You are on page 1of 9

Toxicology Letters 217 (2013) 14–22

Contents lists available at SciVerse ScienceDirect

Toxicology Letters
journal homepage: www.elsevier.com/locate/toxlet

Chlorpyrifos and its metabolites alter gene expression at non-cytotoxic


concentrations in D3 mouse embryonic stem cells under in vitro differentiation:
Considerations for embryotoxic risk assessment
Carmen Estevan ∗ , Eugenio Vilanova, Miguel A. Sogorb
Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Spain

h i g h l i g h t s

 Chlorpyrifos and its metabolites alter gene expression in mouse embryonic stem cells.
 Chlorpyrifos and its metabolites alter gene expression at non-cytotoxic concentrations.
 Chlorpyrifos and its metabolites alter pluripotency of mouse embryonic stem cells.
 Chlorpyrifos and its metabolites alter differentiation of three embryonal lineages.
 Chlorpyrifos might alter differentiation without detectable maternal toxicity.

a r t i c l e i n f o a b s t r a c t

Article history: The effects of organophosphate insecticide chlorpyrifos (CPF) on development are currently under dis-
Received 26 September 2012 cussion. CPF and its metabolites, chlorpyrifos-oxon (CPO) and 3,5,6-trichloro-2-pyridinol (TClP), were
Received in revised form more cytotoxic for D3 mouse embryonic stem cells than for differentiated fibroblasts 3T3 cells. Exposure
27 November 2012
to 10 ␮M CPF and TClP and 100 ␮M CPO for 12 h significantly altered the in vitro expression of biomark-
Accepted 29 November 2012
ers of differentiation in D3 cells. Similarly, exposure to 20 ␮M CPF and 25 ␮M CPO and TClP for 3 days
Available online 7 December 2012
also altered the expression of the biomarkers in the same model. These exposures caused no significant
reduction in D3 viability with mild inhibition of acetylcholinesterase and neuropathy target esterase by
Keywords:
Chlorpyrifos
CPF and severe inhibition by CPO. We conclude that certain in vivo exposure scenarios are possible, which
Chlorpyrifos-oxon cause inhibition of acetylcholinesterase but without clinical symptoms that reach high enough systemic
Embryonic stem cell CPF concentrations able to alter the expression of genes involved in cellular differentiation with poten-
Differentiation tially hazard effects on development. Conversely, the risk for embryotoxicity by CPO and TClP was very
Risk assessment low because the required exposure would induce severe cholinergic syndrome.
© 2012 Elsevier Ireland Ltd. All rights reserved.

Abbreviations: Ache, acetylcholinesterase (gene); AChE, acetylcholinesterase (protein); Afp, ␣-fetoprotein; CPF, chlorpyrifos (O,O-diethyl O-(3,5,6-trichloro-2-
pyridinyl) phosphorothioate); CPO, chlorpyrifos-oxon (O,O-diethyl O-(3,5,6-trichloro-2-pyridinyl) phosphate); DEPC, diethylpyrocarbonate; DMEM, Dulbecco’s Modified
Eagle’s Medium; ECx, concentration needed to cause a reduction in cell viability of X%; EST, Embryonic Stem cell Test; Flk1, foetal liver kinase 1; IC50, concentration needed
to inhibit the enzymatic activity by 50%; LIF, leukaemia inhibition factor; Mhc, myosin heavy chain; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide;
Nanog, nanog homeobox; Nefm, neurofilament medium polypeptide; Nes, nestin; NTE, neuropathy target esterase; OP, organophosphorus compound; PBS, phosphate buffered
saline; Pnpla6, patatin-like phospholipase domain containing 6; PV, phenyl valerate; qRT-PCR, quantitative real-time PCR; TClP, 3,5,6-trichloro-2-pyridinol; TLV, Threshold
Limit Value.
∗ Corresponding author at: Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avenida de la Universidad s/n, 03202 Elche, Spain. Tel.: +34 966658821;
fax: +34 966658511.
E-mail address: cestevan@umh.es (C. Estevan).

0378-4274/$ – see front matter © 2012 Elsevier Ireland Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.toxlet.2012.11.026
C. Estevan et al. / Toxicology Letters 217 (2013) 14–22 15

1. Introduction (used as a differentiated cells model) and in D3 mouse embryonic


stem cells and the capability of the compound to alter the spon-
Chlorpyrifos (CPF) is an organophosphorus compound (OP) used taneous differentiation of D3 cells into beating cardiomyocytes
to control insect pests which has been used for almost 40 years. (Genschow et al., 2004). Nevertheless, the EST needs to be improved
CPF is not authorised for household applications by the USEPA by using molecular endpoints for studying alterations in differen-
since 2002 (USEPA, 2000) and by the European Commission since tiation (Spielmann et al., 2006). In this way, gene expression has
2005 (European Commission, 2005). In the latter case however, it proved a good endpoint to be implemented within conventional
is included in Annexe I to Directive 91/414/EEC for plant protection EST (Romero et al., 2011). Specifically, ␣-fetoprotein (Afp) (Romero
products (European Communities, 1991). et al., 2011; Pamies et al., 2010a) and nestin (Nes) have already
The CPF metabolism consists in the oxidative desulphuration been found suitable for testing embryotoxicity by monitoring their
of the P S group to form chlorpyrifos-oxon (CPO). This reaction is expression, but there are other genes like the patatin-like phospho-
catalysed by cytochrome P450, mainly in the liver, and the oxidised lipase domain containing 6 (Pnpla6), the codifying gene for NTE,
form of OPs (in our case CPO) is always more powerful for inhibiting which seem to play a prominent role in the differentiation of D3
acetylcholinesterase (AChE)1 and neuropathy target esterase (NTE) cells (Pamies et al., 2010b) and might, therefore, be good candidates
than the reduced form (in our case CPF) (WHO, 1986). CPO can be for embryotoxicity testing.
further chemically or enzymatically deactivated by hydrolysis with In this work, we studied the effects of CPF and its metabolites,
the release of diethylphosphate and 3,5,6-trichloro-2-pyridinol CPO and TClP, on D3 mouse embryonic stem cells under sponta-
(TClP). Furthermore, the oxidative dearylation of CPF to directly neous differentiation after short- (12 h) and mid- (3 days) term
release TClP and diethylthiophosphate can also occur in the liver exposure using the gene expression of biomarkers of differentia-
and represents an alternative detoxification pathway (Sogorb and tion of the three main embryonic lineages and the gene expression
Vilanova, 2010). of biomarkers of pluripotency. Most tested genes have previously
CPF is able to induce the well-known cholinergic syndrome, proven their capability to predict embryotoxicity (Romero et al.,
caused by inhibition of AChE, and organophosphorus-induced 2011; Pamies et al., 2010a). Our main aim was to estimate the risk
delayed polyneuropathy, caused by inhibition of NTE (Jokanovic of embryotoxicity to exposure to CPF and we conclude that in cer-
and Kosanovic, 2010). CPF also causes other neurological impair- tain scenarios, exposure to CPF could be enough to become a matter
ments, such as long-term spatial learning dysfunctions (Cañadas of concern.
et al., 2005), which are probably related to loss of dendrite and
spine processes in both the hippocampus and prefrontal cortex 2. Materials and methods
(Ruiz-Muñoz et al., 2011).
There is no consistent evidence for CPF’s capacity to induce ter- 2.1. Chemicals

