. Potassium bromate treatment predominantly causes large deletions, but not GC > TA transversion in human cells. Mutat Res. 2007;619:113–23.

    Article  CAS  PubMed  Google Scholar 

  118. Harrington-Brock K, Collard DD, Chen T. Bromate induces loss of heterozygosity in the thymidine kinase gene of L5178Y/Tk +/−-3.7.2C mouse lymphoma cells. Mutat Res. 2003;537:21–8.

    Article  CAS  PubMed  Google Scholar 

  119. Umemura T, Kanki K, Kuroiwa Y, Ishii Y, Okano K, Nohmi T, et al. In vivo mutagenicity and initiation following oxidative DNA lesion in the kidneys of rats given potassium bromate. Cancer Sci. 2006;97:829–35.

    Article  CAS  PubMed  Google Scholar 

  120. Umemura T, Tasaki M, Kijima A, Okamura T, Inoue T, Ishii Y, et al. Possible participation of oxidative stress in causation of cell proliferation and in vivo mutagenicity in kidneys of gpt delta rats treated with potassium bromate. Toxicology. 2009;257:46–52.

    Article  CAS  PubMed  Google Scholar 

  121. IARC. Chromium, Nickel and Welding. IARC Monogr Eval Carcinog Risks Hum. 1990;49:257–446.

    Google Scholar 

  122. Dunnick JK, Elwell MR, Radovsky AE, Benson JM, Hahn FF, Nikula KJ, et al. Comparative carcinogenic effects of nickel subsulfide, nickel oxide, or nickel sulfate hexahydrate chronic exposures in the lung. Cancer Res. 1995;55:5251–6.

    CAS  PubMed  Google Scholar 

  123. Mayer C, Klein RG, Wesch H, Schmezer P. Nickel subsulfide is genotoxic in vitro but shows no mutagenic potential in respiratory tract tissues of BigBlue rats and muta mouse mice in vivo after inhalation. Mutat Res. 1998;420:85–98.

    Article  CAS  PubMed  Google Scholar 

  124. Kamigaito T, Noguchi T, Narumi K, Takashima R, Hamada S, Sanada H, et al. Evaluation of the in vivo mutagenicity of nickel subsulfide in the lung of F344 gpt delta transgenic rats exposed by intratracheal instillation: a collaborative study for the gpt delta transgenic rat mutation assay. Genes Environ. 2012;34:34–44.

    Article  CAS  Google Scholar 

  125. Hashim D, Boffetta P. Occupational and environmental exposures and cancers in developing countries. Ann Glob Health. 2014;80:393–411.

    Article  PubMed  Google Scholar 

  126. Topinka J, Loli P, Georgiadis P, Dusinska M, Hurbankova M, Kovacikova Z, et al. Mutagenesis by asbestos in the lung of lambda-lacI transgenic rats. Mutat Res. 2004;553:67–78.

    Article  CAS  PubMed  Google Scholar 

  127. Topinka J, Loli P, Dusinska M, Hurbankova M, Kovacikova Z, Volkovova K, et al. Mutagenesis by man-made mineral fibres in the lung of rats. Mutat Res. 2006;595:174–83.

    Article  CAS  PubMed  Google Scholar 

  128. Thornton AS, Oda Y, Stuart GR, Glickman BW, de Boer JG. Mutagenicity of TCDD in Big Blue transgenic rats. Mutat Res. 2001;478:45–50.

  129. McClellan RO. Health effects of exposure to diesel exhaust particles. Annu Rev Pharmacol Toxicol. 1987;27:279–300.

    Article  CAS  PubMed  Google Scholar 

  130. Sato H, Sone H, Sagai M, Suzuki KT, Aoki Y. Increase in mutation frequency in lung of Big Blue rat by exposure to diesel exhaust. Carcinogenesis. 2000;21:653–61.

  131. Muller AK, Farombi EO, Moller P, Autrup HN, Vogel U, Wallin H, et al. DNA damage in lung after oral exposure to diesel exhaust particles in Big Blue rats. Mutat Res. 2004;550:123–32.

