+ All Categories
Home > Documents > Increased frequency of specific genomic deletions resulting from in vitro malathion exposure

Increased frequency of specific genomic deletions resulting from in vitro malathion exposure

Date post: 20-Nov-2023
Category:
Upload: lbl
View: 0 times
Download: 0 times
Share this document with a friend
8
[CANCER RESEARCH 56. 2393-2399. May 15. 1996] Increased Frequency of Specific Genomic Deletions Resulting from in Vitro Malathion Exposure1 Janice M. Pluth,2 Janice A. Nicklas, J. Patrick O'Neill, and Richard J. Albertini Genetics Laboratory, University of Vermont, Burlington. Vermont 05401 ABSTRACT Malathion is a widely used pesticide with high potential for human exposure. Epidemiological studies suggest that individuals with chronic environmental exposures to pesticides have increased risks of various hematological malignancies. The genotoxic data to date have been some what inconclusive with regard to malathion exposure. We have used a cell cloning assay to study the genotoxicity of in vitro exposure of human T lymphocytes to malathion. We exposed cells in G0 to doses of malathion ranging from 10 to 600 fig/ml. Mutant frequencies of treated samples showed both intra- and interindividual variability and, in some cases, slight significant increases over the controls. Molecular analysis of hprt mutants resulting from both in vitro and an in vivo malathion exposure was performed by genomic multiplex PCR. In seven in vitro experiments (using cells from four different individuals) and one experiment on an individual exposed in vivo, one or more independent mutant(s) containing a partial deletion of exon 3 have been isolated from each individual. In five of the seven mutants, the deleted regions overlap extensively, revealing an area within exon 3 exceptionally prone to deletions upon exposure to malathion. This work provides the first evidence of an association between malathion exposure and specific mutations in human T lymphocytes. Additional work is necessary to determine the underlying molecular mechanism for these deletions and how this may relate to agricultural workers' increased risk of cancer. INTRODUCTION Malathion, O,O-dimethyl-S-(l,2-dicarbethoxyethyl) dithiophos- phate (CAS No. 121-75-5), an organophosphorus pesticide, is widely used for both domestic and commercial agricultural purposes. It is considered to be one of the safest organophosphate insecticides, and has been used in large pest eradication programs in Florida, Texas, and California. Technical-grade malathion (the grade usually used for agricultural purposes) is usually 90-95% pure and may contain up to 11 impurities formed during its production and/or storage. Some of these impurities have been found to be significantly more toxic than malathion or to potentiate the toxicity of malathion (1). Malathion's main pharmacological effect is inhibition of acetylcholinesterase en zymes leading to a build up of acetylcholine; many of the symptoms resulting from an acute exposure reflect this and include various muscarinic, nicotinic, and neurological symptoms. Malathion's selec tivity is due primarily to mammals having high levels of carboxyes- terases, which can hydrolyze malathion and malaoxon, the active metabolite, to nontoxic intermediates that can be easily eliminated. Insects lack or have lower levels of these esterases, so are severely affected by malathion exposure. Malathion has been widely studied in a variety of systems, but there is still some controversy over malathion's mutagenic and/or genotoxic potential for humans. No mutagenic effect has been shown in the majority of studies using bacteria and yeast (2-8). However, two gene Received 12/7/95; accepted 3/19/96. The costs of publication of this article were defrayed in pan by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1This research is supported by American Cancer Society Grant CN-141. Department of Energy Grant FG02 87ER60502, and Pathology Training Grant PHS I T3207122. The contents of this article do not necessarily reflect the policies of ACS. DOE or PHS. 2 To whom requests for reprints should be addressed, at Genetics Laboratory. 32 North Prospect, Burlington, VT 05401. Phone: (802) 656-8866; Fax: (802) 656-8333. mutation studies using a mammalian system, the mouse lymphoma thymidine kinase assay, and the active metabolite, malaoxon, did give positive results (9, 10). Degraeve et al. (11) and Degraeve and Moutschen (12) found no increase in chromosome aberrations in studies using mouse bone marrow cells, spermatogonia, and primary spermatocytes after either oral or i.p. exposure, whereas a large number of studies using human cells have shown a significant in crease in chromosome aberrations and/or sister chromatid exchanges with exposure to malathion (13-17). There has also been some evi dence of malathion having a teratogenic effect; the offspring of rats fed with 240 mg/kg/day malathion exhibited growth retardation and elevated mortality, although no negative effects were observed in the parents (18). Many in vivo studies in which individuals were exposed to mala thion and/or other pesticides have shown a genotoxic effect. Yoder et al. (19) found a 5-fold increase in chromatid breaks in farmers during the summer season, when pesticide usage was highest. Van Bao (20) studied individuals who had been exposed only to acute doses of malathion and found a significant increase in the percentage of stable and unstable chromosome aberrations both immediately after expo sure and 1 month later. Lipkowitz et al. (21) showed that exposure to environmental pesticides (one of which was malathion) resulted in a 10-20-fold increased incidence of a particular cytogenetic aberration, inversion 7 [inv (7)], in a population of agricultural workers. Although this aberration itself is not oncogenic, these researchers felt that the elevated frequency of this aberration may relate to an overall increase in genomic instability, perhaps increasing the risk of cancer. Many epidemiological studies have shown a statistically significant associ ation between farming occupations and death from hematological malignancies (22-27). We have used a cell cloning assay to determine whether in vitro exposure of human T lymphocytes to malathion is mutagenic at the hprt locus and whether a particular molecular spectrum is induced. This assay was originally developed to study mutations occurring in vivo in humans (28-33) and has since been modified for use in in vitro studies (34, 35). The hprt clonal assay has been widely used, provid ing a large database with which to compare the molecular spectrum of mutations induced by specific agents (36, 37). MATERIALS AND METHODS Cells and Culture Conditions In Vitro Assay. Informed consent was obtained and peripheral blood samples were drawn from four male individuals, and mononuclear cells were isolated. Equal numbers of T cells were placed in separate 25-cm2 flasks for exposed and control cultures at a density of 1 X 106/ml. A stock solution of malathion was made in DMSO at a concentration of 500 fig/ml; further dilutions before addition to the culture were made in RPMI 1640. All cell culture media contained RPMI 1640 (Mediatech) supplemented with addi tional components for the various uses. The medium used for priming T cells consisted of RPMI 1640 with 10% CBS3 and 20% nutrient medium HL-1 ' The abbreviations used are: CBS, calf bovine serum; CE, cloning efficiency; HPLC. high-performance liquid chromotography; hprt. hypoxanthine (guanine) phosphoribosyl- transferase; MF. mutant frequency: PHA. phytohemagglutinin; TCGF, T-cell growth factor; 6-TG, 6-thioguanine; TCR. T-cell receptor. 2393 Research. on January 27, 2016. © 1996 American Association for Cancer cancerres.aacrjournals.org Downloaded from
Transcript

[CANCER RESEARCH 56. 2393-2399. May 15. 1996]

