Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

NeuroToxicology 25 (2004) 349–363

In Vivo and In Vitro Effects of Acrylamide on


Synaptosomal Neurotransmitter
Uptake and Release
Richard M. LoPachin*, Aron I. Schwarcz, Christopher L. Gaughan,
Shirley Mansukhani, Soma Das
Department of Anesthesiology, Albert Einstein College of Medicine, Montefiore Medical Center,
Moses Research Tower-7, 111 E. 210th Street, Bronx, NY 10467, USA
Received 28 January 2003; accepted 14 July 2003

Abstract

Evidence suggests acrylamide (ACR) neurotoxicity is mediated by impaired presynaptic transmission. To assess the
effects of ACR on nerve terminal function, [3 H]glutamate release and uptake were determined in brain synaptosomes
isolated from intoxicated rats (50 mg/kg per day  8 days, i.p. or 21 mg/kg per day  21 days, p.o.). Regardless of ACR
dose-rate, a significant reduction in synaptosomal Kþ-stimulated, Ca2þ-dependent release was detected, whereas kinetic
analysis of Naþ-dependent uptake did not reveal consistent changes. Immunoblot analysis showed normal protein levels
(e.g. SNAP-25) in dysfunctional synaptosomes isolated from ACR-intoxicated rats. This suggests that defective release
does not involve changes in protein synthesis and/or anterograde delivery of presynaptic constituents. To identify
potential targets, synaptosomes were exposed in vitro to [14 C]-ACR and radiolabeled proteins were separated by
gel electrophoresis and detected by autoradiography. [14 C]-ACR labeling of distinct synaptosomal protein bands
(10.5–154,000 kDa) was blocked by the sulfhydryl alkylating agent, N-ethylmaleimide (NEM; 4 mM) but not by the non-
neurotoxic structural analog propionamide (10 mM). In vitro characterization of synaptosomal [3 H]glutamate uptake
and release showed that ACR, NEM and iodoacetic acid (IAA) produced concentration-dependent decreases in each
parameter that were highly correlated to reductions in free sulfhydryl content. All three chemicals were equiefficacious
with respect to reducing sulfhydryl content and neurotransmitter uptake/release, although the relative potencies differed;
NEM > IAA > ACR. Kinetic analysis of uptake showed that in vitro exposure to ACR, IAA or NEM at their respective
IC50’s caused similar reductions in Vmax. These data suggest that ACR-induced synaptic dysfunction involves adduction of
presynaptic protein thiol groups and subsequent reduction in neurotransmitter release.
# 2003 Elsevier Inc. All rights reserved.

Keywords: Acrylamide; Distal axonopathy; Synaptosomes; Neurotransmitter release; Uptake;


Protein thiol groups; SNARE core complexes

INTRODUCTION ore processing, dye synthesis) and in laboratories for


the electrophoretic separation of macromolecules
Acrylamide (ACR) is a water-soluble, vinyl mono- (Gold and Schaumburg, 2000; Spencer and Schaum-
mer that is used primarily to produce polyacrylamides burg, 1974a; U.S. Environmental Protection Agency,
with different physical and chemical properties. These 1988). ACR monomer is also a contaminant formed by
polymers are used extensively in various chemical an as yet unidentified mechanism during the high-
industries (e.g. water and wastewater management, temperature preparation of certain potato- or grain-
based foods (Tareke et al., 2000). Although the poly-
*
Corresponding author. Tel.: þ1-718-920-5054; mer is non-neurotoxic, long-term, low-level intoxica-
fax: þ1-718-515-4903. tion with monomeric ACR produces ataxia and skeletal
E-mail address: lopachin@aecom.yu.edu (R.M. LoPachin). muscle weakness in occupationally exposed humans

0161-813X/$ – see front matter # 2003 Elsevier Inc. All rights reserved.
doi:10.1016/S0161-813X(03)00149-9
350 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

