@@@@@@
Group A-3 ‰»Šw\‘’@(Chemical Structure)  
œ Acrylic acid iƒAƒNƒŠƒ‹Ž_; 2-ƒvƒƒyƒ“Ž_;@ƒGƒ`ƒŒƒ“ƒJƒ‹ƒ{ƒ“Ž_;@ƒrƒjƒ‹ƒMŽ_jj
@
CASF79-10-7@Industry /Monomer@ MW: 72.06
AM Sal. Max (?)   1)
CA CHL/IU Min (0.75 mg/ml, -S9), 24h: D20=0.25; TR=16 › 2)
1) US/NTP Report (?).
2) Sofuni T. (Ed.): Data Book of Chromosome Aberration Test in vitro, LIC, Tokyo (1998j (Tables in English)

(Additional Ref.)
1) BG, Chemie Toxicol. Evaluations, 2 (1991)
2) Environ.  Molecular Mutagenesis, 17, 264-271 (1991)
3) Environ. Mutagenesis, 9, 1-110 (1987)
4) Fd. Chem. Toxic., 30 (6) (1992)


US-NTP Genotoxicity ScreeningF
›@Ames test:  @@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@ @ @@@@@@@@@@@@@

œ Acrylonitrile iƒAƒNƒŠƒ‹ƒjƒgƒŠƒ‹j(ƒVƒAƒmƒGƒ`ƒŒƒ“G‚Q|ƒvƒƒyƒ“ƒjƒgƒŠƒ‹Gƒrƒj[ƒ‹ƒVƒAƒjƒhj
@
107-13-1@ Industry /Monomer @@53.05
AM Sal. Max ( 1.0 mg/plate, }S9)   1, 2)
AM Sal. Max ( 3.0 mg/plate, }S9)   3)
MY Yeast Min ( 0.8 mg/ml) › 4)
SM Mouse L5178Y Min ( 0.2 mg/ml, }S9), for 2h › 5)
SM Mouse L5178Y Min  0.04 mg/ml, }S9), for 3h › 6)
SM Mouse L5178Y Min ( 0.05 mg/ml, }S9), for 4h › 7)
SM Mouse L5178Y Max ( 0.1 mg/ml, }S9), for 2h   8)
SCE Hum. epitherial Max ( 0.6 mg/ml), for 20h   9)
SCE CHO Min  0.16 mg/ml), }S9), for 2h › 10)
CA CHO Min ( 0.21 mg/ml), }S9), for 1h › 11,)
CA CHI-L Min ( 0.025 mg/ml), +S9), for 1h › 12, 13)
CA CHL/IU Min(0.025 mg/ml), +S9 6-18h; D20= 0.044,TR= 680)  14, 27)
SCE Human lym. Min ( 5 mM, +S9) › 15)
SCE Human lym. Max ( 0.01 mg/ml, }S9)   16)
MN CHO Min ( 0.1 mg/ml, }S9) › 17)
DNA Rat hepatocytes Max ( 10 mM)   18, 19)
CAv Drosophila Min ( 5 mM) › 20)
CAv Drosophila Min ( 8 mM) › 21)
UDSv Rats Min ( 50 mg/kg, or) › 22)
DLv Rats Max ( 60 mg/kg,or), for 5d   23)
CAv Mice, BM Max ( 30 mg/kg,ip)   24)
MNv Mice, BM Min ( 22.5 mg/kg, ip) › 25)
CT Sy. hamster Min ( 50 ƒΚg/ml, +S9) › 26)
1) Lijinsky W & Andrews AN., Teratog. Carcinog. Mutagen., 1, 259-267 (1980
2) Zeiger E & Haworth S., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 187-199 (1985)
3) Matsushima T., et al., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 181-186 (1985)
4) Mehta RD & von Borstel RC., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 271-284 (1985)
5) Lee CG & Webber CD., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 547-554 (1985)
6) Amacher EE & Turner GN, In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 487-496 (1985)

7) Oberly TJ  et al., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5 ,569-582 (1985)
8) Styles JA & Clay P., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 587-596 (1985)
9) Chang C-M., et al., Mutation Res., 241, 355-300 (1990)
10) Gulati DK., et al.., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 413-426 (1985)
11) Natarajan AT., et al., ,In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 433-437 (1985)