atogenicity with exposures below those required to cause maternal


CPF (O,O-diethyl O-(3,5,6-trichloro-2-pyridinyl) phosphorothioate) and TClP
toxicity (Eaton et al., 2008). However, there is contradictory infor- (3,5,6-trichloro-2-pyridinol) were purchased form Sigma–Aldrich Spain with
mation on the capability of CPF to cause neurodevelopmental purities of 99.5% and 98.8%, respectively. CPO (O,O-diethyl O-(3,5,6-trichloro-2-
toxicity since many in vivo studies have reported such effects on pyridinyl) phosphate) was obtained from Dr. Ehrenstorfer with a purity of 98%.
All the other chemicals, materials for cell culture or molecular biology manage-
exposures causing decreases in brain AChE, while other studies
ments were attained from Sigma–Aldrich Spain, Roche or local suppliers, and were
have indicated effects at dose levels which do not cause any appar- of analytical grade.
ent AChE inhibition (Eaton et al., 2008). Indeed, Slotkin et al. (2006)
have suggested that there is a complete dichotomy between the
2.2. Cell cultures and exposures
systemic toxicity of OPs and their capability to cause develop-
mental neurotoxicity. These data suggest that it is necessary to Cells were exposed to all chemicals during either 12 h or 3 days. Twelve hours
assess the risk of exposure to CPF during development by espe- was chosen since this is the earliest point were the expression of the biomarkers
cially considering that exposure occurs in the general (not only in genes could be measured with acceptable accuracy. Indeed, the expression of most
of the tested genes could not be detected after only 10 h of differentiation (data not
an occupationally exposed) population as an epidemiological study shown).
has shown that CPF displayed a prevalence of 11% in meconium at
a mean concentration of 53 ppm (Ostrea et al., 2002).
2.2.1. D3 mouse embryonic stem cells
CPF undergoes intensive biotransformation in live organisms, D3 mouse embryonic stem cells were obtained from the American Type Cul-
mainly the liver. Therefore, it remains unclear if the chemi- ture Collection (Rockville, MD, USA). These cells were firstly isolated from the inner
cal responsible for the above-stated effects is the same CPF or cellular mass of a 129/Sv mouse blastocyst on day 4 of gestation. D3 cells were
some of its metabolites (CPO or TClP). In vitro studies have grown on monolayers in an undifferentiated state on 75-mm plates in Dulbecco’s
Modified Eagle’s Medium (DMEM) supplemented with 15% heat-inactivated foetal
demonstrated that oxidised forms of OPs, such as CPO, display
calf serum, 1% non-essential amino acids, 50 units of penicillin/ml, 100 ␮g strepto-
greater capability (up to 1000 times) to disrupt a number of mycin/ml, 0.1 mM ␤-mercaptoethanol and 1000 units of leukaemia inhibition factor
neurodevelopmental processes such as neuronal proliferation and (LIF)/ml. Undifferentiated cells were incubated at 37 ◦ C in an atmosphere with 1.5%
differentiation, gliogenesis and apoptosis through direct interfer- CO2 and 95% humidity. For culturing D3 cells under spontaneous differentiation, LIF
ence with the morphogenic activity of AChE, receptors and cell was removed from the medium culture and the CO2 concentration was increased to
5%.
signalling molecules and cytoskeletal proteins (Flaskos, 2012). D3 undifferentiated cells were seeded in P100 Petri dishes at a density of
Embryotoxicity can be studied in vitro using the Embryonic Stem 2 × 106 cells/dish (to record gene expression) or in 96-well plates at a density of
cell Test (EST). This methodology is successfully validated and, 2 × 105 cells/well (for cell viability tests and to test the effects on NTE enzymatic
as endpoints, it employs the relationships between the cytotoxi- activities). Afterwards, LIF was removed from the media in order to trigger differen-
tiation, and fresh CPF, CPO and TClP ranging between 10 and 1200 ␮M were added to
city induced by the assessed compound in 3T3 mouse fibroblasts
the media. The exposure performed under differentiation conditions was prolonged
to 12 h.
Similar experiments were performed by prolonging differentiation and expo-
sure for 3 days and by seeding cells at a density of 4 × 105 cells/dish in P100 Petri
1
In this manuscript we quote the abbreviations for the genes italicised with the dishes (to record gene expression) or 3.5 × 104 cells/well in 96-well plates well (for
first letter in the upper case and all the other letters in the lower case. Therefore, cell viability tests and to test the effects on AChE and NTE enzymatic activities). The
the term Ache (italicised) is used to quote the gene codifying for the protein acetyl- CPF, CPO and TClP concentrations ranged between 20 and 300 ␮M, the media of the
cholinesterase, while the term AChE (non-italicised with the upper and lower case in cells exposed to CPO and their correspondent controls were renewed daily, and the
the word) is used to quote the protein acetylcholinesterase or its enzymatic activity. media of the cells exposed to CPF and TClP was left unchanged for 3 days.
16 C. Estevan et al. / Toxicology Letters 217 (2013) 14–22