  132. Bottin MC, Gate L, Rihn B, Micillino JC, Nathalie M, Martin A, et al. Genotoxic effects of bitumen fumes in Big Blue transgenic rat lung. Mutat Res. 2006;596:91–105.

    Article  CAS  PubMed  Google Scholar 

  133. Lehmann L, Esch HL, Kirby PA, Robertson LW, Ludewig G. 4-monochlorobiphenyl (PCB3) induces mutations in the livers of transgenic fisher 344 rats. Carcinogenesis. 2007;28:471–8.

    Article  CAS  PubMed  Google Scholar 

  134. McLean MR, Bauer U, Amaro AR, Robertson LW. Identification of catechol and hydroquinone metabolites of 4-monochlorobiphenyl. Chem Res Toxicol. 1996;9:158–64.

    Article  CAS  PubMed  Google Scholar 

  135. Maddox C, Wang B, Kirby PA, Wang K, Ludewig G. Mutagenicity of 3-methylcholanthrene, PCB3, and 4-OH-PCB3 in the lung of transgenic bigblue rats. Environ Toxicol Pharmacol. 2008;25:260–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Dragsted LO, Daneshvar B, Vogel U, Autrup HN, Wallin H, Risom L, et al. A sucrose-rich diet induces mutations in the rat colon. Cancer Res. 2002;62:4339–45.

    CAS  PubMed  Google Scholar 

  137. Ito N, Tsuda H, Tatematsu M, Inoue T, Tagawa Y, Aoki T, et al. Enhancing effect of various hepatocarcinogens on induction of preneoplastic glutathione S-transferase placental form positive foci in rats—an approach for a new medium-term bioassay system. Carcinogenesis. 1988;9:387–94.

  138. Asamoto M, Ochiya T, Toriyama-Baba H, Ota T, Sekiya T, Terada M, et al. Transgenic rats carrying human c-Ha-ras proto-oncogenes are highly susceptible to N-methyl-N-nitrosourea mammary carcinogenesis. Carcinogenesis. 2000;21:243–9.

    Article  CAS  PubMed  Google Scholar 

  139. Naiki-Ito A, Asamoto M, Hokaiwado N, Takahashi S, Yamashita H, Tsuda H, et al. Gpx2 is an overexpressed gene in rat breast cancers induced by three different chemical carcinogens. Cancer Res. 2007;67:11353–8.

    Article  CAS  PubMed  Google Scholar 

  140. Asamoto M, Ota T, Toriyama-Baba H, Hokaiwado N, Naito A, Tsuda H. Mammary carcinomas induced in human c-Ha-ras proto-oncogene transgenic rats are estrogen-independent, but responsive to d-limonene treatment. Jpn J Cancer Res. 2002;93:32–5.

    Article  CAS  PubMed  Google Scholar 

  141. Ota T, Asamoto M, Toriyama-Baba H, Yamamoto F, Matsuoka Y, Ochiya T, et al. Transgenic rats carrying copies of the human c-Ha-ras proto-oncogene exhibit enhanced susceptibility to N-butyl-N-(4-hydroxybutyl)nitrosamine bladder carcinogenesis. Carcinogenesis. 2000;21:1391–6.

    Article  CAS  PubMed  Google Scholar 

  142. Asamoto M, Toriyama-Baba H, Ohnishi T, Naito A, Ota T, Ando A, et al. Transgenic rats carrying human c-Ha-ras proto-oncogene are highly susceptible to N-nitrosomethylbenzylamine induction of esophageal tumorigenesis. Jpn J Cancer Res. 2002;93:744–51.

    Article  CAS  PubMed  Google Scholar 

  143. National BioResource Project Information Site. National BioResource Project. 2002. http://www.nbrp.jp/.

  144. RIKEN cell bank. RIKEN BRC. http://cell.brc.riken.jp/en/.

  145. Asamoto M, Hokaiwado N, Cho YM, Takahashi S, Ikeda Y, Imaida K, et al. Prostate carcinomas developing in transgenic rats with SV40 T antigen expression under probasin promoter control are strictly androgen dependent. Cancer Res. 2001;61:4693–700.