and in experimental animal models (LeQuesne, 1980, trophysiological studies by Lowndes and Goldstein
1985; LoPachin et al., 2002a; Spencer and Schaum- revealed a defect in neurotransmission at primary
burg, 1974a). Early morphological studies suggested afferent terminals (PAT) in spinal cord of ACR-intoxi-
that this neurotoxicity was mediated by nerve damage cated cats (DeRojas and Goldstein, 1987; Goldstein,
classified as a central–peripheral distal axonopathy 1985; Goldstein and Lowndes, 1979, 1981; Lowndes
(LoPachin and Lehning, 1994; Spencer and Schaum- et al., 1978a,b; see also Tsujihata et al., 1974). This
burg, 1974b; 1976, 1980; Tilson, 1981). Distal axon effect developed prior to PAT degeneration, failure of
swelling and subsequent degeneration were considered corresponding somatosensory receptor function (i.e.
to be the hallmark morphological features of this toxic muscle spindles) and before the onset of neurological
axonopathy and, accordingly, research over the past 25 symptoms. The authors suggested that defective neu-
years has focused on putative axonal sites of action rotransmission might be related to impaired transmitter
(reviewed in LoPachin and Lehning, 1994; LoPachin release and might involve changes in presynaptic
et al., 2002a; Miller and Spencer, 1985; Sabri and uptake, synthesis, or vesicle storage (Goldstein,
Spencer, 1980). However, other contemporary research 1985; Goldstein and Lowndes, 1979, 1981). As mor-
demonstrated that ACR can produce neurological toxi- phological correlates of presynaptic dysfunction, sev-
city in the absence of axonopathy; i.e. whereas equiva- eral studies have shown that peripheral and central
lent neurotoxicity can be induced by intoxication over nerve terminals in ACR-intoxicated laboratory animals
a wide range of daily dose-rates (e.g. 10–50 mg/kg per exhibited early reductions in docked synaptic vesicles
day  28 days), axon degeneration in PNS and (DeGrandchamp and Lowndes, 1990; DeGrandchamp
CNS occurred only during long-term exposure to lower et al., 1990; Prineas, 1969; Tsujihata et al., 1974).
ACR dose-rates (e.g. 21 mg/kg per day  42 days; Moreover, lengthening of the postjunctional endplate
Cavanagh, 1982a; Crofton et al., 1996; Lehning et al., in PNS and increases in receptor number and affinity in
1998, 2002a,b, 2003; LoPachin et al., 2002b). The CNS also occurred and are indicative of postsynaptic
dissociation between neurological dysfunction and denervation hypersensitivity (Agrawal et al., 1981a,b;
the presumed underlying morphological lesion suggests Bondy et al., 1981; DeGrandchamp and Lowndes,
that axonopathy might not be importantly involved 1990; DeGrandchamp et al., 1990; Uphouse and Rus-
in the pathophysiological process leading to ACR neu- sell, 1981). Based on the early development of these
rotoxicity (see reviews by LoPachin et al., 2000, 2002a, structural and functional defects, we have hypothesized
2003). that synaptic dysfunction and eventual nerve terminal
Nerve terminals have been suggested as an alter- degeneration mediate ACR neurotoxicity (LoPachin
native site of ACR action based on early structural and et al., 2002a, 2003). Therefore, as an initial investiga-
functional changes as in PNS and CNS (reviewed in tion of potential presynaptic mechanisms, we have
LoPachin et al., 2002a, 2003). Recently completed determined the in vivo and in vitro effects of ACR
morphological studies (Lehning et al., 2002a,b, exposure on KCl-evoked, Ca2þ-dependent neurotrans-
2003) using a contemporary amino-cupric silver stain- mitter release and on high affinity, Naþ-dependent
ing method to detect degenerating neurons and their uptake in rat brain synaptosomes.
processes (i.e. dendrites, axons, nerve terminals) indi-
cated that intoxication at a higher ACR dose-rate
(50 mg/kg per day  5–11 days) produced selective, MATERIALS AND METHODS
widespread degeneration of nerve terminals in rat brain
and spinal cord regions. Intoxication at a lower dose- Chemicals
rate (21 mg/kg per day  7–38 days) was associated
with initial nerve terminal degeneration followed later ACR (99% purity), propionamide (97% purity), N-
by preterminal axon degeneration. These findings are ethylmaleimide (NEM), iodoacetic acid (IAA, 98%
consistent with other evidence suggesting that, regard- purity), 5,50 -dithiobis(2-nitrobenzoic acid) (DTNB)
less of dose-rate, nerve terminal degeneration was and Percoll (for formation of density gradients) were
an initial consequence of ACR intoxication in both all purchased from the Sigma (St. Louis, MO). [14 C]-
CNS (Cavanagh, 1982a,b; Ghetti et al., 1973; Prineas, ACR (specific activity 5 mCi/mmol) and L-[3 H]gluta-
1969; Spencer and Schaumburg, 1977a,b) and PNS mate (specific activity 60 Ci/mmol) were obtained from
(Cavanagh, 1982a; DeGrandchamp and Lowndes, American Radiolabeled Chemicals (St. Louis, MO).
1990; DeGrandchamp et al., 1990; Schaumburg Antibodies against syntaxin 1 (mouse monoclonal),
et al., 1974; Tsujihata et al., 1974). A series of elec- growth-associated protein of 43 kDa (GAP-43; mouse
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 351

monoclonal) and synatophysin (mouse monoclonal) 4 ¼ severely abnormal gait (inability to support body
were from Calbiochem (SanDiego, CA). Antibodies weight and foot splay). For each ACR dosing regimen,
to synaptobrevin (VAMP; mouse monoclonal) and groups of age-matched control rats (n ¼ 4 rats per
b-tubulin (mouse monoclonal) were obtained from group) were weighed and gait scores were determined.
Chemicon International Inc. (Temecula, CA). Antibo- Control rats for the higher ACR dose-rate group received
dies for synaptosomal-associated protein of 25 kDa daily i.p. injections of 0.9% saline (3 ml/kg). A trained,
(SNAP-25; rabbit polyclonal) and rabphilin 3a (Rab- blinded observer who was not involved in animal care or
3a, rabbit polyclonal) were purchased from Affinity ACR exposure performed the testing.
BioReagents (Golden, CO) and Zymed (San Francisco,
CA), respectively. Preparation of Synaptosomes