12) Danford N,,  In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 397-411 (1985)
13) Parry JM,,  In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5,479-485 (1985)
14) Ishidate MJr & Sofuni T., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 427-432 (1985)
15) Perocco P.,  et al., Scand. J. Work Environ. Health., 2-8, 290-293 (1982)
16) Obe G., et al., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5, 439-442 (1985)
17) Douglas GR., et al,., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5 ,359-368 (1985)
18) Butterworth BE., et al., Environ. Mol. Mutagen., 20, 148-155 (1992)
19) Williams GW.,  et al., In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5 , 341-345 (1985)
20) Vogel. EW.,  In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5 ,313-317 (1985)
21)  Fujikawa.,  In: Ashby J. et al. (Eds) Evaluation of Shor-Term Tests for Canrinogens, IPCS, Rrogress. Mutation Res., 5 , 319-324 (1985)
22) Hogy LL & Guengerich FP., Cancer Res., 46, 3932-3938 (1986)., 
23) Working PK., et al., Mutagenesis, 2, 215-220 (1987)
24) Leonard DL., et al., Toxicol. Lett., 7, 329-334 (1981)
25) Morita T, et al.: Mutation Res., 389, 3-122 (1997)
26) Parent RA. & Casto BC.: J. Natl. Cancer Inst., 62, 1025-1029 (1979)
27) Ministry of Labour, Japan, Mutagen. Test Data of Exist. Chemical Subst. JETOC(Ed.)(1997 Suppl.) (Tables in English)

US-NTP Genotoxicity ScreeningF
›@Ames test:@ 
›@CA test:@
›
›@SCE test:@
›
›@MLA test:@
›
›@Drosophila (SLRL) test:@
 
@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@
yNotez (Cited from IARC Monographs, Suppl. 6, 1987)
@Acrylonitrile did not enhance the frequency of CAs in lymphocytes of exposed workers in one study.
In animals treated in vivo, it did not induced DL mutations, CAs (in bone-marrow cells or spermatogonias), or MNs in mice, or CAs in rat bone marrow cells. It bound. covalently to rat liver DNA in vitro and induced UDS in rat liver but not brain. It induced SCEs, mutation and UDSs but not CAs in human cells in vitro.
@It induced cell transformation in several test systems and inhibited intercellular communication in CH V79 cells. It did not induce aneuploidy but induced CAs, MNs and SCEs in CH cells; it did not induce CAs or SCEs in rat cells in vitro. It induced mutation and DNA strand breaks in rodent cells in vitro. It induced SM in Drosophila and was weakly mutagenic in plants. It induced aneuploidy, mutation, mitotic crossing-over and gene conversion in fungi. It was mutagenic to bacteria. Urine from treated mice and rats, but not bile from rats, was mutagenic to bacteria. It bound covatently to isolated DNA. (IARC Monographs, 19, 73, 1979)

yNotez(Cited from CICADS Document, 39, 2002
In vitro studies
  In the Salmonella assay, acrylonitrile has induced reverse mutations in strains TA1535 (Lijinsky & Andrews, 1980), TA1535, and TA100 (Zeiger & Haworth, 1985), but only when hamster or rat S9 was present. Weak positive results were also reported in several Escherichia coli strains in the absence of metabolic activation (Venitt et al., 1977).

  In mammalian cells, acrylonitrile induced hprt mutations in human lymphoblasts without metabolic activation (Crespi et al., 1985), but not in Chinese hamster V79 cells (Lee & Webber, 1985). In several studies, acrylonitrile was positive at the TK locus in mouse lymphoma L5178 TK+/- cells, either with or without rat S9 (Amacher & Turner, 1985; Lee & Webber, 1985; Myhr et al., 1985; Oberly et al., 1985), and in mouse lymphoma P388F cells with metabolic activation (Anderson & Cross, 1985). It was also mutagenic at the TK locus in human lymphoblasts with metabolic activation (Crespi et al., 1985; Recio & Skopek, 1988).

  Acrylonitrile induced structural chromosomal aberrations either with or without metabolic activation in Chinese hamster ovary cells (Danford, 1985; Gulati et al., 1985; Natarajan et al., 1985) and without metabolic activation in Chinese hamster lung cells (Ishidate & Sofuni, 1985). Results for sister chromatid exchanges in Chinese hamster ovary cells and human lymphocytes both with and without metabolic activation are mixed (Brat & Williams, 1982; Perocco et al., 1982; Gulati et al., 1985; Natarajan et al., 1985; Obe et al., 1985; Chang et al., 1990).