Increased Frequency of Specific Genomic Deletions Resulting from in VitroMalathion Exposure1

Janice M. Pluth,2 Janice A. Nicklas, J. Patrick O'Neill, and Richard J. Albertini

Genetics Laboratory, University of Vermont, Burlington. Vermont 05401

ABSTRACT

Malathion is a widely used pesticide with high potential for humanexposure. Epidemiological studies suggest that individuals with chronicenvironmental exposures to pesticides have increased risks of varioushematological malignancies. The genotoxic data to date have been somewhat inconclusive with regard to malathion exposure. We have used a cellcloning assay to study the genotoxicity of in vitro exposure of human Tlymphocytes to malathion. We exposed cells in G0 to doses of malathionranging from 10 to 600 fig/ml. Mutant frequencies of treated samplesshowed both intra- and interindividual variability and, in some cases,

slight significant increases over the controls. Molecular analysis of hprtmutants resulting from both in vitro and an in vivo malathion exposurewas performed by genomic multiplex PCR. In seven in vitro experiments(using cells from four different individuals) and one experiment on anindividual exposed in vivo, one or more independent mutant(s) containinga partial deletion of exon 3 have been isolated from each individual. In fiveof the seven mutants, the deleted regions overlap extensively, revealing anarea within exon 3 exceptionally prone to deletions upon exposure tomalathion. This work provides the first evidence of an association betweenmalathion exposure and specific mutations in human T lymphocytes.Additional work is necessary to determine the underlying molecularmechanism for these deletions and how this may relate to agriculturalworkers' increased risk of cancer.

INTRODUCTION

Malathion, O,O-dimethyl-S-(l,2-dicarbethoxyethyl) dithiophos-phate (CAS No. 121-75-5), an organophosphorus pesticide, is widely

used for both domestic and commercial agricultural purposes. It isconsidered to be one of the safest organophosphate insecticides, andhas been used in large pest eradication programs in Florida, Texas,and California. Technical-grade malathion (the grade usually used foragricultural purposes) is usually 90-95% pure and may contain up to

11 impurities formed during its production and/or storage. Some ofthese impurities have been found to be significantly more toxic thanmalathion or to potentiate the toxicity of malathion (1). Malathion's

main pharmacological effect is inhibition of acetylcholinesterase enzymes leading to a build up of acetylcholine; many of the symptomsresulting from an acute exposure reflect this and include variousmuscarinic, nicotinic, and neurological symptoms. Malathion's selec

tivity is due primarily to mammals having high levels of carboxyes-

terases, which can hydrolyze malathion and malaoxon, the activemetabolite, to nontoxic intermediates that can be easily eliminated.Insects lack or have lower levels of these esterases, so are severelyaffected by malathion exposure.

Malathion has been widely studied in a variety of systems, but thereis still some controversy over malathion's mutagenic and/or genotoxic

potential for humans. No mutagenic effect has been shown in themajority of studies using bacteria and yeast (2-8). However, two gene

Received 12/7/95; accepted 3/19/96.The costs of publication of this article were defrayed in pan by the payment of page

charges. This article must therefore be hereby marked advertisement in accordance with18 U.S.C. Section 1734 solely to indicate this fact.

1This research is supported by American Cancer Society Grant CN-141. Department

of Energy Grant FG02 87ER60502, and Pathology Training Grant PHS I T3207122. Thecontents of this article do not necessarily reflect the policies of ACS. DOE or PHS.

2 To whom requests for reprints should be addressed, at Genetics Laboratory. 32 North

Prospect, Burlington, VT 05401. Phone: (802) 656-8866; Fax: (802) 656-8333.

mutation studies using a mammalian system, the mouse lymphomathymidine kinase assay, and the active metabolite, malaoxon, did givepositive results (9, 10). Degraeve et al. (11) and Degraeve andMoutschen (12) found no increase in chromosome aberrations instudies using mouse bone marrow cells, spermatogonia, and primaryspermatocytes after either oral or i.p. exposure, whereas a largenumber of studies using human cells have shown a significant increase in chromosome aberrations and/or sister chromatid exchangeswith exposure to malathion (13-17). There has also been some evi

dence of malathion having a teratogenic effect; the offspring of ratsfed with 240 mg/kg/day malathion exhibited growth retardation andelevated mortality, although no negative effects were observed in theparents (18).

Many in vivo studies in which individuals were exposed to malathion and/or other pesticides have shown a genotoxic effect. Yoder etal. (19) found a 5-fold increase in chromatid breaks in farmers during

the summer season, when pesticide usage was highest. Van Bao (20)studied individuals who had been exposed only to acute doses ofmalathion and found a significant increase in the percentage of stableand unstable chromosome aberrations both immediately after exposure and 1 month later. Lipkowitz et al. (21) showed that exposure toenvironmental pesticides (one of which was malathion) resulted in a10-20-fold increased incidence of a particular cytogenetic aberration,

inversion 7 [inv (7)], in a population of agricultural workers. Althoughthis aberration itself is not oncogenic, these researchers felt that theelevated frequency of this aberration may relate to an overall increasein genomic instability, perhaps increasing the risk of cancer. Manyepidemiological studies have shown a statistically significant association between farming occupations and death from hematologicalmalignancies (22-27).

We have used a cell cloning assay to determine whether in vitroexposure of human T lymphocytes to malathion is mutagenic at thehprt locus and whether a particular molecular spectrum is induced.This assay was originally developed to study mutations occurring invivo in humans (28-33) and has since been modified for use in in vitro

studies (34, 35). The hprt clonal assay has been widely used, providing a large database with which to compare the molecular spectrum ofmutations induced by specific agents (36, 37).

MATERIALS AND METHODS

Cells and Culture Conditions

In Vitro Assay. Informed consent was obtained and peripheral bloodsamples were drawn from four male individuals, and mononuclear cells wereisolated. Equal numbers of T cells were placed in separate 25-cm2 flasks forexposed and control cultures at a density of 1 X 106/ml. A stock solution of

malathion was made in DMSO at a concentration of 500 fig/ml; furtherdilutions before addition to the culture were made in RPMI 1640. All cellculture media contained RPMI 1640 (Mediatech) supplemented with additional components for the various uses. The medium used for priming T cellsconsisted of RPMI 1640 with 10% CBS3 and 20% nutrient medium HL-1

' The abbreviations used are: CBS, calf bovine serum; CE, cloning efficiency; HPLC.

high-performance liquid chromotography; hprt. hypoxanthine (guanine) phosphoribosyl-transferase; MF. mutant frequency: PHA. phytohemagglutinin; TCGF, T-cell growthfactor; 6-TG, 6-thioguanine; TCR. T-cell receptor.