    CAS  PubMed  Google Scholar 

  146. Said MM, Hokaiwado N, Tang M, Ogawa K, Suzuki S, Ghanem HM, et al. Inhibition of prostate carcinogenesis in probasin/SV40 T antigen transgenic rats by leuprorelin, a luteinizing hormone-releasing hormone agonist. Cancer Sci. 2006;97:459–67.

    Article  CAS  PubMed  Google Scholar 

  147. Cho YM, Takahashi S, Asamoto M, Suzuki S, Inaguma S, Hokaiwado N, et al. Age-dependent histopathological findings in the prostate of probasin/SV40 T antigen transgenic rats: lack of influence of carcinogen or testosterone treatment. Cancer Sci. 2003;94:153–7.

    Article  CAS  PubMed  Google Scholar 

  148. Tang M, Ogawa K, Asamoto M, Hokaiwado N, Seeni A, Suzuki S, et al. Protective effects of citrus nobiletin and auraptene in transgenic rats developing adenocarcinoma of the prostate (TRAP) and human prostate carcinoma cells. Cancer Sci. 2007;98:471–7.

    Article  CAS  PubMed  Google Scholar 

  149. Asamoto M, Hokaiwado N, Murasaki T, Shirai T. Connexin 32 dominant-negative mutant transgenic rats are resistant to hepatic damage by chemicals. Hepatology. 2004;40:205–10.

    Article  CAS  PubMed  Google Scholar 

  150. Hokaiwado N, Asamoto M, Ogawa K, Shirai T. Transgenic disruption of gap junctional intercellular communication enhances early but not late stage hepatocarcinogenesis in the rat. Toxicol Pathol. 2005;33:695–701.

    Article  CAS  PubMed  Google Scholar 

  151. Hokaiwado N, Asamoto M, Futakuchi M, Ogawa K, Takahashi S, Shirai T. Both early and late stages of hepatocarcinogenesis are enhanced in Cx32 dominant negative mutant transgenic rats with disrupted gap junctional intercellular communication. J Membr Biol. 2007;218:101–6.

    Article  CAS  PubMed  Google Scholar 

  152. Fukamachi K, Tanaka H, Hagiwara Y, Ohara H, Joh T, Iigo M, et al. An animal model of preclinical diagnosis of pancreatic ductal adenocarcinomas. Biochem Biophys Res Commun. 2009;390:636–41.

    Article  CAS  PubMed  Google Scholar 

  153. Ueda S, Fukamachi K, Matsuoka Y, Takasuka N, Takeshita F, Naito A, et al. Ductal origin of pancreatic adenocarcinomas induced by conditional activation of a human Ha-ras oncogene in rat pancreas. Carcinogenesis. 2006;27:2497–510.

    Article  CAS  PubMed  Google Scholar 

  154. Storer RD, French JE, Haseman J, Hajian G, LeGrand EK, Long GG, et al. P53+/− hemizygous knockout mouse: overview of available data. Toxicol Pathol. 2001;29(Suppl):30–50.

    Article  CAS  PubMed  Google Scholar 

  155. Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi VM, et al. Knockout rats via embryo microinjection of zinc-finger nucleases. Science. 2009;325:433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Mashimo T, Takizawa A, Voigt B, Yoshimi K, Hiai H, Kuramoto T, et al. Generation of knockout rats with X-linked severe combined immunodeficiency (X-SCID) using zinc-finger nucleases. PLoS One. 2010;5:e8870.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Tong C, Li P, Wu NL, Yan Y, Ying QL. Production of p53 gene knockout rats by homologous recombination in embryonic stem cells. Nature. 2010;467:211–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Guan Y, Shao Y, Li D, Liu M. Generation of site-specific mutations in the rat genome via CRISPR/Cas9. Methods Enzymol. 2014;546:297–317.

    Article  CAS  PubMed  Google Scholar 

  159. Kawaharada K, Kawamata M, Ochiya T. Rat embryonic stem cells create new era in development of genetically manipulated rat models. World J Stem Cells. 2015;7:1054–63.