2.2.2. 3T3 fibroblast centrifugation (12,000 × g, 15 min, 4 ◦ C) was applied to obtain RNA in the upper
Non-tumourigenic 3T3 fibroblast cells Balb/c clon A31 cells were also obtained (colourless) phase. This phase was collected and transferred to a new Eppendorf
from the American Type Culture Collection (Rockville, MD, USA) and were grown on tube for RNA precipitation by adding 500 ␮l of isopropanol at −20 ◦ C, shaking for
monolayers on 75-mm plates in DMEM medium supplemented with 10% foetal calf 20 s and incubating for at least 3 h at −20 ◦ C. After RNA precipitation, the mixture
serum, 50 units of penicillin/ml and 100 ␮g streptomycin/ml. Cells were incubated was centrifuged (12,000 × g, 10 min, 4 ◦ C) and the pellet was washed with 1 ml
at 37 ◦ C in an atmosphere with 5% CO2 and 95% humidity until 95% confluence. ethanol (75%, v/v) at −20 ◦ C centrifuging (12,000 × g, 5 min, 4 ◦ C). The supernatant
Before starting, exposure cells were seeded on 96-well plates at a density of was finally discarded and the pellet was dried for 3 min at room temperature with
2 × 104 cells/well, and CPF, CPO and TClP ranging between 20 and 200 ␮M were further homogenisation in 25 ␮l of 0.1% (w/v) diethylpyrocarbonate (DEPC) water.
freshly added to the cell media by prolonging exposure to 12 h before testing cell Finally, the tube containing RNA was incubated at 55–60 ◦ C for 15 min and stored
viability. Similar experiments were done by prolonging exposure to 3 days, and by at −80 ◦ C until use. RNA was quantified and its purity was determined according to
seeding cells at a density of 1.5 × 104 cells/well and using CPF, CPO and TClP ranging the 260/280 nm optical density ratio by spectrophotometry (Biowave II WPA).
between 10 and 700 ␮M. In the case of cells exposed during 3 days the CPO content
was daily refreshed because it is extensively degraded (see Section 3.1). Media of 2.5.2. RNA retrotranscription
cells serving as control to CPO-exposed cells was also daily renewed. The media of The extracted RNA was reverse-transcribed using the Expand Reverse Tran-
TClP- and CPF-exposed cells and their respective controls were not renewed during scriptase kit and oligo-dT primers (Roche) according to the supplier’s indications.
the 3 days of exposure. The reaction mixture contained 10 ␮M polydT and 1 ␮g of RNA in a final volume
of 10.5 ␮l of DEPC water. This mixture was heated for 10 min at 65 ◦ C in a Veriti
2.3. Degradation of compounds in the culture system 96-well thermal cycler (Applied Biosystems) to bind polydTs to the tails of polyA
present in messenger RNAs. Afterwards, the following components were added to
It is well known that OPs can be degraded in mammal serum; the D3 and 3T3 the mixture: 2 ␮l of 100 mM dithiothreitol, 2 ␮l of 10 mM dNTPs, 4 ␮l of buffer for
culture media contained up to 15% of foetal calf serum. Therefore, in order to check Expand Reverse Transcriptase, 0.5 ␮l of RNAase inhibitor (Roche) and 1 ␮l (50 units)
the stability of CPF and CPO in culture media, we prepared solutions of 700, 400 and of RNA Expand Reverse Transcriptase. This resulting mixture was further heated at
200 ␮M CPF in fresh culture media for D3 cells and we incubated the solutions for 37 ◦ C for 60 min with a final 5-min step at 93 ◦ C in the thermal cycler.
72 h at 37 ◦ C by extracting aliquots every 24 h. The same procedure was applied for The quality of cDNA was checked by amplifying the cDNA for ␤-actin by PCR and
CPO, but 700, 500 and 100 ␮M solutions were used and aliquots were extracted at 1, by running the PCR product in the Agilent 2100 bioanalyzer (Agilent Technologies).
2, 4, 6, 8 and 24 h. One millilitre of each aliquot was acidified with 0.25 ml of 2 M HCl, It yielded a single DNA fragment of 120 bp corresponding to ␤-actin (Supplementary
and CPF and CPO were extracted with 2 ml of dichloromethane (containing 20 ␮M Data 3).
naphthol as an inner standard) for 30 min. At the end of the process, an organic
layer was separated from the watery layer by centrifugation at 3000 rpm for 5 min.
2.5.3. Quantitative real-time PCR
Finally, 2 ␮l of dichloromethane were injected into a GC–MS system for the further
Quantitative real-time PCR (qRT-PCR) was used to quantify the expression of
analysis of residual CPF, CPO and TClP.
the biomarker genes. All the genes were quantified using the StepOnePlus Real-Time
Chromatographic separation was performed with a ZB-Multiresidue-1 (Zebron)
PCR System (Applied Biosystems) equipment. The Pnpla6 expression was monitored
column with a length of 30 m, and inner diameter of 0.25 mm and a film thickness of
with the specific Taqman kit, while other genes were quantified with the Power SYBR
0.25 ␮m. The gas carrier was He, the injector temperature was 250 ◦ C, the total flow
Green kit (both supplied by Applied Biosystems) with the primers shown in Table 1.
was 24 ml/min and the injections mode was splitless. The temperature programme
For the Power SYBR Green analysis, 96-well reaction plates (Applied Biosystem)
included a gradient from 50 to 250 ◦ C with a rate of 40 ◦ C/min and a 2-min final step at
were utilised, and the following was added to each well: 10 ␮l of master mix, 0.9 ␮l
constant temperature. Under these conditions, the elution times for TClP, naphthol,
of forward primer 25 pmol/␮l, 0.9 ␮l of reverse primer 25 pmol/␮l, 6.2 ␮l of DEPC
CPO and CPF were 4.87, 5.09, 6.83 and 6.86 min, respectively (see Supplementary
water plus 2.0 ␮l of cDNA sample (0.1 ng/ml). The cDNA amplification consisted
Data 1 and 2 for examples of chromatograms).
in an initial step at 95 ◦ C lasting 10 min, followed by 40 cycles of 15 s denaturing at
Residual TClP, CPF and CPO was quantified by recording the mass spectrometric
95 ◦ C, 60 s at the respective annealing temperature (Table 1), 15 s at 95 ◦ C, plus a final
signal in the single ion mode for m/z = 144 (for the inner standard naphthol), 169
melting step of 60 s at 60 ◦ C. Table 1 displays the number of cycles (Ct) of each gene
and a197 (for TClP), 197 and 270 (for CPO) and 197 and 314 (for CPF). The area of
for which the PCR amplification efficiency was considered appropriate. The number
each peak (normalised against the inner standard area) was compared with the area
of Ct needed to quantify the expression of all markers genes never exceeded the
of the standard solutions of known concentrations prepared in water, which were
number of Ct expressed in Table 1.
extracted, chromatographed and quantified in exactly the same way as the samples
The Pnpla6 expression was recorded with 10.0 ␮l of the Taqman master mix,
incubated with the D3 cell culture media. The limits of detection recorded for these
1.0 ␮l of the Taqman assay mix, 2.0 ␮l of sample, plus 7.0 ␮l of DEPC water. The
conditions were 12, 10 and 16 ␮M for CPF, CPO and TClP, respectively.
procedure for the genes analysed by the specific Taqman kit consisted in an initial
step at 50 ◦ C for 2 min, plus 10 min at 95 ◦ C followed by 40 cycles of 15 s at 95 ◦ C and
2.4. Cell viability tests
60 s at 60 ◦ C.
Quantification was performed using 2−Ct calculations (Livak and Schmittgen,
Cell viability and cytotoxicity were assayed with the 3-(4,5-dimethylthiazol-
2001) with the Step-One software, v2.0.1 (Applied Biosystems). ␤-Actin was used
2-yl)-2,5-diphenyltetrazoliumbromide (MTT) test. The MTT test is based on the
as an invariant internal control for each sample. In all cases, qRT-PCR was run with
colorimetric determination of the formazan formed in the mitochondria from the
three independent biological replicates per condition, and the gene expression was
MTT used as a substrate. The amount of formazan formed is directly related to the
estimated in relation to the expression of the same gene in the control plate (not
amount of viable cells present in the media since it is an indication of both mito-
exposed to CPF, CPO or TClP).
chondrial integrity and the level of functionality of mitochondrial dehydrogenases.
D3 under differentiation and 3T3 were exposed to CPF, CPO and TClP for 12 h or
3 days, as described in Section 2.2. Afterwards, chemicals were removed and cells 2.6. Enzymatic activities
were incubated with 200 ␮l of MTT solution (1 mg/ml) for 3 h. After this period, MTT
was removed and cells were washed with phosphate buffered saline (PBS). Finally, 2.6.1. AChE enzymatic activity
200 ␮l dimethylsulphoxide/well were added to each well to lysate cells and to solve The D3 cells under differentiation were exposed for 3 days to CPF, CPO and
the formazan formed in the mitochondria. Formazan absorbance was determined in TClP, as described in Section 2.2. Afterwards, AChE enzymatic activity was recorded
a Beckman Coulter AD340 microplate reader by recording absorbance at 540 nm and according to a procedure based on Ellman’s procedure (Ellman et al., 1961). To that
by correcting the background absorbance with records at 690 nm. Each condition end, chemicals were removed and cells were gently washed with PBS and 200 ␮l
was assayed in 12 independent wells. Absence of cytotoxicity (100% viability) was of 1 mM acethylhtiocholine with 0.25 mM 5,5 -dithiobis (2-nitrobenzoic acid) in
attributed to the controls, and the results were presented as a percentage of these 0.1 M phosphate buffer, pH 7.4, were added to each well to record increments of
controls. EC20 , EC50 and EC80 were estimated as the concentrations of CPF, CPO or absorbance at 405 nm for 30 min in a Beckman Coulter AD340 microplate reader.
TClP causing reductions in cell viability of 20%, 50% and 80%, respectively. AChE activity was expressed as the percentage of activity recorded in the cells not
exposed to CPF, CPO or TClP. Each condition was assayed with 12 independent bio-
2.5. Molecular biology logical replicates. IC50 was estimated as the concentration of CPF, CPO or TClP that
caused a reduction in AChE enzymatic activity by 50%.
2.5.1. RNA extraction
Cells were exposed to CPF, CPO and TClP for 12 h or 3 days as described in 2.6.2. NTE enzymatic activity
Section 2.2. Afterwards cells were trypsinised and pelleted by centrifugation in NTE enzymatic activity was determined as previously described by Pamies
a 1-ml Eppendorf tube. One millilitre of Tripure isolation solution (guanidinium et al. (2010b) as the phenyl valerate (PV) hydrolysing activity resistant to non-
thiocyanate) was added to the Eppendorf tube at room temperature with further neuropathic OP paraoxon and sensitive to neuropathic OP mipafox. B activity was
shaking for 10 s to detach cells and pipetting to mix all the components. Once the defined as the PV hydrolysing activity in the samples pre-incubated for 30 min at
mixture was homogeneous, it was kept for 5 min at room temperature to favour 37 ◦ C with 40 ␮M paraoxon. C activity was defined as the PV hydrolysing activity in
the cellular lysate. Afterwards, 200 ␮l of chloroform were added with shaking the samples pre-incubated with 40 ␮M paraoxon plus 250 ␮M mipafox. Therefore,
until a pink colour appeared. Then after another 10-min rest at room temperature, NTE activity was calculated as B–C.
C. Estevan et al. / Toxicology Letters 217 (2013) 14–22 17

Table 1
Primers sequences and annealing temperatures used for qRT-PCR determinations. It is also displayed the maximum number of amplification cycles allowed for appropriate
quantification.