2393

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

MALATHION EXPOSURE AND GENOMIC DELETIONS

(Ventrex Laboratories, Portland. ME). TCGF was prepared as described previously (32). The medium for subculturing, plating, and growth of cells

contained RPMI 1640 supplemented with 5% CBS, 15% TCGF, 20% nutrientmedium HL-1, and 0.25 mg/ml PHA (HA- 17, Wellcome Diagnostics) and

irradiated human lymphoblastoid feeder cells. Medium used for feeding theplates consisted of 50% TCGF and 50% CBS, with 2 X 106/ml irradiated

feeder cells (and 6-TG when feeding selection plates).

In Vivo Assay. Twelve blood samples (courtesy of Dr. Vincent Garry,University of Minnesota) containing approximately 8 ml of blood each wereobtained from individuals who had been grain workers for different amounts oftime. These workers had received various pesticide exposures through theirwork, which entailed fumigation of stored grain bins. Two control individualblood samples were also obtained. All media used were essentially the same asthose in the in vitro assay, with the exception of the plating medium, which hada PHA concentration of 0.5

Mutation Induction, Expression, and Selection

The conditions for the in vitro Itprt assay were modified slightly from thosedescribed by O'Neill et al. (34). Malathion was added on day 1 to G0 cells and,

after 24 h exposure (day 2), PHA was added at a concentration of 1 mg/ml. Thecultures were incubated for 36-40 h to allow for mitogen stimulation and thenplated for CE determination in the presence and absence of 6-TG (finalconcentration, 10 p.M; ~day 3). From each flask, 2 X IO6 cells were also

subcultured in 20 ml of growth medium and cultivated further to allow formutant phenotype expression. Cells were again subcultured and plated at days5 or 6 and 7 or 8 as on day 3. Cells plated for CE were plated at I and 2cells/well without 6-TG and for calculation of MF at 1.5 x 104/well with

6-TG. Cells were fed 3-4 days after plating with 50 /xl feeder mix medium.Cell growth was scored after 10-14 days.

The in vivo hprt cloning assay was performed basically as described indetail previously (32). The mononuclear cell fractions were isolated, and cellswere counted and direct plated in 10 /J.M 6-TG for selection at 1 x IO4

cells/well and at 2 cells/well without 6-TG for CE determination. Cell growthwas scored after 10-14 days.

For both in vitro and in vivo hprt assays, CEs were calculated by the Poissonrelationship: CE = (-lnP0)/n, where P<>= fraction of negative wells and

n = average number of cells/well. The calculated MF is the ratio of the CEsin the presence and absence of 6-TG. hprt mutants were propagated in vitro,

and DNA was isolated for molecular analysis.

DNA Analysis

In Vitro hprt Assay. Southern blot hybridization was performed on 205mutant clones (from seven experiments) after Pstl and HindlU digestion with

TCR ßand y gene probes to determine clonal relationships of mutants (38).Briefly, clonal relationships are determined by the restriction patterns exhibited

after restriction with HindlU and Pstl (separately) and probing with the TCRgene probes. Clones having the same pattern in all four of the Southern blothybridizations analyzed were considered to have originated from the same celland to be replicate isolates. Conversely, clones with different rearrangementpatterns originated from different cells and, if mutant at hprt, represent independent hprt mutations. Of the mutants. 139 were determined to be independent isolates based on TCR gene analysis (55 control and 84 treated mutants).

Multiplex hprt PCR was performed as described by Gibbs et al. (39).Briefly, eight pairs of primers specific for the exons were added to a reactionmixture containing the PCR buffer [67 niM Tris-HCl (pH 8.8)-16.6 mM(NH4),SO4-5 mM 2-mercaptoethanol-6.8 /UMEDTA], 1.5 mM dNTPs, 10%DMSO, 6.7 mM MgCU, and approximately 250 ng purified DNA or 5-10 ¿¿1of crude cell extract (representing 5-10 X IO3 cells). Samples were incubatedfor 5 min at 94°Cbefore addition of Taq polymerase (Boehringer-Mannheim)and then underwent 33 cycles of the following: 94°Cfor 1 min, 59°Cfor 1 min,68°Cfor 2 min, and a final extension for 5 min at 68°C.

For mutants with alterations of fragment size on multiplex PCR amplification, the fragment was amplified using exon-specific primers, purified usingthe QIA quick-spin PCR purification kit (Qiagen), and sequenced with aninternal primer using the Taq dyedeoxy terminator cycle sequencing kit (Per-kin-Elmer Cetus, Applied Biosytems division). Extension products were elec-

trophoresed and analyzed on an Applied Biosystems 373 A Automated DNAsequencer, version 1.2.1.

In Vivo hprt Assay. Twenty mutant and 22 wild-type clones were picked

from one of the in vivo experiments with the grain workers. This individual hadbeen exposed to pesticides the longest (>5 years; primarily malathion andphosphine). These clones were expanded in vitro and molecularly analyzed, aswere the in v//ro-derived mutants.

HPLC Analysis of Maialinoli and Contaminants. Malathion was analyzed by HPLC with isocratic elution. The mobile phase consisted of 45% 0.05M potassium phosphate with 10 mM 1-hetanesulfonic acid (Sigma Chemical

Co., St. Louis. MO) adjusted to pH 6.5 with concentrated potassium hydroxideand 55% acetonitrile with 0.02% (v/v) triethylamine at a flow rate of 1.0ml/min (at ambient temperature). The HPLC system consisted of a Spectra-Physics model 8800 pump, a Kratos model 757 detector (220 nm). a Spectra-

Physics model 4200 integrator, and a Rheodyne 7125 injector. For HPLCpurity analysis, malathion samples were diluted into mobile phase, and a 20-/¿1injection volume was used. For post-column mass spectral analysis, a 200-filinjection volume was used. All analyses were performed on a Waters Nova-

Pak (3.9 x 300 mm) CI8 column.Mass spectrometry was performed with a Finnigan SSQ 7IOC single quad

ruple liquid chromatography/mass spectrometer with both electrospray and

10.00-

1.00-

Fig. 1. Cytotoxicity of malathion on human T cells over a rangeof doses. Cytotoxicity is represented as the relative CE as comparedto the controls and is graphed on a log scale. Each Cytotoxicity valuefrom the same blood sample is represented with the same symbol(•.donor 1: A. donor 2; •¿�donor 3; V. donor 4).

UJU

ÃŒi

0.10-

0.01100 200 300 400

Dose ug/ml

500 600 700

2394

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

MALATHION EXPOSURE AND GENOMIC DELETIONS

B

Fig. 2. Average induced MFs over a range of maialinoli doses.MF values for days 5-8 are averaged for all four blood samples.Columns, average MF for each dose; bars, SD. The 50-, 450-, and600-fig/ml doses are significantly elevated from the averagedcontrol value (P < 0.05).

0 30 50 80 450

Dose ug/ml

atmospheric pressure chemical ionization. For mass spectrometry analysis,HPLC peaks were collected, acidified with glacial acetic acid, and infused intothe ion source at 5 ¿ti/min.For electrospray ionization, the spray voltage wastypically 4.5 kV, and for atmospheric pressure chemical ionization, the vaporizer tube was kept at 200°C.Putative assignments of unknown contaminant

peaks were based on molecular weight.