Animals and ACR Intoxication Depending upon study conditions, synaptosomes


were isolated from cortical and diencephalon-midbrain
All aspects of this study were in accordance with the regions of brains from ACR-intoxicated rats and their
NIH Guide for Care and Use of Laboratory Animals age-matched controls or from brains of normal rats
and were approved by the Montefiore Medical Center (250–275 g). Previous silver degeneration studies of
Animal Care Committee. Adult male rats (Sprague– ACR-intoxicated rats have shown that these brain
Dawley, 250–275 g; Taconic Farms, Germantown, regions exhibit progressive nerve terminal degenera-
NY) were used in this study. Rats were housed indi- tion (Lehning et al., 2002b, 2003). For each dose-rate
vidually in polycarbonate boxes, and drinking water (50 mg/kg per day  8 days; 21 mg/kg per day  21
and Purina Rodent Laboratory Chow (Purina Mills days), the selected experimental time-points preceded
Inc., St. Louis, MO) were available ad libitum. The the onset of frank nerve terminal degeneration (Lehn-
animal room was maintained at approximately 22 8C ing et al., 2002b, 2003). Synaptosomes were prepared
and 50% humidity with a 12 h light/dark cycle. For by the Percoll gradient method of Dunkley et al.
studies using synaptosomes isolated from different (1988). In brief, brains were rapidly removed and
brain regions of intoxicated rats (see below), randomly specific tissue regions were dissected and minced in
assigned groups of animals (four to six rats per expo- cold (4 8C) buffer containing sucrose 0.32 M, EDTA
sure group) were exposed to ACR at daily dose-rates of 1 mM and dithiothreitol 0.25 mM (SED gradient buf-
either 50 mg/kg per day  8 days (i.p.) or 21 mg/kg per fer; pH 7.4). Tissue was gently homogenized in SED
day  21 days (p.o.). These daily dose-rates and cor- buffer (10 passes in a Teflon-glass homogenizer;
responding routes have been used in mechanistic stu- 700 rpm) and the resulting homogenate was centri-
dies conducted over the past 30 years and are well fuged at 1000  g (10 min, 4 8C). The pellet (P1) was
characterized with respect to neuropathological washed once and supernatants (S1 and S2) were com-
expression (quantitative morphometrics and silver bined. Protein content of the pooled supernatant was
stain analysis), neurological deficits (gait, grip determined by the Bradford assay using bovine serum
strength, hindlimb extensor thrust, foot splay) and albumin as standard. The protein concentration of the
toxicokinetics (Barber et al., 2001; LoPachin et al., supernatant was adjusted with SED to 5 mg/ml and
2002b; Lehning et al., 1998, 2002a,b, 2003; reviewed then layered on top of a freshly prepared four-step
in LoPachin and Lehning, 1994; LoPachin et al., 2000, discontinuous Percoll gradient (3, 10, 15 and 23%
2003). In the present study, body weight and gait scores Percoll in SED, pH 7.4). Gradients were centrifuged
were determined two to three times per week as indices at 32,000  g for 6 min and synaptosomes were col-
of developing neurotoxicity. Gait scoring involved lected at the last interface (15/23%). Characterization
observation of spontaneous open field locomotion, of the Percoll method demonstrated a relative enrich-
which included evaluations of ataxia, hopping, rearing ment of synaptosomal markers (syntaxin 1, synapto-
and hindfoot placement (LoPachin et al., 2002b). To brevin) at the collection interface (data not shown).
assess locomotion, rats were placed in a clear plexiglass Synaptosomes were washed twice in oxygenated
box (90 cm  90 cm) and were observed for 3 min. Kreb’s buffer containing NaCl 140 mM, KCl 5 mM,
Following observations, a gait score was assigned NaHCO3 5 mM, MgCl2 1 mM, NaH2PO4 1.2 mM,
from 1 to 4 where: 1 ¼ a normal gait; 2 ¼ a slightly glucose 10 mM and Hepes 10 mM (pH 7.4), pelleted
abnormal gait (slight ataxia, hopping gait and foot and then resuspended in oxygenated Kreb’s buffer.
splay); 3 ¼ moderately abnormal gait (obvious ataxia ACR intoxication of rats did not alter the final synap-
and foot splay with limb abduction during ambulation); tosomal protein yield; i.e. 21 mg/kg per day  21
352 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

