You are on page 1of 7

Toxicology and Applied Pharmacology 256 (2011) 330–336

Contents lists available at ScienceDirect

Toxicology and Applied Pharmacology


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / y t a a p

Effects of sub-lethal neurite outgrowth inhibitory concentrations of chlorpyrifos oxon


on cytoskeletal proteins and acetylcholinesterase in differentiating N2a cells
J. Flaskos a,⁎, E. Nikolaidis a, W. Harris b, M. Sachana a, A.J. Hargreaves b,⁎
a
Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
b
School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK

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

Article history: Previous work in our laboratory has shown that sub-lethal concentrations (1–10 μM) of chlorpyrifos (CPF),
Received 5 April 2011 diazinon (DZ) and diazinon oxon (DZO) inhibit the outgrowth of axon-like neurites in differentiating mouse
Revised 1 June 2011 N2a neuroblastoma cells concomitant with altered levels and/or phosphorylation state of axonal cytoskeleton
Accepted 3 June 2011
and growth-associated proteins. The aim of the present work was to determine whether chlorpyrifos oxon
Available online 17 June 2011
(CPO) was capable of inhibiting N2a cell differentiation in a similar manner. Using experimental conditions
Keywords:
similar to our previous work, sub-lethal concentrations (1–10 μM) of CPO were found to inhibit N2a cell
Organophosphorothionate pesticide differentiation. However, unlike previous studies with DZ and DZO, there was a high level of sustained
Chlorpyrifos inhibition of acetylcholinesterase (AChE) in CPO treated cells. Impairment of neurite outgrowth was also
Chlorpyrifos oxon associated with reduced levels of growth associated protein-43 and neurofilament heavy chain (NFH), and the
Neurite outgrowth distribution of NFH in cells stained by indirect immunofluorescence was disrupted. However, in contrast to
N2a neuroblastoma previous findings for DZO, the absolute level of phosphorylated NFH was unaffected by CPO exposure. Taken
Cell differentiation together, the findings suggest that sub-lethal concentrations of CPO inhibit axon outgrowth in differentiating
Developmental neurotoxicity
N2a cells and that this effect involves reduced levels of two proteins that play key roles in axon outgrowth and
Neurofilament heavy chain
maintenance. Although the inhibition of neurite outgrowth is unlikely to involve AChE inhibition directly,
Neurofilament phosphorylation
GAP-43 further work will help to determine whether the persistent inhibition of AChE by CPO can account for the
Acetylcholinesterase different effects induced by CPO and DZO on the levels of total and phosphorylated NFH.
© 2011 Elsevier Inc. All rights reserved.

Introduction more strongly than the parent compounds (Chambers, 1992;


Monnet-Tschudi et al., 2000). Thus, they are deemed to be responsible
Organophosphorus esters (OPs) are known to cause acute toxicity for the acute toxicity following organophosphorothionate insecticide
in adults, which is mainly due to the inhibition of neuronal poisoning. On the other hand, the role of these metabolites in the
acetylcholinesterase (AChE). However, a growing body of epidemio- developmental neurotoxicity of CPF and DZ is unclear, due to a lack of
logical and experimental evidence indicates that these compounds appropriate in vivo studies involving direct administration of these
may be also capable of inducing developmental neurotoxicity (Flaskos compounds to animals at sublethal doses.
and Sachana, 2010; Grandjean and Landrigan, 2006). Experimental However, an emerging set of in vitro data from studies using a
studies demonstrate that prenatal or postnatal exposure of rodents to number of cell culture models indicates that CPO and DZO may be able
the organophosphorothionate compounds chlorpyrifos (CPF) and to interfere with the development of the nervous system. For example,
diazinon (DZ), which are two of the most extensively used OP CPO and DZO affect glial cell development in primary cultures and cell
pesticides worldwide, causes considerable changes in a range of lines; CPO also perturbs the expression of astrocyte- and oligoden-
biochemical and morphological/cellular parameters related to ner- drocyte-specific markers at various stages of development in cell
vous system development, leading to persistent defects in behaviour culture aggregates of foetal rat telencephalon (Monnet-Tschudi et al.,
and cognition (Ricceri et al., 2006; Roegge et al., 2008; Slotkin, 2006; 2000). Furthermore, CPO inhibits DNA synthesis in rat C6 glioma and
Timofeeva et al., 2008). human 1321N1 astrocytoma cell lines (Guizzetti et al., 2005; Qiao
These OPs are metabolically converted, mainly in the liver, to their et al., 2001). Apart from its effects on C6 cell replication, CPO also
oxygen (oxon) analogues chlorpyrifos oxon (CPO; Fig. 1) and diazinon interferes with C6 cell differentiation by inhibiting the development of
oxon (DZO), which inhibit AChE activity up to 3 orders of magnitude extensions from these cells under differentiation-promoting condi-
tions and affecting the integrity of the microtubule network and the
⁎ Corresponding authors.
levels of microtubule proteins (Sachana et al., 2008). Similar
E-mail addresses: flaskos@vet.auth.gr (J. Flaskos), alan.hargreaves@ntu.ac.uk morphological and biochemical effects on differentiating C6 cells are
(A.J. Hargreaves). also induced by DZO (Sidiropoulou et al., 2009a).

0041-008X/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.taap.2011.06.002
J. Flaskos et al. / Toxicology and Applied Pharmacology 256 (2011) 330–336 331

were purchased from DakoCytomation (Ely, UK). Electrophoresis gel


and buffer reagents were obtained from Gene Flow (Fradley, UK) or
Sigma, whereas All Blue Precision Plus protein molecular weight
standards were purchased from BioRad Laboratories Ltd (Hemel
Hempstead, UK). Hybond-C nitrocellulose membrane filters and ECL
developing reagents were from GE Healthcare (Amersham, UK).
Chlorpyrifos oxon (purity 98.6%) was supplied by Greyhound
Chromatography (Birkenhead, UK).