RESULTS

Cytotoxicity of Malathion in Cultured Human T Lymphocytes.Peripheral lymphocytes from four male donors were isolated andexposed in G0 phase to a range of doses (10-600 /ig/ml) ofmalathion. Cytotoxicity values were calculated based on CEs fromday 3 platings (36-40 h after addition of PHA) and are graphed inFig. 1 as the CE expressed relative to the controls. No significantcorrelation between Cytotoxicity and dose of malathion was observed. The graph indicates that there may be some individualsensitivity to malathion, as the Cytotoxicity of one individual's

cells (Fig. 1, •¿�)exhibited slightly higher levels of Cytotoxicity ata number of the doses.

Mutagenicity of Malathion in Cultured Human T Lymphocytes. Preliminaryexperimentsrevealed mutationinductionby malathion to have a phenotypic expression time of 5-8 days (data not

shown), as was previously found for 137Cs-induced mutants (34).

Cells were plated on both days 5/6 and 7/8 of phenotypic expressionfor all doses used. Average MFs were calculated (from days 5-8platings) based on 3-11 independent determinations at each dosetested (30-600 /ug/ml), with the SE for each point represented with abar (see Fig. 2). Average treated MFs were slightly elevated from thecontrols but in most cases were not significantly elevated. When alltreated MFs were compared to all the control MFs, a significantincrease in MF was observed (P < 0.05). When analyzed separatelyby dose, only the 450-pig/ml dose showed a significant elevation inMF over the control, without exhibiting a large SD as was found withthe 50- and 600-(xg/ml doses. The largest increase in MF over thecontrols was approximately 10-fold (at 600 /J.g/ml,on day 7 plating),although this large induction was not repeatable, even when the sameindividual was assayed. Overall, mutant frequencies of treated samples showed both intra- and interindividual variability and, in some

cases, slight significant increases.MFs from the in vivoassays performed on the grain workers did not

exhibit a relationship between time worked and MF. In addition, nosignificant elevations in MF over the controls were seen (control MFsranged from 10.9 x 10~6 to 15.8 X 10^6, and MFs of fumigantworkers ranged from 3.3 X 10~6 to 29.6 X 10~6).

Table 1 Summary of multiplex PCR analysis results

Number and kind of alterations Percentage of Total gross structuralvisualized by multiplex PCR total mutants alterations(%)Control

mutants 3 total deletions 5.5 9/55(16.4)3exon 2 and 3 deletions5.51exon 4 and 5 deletion1.82shifts in exon 6"3.6Malathion-exposed

mutants* 3 total deletions 2.922/104(21.0)(In

vilrii doses 30-600 ug/ml) 1 exon 2deletion2

exon 2 and 3deletions7shifts in exon 3<1exon 3deletion1exon deletion4Iexon 4-9deletion1

shift in exon61exon 6-9deletion.0.9i.7.0.0.0.0.03exon 7-9 deletions 2.9

" Shifts are partial deletions of exons that are visualized on multiplex PCR as downward shifts of the band representing the exon indicated.h Included in malathion-exposed group are 20 in viva-derived mutants; no TCR gene analysis for clonality was performed on these mutants, and to be conservative, each was

represented as an independent mutation.

2395

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

MALATHION EXPOSURE AND GENOMIC DELETIONS

Exon 3 begins (bpl6603)

ÚGACTGAACGTCTTGCTCGAGATGTGATGAAGGAGATGGGAGGCCATCACATTG

TAGCCCTCTGTGTGCTCAAGGGGGGCTATAAATTCTTTGCTCACCTGCTGGATT

ACATCAAAGCACTGAATAGAAATAGTGATAGATCCATTCCTATGACTGTACATT..... —¿�- ...... ---- ....... ----- Illlllllllllllllllllllllllllll

Exon } ends (bp 16786)

Fig. 3. Exon 3 region of hprl showing the regions deleted in a number of malathion-

exposed mutants (JP33 M 13. JP34 M2 1 (sibs). and JP34 M 11 deletions exlending from bp16703 lo 16734; JP50B M25 and M26 deletions extending from bp 16703 to 16724; JP66M6 deletion extending from bp 16748 to 16768; JP74 M6, M8. and MI4 (all sibs)deletions extending from bp 16774 to 16796; and in v/vo-mutant LS309 M4 deletionextending from bp 16671 to 16750]. ..... , Deleted in mutants JP33 M13. JP34 M21(sibs), and JP34 M 11; —¿�. deleted in mutants JP50 M25 and M26; 1111. deleted in mutantJP66 M6; ---- deleted in mutants JP74 M6. M8. and M14 (all sibs); - . deleted in invivo mutant LS309 M4.

Molecular Characterization of 6-TG-resistant T-Lymphocyte

Clones. Mutants were isolated from in vitro experiments in whichsome induction in MF over the controls was observed. Molecularanalysis was performed on all mutants picked from the day 5/6 and7/8 platings, which grew to a sufficient number for Southern blotanalysis (~5 X IO6 cells). Twenty mutants were also picked from the

individual with the longest in vivo exposure and propagated in vitrofor molecular analysis. A summary of the multiplex PCR results areshown in Table 1. All in vitro numbers are based on independentmutants as determined by TCR Southern blots (data not shown).Experiments JP33, JP34. and JP35 were performed using the bloodsample of individual 1; JP50A and JP50B with the blood sample ofindividual 2; JP66 with the blood sample of individual 3; and JP74with the blood sample of individual 4. Only two changes were foundin the 20 mutants analyzed from the in vivo experiment, both downward shifts of the exon 3 fragment on the hprt Southern blot, one(LS309 M3) slightly smaller than the other (LS309 M4). But whenmultiplex PCR was performed on these two mutants, one had a partialdeletion of exon 3 (LS309 M4) and the other a total deletion of exon4 (LS309 M3). The breakpoints of the mutant LS309 M4 weredetermined and are depicted in Fig. 3. The exact breakpoints in theother mutant (LS309 M3) are still uncertain, and this mutation mayinvolve some complex rearrangement. Because no TCR analysis wasperformed on the in v/'vo-derived mutants, to be conservative, each

was counted as an independent mutation. The percentage of grossstructural alterations in the malathion-exposed group was slightly

higher than that of the control [21.0 versus 16.1%, respectively (seeTable 1)]. A significant difference was noticed among the types ofdeletions in each group. Thirty-two % (7 of 22) of the mutations fromthe malathion-exposed group showed deletions involving exon 3,

whereas the control group showed none. The frequency of this partialexon 3 deletion is significantly higher in the malathion-treated cellsthan in the in vitro control group's cells (P < 0.05; Table 2).