Gene 5 –3 -oligo 3 –5 -oligo T (◦ C) Maximum Ct

ˇ-actin TGGGAATGGGTCAGAAGGAC TGAAGCTGTAGCCACGCTCG 62 25


Afp GCTGCAAAGCTGACAACAAG GCTTGTTGCCTGGAGGTTTC 63 34
Ache TGAACCTGAAGCCCTTAGAGGTG CCAGAGTATCGGTGGCGCTG 61 32
Flk1 CCGAGGCCACAGACTCCCTGCTT CAGCCAGACAGACAGTGGGATGGTC 61 21
Mhc CCAAACAGTGTCTGCTCTCCACCGG TTTCGCTCGTTGGGAATGATGCA 67 29
Nanog TCTCTCAGGCCCAGCTGTGT CTCAGGTTCAGAATGGAGGAGAGT 63 28
Nefm CAGCAGCTACCAGGACACCATC CCAGGGCCATCTTGACGTTA 64 26
Nes GCTTTCCTGACCCCAAGCTG GGCAAGGGGGAAGAGAAGGA 61 32
Oct4 GCCAGACCACCATCTGTCGCTT CACCAGGTCTCCGATTTGCA 62 25

The D3 cells under differentiation were exposed for either 12 h or 3 days to CPF, the cytotoxicity induced by CPF, CPO and TClP. For CPO, cytotoxicity
CPO and TClP, as described in Section 2.2. Afterwards, chemicals were removed and against D3 and 3T3 cells was similar after 3 days of exposure.
cells were gently washed with PBS before exposing cells for 30 min at 37 ◦ C to either
40 ␮M paraoxon or 40 ␮M paraoxon plus 250 ␮M mipafox, both in PBS. Paraoxon
and mipafox were removed and 100 ␮l/well of 7.5 mM PV in PBS were added by 3.3. Changes in the expression of the gene markers of
prolonging the incubation at 37 ◦ C by an additional 60-min period. The reaction was differentiation
stopped by adding 100 ␮l/well of 4% SDS with 6.15 mM 4-aminoantypirine in 50 mM
TRIS–1 mM EDTA buffer (pH 8.0). After 15 min at room temperature, 50 ␮l/well of 1% The D3 cells under differentiation and 3T3 cells were exposed for
(v/v) potassium ferrocyanide in water were added by leaving the mixture for 10 min
at 37 ◦ C. The released phenol was quantified by recording absorbance at 510 nm and
either 12 h or 3 days to several concentrations of CPF, CPO and TClP,
by comparing with the absorbance of the phenol standards. Six independent wells
were used to test each experimental condition. NTE activity was expressed as the
percentage of activity recorded in the cells not exposed to CPF, CPO or TClP. IC50
was estimated as the concentration of CPF, CPO or TClP that reduced NTE enzymatic
activity by 50%.

3. Results

3.1. Degradation of OPs in cell culture media

Certain OPs can be hydrolysed by phosphotriesterases, such as


paraoxonase, and albumin contained in mammal serum, which is
specially probed in the case of CPO (Sogorb et al., 2008). We checked
the stability of CPF and CPO in the D3 cell culture media (the media
with the highest foetal serum concentration) by incubating CPF and
CPO with cell media and by analysing the residual concentrations
of both OPs by gas chromatography with mass spectrometry, as
described in Section 2.3. No significant decreases were noted in the
residual CPF concentration after 72 h of incubation at 37 ◦ C (Fig. 1).
However, the residual CPO concentration lowered over time (Fig. 2)
to become 20%, 10% and 0% of the initial concentration after 8 h
of incubation with 700, 500 and 100 ␮M CPO, and was negligi-
ble in all cases after 24 h of incubation. In all cases, the drop in
the residual CPO correlated with the appearance of a peak of TClP
(Supplementary Material 2), which supports the idea of a hydrolysis
of CPO by paraoxonase and albumin of foetal serum. No TClP peak
was detected after the incubation of CPF (Supplementary Material
1). These data led us to perform further experiments in the cell
media which were changed to therefore refresh the CPO content
every 24 h, which was not considered necessary for CPF.

3.2. Cytotoxicity of CPF, CPO and TClP

The D3 cells under differentiation and 3T3 cells were exposed


for either 12 h or 3 days to several concentrations of CPF, CPO and
TClP, as described in Section 2.2. The viability of the resulting cul-
tures was further assayed by the MTT test, as described in Section
2.4. The results are displayed in Table 2. The cytotoxicity induced
Fig. 1. Residual organophosphorus concentration after incubation with D3 cell cul-
after 12 h of exposure was very low, and the EC50 for both cell lines ture media. CPF (upper panel) and CPO (lower panel) were incubated at 37 ◦ C with
was always higher than 600 ␮M. CPF and CPO induced a similar D3 culture media for up to 72 h in the case of CPF or 24 h in the case of CPO. The CPF
cytotoxicity to 3T3 cells, although the cytotoxicity of TClP induced and CPO residual concentrations were calculated using gas chromatography cou-
to these cells was lower than that induced by the parental com- pled to mass spectrometry after extracting the compounds with dichloromethane
as described in Section 2.3. It is displayed mean ± s.d. for three independent samples.
pounds. However, the cytotoxicity of CPO and TClP against D3 cells Supplementary Materials 1 and 2 display examples of chromatograms. The initial
was similar and lower than the CPF cytotoxicity against these same CPF concentrations (upper panel) were 700 (); 400 () and 200 () ␮M. The initial
cells. Remarkably, D3 cells were more susceptible than 3T3 cells to CPO concentrations (lower B) were 700 (); 500 () and 100 () ␮M.
18 C. Estevan et al. / Toxicology Letters 217 (2013) 14–22

Table 2
Effect of CPF, CPO and TClP on cellular viability. Cells (12 different biological replicates per each condition) were exposed to different concentrations of each compound as
described in Section 2.2 and afterwards the viability was assayed using the MTT test as described in Section 2.4. It is displayed the effective concentration (EC) needed to
cause viability reductions of 20%, 50% and 80%. The figures in square brackets represent the individual ECs obtained in the different n independent experiments performed
in each case. Each independent experiment was performed assaying twelve biological replicates.

EC20 (␮M) EC50 (␮M) EC80 (␮M)

D3 mouse embryonic stem cells


CPF 12 h 200 [200, 180, 220] 610 [640, 570, 624] 970 [1000, 910, 1010]
3 days 20 [15, 25, 20] 140 [100, 182, 138] 250 [260, 200, 240]
CPO 12 h 820 [860, 770, 874] >1200 [>1200, n = 3] >1200 [>1200, n = 3]
3 days 54 [47, 64, 51] 200 [146, 230, 224] >300 [>300, n = 3]
TClP 12 h >1200 [>1200, n = 3] >1200 [>1200, n = 3] >1200 [>1200, n = 3]
3 days 11 [12, 9, 12] 190 [151, 242, 177] >300 [>300, n = 3]

3T3 mouse fibroblasts


CPF 12 h 390 [400, 380] >1000 [>1000, n = 2] >1000 [>1000, n = 2]
3 days 48 [52, 44] 340 [320, 360] >400 [>400, n = 2]
CPO 12 h 320 [320, n = 2] >2000 [>2000, n = 2] >2000 [>2000, n = 2]
3 days 91 [67, 115] 200 [180, 220] 290 [300, 280]
TClP 12 h 1200 [1230, 1170] >2000 [>2000, n = 2] >2000 [>2000, n = 2]
3 days 260 [290, 230] 620 [610, 630] 970 [1000, 940]

as described in Section 2.2. Afterwards, RNA was extracted, retro-


transcribed to cDNA and the expressions of myosin heavy chain
(Mhc), nanog homeobox (Nanog), neurofilament medium polypep-
tide (Nefm), Nes, Oct4, Pnpla6, acetylcholinesterase (Ache), Afp and
foetal liver kinase 1 (Flk1) were assayed by qRT-PCR, as described
in Section 2.5. The results are displayed in Figs. 2–4. Figs. 2–4 dis-
play the statistically significant alterations (by at least p < 0.05) in
the expression of the biomarker genes together with the grade of
induced variation.