Growth and maintenance of cell lines

Mouse N2a neuroblastoma cells were maintained in the logarith-


mic phase of growth at 37 °C in a humidified atmosphere of 5%
CO2/95% air. Mitotic cells were cultured in Dulbecco's modified Eagles
medium (DMEM) supplemented with 10% (v/v) foetal bovine serum
Fig. 1. Chemical structure of chlorpyrifos oxon. Shown is a schematic diagram of the
chemical structure of CPO. The asterisk indicates that the oxygen atom shown arises
(FBS), L-glutamine (2 mM), penicillin (100 U/ml) and streptomycin
from the bioactivation of the parent compound CPF, which has a sulphur atom in this (100 μg/ml) and passaged at 60–80% confluence.
position, by specific microsomal cytochrome P450 enzymes.

Induction of cell differentiation for measurement of neurite outgrowth


With respect to in vitro effects of oxon metabolites on the
development of neuronal cells, CPO has been shown to affect the Only cells up to passage number 20 were used for differentiation
expression of neuronal-specific markers in brain cell aggregate experiments. Cell differentiation was induced by serum withdrawal and
cultures (Monnet-Tschudi et al., 2000) and the expression of the addition of dibutyryl cAMP (Flaskos et al., 2007; Hargreaves et al.,
developmentally relevant transcription factors in cultures of embry- 2006; Harris et al., 2009a, 2009b; Sidiropoulou et al., 2009b). Briefly,
onic rat cortical and hippocampal neurons (Schuh et al., 2002). mitotic cell monolayers were detached by pipette at 60–80% confluence
However, data from morphological studies are not clear as to whether and harvested by centrifugation. Cell pellets were resuspended in 1 ml
CPO can readily interfere with the process of axonal/neurite growth medium and cell number determined in a haemocytometer
outgrowth. Thus, in the rat PC12 cell line, CPO has no effect during chamber. Cells were diluted to a density of 50,000 cells/ml before being
the initiation phase of neurite outgrowth up to 24 h nor on the seeded into 24-well culture dishes with a total volume of 0.5 ml growth
elaboration phase from 48 to 96 h and an inhibitory effect is only noted medium per well.
following a 7-day cell exposure to CPO (Das and Barone, 1999). After 24 h growth recovery, the medium was carefully removed
Moreover, in primary cultures of embryonic rat superior cervical from each well and replaced with serum free medium (i.e. growth
(Howard et al., 2005) and dorsal root (Yang et al., 2008) ganglia medium minus FBS) containing 0.3 mM cAMP and with or without
neurons, CPO influences axonal length but has no effect on the number CPO (final concentration 1–10 μM). Cells were returned to the CO2
of axons extended. incubator and allowed to differentiate for up to 24 h. CPO was
In this study, we have assessed the capacity of CPO to interfere with prepared as 200-fold concentrated stock solutions in dimethyl
the outgrowth of axon-like neurites in differentiating mouse N2a sulphoxide (DMSO) and added to the serum free medium immedi-
neuroblastoma cells. Using this cell culture system we have recently ately before use. Control cells were treated with serum free medium
found that DZO induces rapid and potent inhibitory effects on neurite containing the same amount of DMSO (0.5% v/v).
outgrowth; following 24 h exposure to a sub-lethal concentration of After 24 h differentiation, serum free medium was carefully
10 μM DZO, the number of long axon-like processes was reduced by removed by aspiration from the edge of the well and cell monolayers
more than 80% compared to the non-DZO-treated control (Sidiropoulou were fixed at −20 °C in 90% (v/v) methanol in Tris buffered saline
et al., 2009b). In addition, in the present study we have investigated the (TBS: 10 mM Tris, 140 mM NaCl pH 7.4). They were then stained for
effects of CPO on cytoskeletal and axon growth-associated proteins, 2 min in Coomassie Brilliant Blue and excess stain removed by
which have been found to be affected in N2a cells after treatment with aspiration followed by 2 rinses in distilled water. The number of
10 μM DZO. Throughout the study, we have employed CPO at non- axon-like neurites per 100 cells was estimated as described previously
cytotoxic concentrations of up to 10 μM, using the same N2a cell growth (Flaskos et al., 2007; Hargreaves et al., 2006; Harris et al., 2009a,
and differentiation conditions used previously to allow valid compar- 2009b; Sidiropoulou et al., 2009b). Cell viability was assessed by
isons to be made between CPO and DZO and also between CPO and the monitoring the reduction of thiazolyl blue tetrazolium bromide (MTT)
parent compound CPF. using the method of Denizot and Lang (1986). Sublethal effects were
confirmed by the inability of CPO treatment to cause a decrease in MTT
Materials and methods reduction compared to the control.

Reagents and materials Measurement of acetylcholinesterase activity

Cell culture reagents and materials were purchased from Scientific For this, cells were induced to differentiate as above, except that 2
Laboratory Supplies (SLS, Wilford, UK) or BioWhittaker UK Ltd million cells were plated into T75 culture flasks in 40 ml of culture
(Wokingham, UK), with the exception of dibutyryl cAMP, which was medium. Cells were harvested by centrifugation, resuspended in 20 ml
obtained from Sigma Aldrich Co Ltd (Sigma: Poole, UK). Mouse of PBS at 4 °C and recentrifuged to remove traces of DMEM. Cell pellets
monoclonal antibodies against GAP-43 (clone GAP-7B10), α-tubulin were resuspended in 0.6 ml of 200 mM sodium phosphate buffer
(clone B512), and neurofilament heavy chain (NFH: clone N52) were (pH 7.4) at 4 °C and sonicated for 15 sec on ice. The activity of AChE in
obtained from Sigma. Monoclonal anti-phosphorylated NFH (pNFH) cell sonicates was monitored by the method of Ellman et al. (1961)
antibody SMI31 and Ta51 were supplied by Cambridge Bioscience modified for a microtitre plate format as described previously (Flaskos
(Cambridge, UK). Secondary antibodies conjugated with either et al., 2007; Sidiropoulou et al., 2009b). Protein was determined in the
fluorescein isothiocyanate (FITC) or horseradish peroxidase (HRP) sonicated extracts by the bicinchoninic acid (BCA) assay (Brown et al.,
332 J. Flaskos et al. / Toxicology and Applied Pharmacology 256 (2011) 330–336