To better evaluate the significance of the partial exon 3 deletionmutation frequency, a larger database was compiled from publishedand unpublished multiplex PCR analysis of hprt mutations from anumber of laboratories (40).4'5 Approximately 40% of the mutants in

this large database were derived from normal non-exposed cells, with

the remainder coming from individuals with various genetic or otherdiseases (diabetics, colon cancer patients, fathers of Prader-Willi

syndrome patients, and AT hétérozygotes)or from various in vivo or

in vitro mutagen-exposed situations (137Cs, radon, amsacrine, cis-

platinum, WOT, and plutonium). Therefore, the larger database,comprising 1654 hprt mutants, is heterogeneous. The frequency ofexon 3 partial deletions in the current 104 mutations derived frommalathion exposure is also significantly elevated from the frequencyfound in this much larger database (P < 0.0001; see Table 2). Of the1654 database mutants, only 6 showed a shift indicating a partialdeletion of exon 3. The mutants that did show the exon 3 shift werederived primarily from in vivo exposures, the Chernobyl incident (3mutants), and in vitro exposures of 137Csradiation (1 mutant) and the

etoposide amsacrine (1 mutant). Genomic sequence analysis wasperformed on the malathion-exposed partial exon 3 deletions. Fig. 3shows the breakpoints for the malathion-exposed partial exon 3 deletions, revealing a 3' region of exon 3 that seems to be exceptionally

breakpoint prone. Included in Fig. 3 is the region of exon 3 founddeleted in a mutant analyzed from an in vivo pesticide-exposed (to

malathion and phosphine. primarily) individual. The region deleted inthis in viYoderived mutant totally encompasses four of the regionsdeleted in the in v;'fro-derived partial exon 3 mutants and overlaps

slightly with the region deleted in another. In most cases, the breakpoints for the partial exon 3 deletions in the larger database of 1654hprt mutations have not yet been determined. Two, however, obtainedin our laboratory after in vitro 137Csexposure, have been sequenced.

These two independent mutations (by TCR gene analysis) have identical breakpoints (bp 16,760-16,813); the region deleted in thesemutants overlaps slightly with the deletion in one of the malathion-

induced mutants (JP66 M6; overlaps 9 bp), and totally encompassesanother (JP74 M6, M8, and M14. all sibs).

An attempt was also made to narrow down the breakpoints for ani/i vitro total exon 3 deletion mutant, isolated from the malathion-

exposed cells. A battery of PCR primers surrounding exon 3 wereused in combination to determine the approximate breakpoints for thisdeletion. On the basis of the size of the products obtained and hprtSouthern blot studies, this mutant appears to have a large insertion(>20 kb), the placement of which has been narrowed down to theregion encompassing base pairs 16.676-16,695 (within exon 3). The

breakpoints for this likely insertion lie within the region deleted in thein v/'vo-derived mutant in Fig. 3. This mutation was not counted in the

total numbers of exon 3 partial deletion mutants because it is likely aninsertion, but it is of interest in that another mutation occurs withinthis particularly malathion-sensitive region of hprt.

Determination of Malathion Purity. Each lot of malathion wasanalyzed using mass spectrometry and HPLC to determine whetherdifferent percentages of contaminants may have affected the mutage-

nicity results. The various lots appeared to be very similar with regardto the percentage and types of contaminants; they contained approximately 1-3% contaminants. There were three lots of malathion pur

chased from Supelco at various time points in the study. The first hadapproximately 2% total contaminants consisting of three primaryspecies (isomalathion, a malathion isomer analogue, and a species thatis likely malaoxon). The second had primarily two species of contaminants that made up the total 3% contamination (the malathionisomer and the malaoxon-like contaminant). The third contained the

Table 2 Frequency of emn 3 shifts as visualized by multiplex PCRin vdr/ííw.vdatabases

Total Number of mutants Percentage of mutantsPopulation analyzed with exon 3 shifts with exon 3 shifts

Multilabs combined database 1654 6In vitro controls 55 0Malathion-exposed" 104 7

0.40

6.7

4 J. Fuscoe. I. Jones. B. Finette, personal communication.s Unpublished data.

" Included in malathion-exposed group are 20 m n'vo-exposed mutants; no TCR gene

analysis for clonality was performed on these mutants, and to be conservative, each wasrepresented as an independent mutation.

2396

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

MALATHION EXPOSURE AND GENOMIC DELETIONS

least amount of contamination (i.e., 1%, all of which was the mala-

thion isomer). These slight differences in contaminants probably didnot affect our results. One trial run comparing lots 2 and 3 did notshow any obvious differences in MF induction or cytotoxicities withrespect to the lot used.

DISCUSSION

A wide range of malathion doses were used in the current study(i.e., 10-600 pig/ml). In extreme cases, a farmer or other agricultural

worker could occupationally experience the lower doses. The estimated lethal oral dose of malathion for humans is 858 mg/kg bodyweight. The concentration of malathion found in the blood of individuals autopsied after malathion overdose ranged from 175-517

/ug/ml (41). Therefore, concentrations considerably less than these(i.e., less than 100 jug/ml) could be achieved in the blood throughvarious nonlethal human exposures. Our studies over a range of dosesreveal malathion as having a low and variable cytotoxicity, even at thehighest dose (600 jag/ml). Also, there may be individual differences insusceptibility to the insecticide as shown in Fig. 1, with one individualshowing higher cytotoxicities over a range of doses.

The mutagenicity of malathion was also variable. Significant increases in MFs in treated versus nontreated cells could be shown onlywhen all treated MFs were averaged and compared to the nontreatedMFs (P < 0.05). A slight dose-response relationship was also seen.Others, too, have failed to find dose-related genotoxic responses with

malathion exposures but were able to detect significant differencesbetween treated and control groups (13, 15, 42). These cytogeneticstudies over a range of malathion doses failed to detect a correlationbetween the frequency of sister chromatid exchanges and/or chromosome aberrations and dose. Several in vivo studies also failed to showa dose-response relationship in mice or humans exposed to malathion(20, 43, 44). Studies examining malathion's effect on cell cycle

progression have shown it to be a very potent cell cycle inhibitor, asreflected by lowered mitotic indices and diminished levels of radio-labeled DNA and RNA (43, 45-48). Therefore, malathion's affect on

the cell cycle is likely to be partially responsible for our lack of adose-related mutagenic response.

An additional source of variability in our studies was the poor

solubility of malathion in the medium used. This could have causedunequal distributions and exposures of cells. In one study of in vitromalathion-exposed T lymphocytes that did show a relationship between sister chromatid exchanges and dose, a surfactant F-127 plu-

ronic, rather than DMSO or ethanol, was used as the initial solvent(14). However, the use of a 5% solution of F-127 pluronic did not

result in reproducibly better results for our mutagenicity studies thanthe use of DMSO, so we continued to use the latter solvent. Theability of Garry et al. (14) to show a dose-response relationship may

reflect different end points studied and/or the temporal aspects of thedifferent assays. The overall effect of the inhibition of the cell cycleby malathion and its unequal dispersion in the culture medium probably resulted in cells at the same exposure level receiving differentdoses, giving the variable and slight responses that we found.