  Results of in vitro assays for DNA single strand breaks (Bradley, 1985; Lakhanisky & Hendrickx, 1985; Bjorge et al., 1996) and DNA repair (unscheduled DNA synthesis) (Perocco et al., 1982; Glauert et al., 1985; Martin & Campbell, 1985; Probst & Hill, 1985; Williams et al., 1985; Butterworth et al., 1992) were mixed but more commonly negative in a range of cell types from rats and humans, with and without activation. Cell transformation in mouse and hamster embryo cells has also been investigated, with mixed results (Lawrence & McGregor, 1985; Matthews et al., 1985; Sanner & Rivedal, 1985; Abernethy & Boreiko, 1987; Yuan & Wong, 1991).

  Binding of 2-cyanoethylene oxide to nucleic acids has also been reported in in vitro studies at high concentrations (Hogy & Guengerich, 1986; Solomon & Segal, 1989; Solomon et al., 1993; Yates et al., 1993, 19946). The formation of acrylonitrile?DNA adducts is increased substantially in the presence of metabolic activation. Under non-activating conditions involving incubation of calf thymus DNA with either acrylonitrile or 2-cyanoethylene oxide in vitro, 2-cyanoethylene oxide alkylates DNA much more readily than acrylonitrile (Guengerich et al., 1981; Solomon et al., 1984, 1993). Incubation of DNA with 2-cyanoethylene oxide yields 7-(2-oxoethyl)-guanine (Guengerich et al., 1981; Hogy & Guengerich, 1986; Solomon & Segal, 1989; Solomon et al., 1993; Yates et al., 1993, 1994) as well as other adducts. Compared with studies with rat liver microsomes, little or no DNA alkylation by acrylonitrile was observed with rat brain microsomes (Guengerich et al., 1981). DNA alkylation in human liver microsomes was much less than that observed with rat microsomes (Guengerich et al., 1981); although there was no glutathione S-transferase activity in cytosol preparations from human liver exposed to acrylonitrile, there was some activity for 2-cyanoethylene oxide (Guengerich et al., 1981).

In vivo studies

@Limitations of the few in vivo studies conducted in which the genotoxicity of acrylonitrile has been investigated preclude definitive conclusions. Data from these studies are also inadequate for characterization of dose-response for comparison between studies or with the cancer bioassays.

  Exposure to acrylonitrile in drinking-water resulted in increased frequency of mutants at the hprt locus in splenic T-cells (Walker & Walker, 1997). Five female F344 rats were exposed to 0, 33, 100, or 500 mg/litre (0, 8, 21, or 76 mg/kg body weight per day; Health Canada, 1994) in drinking-water for up to 4 weeks and serially sacrificed throughout exposure and up to 8 weeks post-exposure. At 4 weeks post-exposure, the average observed mutant frequency in splenic T-cells was increased in a dose-related manner (significant at the two highest doses).

  Results of a range of assays for structural chromosomal aberrations, micronuclei in bone marrow, and micronuclei in peripheral blood cells have been negative or inconclusive, although there was no indication in the published accounts of three of the four studies that the compound reached the target site. These include studies in Swiss (Rabello-Gay & Ahmed, 1980), NMRI (Leonard et al., 1981), and C57B1/6 (Sharief et al., 1986) mice and a colLabourative study following exposure by multiple routes in mice and rats (Morita et al., 1997).

  Results of dominant lethal assays were inconclusive in mice (Leonard et al., 1981) and negative in rats (Working et al., 1987).

  In assays for unscheduled DNA synthesis in rats, results were positive only for the liver (Hogy & Guengerich, 1986), equivocal in lung, testes, and gastric tissues (Ahmed et al., 1992a,b; Abdel-Rahman et al., 1994), and, notably, negative in the brain (Hogy & Guengerich, 1986). In these studies, however, unscheduled DNA synthesis was measured by liquid scintillation counting to determine [3H]thymidine uptake in the cell population, which does not discriminate between cells undergoing repair and those that are replicating. Results for unscheduled DNA synthesis in rat liver and spermatocytes were negative when [3H]thymidine uptake in individual cells was determined by autoradiography, which eliminates replicating cells from the analysis (Butterworth et al., 1992).