1989) and specific activity was expressed as absorbance change /mg concentration-dependent reduction in the outgrowth of axon-like
protein/h. neurites following 24 h exposure to the same levels of CPO (Fig. 2).
The acute cholinergic effects of CPO were investigated by performing
Gel electrophoresis and Western blotting assays of AChE activity following exposure of differentiating N2a cells to
the same concentrations of CPO for 4 h and 24 h. As indicated in Fig. 3,
For Western blotting analysis, cells were seeded and induced to there was a statistically significant dose-dependent reduction in the
differentiate in the presence and absence of CPO in T75 culture flasks, specific activity of AChE following both 4 h and 24 h exposure, with
as described above. Intact cell monolayers were then solubilised by values falling to approximately 30% of control values at 5 and 10 μM
boiling in 2 ml of 0.5% SDS in TBS prior to protein determination using (p b 0.02). After 24 h exposure, there appeared to be slightly less
the BCA assay. The resultant cell lysates were subsequently subjected inhibition at the lower concentrations of CPO than at 4 h, although this
to gel electrophoresis in the presence of sodium dodecyl sulphate effect was not deemed to be statistically significant when tested by two-
(SDS-PAGE) employing a 10% w/v polyacrylamide resolving gel, way ANOVA.
overlaid with a 4% w/v polyacrylamide stacking gel (Laemmli, 1970). The molecular basis of the observed ability of CPO to inhibit neurite
The separated proteins were then electrophoretically transferred onto outgrowth at sub-lethal concentrations was further investigated by
nitrocellulose membrane filters (Towbin et al., 1979). Western blotting analysis using antibodies to α-tubulin, GAP-43, NFH
The resultant Western blots were blocked with 3% w/v BSA in TBS and pNFH. As shown in the panel of blots in Fig. 4, the cross-reactivity
(BSA/TBS) for at least 1 h at room temperature. Blots were then of cell lysates with monoclonal anti-α-tubulin antibody B512 was
probed overnight at 4 °C with appropriate dilutions of primary apparently unaffected by exposure to CPO. Anti-GAP-43 (GAP7B10)
antibodies in BSA/TBS, including anti-total NFH (N52; 1/500), anti- cross-reactivity appeared to be reduced compared to the control
pNFH (SMI34; 1/500), anti-GAP-43 (GAP7B10; 1/500), anti-α-tubulin following exposure to higher concentrations of CPO. Anti-NFH (N52)
(B512; 1/2000). After six 10-min washes in TBS containing 0.05% v/v also showed a trend of reduced cross-reactivity at increasing
Tween-20 (TBS/Tween), blots were probed with HRP-conjugated concentrations of CPO, whereas reactivity of blots with anti-pNFH
anti-mouse immunoglobulin (1/2000) for 3 h at room temperature. (SMI34) showed slightly raised or similar levels to the control in
Following 6 further washes with TBS/Tween, antibody reactivity was lysates of CPO treated cells.
visualised with enhanced chemiluminescence reagent (ECL). For Quantification of the observed changes was then performed using
quantification of antibody reactivity, band intensities on images of AIDA software on blots obtained from 4 independent experiments. As in
developed Western blots were determined using Advanced Image previous work on OP treatment of N2a cells, anti-tubulin reactivity
Data Analysis software (Fuji). Anti-tubulin reactivity levels were used showed no significant change following treatment with CPO (104% of
for normalisation. the control values at 10 μM CPO). Since tubulin was used for normal-
isation in previous studies on N2a cells, the densitometric values of all
Indirect immunofluorescence staining of cell monolayers other bands were normalised to tubulin, which served as a loading
control. The observed reductions in reactivity of cell lysates with anti-
The intracellular distributions of tubulin, GAP-43 and NFH were GAP-43 and anti-NFH were confirmed by densitometric analysis, which
further studied by indirect immunofluorescence staining. Typically, indicated a dose-dependent decrease in peak area in both cases that was
300 μl of N2a cell suspension (50,000 cells/ml) were seeded into each statistically significant compared to the non-CPO-treated control at the
well of an 8-well cell culture chamber slide. After 24 h differentiation highest concentration tested (Table 1).
in the presence or absence of CPO, cells were fixed for 10 min at In contrast to the decrease in reactivity with anti-NFH, cross-
−20 °C in 90% v/v methanol in TBS. They were then blocked with reactivity with anti-pNFH was not significantly different from control
BSA/TBS for 45 min at room temperature, extracted in TBS/Tween for values (p N 0.8). In order to determine whether the different responses
5 min followed by incubation with TBS for a further 5 min at room observed for anti-NFH and anti-pNFH reflected altered phosphoryla-
temperature. Cells were then incubated overnight at 4 °C with tion status of NFH, band intensities relative to the control were
primary antibodies against tubulin (B512; 1/100), GAP-43 calculated as a ratio, where a value of 1 would indicate no overall
(GAP7B10; 1/50) or NFH (N52 and SMI34; both diluted 1/50 in change in phosphorylation status. As indicated in the right hand
BSA/TBS). After three 5-min washes with TBS, they were incubated for column of Table 1, there was a consistent dose-dependent decrease in
2 h at room temperature with FITC-conjugated rabbit anti-mouse IgG
secondary antibodies. After 3 further washes in TBS, cells were
mounted under glass cover slips using VectaShield® mounting
medium for fluorescence (Vector Laboratories Ltd., Peterborough,
UK) and viewed using a Leica CLSM confocal laser scanning
microscope fitted with epifluorescence optics, as described previously
(Flaskos et al., 2007; Hargreaves et al., 2006; Harris et al., 2009b).