3.3.1. Changes in the CPF-induced gene expression


After a 12-h exposure, all the genes except Afp were down-
regulated, while the 3-day exposure up-regulated Flk1, Mhc, Nefm,
Ache, Oct4 and Nes, with slight variations or slight down-regulations
of Nanog, Afp and Pnpla6 (Fig. 2A). After the 12-h exposure to 10 ␮M
CPF, the expressions of Flk1, Oct4 and Ache statistically reduced
by 40%, 80% and 80%, respectively, while this same concentration
increased the expression of Pnpla6 by 70% (Fig. 2). Higher concen-
trations (100 and 200 ␮M) also caused alterations to the expression
of all the other tested genes (Fig. 2).
The lowest concentration to cause significant alterations in the
expressions of 6 of the 9 tested genes after the 3-day exposure was
20 ␮M, which was able to induce an overexpression of the genes
of a range between 3 and 29 times the expression of the control
cells (Fig. 2). None of the tested concentrations was able to induce
significant changes in the expressions of Nes and Ache (Fig. 2). The
overexpression of the altered genes reached a maximum at 50 ␮M
CPF, while higher concentrations lowered the expression (Fig. 2B).
The 3-day exposure to CPF led to an increased expression of the
pluripotency (Oct4 and Nanog) and mesoderm (Flk1) markers, while
the 12-h exposure caused the opposite effect (Fig. 2).

3.3.2. Changes in the CPO-induced gene expression


The lowest CPO concentration capable of causing statistically
significant alterations in some of the tested batteries of genes was
100 ␮M, which was able to increase the expression of Flk1 by 180%
while, at the same time, it reduced the expression of Nes (Fig. 3).
Higher CPO concentrations (300 ␮M) brought about increases in
the expressions of Nefm and Afp, while 600 ␮M and 700 ␮M were
Fig. 2. Effect of CPF on the gene expression of biomarkers of differentiation. D3
mouse embryonic stem cells under spontaneous differentiation were exposed dur-
required to statistically alter the expressions of Ache and Oct4,
ing 12 h (upper panel) or 3 days (lower panel) to several CPF concentrations. At the respectively (Fig. 3).
end of the exposure RNA was extracted, retrotranscribed and the expression of the The expressions of Mhc and Nanog remained unaltered after the
biomarker genes assayed by qRT-PCR and expressed regarding control non-exposed 12-h exposure, but the expression of these genes increased by 8 and
cells as described in Section 2.5. It is displayed mean ± s.d. for 3 independent bio-
6 times, respectively, after the 3-day exposure to 25 ␮M CPO. A sim-
logical replicates assayed per each condition. Statistical analyses were performed
comparing the expression of each point with expression of control cells by t Student ilar increase (5 times) in the expression of Flk1 was also observed
test. at this same concentration (Fig. 3).
C. Estevan et al. / Toxicology Letters 217 (2013) 14–22 19

Fig. 3. Effect of CPO on the gene expression of biomarkers of differentiation. D3 Fig. 4. Effect of TClP on the gene expression of biomarkers of differentiation. D3
mouse embryonic stem cells under spontaneous differentiation were exposed dur- mouse embryonic stem cells under spontaneous differentiation were exposed dur-
ing 12 h (upper panel) or 3 days (lower panel) to several CPO concentrations. At the ing 12 h (upper panel) or 3 days (lower panel) to several TClP concentrations. At the
end of the exposure RNA was extracted, retrotranscribed and the expression of the end of the exposure RNA was extracted, retrotranscribed and the expression of the
biomarker genes assayed by qRT-PCR and expressed regarding control non-exposed biomarker genes assayed by qRT-PCR and expressed regarding control non-exposed
cells as described in Section 2.5. It is displayed mean ± s.d. for 3 independent bio- cells as described in Section 2.5. It is displayed mean ± s.d. for 3 independent bio-
logical replicates assayed per each condition. Statistical analyses were performed logical replicates assayed per each condition. Statistical analyses were performed
comparing the expression of each point with expression of control cells by t Student comparing the expression of each point with expression of control cells by t Student
test. test.

3.3.3. Changes in the TClP-induced gene expression


The only gene that was up-regulated after exposure to TClP for we studied the alterations on the enzymatic expression caused by
12 h was Afp (Fig. 4). TClP exhibited a potent effect on gene expres- exposure to CPF, CPO and TClP for 3 days. CPO inhibited AChE with
sion since 10 ␮M was capable of down-regulating the expressions a power of 17 times higher than CPF (Table 3), which was expected
of Nanog, Nefm, Nes, and Pnpla6 by between 50% and 90% after a
12-h exposure (Fig. 4). Table 3
Pnpla6 was the only gene to be underexpressed after exposure Effect of CPF, CPO and TClP on enzymatic activities of target esterases. D3 mouse
to TClP for 3 days, although the effects noted were statistically sig- embryonic stem cells under differentiation were exposed to CPF, CPO and TClP as
nificant at lower concentrations (25 ␮M) than the concentrations described in Section 2.2 and afterwards the AChE and NTE enzymatic activities were
recorded as described in Section 2.6. It is displayed the concentration needed to
needed to alter the expression of other overexpressed genes, such
cause an inhibition of 50% of each activity for the indicated time of exposure. AChE
as Flk1, Ache and Mhc, whose expression increased by between 13 could not be detected in control D3 cells differentiated during 12 h. Each AChE and
and 47 times (Fig. 4). NTE determination was assayed with 12 and 6 independent biological replicates,
respectively. The figures in square brackets represent the individual ECs obtained
in the different n independent experiments performed in each case.
3.4. Effect of CPF, CPO and TClP on AChE and NTE enzymatic
activities AChE (3 days) NTE (12 h) NTE (3 days)
IC50 (␮M) IC50 (␮M) IC50 (␮M)

3.4.1. Effects on AChE CPF 35 [30, 40] 200 [200, 200] >135 [>135, >135]
AChE enzymatic activity was not detected in the control cells CPO 2 [2, 2] 6 [<10, 6] 35 [<50, 35]
TClP >120 [>120, >120] 9 [<10, 9] 280 [270, 290]
by the experimental procedure described in Section 2.6. Therefore,
20 C. Estevan et al. / Toxicology Letters 217 (2013) 14–22

because CPO is more toxic in vivo than CPF. TClP displayed a certain at least on the basis of comparing relative cytotoxicity to embry-
capability to inhibit AChE, but with the lowest potency. onic/differentiated cells.