  Urine from acrylonitrile-exposed rats and mice was also mutagenic in Salmonella typhimurium following intraperitoneal administration of acrylonitrile to rats and mice (Lambotte-Vandepaer et al., 1980, 1981). In both species, mutagenic activity occurred without activation. Mutagenic activity was also observed in urine of rats administered acrylonitrile by stomach intubation (Lambotte-Vandepaer et al., 1985). Thiocyanate, hydroxyethylmercapturic acid, and cyanoethylmercapturic acid were not believed to be responsible for urinary mutagenicity.

  In in vivo studies in F344 rats administered 50 mg acrylonitrile/kg body weight intraperitoneally, 7-(2-oxoethyl)-guanine adducts were detected in liver (Hogy & Guengerich, 1986). Incorporation of acrylonitrile into hepatic RNA was observed following intraperitoneal administration to rats (Peter et al., 1983). However, no DNA adducts were detected in the brain, which is the primary target for acrylonitrile-induced tumorigenesis, in this or a subsequent study in which F344 rats received 50 or 100 mg acrylonitrile/kg body weight by subcutaneous injection (Prokopczyk et al., 1988). In contrast, in three studies from one Labouratory, exposure of SD rats to 46.5 mg [14C]acrylonitrile/kg body weight (50 ƒΚCi/kg body weight) resulted in apparent binding of radioactivity to DNA from liver, stomach, brain (Farooqui & Ahmed, 1983), lung (Ahmed et al., 1992a), and testicles (Ahmed et al., 1992b). In each tissue, there was a rapid decrease in radioactivity of DNA samples collected up to 72 h following treatment.

  It is not clear why acrylonitrile-DNA binding was detected in the brain in these studies and not in those by Hogy & Guengerich (1986) or Prokopczyk et al. (1988). The DNA isolation protocols and method for correcting for contaminating protein in the DNA sample used by Hogy & Guengerich (1986) may have allowed a more stringent determination of DNA-bound material. Alternatively, the methods used to achieve greater DNA purity might have caused the loss of adducts or inhibited the recovery of adducted DNA; more likely, however, 7-oxoethylguanine and cyanoethyl adducts are of little consequence in the induction of acrylonitrile-induced brain tumours. Indeed, investigation of the role of cyanohydroxyethylguanine and other adducts in the induction of these tumours seems warranted.

œ Actionmycin D iƒAƒNƒ`ƒmƒ}ƒCƒVƒ“ D)
   
50-76-0@ Medicine @1255.43
AM Sal/E. coli Max (6.1 mg/ml, }S9)   1)
BM N. crassa Min ( 5ƒΚg/m‚Œ) › 2)
CA CHL/IU 0.000013 mg/ml, -S9, 24h; D20= 0.00002; TR= 720000):  3)
PL Soy beans (Specific Loci Test) › 4)
SCE Sy Ham Min ( 0.005ƒΚg/m‚Œ)  5)
SCE Human Ly Max (.0.009 mg/kg)   6)
SPM Mice Min ( 0.21 mg/kg x 5), ip › 7)
1) Seine Y, et al: Cancer Rest., 38, 2148-2156 (1978)
2) Fisher CRY, et al.: Mutation Rest., 33, 187-192 (1975)
3) Sound T (Ed.): Data Book of Chromosome Aberration Test in victor LICK, Tokyo (1998) (Tables in English)
4) Via BK : Mutation Rest., 31, 49-56 (1975)
5) Banners A. & Benedict WE: Cancer Rest., 39, 797-799 (1979)
6) Lambert B, et al: Mutation Rest., 59, 295-300 (1979)
7) AGE & Bruce WRY : Pro. Natal. Aced. Sigh. (USA), 72, 4425-4429 (1975)

yReviewz
1) Via BC: Mutation Rest., 49, 189-238 (1977)

yNotez (Cited from IRAQ Monographs, Supple. 6, 1987)
@Actinomycin D did not induce SCEs in peripheral blood Ly. of treated patients in one study.@It induced CAs and DNA strand breaks in human cells in vitro. It transformed mouse C3H 10T1/2 cells and induced CAs, SCEs mutation, DNA strand breaks and UDS, but not aneuploidy, in rodent cells in vitro.
@It induced SLRM in Drosophila. It did not cause CAs in plants. It was mutagenic to Neurospora crassa but not to Saccharomyces cerevisiae, and conflicting results were obtained for gene conversion and mitotic recombination. It did not induce DNA damage in Schizosaccharomyces pombe. It was not mutagenic to bacteria and did not induce prophage. (IARC Monographs, 10, 29, 1976).