Statistical analysis of data

All sets of data are based on a minimum of 3 independent


experiments and expressed as mean ± SEM. Average values for each
treatment were compared to the corresponding control by one-way
or two-way ANOVA, as appropriate, using 95% confidence limits.

Results

In order to obtain data comparable to our previous work on Fig. 2. Effects of chlorpyrifos oxon on neurite outgrowth in differentiating N2a cells.
organophosphorothionate toxicity, N2a cells were induced to differen- Mouse N2a neuroblastoma cells were induced to differentiate in the absence and
presence of CPO, fixed, stained and assessed for the outgrowth of axon-like neurites as
tiate for up to 24 h in the presence of 0–10 μM CPO. Measurement of described in Materials and methods. Shown are the mean numbers of axon-like
MTT reduction indicated no significant decrease in CPO treated cells neurites per 100 cells ± SEM from 4 independent experiments. Asterisks indicate
compared to the controls (not shown). However, there was a statistically significant differences compared to the untreated control (p b 0.001).
J. Flaskos et al. / Toxicology and Applied Pharmacology 256 (2011) 330–336 333

Table 1
Densitometric analysis of probed western blots. Lysates of N2a cells induced to
differentiate in the absence and presence of CPO were subjected to SDS-PAGE and
Western blotting analysis as described in Materials and methods. Shown are the
average band intensities ± SEM from a minimum of 4 independent experiments,
showing cross-reactivity with antibodies to GAP-43 (GAP7B10), total NFH (N52) and
pNFH (SMI34). The phosphorylation ratio of the change in NFH:pNFH is shown in the
right hand column of the table. Asterisks indicate changes that are statistically
significant compared to the non-CPO-treated control (p b 0.05).

CPO concentration Antibody reactivity (% control ± SEM)


(μM)
GAP43 NFH pNFH NFH:pNFH ratio

1 115.2 ± 35.4 84.5 ± 20.6 137.5 ± 34.4 0.64 ± 0.12


5 74.2 ± 17.5 58.8 ± 15.1 102.0 ± 22.4 0.59 ± 0.11*
10 23.6 ± 7.5* 42.3 ± 11.5* 119.0 ± 45.3 0.32 ± 0.06*

patterns for all antibodies in Fig. 5 showed a gradual decrease in


neurite outgrowth as the concentration of CPO increased. Staining
with anti-α-tubulin antibody was strong in both cell bodies and
neurites (when present) even at the highest concentration of CPO
(Figs. 5a–c). In the case of anti-GAP-43 staining, a general reduction in
staining intensity occurred at increasing CPO concentration with
weaker staining of neurites when present in CPO-treated cell
monolayers (Figs. 5d and e). Anti-NFH antibody N52 stained long
axon-like neurites strongly in control cells but much more weakly in
CPO-treated cells (Figs. 5g–i). The most predominant feature of CPO
treated cells stained with this antibody was the presence of
aggregates of anti-NFH staining (upper arrows in Figs. 5h and i).
The axonal staining pattern for ant-pNFH was similar to that of anti-
NFH in control cells; however, the cell body was much more evenly
stained with this antibody than with anti-NFH (Figs. 5j–l). The
staining intensity of anti-pNFH and its diffuse distribution in cell
Fig. 3. Effects of chlorpyrifos oxon on acetylcholinesterase activity in differentiating N2a
bodies remained constant for all treatments, although as previously
cells. Mouse N2a neuroblastoma cells were induced to differentiate for 4 h or 24 h in the mentioned the number of stained neurites decreased.
absence and presence of CPO, harvested and cell sonicates assayed for AChE activity as
described in Materials and methods. Results are expressed as mean specific activity
(Absorbance change/mg protein/h) ± SEM for 3 (24 h) or 4 (4 h) independent Discussion
experiments. Asterisks indicate significant differences compared to the untreated
control (p b 0.02).
In previous work we found that sub-lethal concentrations of
organophosphorothionate and organophosphonothionate pesticides
the phosphorylation ratio (NFH: pNFH), which was statistically such as CPF, DZ and leptophos, inhibited the outgrowth of axon-like
significant following exposure to both 5 μM and 10 μM CPO. processes by differentiating N2a cells (Flaskos et al., 2007; Sachana et
The effects of treatment with CPO on the intracellular distribution al., 2001, 2003). More recently, DZO was also found to inhibit neurite
of tubulin, NFH and GAP-43 were then studied by indirect immuno- outgrowth over the same concentration range. The aim of this study
fluorescence staining of cells following 24 h exposure. The staining was to determine whether CPO behaved in a comparable fashion and
to determine its effects on a similar panel of protein biomarkers to
that used in studies of the other compounds.
The inability of CPO to cause a decrease in MTT reduction by
differentiating N2a monolayers suggests that 1–10 μM CPO has no
significant effect on cell viability under the conditions tested. This
finding is in good agreement with our previous work, in which similar
concentrations of CPF, DZ and DZO were also found to be sub-lethal
towards differentiating N2a cells after 24 h exposure (Flaskos et al.,
2007; Sachana et al., 2001, 2005; Sidiropoulou et al., 2009b). However,
morphological analysis of Coomassie blue stained cells clearly in-
dicates that these concentrations of CPO caused inhibition of neurite
outgrowth in a dose dependent manner, which is again consistent with
our previous findings for CPF, DZ, leptophos and DZO (Flaskos et al.,
2007; Sachana et al., 2001, 2003, 2005; Sidiropoulou et al., 2009b).
The data obtained in this study showed significant inhibition of the
enzymatic activity of AChE both concomitantly and prior to the
morphological and biochemical effects of CPO. In a previous study with
Fig. 4. Detection of cytoskeletal and growth associated proteins on Western blots of N2a N2a cells and the oxon metabolite of DZ, a significant AChE inhibition
cell lysates. Mouse N2a neuroblastoma cells were induced to differentiate for 24 h in was also noted, but only at the earlier time point (Sidiropoulou et al.,
the absence and presence of CPO and cell lysates subjected to SDS-PAGE and Western
blotting as described in Materials and methods. Shown are typical blots probed with
2009b). The demonstration of a significant reduction in AChE activity
antibodies to NFH (N52), pNFH (SMI34), GAP-43 (GAP7B10) and α-tubulin (B512), by CPO in this study both at 4 and 24 h is in line with previous data
followed by HRP-conjugated secondary antibodies and development using ECL reagent. showing that administration of CPF, compared to DZ under the same
334 J. Flaskos et al. / Toxicology and Applied Pharmacology 256 (2011) 330–336