The above complications of the in vitro mutagenicity assays makethe molecular findings of the current study even more striking. Whenmutants from the treated cultures were analyzed, a clear difference inthe molecular mutational spectrum as compared with that of untreatedcells became evident. Several mutants from the treated culturesshowed partial deletion in hprt exon 3, whereas none with this DNAchange were found any nontreated mutants. There was also specificityin the deleted region, with four of the six in vi'/ro-derived treated

mutants showing overlapping deletions in the 3' region of exon 3.

Furthermore, an additional in wVo-derived mutant had the largest

partial deletion of exon 3 (i.e., 80 bp), which encompassed the regiondeleted in four of the in vifro-derived mutants and overlapped with a

region deleted in another. The finding of seven independent mutations(at least one from each individual's in w'/ro-exposed blood sample),

represented by one or more mutants, in the malathion-treated cells

with partial exon 3 deletions and none in 55 independent in vitromutations from untreated cells, strongly supports the role of malathionin inducing these deletions.

To determine the extent to which this intra-exon 3 deletion couldserve as a "signature" of a malathion-induced genotoxic event, we

compiled a larger database of hprt mutation determinations by multiplex PCR, using published and unpublished results from severallaboratories, including our own. In a total of 1654 mutants analyzedby multiplex PCR, only 6 showed a partial deletion of exon 3 (see

Fig. 4. Hairpin-loop structures that may form in theexon 3 region of hprt. Breakpoints for each independentmalathion-induced mutant are shown, and 5' or 3' break

point is indicated |JP33 M13, JP34 M21 (sibs). and JP34Mil deletions extending from bp 16703 to 16734;JP50B M25 and M26 deletions extending from bp 16703to 16724; JP66 M6 deletion extending from bp 16748 to16768; JP74 M6. MS. and MI4 (all sibs) deletions extending from bp 16774 to 16796; and in vim mutantLS309 M4 deletion extending from bp 16671 to 16750].The number of deletion breakpoints at each of these sitesis shown in parentheses; *. in w'w-deletion breakpoints.

o1G-C"T-AA-TG-CT-AG-CT-AA-T

0¿^C

n...CTTG-C\ftV»,•if-

^><on.G5'

x^G-C i ' ~_ 3'

16703 T-A S 16796(4) C C £ (1)

CI/jAG<G

A 3'£T-A"*16724f?C

T (2)PGATG

ST-A S

l\SÇA5T T 35'3'

O4C-T7ATA * ^"^^B~—J 6774A rv 16734 ^(DAr* —¿�(2)/A-l^A

G /S'^^.31VS' T-A 5. 3' /y16768^>

16671 A-T Ì6743 16750 ^(1,G^^-(1*) T-A (^ y<1 I (£•

2397

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

MALATHION EXPOSURE AND OENOMIC DELETIONS

Table 3 Short nomologie* surrounding nialalhìan-ìnducedpartial exon 3 deletions

Mutants Region surrounding partial exon 3 deletion

LS 309 M4(in vivo derived)JP33 M13 and JP34 M21 (sibs)0

(individual 1)

JP34M11(individual I)

JP50B M25(individual 2)

JP50B M26(individual 2)

JP66 M6(individual 3)

JP74 M6, M8. and M14 (all sibs)(individual 4)

[5'region//. . .deleted region. . .//3'region]

GTGTGC//TCAAGG //ATGACGTGTGC// ATTCCT//ATGAC

GACCTG//CTGGAT //TGATAGGACCTG// AAAT AG//TGATAG

GACCTG//CTGGAT //TGATAGGACCT// AAAT AG//TGATAG

GACCTG//CTGGAT //ATAOAAGACCTG //CACTGA//ATAGAA

GACCTG/ /CTGGAT //ATAQAAGACCTG// CACTGA//ATAGAA

o*ATCCATT//CCTATG.ATCCATT//. . .G6.

. . G. ..//CAGACTTTTTATC//CAGACT

TCAGAC//TGAAGA //TTAATATCAGAC//. .GTATAT//TTAATA

" Sibs, replicate isolates of the same in vitro mutation.

This nucleotide found inserted between the deletion breakpoints.

Table 2). None of these intra-exon 3 deletions in this larger database

were from control individuals or cells; all were associated with in vivoor in vitro exposures to known clastogenic agents. Thus, the percentage (6.7%) of intra-exon 3 deletions in the malathion-exposed cells is

significantly greater than even that found in this larger database(0.4%; P < 0.0001).

Mechanistically, it is unknown how malathion produces genomicdeletions and, moreover, why they are specific and restricted to afairly small region (125 bp). An analysis of the exon 3 sequenceshows that it has the potential to form up to four hairpin-like struc

tures, each with a stem of at least 4 bp, the AG values of which rangefrom 0 to -5.8 kcal/mol. The two most stable hairpin structures are

depicted in Fig. 4 with the breakpoints for the malathion inducedmutants labeled. It is possible that malathion stabilizes these hairpin-

loop structures leading to the characteristic breakpoints. There appearsto be some association between the breakpoints and these hairpinstructures because four of the six deletion breakpoints of the inwire-derived mutant reside in the larger second hairpin structure, andthe deletion breakpoints of the in vivo-derived mutant are near the

base on either side of this structure.Small direct repeats of 2-5 bp have been associated with the

breakpoints in spontaneous deletion mutants (49-51). Although smalldirect repeats (2-5 bp) were found at or near the breakpoints in all thein vi/ra-derived malathion-associated mutants (shown in bold type in

Table 3), it is difficult to understand why they would only increase thefrequency of deletions at these sites in the treated cells. It is possiblethat malathion affects the ability of one or more DNA repair enzymesto function accurately, leading to an increased frequency of deletionsin the treated cells, but again, this explanation does not explain theincreased frequency of deletions in such a small region of hprt.

Previous studies of malathion's interaction with DNA have estab

lished only a possible weak interaction. Malathion was able to inducebreakage in Col El plasmid DNA from E. coli at a concentration of0.01 mg/ml (8). Malathion was also able to alkylate nitrobenzylpyri-

dine [a synthetic substrate (52)] and methylate DNA bases in vitro(53). However, the ability to methylate and alkylate DNA does notexplain malathion's apparent ability to cause genomic deletions,

because other alkylating agents normally induce primarily pointmutations.

The results of this study have potential health implications forhumans. First, we have shown that malathion mutagenicity can bedetected at the molecular level. The molecular changes at hprt wereseen at doses of malathion that produced no cytotoxicity in vitro (:£

50 mg/ml) and at exposure levels experienced by the individual fromwhich the in vivo mutant was obtained. Inasmuch as several epide-

miological studies have found an increase in hematological malignancies in individuals occupationally exposed to pesticides, our finding ofmalathion genotoxicity may be especially relevant. Similar changes tothose reflected in hprt in our study may be occurring also at other loci,especially sites of oncogenes or tumor suppressor genes, and may playa role in the induction of malignancies in individuals exposed to thisor a similar agent. Finally, our finding of specificity of the malathioninduced genomic deletions at hprt may have a practical relevance inallowing the development of a molecular-based assay for the rapid

detection of in vivo genotoxicity in individuals exposed to pesticides.Molecular epidemiology can then supplement traditional epidemiology in elucidating the true health effects of these potentially widespread human exposures.