œ Acylase iƒAƒVƒ‰[ƒ[j
@
9012-37-7@Food/Natural@
AM Sal Min (?)(-S9) › 1)
CA CHL/IU Max (0.25 mg/ml, -S9), 48h   2)
1) Ishidate M.Jr et al : Toxicol. Forum, 8, 705-708 (1985) (Japanese)
2) Sofuni T. (Ed.): Data Book of Chromosome Aberration Test in vitro LIC, Tokyo (1998) (Tables in English)

œ Adipic acid@iƒAƒfƒBƒsƒbƒNŽ_j
@
124-04-9@@Industry@@146.14
SM Sal./E.coli Max ( 10.0 mg/plate, }S9)
  1)
SM Human Lung Max (?)   2)
CAv Rat, BM Max (?)   2)
1) Ministry of Labour, Japan, Mutagen. Test Data of Exist. Chemical Subst, JETOC(Ed.)(1996.) (Tables in English)
2) Litton Bionetics. Inc.: NTIS Pub. No. PB-466/8 ST., Natl Inform. Service (1974)

œ Adiponitrile @iƒAƒfƒBƒ|ƒjƒgƒŠƒ‹j
@
111-69-3 @Industry/Intermediate@108.1@
@@@@@@@@@@@@@@@@@@@@@@@@@@@@@
AM Sal Max@(?)   1, 2)
1) E.I. du Pont de Nemoures & Co.: DuPount Co., Inc., Wilmington, DE (Feb., 24, 1976)
2) Natl Inst. Occup. Safety and Health: (Unpublished data); NTIS Pub. No. PB-81-225-534. @Natl Tech. Inform. Service, Springfield, VA (1978)@
@
@@@@@@@@@@@@@@@@@@@@@@@@@@@@
US-NTP Genotoxicity Screening:
›@Ames test:@
 

œ Adriamycin iƒAƒhƒŠƒAƒ}ƒCƒVƒ“j@
@
23214-92-8@Medicine/Natural 543.53
AM Sal Min (?)
›
1)
SCE Human Ly Min ( 1ƒΚg/m‚Œ)

2)
UDS Rat Cardiac Cells Min ( 1ƒΚg/m‚Œ)

3)
SCEv Mice Min ( 12 mg/kg), iv
›
4)
MNv Mice Min ( 1mg/kg), ip

5)
SCEv Mice, spematogonia Min (0.125 mg/kg) ip)
›
6)
1) McCann J, et al. : Proc. Natl Acad. Sci. (USA), 72, 5135-5139 (1975)
2) Nebstad NP: Mutation Res., 57, 253-258 (1978)
3) Fialkoff H,et al.: Cancer Res., 39, 1321-1327 (1979)
4) Nakanishi Y. & Schneider EL: Mutation Res., 60, 329-337 (1979)
5) Maier P & Schmid W: Mtutation Res., 40, 325-338 (1976)
6) Abraham SK & Fraenz J: Mutation Res., 108, 373-381 (1983)

US-NTP Genotoxicity ScreeningF
›@Ames test:@ 

yNotez (Cited from IARC Monographs, Suppl. 6, 1987)
@@Adriamycin induced CAs in treated patients in one of two studies and SCEs in both studies. In another study, cisplatin-adriamycin combination chemotherapy induced SCEs in peripheral blood Ly. of treated patents. DNA strand breaks were induced in the cells of treated patients in one study.
@@It has been tested extensively fro genetic effects in a wide variety of tests in vivo and in vitro, giving consistently positive results. It induce CAs, MNs SCEs and DNA damage in human cells in vitro. @@It transformed virus-infected Fischer rat embryo cells and induced CAs , SCEs, mutation and DNA damage in cultured rodent cells. It induced SLR mutation in Drosophila, CAs in plants and mutation in fungi. It was mutagenic to bacteria and induced DNA damage. (IARC Monographs, 10, 43, 1976).