    PubMed  PubMed Central  Google Scholar 

  160. Matsushita K, Kijima A, Ishii Y, Takasu S, Jin M, Kuroda K, et al. Development of a medium-term animal model using gpt delta rats to evaluate chemical carcinogenicity and genotoxicity. J Toxicol Pathol. 2013;26:19–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Akagi J, Toyoda T, Cho YM, Mizuta Y, Nohmi T, Nishikawa A, et al. Validation study of the combined repeated-dose toxicity and genotoxicity assay using gpt delta rats. Cancer Sci. 2015;106:529–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Verdin E, Ott M. 50 years of protein acetylation: from gene regulation to epigenetics, metabolism and beyond. Nat Rev Mol Cell Biol. 2015;16:258–64.

    Article  CAS  PubMed  Google Scholar 

  163. Alabert C, Groth A. Chromatin replication and epigenome maintenance. Nat Rev Mol Cell Biol. 2012;13:153–67.

    Article  CAS  PubMed  Google Scholar 

  164. Salnikow K, Costa M. Epigenetic mechanisms of nickel carcinogenesis. J Environ Pathol Toxicol Oncol. 2000;19:307–18.

    CAS  PubMed  Google Scholar 

  165. Beal MA, Gagne R, Williams A, Marchetti F, Yauk CL. Characterizing Benzo[a]pyrene-induced lacZ mutation spectrum in transgenic mice using next-generation sequencing. BMC Genomics. 2015;16. doi:10.1186/s12864-015-2004-4.

  166. Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M, Nohmi T. Estimation of the frequency of inherited germline mutations by whole exome sequencing in ethyl nitrosourea-treated and untreated gpt delta mice. Genes Environ. 2016;38. doi:110.1186/s41021-016-0035-y.

  167. Kanki K, Nishikawa A, Masumura K, Umemura T, Imazawa T, Kitamura Y, et al. In vivo mutational analysis of liver DNA in gpt delta transgenic rats treated with the hepatocarcinogens N-nitrosopyrrolidine, 2-amino-3-methylimidazo[4,5-f]quinoline, and di(2-ethylhexyl)phthalate. Mol Carcinog. 2005;42:9–17.

    Article  CAS  PubMed  Google Scholar 

  168. Mei N, McDaniel LP, Dobrovolsky VN, Guo X, Shaddock JG, Mittelstaedt RA, et al. The genotoxicity of acrylamide and glycidamide in big blue rats. Toxicol Sci. 2010;115:412–21.

    Article  CAS  PubMed  Google Scholar 

  169. Koyama N, Yasui M, Kimura A, Takami S, Suzuki T, Masumura K, et al. Acrylamide genotoxicity in young versus adult gpt delta male rats. Mutagenesis. 2011;26:545–9.

    Article  CAS  PubMed  Google Scholar 

  170. Davies R, Oreffo VI, Martin EA, Festing MF, White IN, Smith LL, et al. Tamoxifen causes gene mutations in the livers of lambda/lacI transgenic rats. Cancer Res. 1997;57:1288–93.

    CAS  PubMed  Google Scholar 

  171. Bol SA, Horlbeck J, Markovic J, de Boer JG, Turesky RJ, Constable A. Mutational analysis of the liver, colon and kidney of Big Blue rats treated with 2-amino-3-methylimidazo[4,5-f]quinoline. Carcinogenesis. 2000;21:1–6.

    Article  CAS  PubMed  Google Scholar 

  172. Moller P, Wallin H, Vogel U, Autrup H, Risom L, Hald MT, et al. Mutagenicity of 2-amino-3-methylimidazo[4,5-f]quinoline in colon and liver of Big Blue rats: role of DNA adducts, strand breaks, DNA repair and oxidative stress. Carcinogenesis. 2002;23:1379–85.

    Article  CAS  PubMed  Google Scholar 

  173. Yang H, Stuart GR, Glickman BW, de Boer JG. Modulation of 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine-induced mutation in the cecum and colon of big blue rats by conjugated linoleic acid and 1,2-dithiole-3-thione. Nutr Cancer. 2001;39:259–66.

    Article  CAS  PubMed  Google Scholar 

  174. Yang H, Glickman BW, de Boer JG. Effect of conjugated linoleic acid on the formation of spontaneous and PhIP-induced mutation in the colon and cecum of rats. Mutat Res. 2002;500:157–68.