ACKNOWLEDGMENTS

We gratefully thank Dr. Vincent Garry (University of Minnesota) for thegrain fumigant workers' blood samples and Drs. James C. Fuscoe of EPA

(Research Triangle Park, NC), Irene Jones (Lawrence Livermore Laboratory,Livermore, CA), Barry Finette, and Scott Clark (University of Vermont.Burlington, VT) for sharing their multiplex PCR data bases. We also thankLinda Sullivan, Tim Hunter, and Dr. James Bigelow for technical assistance.

REFERENCES

1. Umetsu, N., Grose, F. H.. Allahyari. R., Abu-El-Haj. S.. and Fukuto, T. R. Effect of

impurities on mammalian toxicity of technical malathion and acéphale.J. Agrie. FoodChem.. 25: 946-953. 1977.

2. Shirasu, Y., Moriya, M., Furuhashi. A., and Kada. T. Mutagenicity screening ofpesticides in the microbial system. Mutât.Res.. 40: 19-30. 1976.

3. McCann, J., Choi. E.. Yamasaki. E.. and Ames. B. Detection of carcinogens asmutagens in the Salmonella/microsome test: assay of 300 chemicals. Proc. Nati.Acad. Sci. USA, 72: 5135-5139, 1975.

4. Gilot-Delhalle. J.. Colizzi, A.. Moutschen. J.. and Moutschen-Dahmen. M. Mutagenicity of some organophosphorus compounds at the adeo locus of Schizo.wcharo-m\ces pombe. Teratog. Carcinog. Mutagen., 117: 139-148, 1983.

5. Waters, M., Sandu. S., Simmon. V., Mortelmans, K. E., and Mitchell, A. P. Study ofpesticide genotoxicily. Basic Life Sci., 21: 275-326, 1982.

6. Wong, P.. Wai, C.. and Liong. E. Comparative study on mutagenicities of organophosphorus insecticides in Sa/monella. Chemosphere, 18: 2413-2422. 1989.

7. Mohn, G. 5-methyltryptophan resistance mutations in Eacherichia coli K-12: muta-

genie activity of monofunctional alkylating agents including organophosphorus insecticides. Mutât.Res.. 20: 7-15. 1973.

8. Wild. D. Mutagenicity studies on organophosphorus insecticides. Mutât.Res.. 32:133-150, 1975.

9. Tennant. R.. Margolin. B., Shelby. M.. Zeiger. E.. Haseman, J. K.. and Spalding. J.Prediction of chemical carcinogenicity in rodents from in vitro genetic loxicity assays.Science (Washington DC), 236: 933-941, 1987.

2398

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

MALATHION EXPOSURE AND GENOMIC DELETIONS

10. Myhr, B., and Caspary, W. Chemical mutagenesis at the thymidine kinase locus inL5178Y mouse lymphoma cells: results for 31 coded compounds in the NationalToxicology Program. Environ. Mol. Mutagen., 18: 51-83, 1991.

11. Degraeve, N., Chollet, M. C, and Moutschen, J. Cytogenetic and genetic effects ofsubchronic treatment with organophosphorus insecticides. Arch. Toxicol., 56: 66-67,

1984.12. Degraeve, N., and Moutschen. J. Genetic and cytogenetic effects induced in the

mouse by an organophosphorus insecticide: malathion. Environ. Res., 34: 170-174,

1984.13. Herath, J. F., Jala, S. M., Ebert, M. J., and Martsolf, J. T. Genotoxicity of the

organophosphorus insecticide malathion based on human lymphocytes in culture.Cytologia, 54: 191-195, 1989.

14. Garry, V., Nelson, R., Griffith, J., and Harkins, M. Preparation for human study ofpesticide applicators: sister chromatid exchanges and chromosome aberrations incultured human lymphocytes exposed to selected fumigants. Teratog. Carcinog.Mutagen., 10: 21-29, 1990.

15. Walter, Z., Czajkowska, A., and Lipecka, A. Effect of malathion on the geneticmaterial of human lymphocytes stimulated by phytohemagglutinin (PHA). Hum.Genet., 55:375-381, 1980.

16. Sobti, R. C., Krishan, A., and Pfaffenberger, C. D. Cytokinetic and cytogenetic effectsof some agricultural chemicals on human lymphoid cells in vitro: organophosphates.Mutât.Res., 102: 89-102, 1982.

17. Nicholas, A., Vienne, M., and van den Berghe, H. Induction of sister-chromatidexchanges in cultured human cells by an organophosphorous insecticide: malathion.Mutât.Res., 67: 167-172, 1979.

18. Kalow, W., and Marlon, A. Second-generation toxicity of malathion in rats. Nature(Lond.), 192: 464-465, 1961.

19. Yoder. J., Watson, M., and Benson, W. W. Lymphocyte chromosome analysis ofagricultural workers during extensive occupational exposure to pesticides. Mutât.Res., 21: 335-340, 1973.

20. van Bao, T., Szabo, I., Ruzicska, P., and Czeizel, A. Chromosome aberrations inpatients suffering acute organic phosphate insecticide intoxication. Humangenetik.24: 33-57, 1974.

21. Lipkowitz, S., Garry, V. F., and Kirsch, I. R. Imerlocus V-J recombination measures

genomic instability in agriculture workers at risk for lymphoid malignancies. Proc.Nat. Acad. Sci. USA., 89: 5301-5305, 1992.

22. Milham, S. Leukemia and multiple myeloma in farmers. Am. J. Epidemic!., 94:307-310, 1971.

23. Cantor. K. P. Farming and mortality from non-Hodgkin's lymphoma: a case-control

study. Int. J. Cancer, 29: 239-247, 1982.

24. Blair, A., Malker, H., Cantor, K. P., Burmeister, L., and Wiklund, K. Cancer amongfarmers. Scand. J. Work Environ. Health, //: 397-407, 1985.

25. Hoar, S. K., Blair, A., Holmes, F. F., Boysen, C. D., Robel, R. J., Hoover. R., andFraumeni, J. F., Jr. Agricultural herbicide use and risk of lymphoma and soft-tissuesarcoma. J. Am. Med. Assoc., 256: 1141-1147, 1986.

26. Brown, L. M., Blair, A., Gibson, R., Everett. G. D., Cantor, K. P., Schuman, L. M.,Burmeister, L. F., Van Lier, S. F., and Dick, F. Pesticide exposures and otheragricultural risk factors for leukemia among men in Iowa and Minnesota. CancerRes., 50: 6585-6591, 1990.

27. Pasqualetti, P., Casale, R., Colantonio, D., and Collacciani. A. Occupational risk forhematological malignancies. Am. J. Hematol., 38: 147-149, 1991.