4.2. Expression of biomarkers of differentiation


3.4.2. Effects on NTE enzymatic activity
Once again, CPO was seen to be a more potent inhibitor of NTE The battery of genes considered for testing changes in the differ-
than CPF, although the differences in inhibitory power of CPF and entiation of D3 embryonic stem cells include (Figs. 2–4): markers
CPO were lower for 3 days than for 12 h (Table 3). TClP exhibited a of mesoderm (Flk1, Mhc and Pnpla6); markers of ectoderm (Nefm,
surprising capability to inhibit NTE with a similar power to CPO for Nes and Ache), markers of endoderm (Afp) and markers of pluripo-
the 12-h exposure, while the inhibitory potency of TClP was similar tency (Oct4 and Nanog). These genes codify for proteins involved
to that exhibited by CPF after a 3-day exposure (Table 3). The IC50 either in the development or maintenance of vascular (Flk1, Ache
for NTE after 3 days of exposure to CPO was 17 times higher than and Pnpla6), nervous (Nefm and Nes) and muscular (Mhc) systems
IC50 for AChE with the same chemical (Table 3). or in the distribution of nutrients (fatty acids) and other factors
(nickel, copper) in the embryo (Afp). The intensity of the induced
4. Discussion changes was also remarkable, ranging between under-expressions
of up to 99 times and over-expressions of up to 400 times the
In this paper, we demonstrate that CPF and its metabolites, CPO expressions recorded in the non-exposed control cells. Although
and TClP, are more cytotoxic for embryonic stem cells than for this work did not aim to study embryotoxicity mechanisms of the
fibroblasts cells, and that all three compounds are able to signifi- analysed chemicals, it is expected that all these changes working
cantly alter the in vitro expressions of nine different genes (most of together may potentially cause severe disturbances in differentia-
which have a proven capability to detect exposure to embryotox- tion, especially if these alterations are combined with impairments
icants) belonging to all three main cellular lineages in D3 mouse in placental function, such as pregnancy maintenance, trophoblast
embryonic stem cells after exposures lasting 12 h or 3 days. In addi- differentiation and membrane drug transport, as recently reported
tion, alterations to expressions were detected at concentrations for in vitro (Ridano et al., 2012).
which low or no cytotoxicity was expected. Among all the tested genes, Afp, Nes and Pnpla6 have
already been shown their capacity to discriminate among the
embryotoxicity powers of 5-fluorouracil (strong embryotoxicant),
4.1. Cytotoxicity 5,5-diphenylhydantoin (weak embryotoxicant) and saccharin
(non-embryotoxicant) in a modified EST protocol (Romero et al.,
The substances tested in this study have been shown to be more 2011). Afp has also been demonstrated to be an extraordinary sen-
cytotoxic for the embryonic D3 stem cells than for the differentiated sitive biomarker of exposure to 5-fluorouracil since the expression
3T3 fibroblast cells at the two exposure periods tested (Table 2). of this gene was severely altered after exposing differentiating D3
These results are consistent with some in vivo studies (Atterberry cells at concentrations of several orders of magnitude below those
et al., 1997) showing more susceptibility to CPF in young rats than causing cytotoxicity (Pamies et al., 2010a). Therefore, all these data
in adult rats, probably due to the reduced metabolic capacity to support the hypothesis that the detected alterations might actually
hydrolyse the oxon form of OPs or other mechanisms to detoxify entail embryotoxicity in developing embryos exposed to CPF, CPO
chemicals. or TClP.
CPF and other OPs have showed to cause cytotoxicity through Changes in gene expression in cells exposed to CPF for 12 h
induction of oxidative stress with concurrent DNA damage and always displayed an overexpression or underexpression; however,
apoptosis (Lu et al., 2012; Lee et al., 2012; Ojha et al., 2011). Never- while the changes reported in the cells exposed to CPF for 3 days
theless, in the specific case of CPF other cytotoxicity mechanisms showed overexpressions for certain genes at low concentrations,
have been also described, as the extracellular signal-regulated with further decreased expressions at higher concentrations. This
kinase inhibition in primary rat hippocampal neurons (Tan et al., might be explained by the cytotoxicity basis. Indeed, the highest
2009) or the up-regulation of protein kinase C in PC12 cells (Slotkin concentration tested for the 12-h exposure for all three compounds
and Seidler, 2009). Nevertheless, 12 h of exposure is too short time was expected to cause low or no cytotoxicity, while the highest
to develop the cytotoxic effects through the specific mechanisms concentrations employed in the 3-day exposure experiments came
and our results are probably the reflection of oxidative stress, while close to EC50 . Therefore, certain cytotoxicity may be expected for
we cannot refuse the possibility of other effects, as apoptosis, when the mid dose-response curves.
cells were exposed during three days. The lowest in vitro CPF concentration to cause statistically sig-
The cytotoxicity of CPF and TClP was higher for D3 cells (the nificant alterations in the expression of the tested genes ranged
embryonic differentiating cells model) than for 3T3 cells (the non- between 10 and 200 ␮M for the 12-h exposure (Fig. 2), which is
differentiating cells model) (Table 2). The validation study of the between 3 and 61 times lower than the estimated EC50 for the
Embryonic Stem cell Test, sponsored by the European Centre for viability of D3 cells. Likewise for the 3-day exposure, the lowest
Validation of Alternative Methods, classifies the embryotoxicity of CPF concentration causing significant alterations in gene expres-
chemicals into three categories according to the following criteria: sion ranged between 20 and 50 ␮M, which is between 2.8 and 7
(a) strong embryotoxicants are chemicals causing toxic effects to times lower than the estimated EC50 for the viability of D3 cells.
the embryo of all the tested species with very low, or absence of, The same analysis showed that CPO is capable of significantly alter-
maternal toxicity; (b) weak embryotoxicants are chemicals causing ing the expression of certain genes at concentrations of between
toxic effects to the embryo of the multiple tested species with very 12 (for 12-h exposure) and 8 (for 3-day exposure) times lower
low, or absence of, maternal toxicity; and (c) non-embryotoxicants than the corresponding EC50 records for the viability of D3 cells
are those chemicals causing toxic effects to the embryo of mul- (Table 2 and Fig. 3). Similarly, concentrations of TClP of between
tiple tested species with severe maternal toxicity (Brown, 2002). 12 and 120 times lower than the EC50 for the viability of D3 cells
Therefore, although the main goal of this work was not to set a also significantly altered the expression of several genes included
classification for CPF, CPO and TClP, our results (more cytotoxic- in the array (Tables 2 and 4). Table 2 and Fig. 4 in conclusion, all
ity for embryonic-differentiating cells than for differentiated cells) these data suggest that the alterations detected in gene expression
suggest that they may match the weak embryotoxicants category, might address alterations in differentiation mechanisms, but not
C. Estevan et al. / Toxicology Letters 217 (2013) 14–22 21