days ¼ 8:74  0:64 mg protein/ml; 50 mg/kg per NaH2PO4 1 mM, CaCl2 2.5 mM, glucose 10 mM,
day  8 days ¼ 8:47  0:66 mg protein/ml; combined Hepes 25 mM, pH 7.4) and, following an equilibration
age-matched control ¼ 7:96  0:96 mg protein/ml. period (60 min), four 2 min fractions were collected as
a measure of basal [3 H]-neurotransmitter efflux. Ca2þ-
Binding of [14C]-ACR to Synaptosomal Proteins dependent, [3 H]glutamate release was stimulated by a
90 s pulse of 40 mM KCl in modified Krebs buffer.
As an initial investigation of putative targets and Preliminary studies established the optimal pulse dura-
chemical mechanisms, [14 C]-ACR–protein binding tion, KCl concentration and Ca2þ-dependency of
was characterized in synaptosomes prepared from evoked neurotransmitter release from synaptosomes
cerebrocortical or midbrain-diencephalon regions of (data not shown). Superfusate fractions were collected
control rat brains. Synaptosomes (30 mg protein) were at 2 min intervals (1.2 ml total volume) throughout the
preincubated for 30 min at 37 8C in oxygenated control entire experiment; i.e. prestimulus, stimulus and post-
Krebs–Hepes buffer or in buffer containing either the stimulus periods. The isotope contents of each col-
sulfhydryl alkylating agent, N-ethylmaleimide (4 mM) lected fraction and of the corresponding filter-trapped
or the non-neurotoxic ACR analog, propionamide synaptosomes were determined by scintillation count-
(10 mM). At the end of the preincubation period, ing. [3 H]glutamate exocytosis was calculated as per-
[14 C]-labeled ACR (7 mCi per tube or 58 nM ACR) cent fractional release, which is integrated area under
was added to all tubes, which were then incubated at the release curve (AURC) divided by total synaptoso-
37 8C for 60 min. Following incubation, samples were mal counts (filter þ effluent fractions). The effects of
snap-frozen in liquid nitrogen and stored at 180 8C ACR on neurotransmitter release were expressed as
until analyzed. For each synaptosomal sample, labeled mean percent of control  S:E:M: and mean group
and unlabeled proteins were resolved by sodium dode- data were analyzed statistically (P < 0:05) by a paired
cyl sulfate-polyacrylamide gel electrophoresis (SDS- Student’s t-test.
PAGE; 7.5, 10.0 or 16.5% gels) and then transferred Our studies of synaptosomes derived from brains or
overnight to nitrocellulose membranes. Autoradio- ACR-intoxicated rats revealed a significant defect in
graphs of radiolabeled proteins were obtained by neurotransmitter release (see ahead). To investigate the
exposure of the dried membranes to X-ray film (Super possibility that this effect was due to decreased synth-
RX Fuji film) for 6 weeks (80 8C). Molecular weights esis and/or impaired kinesin-based fast anterograde
(kDa) of radiolabeled proteins were estimated based on axonal transport of presynaptic proteins involved in
the migration of standard proteins. Fig. 2 shows a the release process (Sickles et al., 2002), the levels of
representative PAGE (7.5% gel) and corresponding selected proteins were determined in synaptosomes
autoradiographic evidence from a total of three sepa- isolated from brain regions of ACR-intoxicated rats
rate trials. (see above ‘‘Animals and ACR Intoxication’’). This
experimental approach was based on previous studies
Determination of Synaptosomal L-[3H]glutamate showing that impairment of rapid axonal transport
Release, Free Sulfhydryl Concentrations and (e.g. antisense-reduced kinesin synthesis; kif1Bþ/
Protein Content knockout) resulted in decreased delivery of proteins
(e.g. synaptophysin, synaptotagmin) to distal axons and
Effects of In Vivo ACR on Neurotransmitter nerve terminals as detected by immunoblot analysis
Release and Protein Content (e.g. see Amaratung et al., 1995; Zhao et al., 2001).
Synaptosomes were prepared from brains of Synaptosomal proteins were resolved by SDS-PAGE
ACR-intoxicated rats or their age-matched controls and then transferred to nitrocellulose membranes.
(see above ‘‘Preparation of Synaptosomes’’) and were After transfer, membranes were blocked with 5.0%
loaded with L-[3 H]glutamate (10 mCi per tube or dried non-fat milk in TBS (Tris–HCl 20 mM, NaCl
167 nM glutamate) for 10 min at 36 8C. Labeled 0.5 M, pH 8.3) for 45 min and then rinsed. Membranes
synaptosomes (250 mg protein) were trapped on glass were incubated for 2 h at 25 8C with the appropriate
fiber filters (Whatman GF/B filter disc) in a 12-well primary antibody diluted in 5.0% dried milk/TBS.
superfusion chamber (Brandel Suprafusion 1000 sys- Following primary antibody incubation, membranes
tem, Gaithersburg, MD) interfaced with a fraction were washed in TBS and incubated for 1 h at 25 8C
collector. Synaptosomes were superfused at 0.6 ml/ with an appropriate alkaline phosphatase-conjugated
min with oxygenated (95% O2/5% CO2) Krebs–Hepes secondary antibody. Membranes were washed with
buffer (NaCl 124 mM, MgSO4 2.4 mM, KCl 4 mM, TBS and bound secondary antibody was visualized
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 353

with the ProtoBlot II AP system (Promega, Madison, dence intervals were calculated by the Cheng–Prusoff
WI). Immunoreactive synaptosomal protein bands were equation and were compared statistically by Student’s
scanned with a densitometer, digitized and quantified t-test (PrismTM 3.0, GraphPad Software, San Diego,
using the NIH Imaging Program. Statistical differences CA). The effects of each chemical on free sulfhydryl
(P < 0:05) between control and experimental group groups and [3 H]glutamate release were compared by
means were determined by one-tailed Student’s t-test linear regression analysis. Corresponding coefficients
(InStatTM, GraphPad Software, San Diego, CA). Data of determination (r2) were calculated from the Pearson
are expressed as mean percent of control  S:E:M. correlation coefficient (InStatTM 3.0, GraphPad Soft-
ware, San Diego, CA).
Effects of In Vitro ACR, Propionamide and
Sulfhydryl Reagents on Neurotransmitter Release Determination of Synaptosomal Glutamate
and Free Sulfhydryl Concentrations Uptake and Kinetic Analysis
As a potential model for exploring the mechanism of
nerve terminal dysfunction, the in vitro concentration- Effects of In Vivo ACR on Neurotransmitter
dependent effects of ACR and two sulfhydryl reagents Uptake
(N-ethylmaleimide or iodoacetic acid, IAA) on synap- Glutamate uptake was measured in synaptosomes
tosomal Ca2þ-dependent, KCl-evoked [3 H]glutamate isolated from ACR-intoxicated rats and their age-
release were determined. Synaptosomes were prepared matched controls (see above ‘‘Animals and ACR Intox-
from control rats, equilibrated at 36 8C in oxygenated ication’’) according to the method of Robinson et al.
Krebs–Hepes buffer and then loaded with [3 H]gluta- (1991). Synaptosomes were trapped on filters in a
mate and placed in a superfusion chamber as described superfusion system, equilibrated at 36 8C and then
above. Following equilibration, synaptosomes were superfused with Krebs–Hepes buffer containing
exposed to varying concentrations of N-ethylmalei- labeled and unlabeled glutamate at final concentrations
mide, iodoacetic acid or ACR and, at the end of ranging from 1 to 500 mM. To correct for Naþ-inde-
toxicant exposure (30 min), [3 H]glutamate release pendent, low affinity transport (Robinson et al., 1991),
was evoked by a 90 s pulse of 40 mM KCl Krebs– uptake was measured in the presence and absence
Hepes buffer. Synaptosomes were also exposed in vitro (equimolar choline chloride substitution) of sodium
to equimolar concentrations of the structural analog ions. Following the uptake period (3 min), filter-
propionamide. Fractional release was calculated as trapped synaptosomes were washed (5 min) with ice-
previously described. In parallel studies, the concen- cold buffer and radioactivity was determined by liquid
tration-dependent effects of ACR, propionamide, NEM scintillation counting. Kinetic parameters (Km, Vmax)
or IAA on free synaptosomal sulfhydryl content were for synaptosomal glutamate uptake were determined
determined by the method of Ando and Steiner (1973). by nonlinear regression analysis (PrismTM, GraphPad
Following incubation with ACR, propionamide or the Software, San Diego, CA). Respective control and
sulfhydryl chemicals (see above in vitro exposure ACR kinetic parameters were compared statistically
protocol), synaptosomes (250 mg protein) were solu- (P < 0:05) by two-tailed Student’s t-test (InStatTM;
bilized with 1% SDS (5 min). 5,50 -Dithiobis(2-nitro- GraphPad Software, San Diego, CA).
benzoic acid) (3 mM) was added and, following
equilibration (5 min, 25 8C), absorbance was read at Effects of In Vitro ACR, Propionamide, NEM
412 nm using a Jenway 6305 spectrophotometer. A and IAA on Neurotransmitter Uptake
reagent blank without DTNB was used to zero the To characterize effects of in vitro toxicant exposure,
spectrophotometer. The concentration of 3-carboxy- synaptosomes were incubated with varying concentra-
lato-4-nitrothiophenolate, the thiol anion released dur- tions of ACR, propionamide, NEM or IAA (see above
ing adduction of sulfhydryl groups by the disulfide exposure protocols for neurotransmitter release). Fol-
reagent DTNB, was calculated by the molar extinction lowing chemical incubation, Naþ-dependent synapto-
coefficient, 1:36  104 M1 cm1. Sulfhydryl data somal uptake was initiated by incubation with Krebs–
were calculated as nmol/mg synaptosomal protein Hepes buffer containing [3 H]glutamate (0.417 mM
and the effects of ACR, propionamide, NEM or IAA [3 H]glutamate). Uptake was terminated by rapid super-
were expressed as mean percent of control  S:E:M. fusion of the filter-trapped synaptosomes with normal
Both the release and sulfhydryl curves (Fig. 3A) were ice-cold Krebs–Hepes buffer and radioactivity was
fitted by nonlinear regression analysis (r2 for all determined by liquid scintillation counting. Coeffi-
curves > 0:98) and respective IC50’s and 95% confi- cients of determination, IC50’s and 95% confidence
354 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