Fig. 5. Indirect immunofluorescence staining of differentiating N2a cells. Mouse N2a neuroblastoma cells induced to differentiate for 24 h in the absence and presence of CPO, were
fixed and stained by indirect immunofluorescence as described in Materials and methods. Shown are typical images of control cells (a, d, g, k) and cells treated with 5 μM (b, e, h, l) or
10 μM (c, f, i, m) CPO stained with anti-α-tubulin (a–c), anti-GAP43 (d–f), anti-NFH (g–i) and anti-pNFH (k–m). Horizontal arrows indicate typical axon-like neurites in controls and
vertical arrows indicate typical aggregates detected by anti-NFH staining in CPO treated cells. Bar represents 20 μm.

conditions, produces greater inhibition of AChE (Jameson et al., 2007) (Flaskos et al., 2007; Sachana et al., 2001, 2003, 2005). Densitometric
and higher mortality (Slotkin et al., 2006). However, on the basis of our analysis of Western blots in the current work suggests that this is also
previous data on differentiating N2a cells, it is unlikely that the the case for CPO exposure, in contrast to DZO which was previously
inhibition of axon-like outgrowth is aetiologically related to AChE shown to have no overall effect on total NFH levels (Sidiropoulou et
inhibition. For example, although the OP trio-ortho-cresyl phosphate al., 2009b). Reduced levels of NFH detected on Western blots could
is a weak inhibitor of AChE (Lock and Johnson, 1990), it inhibits reflect increased degradation by proteases such as cathepsin D or
outgrowth of axon-like processes in N2a cells to an equal or even calpain (James et al., 1998; Nixon and Marotta, 1984). In this respect,
greater extent than the AChE-inhibiting OPs, DZ and CPF (Flaskos et al., activation of calpain has been shown to occur in response to OP
2007; Fowler et al., 2001; Sachana et al., 2005). That the inhibition of treatment in vivo (El-Fawal and Ehrich, 1993). However, the reduced
the enzymatic activity of AChE by CPF is not related to its neurite antibody reactivity could also be caused by decreased synthesis of
inhibitory effect has also been demonstrated in a number of cell NFH, which could be related to the known ability of CPF to inhibit the
culture studies (Das and Barone, 1999; Howard et al., 2005) as well as synthesis of both DNA and proteins in vivo (Song et al., 1998).
in vivo (Slotkin et al., 2006). Neurofilaments are known to be important in the regulation of
Previous studies showed that exposure to CPF, DZ and leptophos axon growth and stability and NFH becomes more highly expressed
was associated with reduced levels of NFH compared to the control and increasingly phosphorylated as axons mature (Lee et al., 1988; Lee
J. Flaskos et al. / Toxicology and Applied Pharmacology 256 (2011) 330–336 335