28. Albenini, R. J., Sylwester, D. L., and Allen. E. F. The 6-thioguanine resistantperipheral blood lymphocyte assay for direct mutagenicity testing in man. In: J. A.Heddle (ed.), Mutagenicity: New Horizons in Genetic Toxicology, pp. 130-145. New

York: Academic Press, 1982.29. Morley, A. A., Cox, S., Wismore, D., Seshadri, R., and Dempsey, J. L. Enumeration

of thioguanine-resistant lymphocytes using autoradiography. Mutât.Res., 95: 363-

375, 1983.30. Albertini, R. J.. Sullivan. L. S., Berman, J. K.. Greene, C. J., Stewart, J. A., Silveira,

J. M., and O'Neill, J. P. Mutagenicity monitoring in humans by autoradiographic

assay for mutant T-lymphocytes. Mutai. Res., 204: 481-492, 1988.31. Albertini, R. J., Nicklas, J. A., O'Neill, J. P., and Robison, S. H. In vivo somatic

mutations in humans: measurement and analysis. Annu. Rev. Genet., 24: 305-326,

1990.32. O'Neill, J. P., McGinniss, M. J., Berman. J. K.. Sullivan, L. M.. Nicklas, J. A., and

Albertini, R. J. Refinement of a T-lymphocyte cloning assay to quantify the in vivothioguanine-resistant mutant frequency in humans. Mutagenesis, 2: 87-94. 1987.

33. O'Neill, J. P., Sullivan. L. M., Booker, J. K., Pomelos, B. S., Falla, M. F., Greene,

C. J., and Albertini, R. J. Longitudinal study of the in vim hprt mutant frequency inhuman T-lymphocytes as determined by a cell cloning assay. Environ. Mol. Mutagen., 13: 289-293, 1989.

34. O'Neill, J. P., Sullivan, L. M., and Albertini. R. J. In vitro induction, expression and

selection of thioguanine resistant mutants with human T-lymphocytes. Mutât.Res.,240: 135-142, 1990.

35. O'Neill, J. P., Sullivan, L. M., Hunter, T. C., and Nicklas, J. A. Cytosine arabinoside

enhancement of gamma irradiation induced mutations in human T-lymphocytes.Environ. Mol. Mutagen., 17: 231-237, 1991.

36. Cartello. N. F.. and Skopek. T. R. Analysis of mutation occurring at the human hprtlocus. J. Mol. Biol., 231: 41-57, 1993.

37. Cartello, N. F., Craft, T. R., Vrieling. H.. van Zeeland, A. A., Adams, T., and Skopek.T. R. Human HPRT mutant database. Software for data entry and retrieval. Environ.Mol. Mutagen., 20: 81-83, 1992.

38. Nicklas, J. A., Hunter, T. C., O'Neill. J. P., and Albertini, R. J. Molecular analyses

of in vivo hprt mutations in human T-lymphocytes: III. Longitudinal study oíhprtgene structural alterations and T-cell clonal origins. Mutât.Res., 275: 147-160,

1989.39. Gibbs, R. A.. Nguyen, P. N.. Edwards, A., Civitello, A. B., and Caskey, C. T.

Multiplex DNA deletion detection and exon sequencing of the hypoxanthinephosphoribosyltransferase gene in Lesch-Nyhan families. Genomics, 7: 235-244,

1990.40. Fuscoe, J. C., Zimmerman, L. J., Harrington-Brock, K.. and Moore, M. M. Deletion

mutations in the hprt gene of T-lymphocytes as a biomarker for genomic rearrangements important in human cancers. Carcinogenesis (Lond.), /5: 1463-1466, 1994.

41. Jadhav, R. K., Sharma. V. K.. Rao. G. J., Saraf. A. K., and Chandra, H. Distributionof malathion in body tissues and fluids. Forensic Sci. Int., 52: 223-229, 1992.

42. Bradley, M. O., Bhuyan, B., Francis, R., Langenbach. R., Peterson, R., andHuberman, E. Mutagenesis by chemical agents in V79 Chinese hamster cells: areview and analysis of the literature. Mutât.Res., 87: 81-92, 1981.

43. Dulout, F. N., Olivero, O. A., von Guradze. H., and Pastori, M. C. Cytogenetic effectof malathion assessed in bone-marrow cells of mice: dose-response relationships.Mutât.Res., /05: 413-416. 1982.

44. Dulout, F. N., Pastori, M. C., Olivero, O. A., Gonzalez, C. D., Loria, D., Matos. E.,Sobel, N., de Bujan, E. C., and Albiano, N. Sister-chromatid exchanges and chromosomal aberrations in a population exposed to pesticides. Mutât.Res., 143: 237-

244, 1985.45. Czajkowska, A., and Walter, Z. Effect of malathion on nucleic acid synthesis in

phytohemagglutinin-stimulated human lymphocytes. Hum. Genet.. 56: 189-194,

1980.46. Salvadori, D., Ribeiro. L.. Pereira, C.. and Becak. W. Cytogenetic effects of mala

thion insecticide on somatic and germ cells of mice. Mutât.Res., 204: 283-287, 1988.

47. Rupa, D. S., Reddy. P. P., Sreemannarayana. K.. and Reddi, O. S. Frequency of sisterchromatid exchanges in peripheral lymphocytes of male pesticide applicators. Environ. Mol. Mutagen., 18: 136-138, 1991.

48. Chen. H.. Hsueh. J., Sirianni, S., and Huang, C. Induction of sister chromalidexchanges and cell cycle delay in cultured mammalian cells treated with eightorganophosphorus pesticides. Mutât.Res.. 88: 307-316. 1981.

49. Thacker. J., Chalk, J., Ganesh, A., and North, P. A. Mechanism for deletion formationin DNA by human cell extracts: the involvement of short sequence repeats. NucleicAcids Res., 20: 6183-6188, 1992.

50. Monnat, R. J., Hackmann, A. F. M., and Chiaverotti, T. A. Nucleotide sequenceanalysis of the human hypoxanthine phosphoribosyltransferase (HPRT) gene deletions. Genomics, 13: 777-787, 1992.

51. Meuth, M. The structure of mutation in mammalian cells. Biochim. Biophys. Acta,1032: 1-17, 1990.

52. Imamura. T.. and Talcott. R. E. Mutagenic and alkylaling activities of organophos-phate impurities of commercial malathion. Mutât.Res., 755: 1-6, 1985.

53. Wiaderkiewicz, R.. Walter, Z.. and Reimschussel, W. Sites of methylation of DNAbases by the action of organophosphorus insecticides in vitro. Acta Biochim. Pol., 33:73-85, 1986.

2399

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from

1996;56:2393-2399. Cancer Res   Janice M. Pluth, Janice A. Nicklas, J. Patrick O'Neill, et al.  

Malathion Exposurein Vitrofrom Increased Frequency of Specific Genomic Deletions Resulting

  Updated version

  http://cancerres.aacrjournals.org/content/56/10/2393

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

Research. on January 27, 2016. © 1996 American Association for Cancercancerres.aacrjournals.org Downloaded from


Recommended