intervals were calculated as described above. To deter- Table 1


mine how ACR and the other sulfhydryl reagents Effects of in vivo ACR intoxication on synaptosomal neurotrans-
mitter release
affected the kinetic parameters for in vitro glutamate
uptake, synaptosomes were exposed to the correspond- Parameter 21 mg/kg per 50 mg/kg per
ing IC50’s for each toxicant (i.e. ACR 187 mM; NEM day  21 days day  8 days
35 mM; IAA 9.5 mM) followed by incubation with AURC 65  5 49  8
varying glutamate concentrations (see above). Kinetic Fractional release 81  1 67  9
constants (Km, Vmax) were computed and statistically Synaptosomes were prepared from diencephalon-midbrain region of brains
compared as described above. from ACR-intoxicated rats or their age-matched controls (n ¼ 4–6 rats per
group). Isolated synaptosomes were loaded with [3 H]glutamate, placed in
a superfusion system and Ca2þ-dependent, release was stimulated by a 90 s
pulse of 40 mM KCl-containing Krebs buffer. Superfusate was collected
RESULTS continuously and radioactivity was determined by liquid scintillation
counting. Data are presented as mean percent of control  S:E:M: and ()
indicates significantly (P < 0:05) different from control. AURC: inte-
Neurological Evaluation grated area under the release curve (control ¼ 9023  436); fractional
release ¼ percent fractional release calculated as AURC/total counts
ACR intoxication at 50 mg/kg daily dose-rate pro- (control ¼ 21  1%).
duced a progressive loss in body weight and abnormal
changes in gait (LoPachin et al., 2002b). Rats in this In Vivo Effects of ACR on Synaptosomal Protein
dose-rate group had a mean (S.E.M.) starting body Content and Neurotransmitter Release and Uptake
weight of 274  4 g and after 8 days of ACR intoxica-
tion, mean weight decreased by 14% to 236  8 g. Gait We measured several parameters of Ca2þ-dependent,
scores for the higher exposure group increased from [3 H]glutamate release in synaptosomes prepared from
1:3  0:4 on day 5 to 2:3  0:1 on day 8 of intoxica- ACR-intoxicated and age-matched control rats. Mean
tion. This score represents statistically significant, (S.E.M.) fractional release evoked by superfusion of
slight-to-moderate changes in gait (see numerical gait control synaptosomes with a KCl (40 mM) pulse was
scales in ‘‘MATERIALS AND METHODS’’). During 21  1%, which is consistent with previous release
the same time period, age-matched, saline-injected studies (Nedvetsky et al., 2000). Results presented in
control rats exhibited no significant changes in gait Table 1 indicate that intoxication at either ACR dose-
(data not shown) and gained approximately 19% in rate produced significant decreases in percent release
body weight; i.e. starting body weight for the control and corresponding integrated area under the release
group was 266  3 g and after 8 days the body weight curve. Fig. 1 shows Naþ-dependent, [3 H]glutamate
increased to 316  4 g. ACR intoxication at the lower uptake in synaptosomes isolated from brain regions
daily dose-rate (21 mg/kg) slowed the normal rate of of ACR-intoxicated and age-matched control rats. The
daily weight gain and induced significant increases in hyperbolic substrate–velocity graphs for control data
gait scores (LoPachin et al., 2002b). Age-matched (Fig. 1A and B) demonstrates typical substrate satura-
control rats had a starting mean body weight of tion kinetics expected of Naþ-dependent, glutamate
269  3 g, which increased to 358  4 g by day 21 transport in synaptosomes (Robinson et al., 1991). The
of the exposure paradigm. This represents a 35% data could be closely fit to a Michaelis–Menten model
increase in body weight at a daily rate of approximately as indicated by an r2 value of 0.98. Intoxication at the
4.5 g per day. In contrast, rats intoxicated at the higher ACR dose-rate (Fig. 1A) was not associated
21 mg/kg dose-rate exhibited a significantly slower rate with statistically significant changes in uptake kinetic
of weight gain; i.e. rats in this exposure group had a parameters. The lower dose-rate (Fig. 1B) produced a
starting weight of 264  4 g and after 21 days of ACR significant decrease in Km but did not affect Vmax.
intoxication body weight rose to 312  5 g. This repre- Table 2 presents the levels of several proteins in
sents an 18%increase in weight at a daily rate of synaptosomes isolated from ACR-intoxicated rats.
approximately 2.3 g per day. Age-matched control rats Except for b-tubulin (see ahead), the proteins measured
did not exhibit changes in gait during the 21-day experi- in this study are delivered to nerve terminals by fast
mental period, whereas the gait score for ACR-intoxi- anterograde transport (FAT) (Li et al., 1995, 1996;
cated rats (21 mg/kg per day) increased steadily from an Shiff and Morel, 1997; Skene and Willard, 1981)
initial score of 1:1  0:1 to 2:8  0:2 on day 21 of and they participate in synaptic vesicle cycling/exocy-
exposure. This gait score represents a slight-to-moderate tosis (Calakos and Scheller, 1996; Lin and Scheller,
level of neurotoxicity. 2000). In contrast, b-tubulin is a polypeptide subunit of
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 355