the general phenomena relating to severe loss in cell viability of 1999). If we use the same toxicokinetic parameters as those above
the culture. to estimate the systemic concentration needed to reach the acute
reference dose, we can conclude that it may be around 0.5 ␮M.
4.3. Alterations in the enzymatic activities of AChE and NTE It is remarkable that the above-stated reference values (TLV and
acute reference dose) were obtained on the basis of statistically
Exposure of cells to CPF, CPO and TClP during differentiation led significant alterations of blood AChE and with a safety factor of
to severe reductions in the enzymatic activities of AChE and NTE 10. However, statistically significant alterations of AChE as regards
(Table 3). As expected, CPO was a more potent inhibitor of AChE controls do not necessarily mean clinical signs since they do not
and NTE than CPF. It is also remarkable to note than the IC50 for appear below the 70% inhibition of brain cholinesterase (Clegg and
NTE was always higher than that for AChE, which was expected van Gemert, 1999). Therefore, exposure scenarios where occupa-
given the low capability of CPF to induce delayed polyneuropathy tional hygiene is not strictly observed may cause CPF exposures
(Albers et al., 2004). The differences in the IC50 for NTE inhibition which, without clinical maternal symptoms because AChE is not
after the 12-h and 3-day exposures were not as high as expected; high enough, reach similar systemic concentrations to those dis-
even for CPO, the IC50 for the 3-day exposure was higher than for played in Figs. 2–4, and cause significantly altered gene expressions.
the 12-h exposure, which is not logical for irreversible time- and The highest urinary excretion of TClP reported for workers
concentration-dependent inhibition (Table 3). We cannot provide involved in manufacturing CPF was 325 ␮g/day (Chen et al., 2011).
a clear explanation for this phenomenon, but it might be related to By assuming 1 l of urine/day and the above-stated toxicokinetic
a mixture of effects which simultaneously affect cells, such as the and physiological parameters (70 kg, 38 l of corporal volume and no
chemical inhibition caused by CPO through a direct effect on the bioaccumulation), the estimated systemic TClP concentration could
protein and the effect of CPO on the expression of Pnpla6. be around 43 nM. This figure is several orders of magnitude lower
Surprisingly, TClP also displayed a certain inhibition power of than that which caused significantly altered gene expressions. Thus,
enzymes. This inhibition cannot be accounted for as it can for CPF the risk of embryotoxicity due to CPF exposure is not expected
and CPO because this compound has not been reported to be an through its metabolite. Regarding CPO, it is also remarkable to note
irreversible inhibitor of esterases. The observed effects might be that gene expressions alter only at concentrations that cause severe
due to a mixture of complex effects ranging between some type AChE inhibition and, therefore, severe maternal toxicity.
of interaction between the protein and the compound that has not
been described to date and alterations in the level of expression of
4.5. Final remarks
both the genes reported in Fig. 4.
In all cases, the greatest effects of CPO and CPF on gene expres-
The information available on the embryotoxicity of CPF is con-
sion were reached at concentrations at which a high inhibition of
tradictory. Most of the studies performed, which follow the OECD
both esterases was expected. With CPF, the lowest concentrations
Guidelines for development or toxicity to reproductions, have
causing alterations in gene expression were lower than, or in the
reported embryotoxicity only at doses with high maternal toxic-
same order of magnitude as, IC50 for AChE and NTE (Tables 3 and 4).
ity (Eaton et al., 2008; FAO/WHO, 1999)]. However, other studies
However, the lowest CPO concentrations to alter gene expression
have reported that subcutaneously injections of rats with CPF dur-
were much higher, or in the same order of magnitude as, IC50 for
ing different gestational periods irreversible reduce the dopamine
AChE and NTE (Tables 3 and 4). These findings suggest that alter-
contents in both the cerebral cortex and hippocampus, two key
ations in gene expression do not relate to esterase inhibition. It is
brain regions involved in learning and memory (Chen et al., 2011).
also remarkable to note that EC50 for the viability in D3 cells is far
Furthermore, epidemiological studies have not proved conclusive
from the IC50 for the inhibition of AChE and NTE, suggesting that
since Mink et al. (2012) have recently reviewed four independent
cytotoxicity is not caused by a mechanism mediated by inhibition
epidemiological cohort studies, and found no causal association
of esterases.
between CPF exposures during pregnancy and measures of foetal
growth.
4.4. Considerations for the risk assessment of embryotoxicity of
The above-stated discrepancies might be explained on the basis
CPF
of the exposure route. Indeed, the OECD Guidelines exclusively con-
sider oral exposure (Estevan et al., 2011), where the first passage of
The correlation found between in vivo exposures and in vitro
administered CPF through the liver of the mother would massively
doses is always hard to estimate. Nonetheless, Albers et al. (2007)
bioactivate CPF to CPO, which is a more powerful AChE inhibitor and
have suggested that a person occupationally exposed to the Thresh-
would cause clinical cholinergic signs before reaching embryotoxic
old Limit Value (TLV) (200 ␮g/m3 ) at a respiratory rate of 10 m3
concentrations. However, other animal studies have considered
air/working day via the inhalatory route, by assuming 100% of
different routes, such as subcutaneous exposure, where less CPF
pulmonary absorption, would be exposed to a systemic dose of
reaches the liver and CPO appears more slowly than in oral expo-
2000 ␮g CPF/day. Assuming that CPF is distributed according to a
sure, which allows, in this case, higher and potentially toxic CPF
monocompartimental model, which seems logical since OPs do not
concentrations to reach the embryo with no significant maternal
bioaccumulate (WHO, 1986), by contemplating a corporal volume
toxicity signs. Similar situations may appear during human occu-
of 38 l for a person weighing 70 kg (Lehman-McKeeman, 2009), the
pational exposure, which can essentially be inhalatory or dermal,
expected systemic concentration of CPF would be around 0.2 ␮M.
therefore entailing greater risk.
On the other hand, USEPA estimates that adult exposures to CPF
Finally, it is remarkable that the reported effect of CPF might
could be around 11.4 ␮g/kg/day (Cochran, 2002). If we assume
address it, but not its metabolites CPO or TClP, which despite
the considerations as above, then the systemic CPF concentra-
also displaying embryotoxic hazards, do not appear for compatible
tions could be around 0.1 ␮M. To conclude, the estimated systemic
exposures with mother survival.
concentration in both cases is still far from the lowest CPF con-
centrations causing significant alterations to gene expressions for
exposures of either 12 h (10 ␮M) or 3 days (20 ␮M). 5. Conclusions
Other studies have considered that the acute reference dose
for CPF is 0.1 mg/kg, estimated based on inhibition of blood The short- and mid-term exposure of embryonic stem cells
cholinesterases and by assuming a safety factor of 10 (FAO/WHO, to CPF and its metabolites significantly modify the expression of
22 C. Estevan et al. / Toxicology Letters 217 (2013) 14–22