and Cleveland, 1994; Veeranna et al., 1998). Previous work indicated which other OPs also caused reduced levels of total NFH and the
that DZO exposure was associated with increased phosphorylation of formation of aggregates in cell bodies (Flaskos et al., 1998, 2007;
NFH. However, the lack of a significant change in reactivity of lysates Hargreaves et al., 2006), indicating that the two phenomena may be
with SMI34 suggests that, in contrast to the effects of DZO, there is no closely related.
overall change in the absolute levels of pNFH in CPO treated cells. The above morphological and biochemical effects of CPO noted in
Although a significant increase was not observed in CPO treated cells in vitro are caused by concentrations similar to those occurring in vivo in
the current work, the lack of change in SMI34 antibody reactivity the developing organism. In humans, foetal concentrations of the
relative to the control is consistent with a possible alteration to the parent phosphorothionate CPF may commonly reach levels of 8 μg/ml
overall phosphorylation status of NFH in CPO-treated cells. This notion (22.8 μM) (Ostrea et al., 2002). As shown recently, the steady state
was confirmed by calculating the ratio of CPO induced change in NFH ratio of CPO/CPF in human plasma varies widely among adults and in
to that in pNFH, which clearly shows a significant reduction to 59% and several cases can be higher than 0.04–0.05 (Eyer et al., 2009). Due to
32% of normal levels with 5 and 10 μM CPO, respectively, further the defective hydrolysis of the oxons by A- and B-esterases during
confirming the reduction in total NFH levels relative to pNFH. These mammalian development (Furlong et al., 2006; Vidair, 2004), this
findings are consistent with the possibility that NFH is more prone to ratio is expected to be higher in the developing organism. Thus, levels
proteolytic degradation when it is not phosphorylated and/or that the of CPO in the low micromolar range are attainable in developing
non-degraded NFH in CPO-treated cells has an increased level of humans.
phosphorylation compared to the control. Further work will help to Although CPO is less lipophilic than CPF, which might affect its
determine whether CPO is able to activate a protease or a protein penetration into the foetus, data showing considerable inhibition of
kinase for which NFH is a substrate and/or inhibit the activity of a cholinesterase in foetal tissues after in vivo administration of
protein phosphatase that regulates the phosphorylation status of NFH. phosphorothionate insecticides (Gupta, 1995) indicate that the foetus
Data obtained from the quantitative analysis of blots probed with is exposed to the oxon. In humans, the foetus is exposed to CPO
monoclonal antibody GAP7B10 suggested that there was a dose generated in maternal tissues. In contrast, placental metabolism is
dependent reduction in the levels of GAP-43 protein in lysates of CPO unlikely to contribute significantly to CPO formation, since CYP2B6,
treated cells. This finding is in good agreement with our previous work the main enzyme responsible for converting CPF to CPO in humans
in which N2a cells were exposed to a number of different OPs (Fowler et (Croom et al., 2010), is expressed to a very low extent in human
al., 2001; Harris et al., 2009a; Sachana et al., 2001, 2003, 2005; placenta (Pelkonen et al., 2006).
Sidiropoulou et al., 2009b), suggesting that GAP-43 represents a In conclusion, the data presented here suggest that exposure of
common molecular marker of sub-lethal neurite inhibitory effects of differentiating N2a cells to sub-lethal neurite inhibitory levels of CPO
OPs. These changes in GAP-43 could have a significant bearing on the is associated with persistent inhibition of AChE, reduced levels of
morphological effects of CPO and other OPs, since GAP-43 is known to GAP-43 and NFH protein but that the level of NFH phosphorylation is
exhibit increased synthesis and play a key role during axon outgrowth unaffected.
(Meiri et al., 1986, 1998; Skene, 1989). Consistent with the aforemen-
tioned studies on N2a cells, tubulin levels were unaffected by any of the Conflict of interest statement
CPO concentrations in the present study, which is in agreement with
findings in the above mentioned studies on N2a cells, suggesting that The authors do not have any conflicts of interest to disclose.
microtubules are not a major target for these compounds in differen-
tiating N2a cells. References
The CPO induced reduction in the number of neurites stained by
Brown, R.E., Jarvis, K.L., Hyland, K.J., 1989. Protein measurement using bicinchoninic
indirect immunofluorescence with antibodies to cytoskeletal proteins acid: elimination of interfering substances. Anal. Biochem. 180, 136–139.
confirmed the dose-dependent reduction shown in the morphological Chambers, J.E., 1992. The role of target site activation of phosphorothionates in acute
analysis presented in Fig. 2. The staining patterns for monoclonal toxicity. In: Chambers, J.E., Levi, P.E. (Eds.), Organophosphates: Chemistry, Fate and
Effects. Academic Press Inc., San Diego, pp. 229–239.
antibodies B512, GAP7B10 and SMI34 suggested that the distribution
Croom, E.L., Wallace, A.D., Hodgson, E., 2010. Human variation in CYP-specific
of tubulin, GAP-43 and pNFH, respectively, within the cell body of N2a chlorpyrifos metabolism. Toxicology 276, 184–191.
cells was not significantly affected by CPO treatment. The fact that, Das, K.P., Barone Jr., S., 1999. Neuronal differentiation in PC12 cells is inhibited by
chlorpyrifos and its metabolites: is acetylcholinesterase inhibition the site of
even following exposure to 10 μM CPO, axon like neurites (when
action? Toxicol. Appl. Pharmacol. 160, 217–230.
present) were still strongly stained with the anti-tubulin antibody Denizot, F., Lang, R., 1986. Rapid colorimetric assay for cell growth and survival—
B512 further supports the view that tubulin distribution and the modification to the tetrazolium dye procedure giving improved sensitivity and
microtubule network are not significantly affected in this cellular reliability. J. Immunol. Methods 89, 271–277.
El-Fawal, H.A., Ehrich, M.F., 1993. Calpain activity in organophosphorus-induced
model. However, Grigoryan and Lockridge (2009) demonstrated that delayed neuropathy (OPIDN): effects of a phenylalkylamine calcium channel
CPO could bind to tubulin and disrupt the polymerisation of purified blocker. Ann. N. Y. Acad. Sci. 679, 325–329.
bovine brain microtubules in vitro, suggesting that a direct interaction Ellman, G.L., Courtney, K.D., Andres Jr., V., Featherstone, R.M., 1961. A new and rapid
colorimetric determination of acetylcholinesterase. Biochem. Pharmacol. 7, 88–95.
of this compound with tubulin was possible. This apparent discrep- Eyer, F., Roberts, D.M., Buckley, N.A., Eddeston, M., Thiermann, H., Worek, F., Eyer, P.,
ancy may be due to differences in the experimental procedures used; 2009. Extreme variability in the formation of chlorpyrifos oxon (CPO) in patients
thus, under the conditions tested, microtubules in differentiating N2a poisoned by chlorpyrifos (CPF). Biochem. Pharmacol. 78, 531–537.
Flaskos, J., Sachana, M., 2010. Developmental neurotoxicity of anticholinesterase
cells may be less accessible to CPO binding than the purified bovine pesticides. In: Satoh, T., Gupta, R.C. (Eds.), Anticholinesterase Pesticides: Metabo-
brain microtubules used in the study of Grigoryan and Lockridge lism, Neurotoxicity, and Epidemiology. John Wiley and Sons, Hoboken, pp. 203–223.
(2009). Further work on the identification of CPO binding targets in Flaskos, J., McLean, W.J., Fowler, M.J., Hargreaves, A.J., 1998. Tricresyl phosphate inhibits
the formation of axon-like processes and disrupts neurofilaments in cultured
N2a cells would be worthwhile.
mouse N2a and rat PC12 cells. Neurosci. Lett. 242, 101–104.
The observed decrease in relative staining intensity in axons for Flaskos, J., Harris, W., Sachana, M., Muñoz, D., Tack, J., Hargreaves, A.J., 2007. The effects
GAP-43 and NFH further confirms that the levels of GAP-43 and NFH of diazinon and cypermethrin on the differentiation of neuronal and glial cell lines.
Toxicol. Appl. Pharmacol. 219, 172–180.
in axons may be reduced in CPO-treated cells compared to the control.
Fowler, M.J., Flaskos, J., McLean, W.G., Hargreaves, A.J., 2001. Effects of neuropathic
Disruption of the neurofilament network is further confirmed by isomers of tricresyl phosphate and their microsomal activation on the production
major changes in the staining pattern of anti-NFH, which indicates a of axon-like processes by differentiating mouse N2a neuroblastoma cells. J.
major reorganisation of total NFH from axon staining in control cells Neurochem. 76, 671–678.
Furlong, C.E., Holland, N., Richter, R.J., Bradman, A., Ho, A., Eskenazi, B., 2006. PON1
to cell body localised aggregates in CPO-treated cells. This type of status of farmworker mothers and children as a predictor of organophosphate
disruption is consistent with previous studies in our laboratory, in sensitivity. Pharmacogenet. Genomics 16, 183–190.
336 J. Flaskos et al. / Toxicology and Applied Pharmacology 256 (2011) 330–336