200 (A) ACR (50mg/kg/d x 8d) Table 2


Relative protein contents of synaptosomes isolated from ACR-
intoxicated rats
nmol/mg/min

150
Protein 21 mg/kg per 50 mg/kg per
day  21 days day  8 days
100
Cerebral cortex
( )Control ( ) ACR b-Tubulins 89  7 96  11
50 Gap-43 121  11 111  7
V m ax 131.0 134.7
SNAP-25 105  9 103  5
Km 45.8 67.4
0
Rab-3a 94  7 109  8
0 100 200 300 400 500 600 Synaptophysin 103  4 101  10
Synaptobrevin 100  6 93  6
Gl ( M)
Midbrain-diencephalon
b-Tubulin 91  11 90  6
300 (B) ACR (21mg/kg/d x 21d) Gap-43 107  2 92  5
SNAP-25 111  11 103  4
250 Rab-3a 100  13 98  3
nmol/mg/min

Synaptophysin 104  9 95  5
200
Synaptobrevin 116  10 101  5
150 Synaptosomes were isolated from brain regions of ACR-intoxicated rats
and their age-matched controls (n ¼ 4–6 rats per group). Synaptosomal
100 proteins were separated by SDS-PAGE and were then transferred to
( )Control ( ) ACR
nitrocellulose membranes and probed with indicated antibody. Immunor-
50 V m ax 219.8 236.3
eactive bands were scanned by a densitiometer, digitized and quantified by
Km 129.6 70.8 * image analysis (NIH Imaging Program). Data are presented as mean
0 percent ofcontrol  S:E:M. Statistical analysis did not indicate statistically
0 100 200 300 400 500 600
significant (P < 0:05) changes in protein content relative to respective
Glu ( M) control. Gap-43: growth-associated protein of 43 kDa; SNAP-25: synap-
tosomal-associated protein of 25 kDa; Rab-3a: Rabphilin 3a.
Fig. 1. Kinetic analysis of high affinity, Naþ-dependent transport
of [3 H]glutamate by synaptosomes isolated from ACR-intoxicated
rat brains. Rats were intoxicated with ACR at two different daily
dose-rates [(A) 50 mg/kg per day, i.p. or (B) 21 mg/kg per day
p.o.] until a slight-to-moderate level of neurotoxicity was attained
(days 8 or 21, respectively). Graphed data in A and B are
presented as mean nmol glutamate/mg synaptosomal protein/
min  S:E:M. Corresponding kinetic analyses (Vmax ¼ nmol/mg;
Km ¼ mM) are provided in the insert table. Asterisk indicates
significantly different from age-matched control (P < 0:05).

cytoskeletal microtubules and is transported in slow


component a (SCa; Sullivan, 1988). Data in Table 2
indicate that, despite changes in neurotransmitter
release, neither ACR dosing-regimen altered synapto- Fig. 2. This figure shows autoradiographic patterns of [14 C]-
somal protein content as measured by immunoblot labeled synaptosomal proteins associated with different experi-
analysis. mental conditions. Lane 1: molecular weight standards; lane 2:
distribution of Coomassie blue-stained proteins in 7.5% poly-
In Vitro Chemical Adduction of Synaptosomal acrylamide gel, proteins were isolated from synaptosomes
incubated in control conditions; lane 3: polyacrylamide gel
Proteins (7.5%) showing the migration pattern of proteins from synapto-
somes incubated with ACR (10 mM); lanes 4 and 5: autoradio-
Lanes 2 and 3 of Fig. 2 show the electrophoretic graph showing labeled protein bands following incubation of
separation (7.5% gel) of proteins from cerebrocortical synaptosomes with [14 C]-ACR (58 nM, 7 mCi); lanes 6 and 7:
synaptosomes. Lane 3 is derived from synaptosomes autoradiograph showing an absence of labeled protein bands
following incubation of synaptosomes with NEM (4 mM) and
exposed to non-labeled ACR in vitro (10 mM  60 min).
[14 C]-ACR (dark band at the top of the autoradiograph represents
The comparable Coomassie blue staining patterns non-migrating [14 C]-ACR); lanes 8 and 9: autoradiograph showing
(compare lanes 2 and 3) indicate that in vitro ACR labeled protein bands following incubation of synaptosomes with
exposure did not affect migration of synaptosomal propionamide (10 mM) and [14 C]-ACR.
356 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