    Article  CAS  PubMed  Google Scholar 

  175. Meng F, Li Z, Yan J, Manjanatha M, Shelton S, Yarborough S, et al. Tissue-specific microRNA responses in rats treated with mutagenic and carcinogenic doses of aristolochic acid. Mutagenesis. 2014;29:357–65.

    Article  CAS  PubMed  Google Scholar 

  176. McDaniel LP, Elander ER, Guo X, Chen T, Arlt VM, Mei N. Mutagenicity and DNA adduct formation by aristolochic acid in the spleen of Big BlueR rats. Environ Mol Mutagen. 2012;53:358–68.

    Article  CAS  PubMed  Google Scholar 

  177. Kawamura Y, Hayashi H, Tajima O, Yamada S, Takayanagi T, Hori H, et al. Evaluation of the genotoxicity of aristolochic acid in the kidney and liver of F344 gpt delta transgenic rat using a 28-day repeated-dose protocol: a collaborative study of the gpt delta transgenic rat mutation assay. Genes Environ. 2012;34:18–24.

    Article  CAS  Google Scholar 

  178. Unfried K, Schurkes C, Abel J. Distinct spectrum of mutations induced by crocidolite asbestos: clue for 8-hydroxydeoxyguanosine-dependent mutagenesis in vivo. Cancer Res. 2002;62:99–104.

    CAS  PubMed  Google Scholar 

  179. He Z, Kosinska W, Zhao ZL, Wu XR, Guttenplan JB. Tissue-specific mutagenesis by N-butyl-N-(4-hydroxybutyl)nitrosamine as the basis for urothelial carcinogenesis. Mutat Res. 2012;742:92–5.

    Article  CAS  PubMed  Google Scholar 

  180. Mei N, Guo L, Fu PP, Heflich RH, Chen T. Mutagenicity of comfrey (Symphytum Officinale) in rat liver. Br J Cancer. 2005;92:873–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Topinka J, Oesterle D, Reimann R, Wolff T. No-effect level in the mutagenic activity of the drug cyproterone acetate in rat liver. Part II. Multiple dose treatment. Mutat Res. 2004;550:101–8.

    Article  CAS  PubMed  Google Scholar 

  182. Recio L, Saranko CJ, Steen AM. 1,3-butadiene: cancer, mutations, and adducts. Part II: Roles of two metabolites of 1,3-butadiene in mediating its in vivo genotoxicity. Res Rep Health Eff Inst. 2000;(92):49–87.

  183. Lefevre PA, Tinwell H, Ashby J. Mutagenicity of the potent rat hepatocarcinogen 6BT to the liver of transgenic (lacI) rats: consideration of a reduced mutation assay protocol. Mutagenesis. 1997;12:45–7.

    Article  CAS  PubMed  Google Scholar 

  184. Manjanatha MG, Shelton SD, Culp SJ, Blankenship LR, Casciano DA. DNA adduct formation and molecular analysis of in vivo lacI mutations in the mammary tissue of Big Blue rats treated with 7, 12-dimethylbenz[a]anthracene. Carcinogenesis. 2000;21:265–73.

    Article  CAS  PubMed  Google Scholar 

  185. Manjanatha MG, Shelton SD, Rhodes BS, Bishop ME, Lyn-Cook LE, Aidoo A. 17 Beta-estradiol and not genistein modulates lacI mutant frequency and types of mutation induced in the heart of ovariectomized big blue rats treated with 7, 12-dimethylbenz[a]anthracene. Environ Mol Mutagen. 2005;45:70–9.

    Article  CAS  PubMed  Google Scholar 

  186. Hobbie KR, Deangelo AB, King LC, Winn RN, Law JM. Toward a molecular equivalent dose: use of the medaka model in comparative risk assessment. Comp Biochem Physiol C Toxicol Pharmacol. 2009;149:141–51.

    Article  PubMed  CAS  Google Scholar 

  187. McDaniel LP, Ding W, Dobrovolsky VN, Shaddock Jr JG, Mittelstaedt RA, Doerge DR, et al. Genotoxicity of furan in Big Blue rats. Mutat Res. 2012;742:72–8.