Grandjean, P., Landrigan, P.J., 2006. Developmental neurotoxicity of industrial Pelkonen, O., Vahakangas, K., Gupta, R.C., 2006. Placental toxicity of organophosphate
chemicals. Lancet 368, 2167–2178. and carbamate pesticides. In: Gupta, R.C. (Ed.), Toxicology of Organophosphate and
Grigoryan, H., Lockridge, O., 2009. Nanoimages show disruption of tubulin polymer- Carbamate Compounds. Elsevier Academic Press, Burlington, pp. 463–479.
isation by chlorpyrifos oxon: implications for neurotoxicity. Toxicol. Appl. Qiao, D., Seidler, F.J., Slotkin, T.A., 2001. Developmental neurotoxicity of chlorpyrifos
Pharmacol. 240, 143–148. modelled in vitro: comparative effects of metabolites and other cholinesterase
Guizzetti, M., Pathak, S., Giordano, G., Costa, L.G., 2005. Effect of organophosphorus inhibitors on DNA synthesis in PC12 and C6 cells. Environ. Health Perspect. 109,
insecticides and their metabolites on astroglial cell proliferation. Toxicology 215, 909–913.
182–190. Ricceri, L., Venerosi, A., Capone, F., Cometa, M.F., Lorenzini, P., Fortuna, S., Calamandrei,
Gupta, R.C., 1995. Environmental agents and placental toxicity: anticholinesterases and G., 2006. Developmental neurotoxicity of organophosphorus pesticides: fetal and
other agents. In: Sastry, B.V.R. (Ed.), Placental Toxicology. CRC Press, Boca Raton, neonatal exposure to chlorpyrifos alters sex-specific behaviors at adulthood in
pp. 257–278. mice. Toxicol. Sci. 93, 105–113.
Hargreaves, A.J., Fowler, M.J., Sachana, M., Flaskos, J., Bountouri, M., Coutts, I.C., Glynn, Roegge, C.S., Timofeeva, O.A., Seidler, F.J., Slotkin, T.A., Levin, E.D., 2008. Developmental
P., Harris, W., McLean, W.G., 2006. Inhibition of neurite outgrowth in diazinon neurotoxicity in rats: later effects on emotional response. Brain Res. Bull.
differentiating mouse N2a neuroblastoma cells by phenyl saligenin phosphate: 75, 166–172.
effects on neurofilament heavy chain phosphorylation, MAP kinase (ERK 1/2) Sachana, M., Flaskos, J., Alexaki, E., Glynn, P., Hargreaves, A.J., 2001. The toxicity of
activation and neuropathy target esterase activity. Biochem. Pharmacol. 71, chlorpyrifos towards differentiating mouse N2a neuroblastoma cells. Toxicol. In
1240–1247. Vitro 15, 369–372.
Harris, W., Sachana, M., Flaskos, W., Hargreaves, A.J., 2009a. Neuroprotection from Sachana, M., Flaskos, J., Alexaki, E., Hargreaves, A.J., 2003. Inhibition of neurite
diazinon-induced toxicity in differentiating murine N2a neuroblastoma cells. outgrowth in N2a cells by leptophos and carbaryl: effects on neurofilament heavy
Neurotoxicology 30, 958–964. chain, GAP-43 and HSP-70. Toxicol. In Vitro 17, 115–120.
Harris, W., Sachana, M., Flaskos, W., Hargreaves, A.J., 2009b. Proteomic analysis of Sachana, M., Flaskos, J., Hargreaves, A.J., 2005. Effects of chlorpyrifos and chlorpyrifos-methyl
differentiating neuroblastoma cells treated with sub-lethal neurite inhibitory on the outgrowth of axon-like processes, tubulin and GAP-43 in N2a cells. Toxicol. Mech.
concentrations of diazinon: identification of novel biomarkers of effect. Toxicol. Methods 15, 405–410.
Appl. Pharmacol. 240, 159–165. Sachana, M., Flaskos, J., Sidiropoulou, E., Yavari, C.A., Hargreaves, A.J., 2008. Inhibition of
Howard, A.S., Bucelli, R., Jett, D.A., Bruun, D., Yang, D., Lein, P.J., 2005. Chlorpyrifos exerts extension outgrowth in differentiating rat C6 glioma cells by chlorpyrifos and
opposing effects on axonal and dendritic growth in primary neuronal cultures. chlorpyrifos oxon: effects on microtubule proteins. Toxicol. In Vitro 22, 1387–1391.
Toxicol. Appl. Pharmacol. 207, 112–124. Schuh, R.A., Lein, P.J., Beckles, R.A., Jett, D.A., 2002. Noncholinesterase mechanism of
James, T., Matzelle, D., Bartus, R., Hogan, E.L., Banik, N.L., 1998. New inhibitors of calpain chlorpyrifos neurotoxicity: altered phosphorylation of Ca+ 2/cAMP response element
prevent degradation of cytoskeletal and myelin proteins in spinal cord in vitro. J. binding protein in cultured neurons. Toxicol. Appl. Pharmacol. 182, 176–185.
Neurosci. Res. 51, 218–222. Sidiropoulou, E., Sachana, M., Flaskos, J., Harris, W., Hargreaves, A.J., Woldehiwet, Z.,
Jameson, R.R., Seidler, F.J., Slotkin, T.A., 2007. Nonenzymatic functions of acetylcholin- 2009a. Diazinon oxon interferes with differentiation of rat C6 glioma cells. Toxicol.