proteins through the gel. Additional studies showed (A) Release


110
that preincubation with NEM or propionamide also did 100

% Control ± SEM
not alter gel migration of synaptosomal proteins (data 90
not shown). The corresponding autoradiographs (lanes 80
70
4 and 5) show that [14 C]-ACR labeled several distinct 60 NEM IAA ACR
SH Content SH Content SH Content
protein bands ranging from an estimated MW of 10.5– 50 (31µM) (2.7mM) (356mM)
Release Release Release
154,000 kDa (Fig. 2). ACR labeling was selective since 40 (13µM) (2.1mM) (480mM)

many protein bands on the gels were not associated 30


20
with autoradiographic counterparts (e.g. lane 3; band at 10
31.7 kDa). Certain protein bands that were heavily 0
stained by Coomassie blue (e.g. lane 3; band at -8 -7 -6 -5 -4 -3 -2 -1 0 1
110,000 kDa) exhibited correspondingly dense [14 C]- Log Concentrations
labeled autoradiograph bands (lane 4). Other heavily
stained proteins (e.g. lane 3; band at 38,000 kDa) were 100 (B) Regression Analysis
matched to lightly labeled autoradiographic bands 90

Fractional Release
80
(lane 4) suggesting limited ACR binding affinity for
r 2 = 0.9594

% Control
70
these proteins. When synaptosomes were preincubated
60
with the sulfhydryl alkylating agent, NEM, followed 50
by [14 C]-ACR exposure, the corresponding autoradio- 40
graphs (lanes 6 and 7) lacked protein bands (compare 30
lanes 5 and 6) and a broad dark stain was present at the 20
top of the respective lanes. Corroborative gel electro- 10
phoresis studies with [14 C]-ACR alone (no protein) 0
0 10 20 30 40 50 60 70 80 90 100
revealed similar positioning of a dark stain on corre-
sponding autoradiographs indicating that unbound % Control SH Content
radiolabel did not migrate during electrophoresis (data Fig. 3. Concentration-dependent effects of NEM, IAA and ACR
not shown). This, and the observation that NEM on synaptosomal KCl-evoked, Ca2þ-dependent [3 H]glutamate
adduction does not affect the gel migration pattern release and free sulfhydryl group content are presented in (A)
(see above), suggest that preincubation with NEM- Data are presented as mean percent control  S:E:M:; control free
sulfhydryl content: 88  14 pmol/mg synaptosomal protein; con-
blocked binding of [14 C]-ACR to synaptosomal pro- trol fractional release: 21  1%. Calculated IC50’s are provided in
teins. Accordingly, the dark band at the top of lanes 6 parentheses. Regression analysis of pooled neurotransmitter
and 7 represents unbound, non-migrating [14 C]-ACR. release and sulfhydryl content data is presented in (B). Coefficient
In contrast, preincubation with propionamide, a non- of determination (r2) is provided and dashed lines denote the 95%
neurotoxic structural analog of ACR, did not affect confidence interval.
ACR–protein binding; i.e. compare lanes 5 and 8
(Fig. 2). Similar results were obtained when the same sulfhydryl contents. In contrast, propionamide did
[14 C]-ACR incubation conditions were followed by not affect synaptosomal [3 H]glutamate release or sulf-
separation of synaptosomal proteins on 10 or 16.5% hydryl content (data not shown). Specifically, ACR
gels (n ¼ 3 per gel; data not shown). exposure produced a concentration-dependent decrease
in neurotransmitter release that was nearly identical to
In Vitro Effects of ACR, Propionamide, NEM or the effects on sulfhydryl content over a wide concen-
IAA on Synaptosomal Neurotransmitter Release tration range (Fig. 3A; 50 mM–2 M). Linear regression
and Free Sulfhydryl Groups analysis revealed that the reduction of synaptosomal
neurotransmitter release induced by in vitro ACR
In this series of studies, we determined the acute in exposure was closely correlated to the decrease in free
vitro effects of ACR, propionamide, NEM and IAA sulfhydryl groups (r 2 ¼ 0:9705). In addition, the
on both synaptosomal [3 H]glutamate release and free respective IC50’s for release and sulfhydryl content,
sulfhydryl content. Results (Fig. 3A) show that ACR, 480 mM versus 356 mM, did not differ significantly
NEM and IAA each produced concentration-depen- (Fig. 3A, P ¼ 0:9331). NEM was the most potent of
dent decreases in synaptosomal neurotransmitter the three chemicals tested and decreased both release
release and, over the same concentration ranges, all and sulfhydryl content over a concentration range of
chemicals produced highly correlated reductions in 100 nM–500 mM (Fig. 3A). The respective IC50’s for
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 357

the NEM-induced effects on release and sulfhydryl 110 (A) Uptake


content were not statistically different; i.e. 13 and 100

% Control ± SEM
31 mM (P ¼ 0:7162). The concentration-dependent 90
80
inhibition of [3 H]glutamate release induced by NEM
70
was closely correlated to the reduction of sulfhydryl 60 NEM IAA ACR
content (r 2 ¼ 0:9425). IAA was of intermediate 50 SH Content
(31µM)
SH Content
(2.7mM)
SH Content
(356mM)
potency and reduced both release and sulfhydryl con- 40 Uptake
(35µM)
Uptake
(9.5mM)
Uptake
(187mM)
tent over a dose-range of 10 mM to 100 mM (Fig. 3). 30
20
The respective IC50’s, 2.7 and 2.1 mM, did not differ
10
statistically (P ¼ 0:6117), and the effects of IAA on 0
release and sulfhydryl content were highly correlated -8 -7 -6 -5 -4 -3 -2 -1 0 1
(r 2 ¼ 0:9625). Linear regression analysis of the pooled Log Concentrations
release and sulfhydryl content data for each chemical
tested indicated a high degree of correlation among the (B) Regression Analysis
100
respective data sets (Fig. 3B; r 2 ¼ 0:9594). 90