    Article  CAS  PubMed  Google Scholar 

  188. Topinka J, Loli P, Hurbakova M, Kovacikova Z, Volkovova K, Wolff T, et al. Benzo[a]pyrene-enhanced mutagenesis by man-made mineral fibres in the lung of lamda-lacI transgenic rats. Mutat Res. 2006;595:167–73.

    Article  CAS  PubMed  Google Scholar 

  189. Thompson CM, Young RR, Suh M, Dinesdurage HR, Elbekai RH, Harris MA, et al. Assessment of the mutagenic potential of Cr(VI) in the oral mucosa of Big BlueR transgenic F344 rats. Environ Mol Mutagen. 2015;56:621–8.

    Article  CAS  PubMed  Google Scholar 

  190. Chen T, Mittelstaedt RA, Shelton SD, Dass SB, Manjanatha MG, Casciano DA, et al. Gene- and tissue-specificity of mutation in Big Blue rats treated with the hepatocarcinogen N-hydroxy-2-acetylaminofluorene. Environ Mol Mutagen. 2001;37:203–14.

    Article  CAS  PubMed  Google Scholar 

  191. da Gamboa CG, Manjanatha MG, Marques MM, Beland FA. Induction of lacI mutations in Big Blue rats treated with tamoxifen and alpha-hydroxytamoxifen. Cancer Lett. 2002;176:37–45.

    Article  Google Scholar 

  192. White IN, Carthew P, Davies R, Styles J, Brown K, Brown JE, et al. Short-term dosing of alpha-hydroxytamoxifen results in DNA damage but does not lead to liver tumours in female Wistar/Han rats. Carcinogenesis. 2001;22:553–7.

    Article  CAS  PubMed  Google Scholar 

  193. Vogel U, Danesvar B, Autrup H, Risom L, Weimann A, Poulsen HE, et al. Effect of increased intake of dietary animal fat and fat energy on oxidative damage, mutation frequency, DNA adduct level and DNA repair in rat colon and liver. Free Radic Res. 2003;37:947–56.

    Article  CAS  PubMed  Google Scholar 

  194. Theisen J, Peters JH, Fein M, Hughes M, Hagen JA, Demeester SR, et al. The mutagenic potential of duodenoesophageal reflux. Ann Surg. 2005;241:63–8.

    PubMed  PubMed Central  Google Scholar 

  195. Boyiri T, Guttenplan J, Khmelnitsky M, Kosinska W, Lin JM, Desai D, et al. Mammary carcinogenesis and molecular analysis of in vivo cII gene mutations in the mammary tissue of female transgenic rats treated with the environmental pollutant 6-nitrochrysene. Carcinogenesis. 2004;25:637–43.

    Article  CAS  PubMed  Google Scholar 

  196. Young RR, Thompson CM, Dinesdurage HR, Elbekai RH, Suh M, Rohr AC, et al. A robust method for assessing chemically induced mutagenic effects in the oral cavity of transgenic Big BlueR rats. Environ Mol Mutagen. 2015;56:629–36.

    Article  CAS  PubMed  Google Scholar 

  197. Zeng H, Uthus EO, Ross SA, Davis CD. High dietary intake of sodium selenite does not affect gene mutation frequency in rat colon and liver. Biol Trace Elem Res. 2009;131:71–80.

    Article  CAS  PubMed  Google Scholar 

  198. Hully JR, Su Y, Lohse JK, Griep AE, Sattler CA, Haas MJ, et al. Transgenic hepatocarcinogenesis in the rat. Am J Pathol. 1994;145:384–97.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is partly supported by a grant-in-aid for Scientific Research (B) (JSPS KAKENHI Grant 26281029).

Authors’ contributions

TN drafted the manuscript and part of Table 1. KM collected reference data and drafted Table 1. NT drafted the manuscript of transgenic rats for carcinogenesis and Table 2. All of the authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Takehiko Nohmi.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nohmi, T., Masumura, K. & Toyoda-Hokaiwado, N. Transgenic rat models for mutagenesis and carcinogenesis. Genes and Environ 39, 11 (2017). https://doi.org/10.1186/s41021-016-0072-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41021-016-0072-6

Keywords