œ Aflatoxin B1 iƒAƒtƒ‰ƒgƒLƒVƒ“B1j
@
1162-65-8@Natural/Carcinogen@@1255.43

AM Sal Min ( 0.1ƒΚg/plate, +S9)
›
1), 2)
BM N. crassa
(ƒAƒJƒpƒ“ƒJƒr)
Min ( 0.67 mM, +S9)
›
3)
SM V79 (6TG) Min (0.4 nM, -S9)
›
4)
UDS HeLa Cells Min ( 10-8 M)
›
5)
CT Balb/3T3 Min ( 0.5ƒΚg/ml)
›
6)
CA V79 Min ( 0.5 ƒΚg/ml, +S9,) 1-26h
›
7)
MNv Rat Bone Marrow Min ( 2.5 mg/kg) ip
›
8)
HLAv Fi-Mice Max ( 5 mg/kg) ip
 
9)
1) Ames BN, et al.: Mutation Res., 31, 347-364 (1971)
2) Wehner FC, et al.: Mutation Res., 58, 193-203 (1978)
3) Matzinger PK. & Ong T.: Mutation Res., 37, 27-32 (1976)
4) Krahn DF & Heiderberger C: Mutation Res., 46, 27-44 (1977)
5) Martin CN, et al.: Cancer Res., 38, 2621-2627(1978)
6) DiPaolo JA, et al.: Cancer Res. 32, 2686-2695 (1972)
7) Batt TR, et al.: Carcinogenesis, 1, 759-763 (1980)
8) Trzos RJ, et al.: Mutation Res., 58, 79-86 (1978)
9) Leonard A, et al., Mutation Res., 28, 137-139 (1975)

yNotez (Cited from IARC Monographs, Suppl. 6, 1987)@@@@@@@@@@@@@@@@@@@@@@@@@@@
@ In one study, aflatoxin B1-DNA adducts were excreted in human urine.
@Aflatoxin B1 has been tested extensively for genetic effects in a wide variety of tests in vivo and in vitro,, giving consistently positive results. It induced CAs, MNs, SCEs, UDS and DNA strand breaks, and bound covalently to DNA in cells of rodents treated in vivo; it was reported to be weakly active in a DL assay in mice.
@In human cells in vitro, it induced CAs, MNs, SCEs and UDS and bound covalently to DNA. @It induced CT in several test systems, and induced CAs, SCEs, mutation, UDS and DNA strand breaks in rodent cells in vitro. It induced SLRL and SA and recombination in Drosophila. In Fungi, it was mutagenic and induced gene conversion and mitotic recombination. It was mutagenic and induced DNA damage in bacteria and bound convalently to isolated DNA.

œ Agropine (ƒAƒOƒƒpƒCƒ“)@
@
70699-77-3 @Natural@@292.33
CA CHL/IU Min ( 0.75 mg/ml, -S9), 24h; D20= 0.52, TR= 16)
›@
1)
1) Sofuni T. (Ed.): Data Book of Chromosome Aberration Test in vitro, LIC, Tokyo (1998) (Tables in English)

œ
Alachlor (ƒAƒ‰ƒNƒ[ƒ‹)@(2-chloro-2', 6'-diethyl-N-(methoxymethyl)
@acetanilide)
@
15972-60-8   Pesticide @@269.77

YM Gene Conversion (Plant mediated) Min (10 ppm) @›@ 1)
CA CHL/IU Min ( 0.01 mg/ml, -S9), 24h; D20= 0.013, TR= 2300  2)
1) Gentile, JM. et al.: Mutation Res., 48, 113-116 (1977)
2) Sofuni T. (Ed.): Data Book of Chromosome Aberration Test In Vitro, LIC, Tokyo (1998) (Tables in English)

œTop Page@iƒgƒbƒvƒy[ƒWj
œAbbreviation @iΘ—ͺ‹L†j@
œMutagenicityi•ΟˆΩŒ΄«j
œTest Systems@(ŽŽŒ±–@‚ΜŽν—ށj
œTechnical Problems@i‹Zp“I–β‘θ“_j
œList of@Compoundsi‰»‡•¨ƒŠƒXƒgj
œEvaluation of Results@iŽŽŒ±Œ‹‰Κ‚Μ•]‰Ώj
@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@