marker genes of differentiation during early in vitro differentiation Genschow, E., Spielmann, H., Scholz, G., Pohl, I., Seiler, A., Clemann, N., Bremer, S.,
at concentrations that do not affect viability. However, alterations Becker, K., 2004. Validation of the embryonic stem cell test in the international
ECVAM validation study on three in vitro embryotoxicity tests. Alternatives to
to marker genes of differentiation occur at concentrations which Laboratory Animals 32, 209–244.
cause severe AChE inhibition. Therefore, embryotoxicity is possi- Jokanovic, M., Kosanovic, M., 2010. Neurotoxic effects in patients poisoned with
ble only under exposure scenarios which allow low bioactivation organophosphorus pesticides. Environmental Toxicology and Pharmacology 29,
195–201.
of CPF to avoid neurotoxicity. Lee, J.E., Park, J.H., Shin, I.C., Koh, H.C., 2012. Reactive oxygen species regulated
mitochondria-mediated apoptosis in PC12 cells exposed to chlorpyrifos. Tox-
Conflict of interest statement icology and Applied Pharmacology 263 (September (2)), 148–162, http://dx.
doi.org/10.1016/j.taap.2012.06.005 (Epub 2012 June 17).
Lehman-McKeeman, L.D., 2009. Absorption, distribution and excretion of toxicants.
The authors report no conflicts of interest. In: Klaassen, K. (Ed.), Casarett & Doull’s Toxicology the Basic Science of Poisons.
McGraw Hill, Kansas. USA, pp. 31–159.
Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using
Appendix A. Supplementary data real-time quantitative PCR and the 2−CT method. Methods 25, 402–408.
Lu, X.T., Ma, Y., Wang, C., Zhang, X.F., Jin da, Q., Huang, C.J., 2012. Cytotoxicity and
Supplementary data associated with this article can be DNA damage of five organophosphorus pesticides mediated by oxidative stress
in PC12 cells and protection by vitamin E. Journal of Environment Science and
found, in the online version, at http://dx.doi.org/10.1016/
Health. Part B: Pesticides 47 (5), 445–454.
j.toxlet.2012.11.026. Mink, P.J., Kimmel, C.A., Li, A.A., 2012. Potential effects of chlorpyrifos on fetal growth
outcomes: implications for risk assessment. Journal of Toxicology and Environ-
mental Health. Part B: Critical Reviews 15, 281–316.
References
Ojha, A., Yaduvanshi, S.K., Pant, S.C., Lomash, V., Srivastava, N., 2011. Evaluation of
DNA damage and cytotoxicity induced by three commonly used organophos-
Albers, J.W., Garabrant, D.H., Schweitzer, S.J., Garrison, R.P., Richardson, R.J., Berent, phate pesticides individually and in mixture, in rat tissues. Environmental
S., 2004. The effects of occupational exposure to chlorpyrifos on the peripheral Toxicology July, http://dx.doi.org/10.1002/tox.20748.
nervous system: a prospective cohort study. Occupational and Environmental Ostrea, E.M., Morales, V., Ngoumgna, E., Prescilla, R., Tan, E., Hernandez, E., Ramirez,
Medicine 61, 201–211. G.B., Cifra, H.L., Manlapaz, M.L., 2002. Prevalence of fetal exposure to envi-
Albers, J.W., Garabrant, D.H., Mattsson, J.L., Burns, C.J., Cohen, S.S., Sima, C., Garrison, ronmental toxin as determined by meconium analysis. Neurotoxicology 23,
R.P., Richardson, R.J., Berent, S., 2007. Dose-effect analyses of occupational chlor- 329–339.
pyrifos exposure and peripheral nerve electrophysiology. Toxicological Sciences Pamies, D., Vicente-Salar, N., Sogorb, M.A., Roche, E., Reig, J.A., 2010a. Specific effect
97, 196–204. of 5-fluorouracil on alpha-fetoprotein gene expression during the in vitro mouse
Atterberry, T.T., Burnett, W.T., Chambers, J.E., 1997. Age-related differences in embryonic stem cell differentiation. International Journal of Toxicology 29,
parathion and chlorpyrifos toxicity in male rats: target and nontarget esterase 297–304.
sensitivity and cytochrome P450-mediated metabolism. Toxicology and Applied Pamies, D., Reig, J.A., Vilanova, E., Sogorb, M.A., 2010b. Expression of Neuropathy
Pharmacology 147, 411–418. Target Esterase (NTE) in mouse embryonic stem cells during differentiation.
Brown, N.A., 2002. Selection of test chemicals for the ECVAM international validation Archives of Toxicology 84, 481–491.
study on in vitro embryotoxicity tests. European Centre for the Validation of Ridano, M.E., Racca, A.C., Flores-Martín, J., Camolotto, S.A., de Potas, G.M., Genti-
Alternative Methods. Alternatives to Laboratory Animals 30, 177–198. Raimondi, S., Panzetta-Dutari, G.M., 2012. Chlorpyrifos modifies the expression
Cañadas, F., Cardona, D., Dávila, E., Sánchez-Santed, F., 2005. Long-term neurotox- of genes involved in human placental function. Reproductive Toxicology 33,
icity of chlorpyrifos: spatial learning impairment on repeated acquisition in a 331–338.
water maze. Toxicological Sciences 85, 944–951. Romero, A.C., Vilanova, E., Sogorb, M.A., 2011. Shortening and improving the embry-
Chen, X.P., Wang, X., Dong, J.Y., 2011. Different reaction patterns of dopamine con- onic stem cell test through the use of gene biomarkers of differentiation. Journal
tent to prenatal exposure to chlorpyrifos in different periods. Journal of Applied of Toxicology 2011, 286034.
Toxicology 31, 355–359. Ruiz-Muñoz, A.M., Nieto-Escamez, F.A., Aznar, S., Colomina, M.T., Sanchez-Santed,
Clegg, D.J., van Gemert, M., 1999. Determination of the reference dose for chlorpyri- F., 2011. Cognitive and histological disturbances after chlorpyrifos exposure and
fos: proceedings of an expert panel. Journal of Toxicology and Environmental chronic A␤ (1–42) infusions in Wistar rats. Neurotoxicology 32, 836–844.
Health. Part B: Critical Reviews 2, 211–255. Slotkin, T.A., Levin, E.D., Seidler, F.J., 2006. Comparative developmental neurotoxicity
Cochran, R.C., 2002. Appraisal of risks from nonoccupational exposure to chlorpyri- of organophosphate insecticides: effects on brain development are separable
fos. Regulatory Toxicology and Pharmacology 35, 105–121. from systemic toxicity. Environmental Health Perspectives 114 (5), 746–751.
Council of the European Communities, 1991. Council Directive 91/414/EEC of 15 Slotkin, T.A., Seidler, F.J., 2009. Protein kinase C is a target for diverse developmental
July 1991 concerning the placing of plant protection products on the market. neurotoxicants: transcriptional responses to chlorpyrifos, diazinon, dieldrin and
Official Journal 230, 1–32, Available from: http://eur-lex.europa.eu/LexUriServ/ divalent nickel in PC12 cells. Brain Research 1263 (March), 23–32.
LexUriServ.do?uri=CELEX:31991L0414:EN:NOT (last accessed: 20.09.12). Sogorb, M.A., García-Argüelles, S., Carrera, V., Vilanova, E., 2008. Serum albumin
Eaton, D.L., Daroff, R.B., Autrup, H., Bridges, J., Buffler, P., Costa, L.G., Coyle, J., McK- is as efficient as paraxonase in the detoxication of paraoxon at toxi-
hann, G., Mobley, W.C., Nadel, L., Neubert, D., Schulte-Hermann, R., Spencer, cologically relevant concentrations. Chemical Research in Toxicology 21,
P.S., 2008. Review of the toxicology of chlorpyrifos with an emphasis on human 1524–1549.
exposure and neurodevelopment. Critical Reviews in Toxicology 38, 1–125. Sogorb, M.A., Vilanova, E., 2010. Detoxication of anticholinesterase pesticides. In:
Ellman, G.L., Courtney, K.D., Andres Jr., V., Featherstone, R.M., 1961. A new and Satoh, T., Gupta, R.G. (Eds.), Anticholinesterase Pesticides: Metabolism, Neuro-
rapid colorimetric determination of acetylcholinesterase activity. Biochemical toxicity, and Epidemiology. John Wiley & Sons, New Jersey. USA, pp. 121–133.
Pharmacology 7, 88–95. Spielmann, H., Seiler, A., Bremer, S., Hareng, L., Hartung, T., Ahr, H., Faustman, E., Haas,
Estevan, C., Pamies, D., Sogorb, M.A., Vilanova, E., 2011. OECD guidelines and vali- U., Moffat, G.J., Nau, H., Vanparys, P., Piersma, A., Sintes, J.R., Stuart, J., 2006. The
dated methods for in vivo testing of reproductive toxicity. In: Gupta, R.G. (Ed.), practical application of three validated in vitro embryotoxicity tests. The report
Reproductive and Developmental Toxicology. Elsevier, Burlington, MA. USA, pp. and recommendations of an ECVAM/ZEBET workshop (ECVAM workshop 57).
123–133. Alternatives to Laboratory Animals 34, 527–538.
European Commission, 2005. Commission Directive 2005/72/EC of 21 October 2005 Tan, D.H., Peng, S.Q., Wu, Y.L., Wang, Y.M., Lu, C.F., Ding, W., Wang, Q.X., Yan, C.H.,
amending Council Directive 91/414/EEC to include chlorpyrifos, chlorpyrifos- 2009. Chlorpyrifos induces delayed cytotoxicity after withdrawal in primary
methyl, mancozeb, maneb, and metiram as active substances. Official hippocampal neurons through extracellular signal-regulated kinase inhibition.
Journal 279, 0063–0069, Available from: http://eur-lex.europa.eu/LexUriServ/ Biological and Pharmaceutical Bulletin 32 (October (10)), 1649–1655.
LexUriServ.do?uri=CELEX:32005L0072:EN:HTML (last accessed: 20.09.12). United States Environmental Protection Agency Administrator Announcement,
Food and Agriculture Organization of the United Nations and World Health Organi- 2000. Prevention Pesticides and Toxic Substances (7506C): Chlorpyrifos Revised
zation, 1999. Chlorpyrifos. In: Food and Agriculture Organization of the United Risk Assessment and Agreement with Registrants. U.S. Environmental Protec-
Nations and World Health Organization (Eds.), Pesticides Residues in Food. tion Agency, Washington, DC, Available from: http://aspcro.ceris.purdue.edu/
Available from: http://www.inchem.org/documents/jmpr/jmpmono/v99pr03. htm/pr/chlorpyrifos/riskassessment.htm (last accessed 20.09.12).
htm World Health Organization, 1986. Organophosphorous insecticides a general intro-
Flaskos, J., 2012. The developmental neurotoxicity of organophosphorus insec- duction. In: World Health Organization (Eds.), Environmental Health Criteria,
ticides: a direct role for the oxon metabolites. Toxicology Letters 209, vol. 63. Available from: http://www.inchem.org/documents/ehc/ehc/ehc63.htm
86–93. (last accessed 20.09.12).

You might also like