esterase splice variants in the developmental neurotoxicity of organophosphates: In Vitro 23, 1548–1552.
chlorpyrifos, chlorpyrifos oxon and diazinon. Environ. Health Perspect. 115, 65–70. Sidiropoulou, E., Sachana, M., Flaskos, J., Harris, W., Hargreaves, A.J., Woldehiwet, Z.,
Laemmli, U.K., 1970. Cleavage of structural proteins during the assembly of the head of 2009b. Diazinon oxon affects the differentiation of mouse N2a neuroblastoma cells.
bacteriophage T4. Nature 227, 680–685. Arch. Toxicol. 83, 373–380.
Lee, M.K., Cleveland, D.W., 1994. Neurofilament function and dysfunction: involvement Skene, J.H.P., 1989. Axonal growth-associated proteins. Annu. Rev. Neurosci. 12, 127–156.
in axonal growth and neuronal disease. Curr. Opin. Cell Biol. 6, 34–40. Slotkin, T.A., 2006. Developmental neurotoxicity of organophosphates: a case study of
Lee, V.M.Y., Otvos Jr., L., Carden, M.J., Hollosi, M., Dietzchold, B., Lazzarini, R.A., 1988. chlorpyrifos. In: Gupta, R.C. (Ed.), Toxicology of organophosphate and carbamate
Identification of the major multiphosphorylation site in mammalian neurofila- compounds. Academic Press Inc., San Diego, pp. 293–314.
ments. Proc. Natl Acad. Sci. U. S. A. 85, 1998–2002. Slotkin, T.A., Levin, E.D., Seidler, F.J., 2006. Comparative developmental neurotoxicity of
Lock, E.A., Johnson, M.K., 1990. Delayed neuropathy and acute toxicity studies with organophosphate insecticides: effects on brain development are separable from
pirimiphos-methyl in the hen. J. Appl. Toxicol. 10, 17–21. systemic toxicity. Env. Health Perspect. 114, 746–751.
Meiri, K.F., Pfenninger, K.H., Willard, M., 1986. Growth associated protein, GAP-43, a Song, X., Violin, J.D., Seidler, F.J., Slotkin, T.A., 1998. Modeling the developmental
polypeptide that is induced when neurons extend axons, is a component of growth neurotoxicity of chlorpyrifos: macromolecule synthesis in PC12 cells. Toxicol. Appl.
cones and correspond to pp 46, a major polypeptide of a subcellular fraction Pharmacol. 151, 182–191.
enriched in growth cones. Proc. Natl Acad. Sci. U. S. A. 83, 3537–3541. Timofeeva, O.A., Roegge, C.S., Seidler, F.J., Slotkin, T.A., Levin, E.D., 2008. Persistent
Meiri, K.F., Saffell, J., Walsh, F.S., Doherty, P., 1998. Neurite outgrowth stimulated by cognitive alterations in rats after early postnatal exposure to low doses of the
neural cell adhesion molecules requires growth-associated protein-43 (GAP-43) organophosphate pesticide, diazinon. Neurotoxicol. Teratol. 30, 38–45.
function and is associated with GAP-43 phosphorylation in growth cones. J. Towbin, S., Staehelin, T., Gordon, J., 1979. Electrophoretic transfer of proteins from
Neurosci. 18, 10429–10437. polyacrylamide gels to nitrocellulose sheets: procedure and some applications.
Monnet-Tschudi, F., Zurich, M.G., Schilter, B., Costa, L.G., Honegger, P., 2000. Proc. Natl Acad. Sci. U. S. A. 76, 4350–4354.
Maturation-dependent effects of chlorpyrifos and parathion and their oxygen Veeranna, Amin, N.D., Ahn, N.G., Jaffe, H., Winters, C.J., Grant, P., Pant, H.C., 1998.
analogs on acetylcholinesterase and neuronal and glial markers in aggregating Mitogen-activated protein kinases (ERK 1/2) phosphorylate Lys-Ser-Pro (KSP)
brain cell cultures. Toxicol. Appl. Pharmacol. 165, 175–183. repeats in neurofilament proteins NFH and NFM. J. Neurosci. 18, 4008–4021.
Nixon, R.A., Marotta, C.A., 1984. Degradation of neurofilament proteins by purified Vidair, C.A., 2004. Age dependence of organophosphate and carbamate neurotoxicity in
human brain cathepsin D. J. Neurochem. 43, 507–516. the postnatal rat: extrapolation to the human. Toxicol. Appl. Pharmacol. 196, 287–302.
Ostrea, E.M., Morales, V., Ngoumgna, E., Prescilla, R., Tan, E., Hernandez, E., Ramirez, Yang, D., Howard, A., Bruun, D., Ajua-Alemanj, M., Pickart, C., Lein, P.J., 2008.
G.B., Cifra, H.L., Manlapaz, M.L., 2002. Prevalence of fetal exposure to environmental Chlorpyrifos and chlorpyrifos-oxon inhibit axonal growth by interfering with the
toxins as determined by meconium analysis. Neurotoxicology 23, 329–339. morphogenic activity of acetylcholinesterase. Toxicol. Appl. Pharmacol. 228, 32–41.

You might also like