% Control Uptake
80
In Vitro Effects of ACR, Propionamide, NEM or 70 r2= 0.9266
IAA on Synaptosomal Neurotransmitter Uptake 60
and Free Sulfhydryl Groups 50
40
Our analysis indicates that ACR, NEM and IAA 30
each produced concentration-dependent decreases in 20
synaptosomal glutamate uptake, whereas propiona- 10
mide was without effect (data not shown). Fig. 4A 0
0 10 20 30 40 50 60 70 80 90 100
shows that in vitro exposure of synaptosomes to ACR
produced progressive decreases in both Naþ-depen- % Control SH Content
dent, [3 H]glutamate uptake and free sulfhydryl content Fig. 4. Concentration-dependent effects of NEM, IAA and ACR
over the same concentration range. The respective on synaptosomal high affinity, Naþ-dependent [3 H]glutamate
IC50’s, 356 and 187 mM, did not differ statistically uptake and free sulfhydryl group content are presented in (A).
(P ¼ 0:0634). Moreover, the concentration-dependent Data are presented as mean percent control  S:E:M:; control free
reduction in free sulfhydryl groups produced by ACR sulfhydryl content ¼ 88  14 pmol/mg synaptosomal protein;
control uptake ¼ 57:4  6:1 pmol glutamate/mg protein/min.
was closely correlated to the decrease in uptake Calculated IC50’s are provided in parentheses. Regression analysis
(r 2 ¼ 0:9705). NEM produced a graded reduction of of pooled neurotransmitter release and sulfhydryl content data is
both synaptosomal [3 H]glutamate uptake and sulfhy- presented in (B). Coefficient of determination (r2) is provided and
dryl groups over the same concentration range dashed lines denote the 95% confidence interval.
(Fig. 4A). The respective IC50’s were 35 and 31 mM
and were not statistically different (P ¼ 0:7162). Like the respective IC50’s for each chemical and transport
ACR, the concentration-dependent reduction in sulf- was measured as a function of [3 H]glutamate concen-
hydryl content induced by NEM was closely correlated tration. Results shown in Fig. 5 indicate that each
to the progressive inhibition of [3 H]glutamate uptake chemical produced a significant comparable decrease
(r 2 ¼ 0:9661). The ability of IAA to produce concen- in Vmax, whereas, with the exception of NEM, these
tration-dependent inhibition of uptake was also similar chemicals did not affect Km.
to the corresponding relative potency for reduction of Data presented in Figs. 3A and 4A indicate that in
sulfhydryl content (Fig. 4A). The respective IC50’s, 9.5 vitro exposure of synaptosomes to ACR, NEM or IAA
and 2.7 mM, did not differ statistically (P ¼ 0:0652) caused graded, concentration-dependent decreases in
and the effects of IAA on sulfhydryl content and uptake synaptosomal neurotransmitter release, uptake and free
were highly correlated (r2 ¼ 0:9276). When the sulf- sulfhydryl groups. For each parameter measured, the
hydryl and uptake data for ACR, NEM and IAA were rank order of respective IC50’s for these chemicals (e.g.
pooled and analyzed by linear regression, the r2 value Fig. 3A) indicates relative differences in potency; i.e.
was 0.9266 (Fig. 4B). To determine how in vitro NEM > IAA > ACR. However, it is important to note
exposure to ACR, NEM and IAA affected the kinetic that NEM, IAA and ACR were equivalent with respect
properties of uptake, synaptosomes were exposed to to maximal efficacy; i.e. all chemicals were capable of
358 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

300 fast anterograde transport of materials to distal axon


regions (Cavanagh, 1964, 1979; Sickles et al., 1996;
250
Stone et al., 2000). Subsequent depletion of nerve
nmol/mg/min

200 terminal components critically involved in the synaptic


vesicle cycle (e.g. SNAP-25, synaptobrevin) could be
150
responsible for synaptic dysfunction. Based on this
100 possibility, we measured the levels of several proteins
V m ax K m
Control 259.7 55.33 in release incompetent synaptosomes isolated from
50 ACR 122.8 * 27.83
NEM 124.7* 16.47 * different brain regions of ACR-intoxicated rats.
IAA 154.4* 45.81
0 Although these proteins were synthesized in the nerve
0 100 200 300 400 500 cell body and the majority were delivered to nerve
Glu (µM) terminals by FAT, we found no changes in correspond-
ing content (Table 2). These findings are consistent
Fig. 5. Kinetic analysis of high affinity, Naþ-dependent transport
of [3 H]glutamate by synaptosomes exposed in vitro to the res-
with previous research from this and other laboratories,
pective IC50 concentrations of ACR, NEM or IAA. Graphed data which showed that induction of ACR neurotoxicity was
are presented as mean nmol glutamate/mg synaptosomal protein/ not associated with significant changes in peripheral
min  S:E:M. Corresponding kinetic analyses (Vmax ¼ nmol/mg; nerve axonal Naþ/Kþ-ATPase protein content/activity,
Km ¼ mM) are provided in the insert table. Asterisk indicates subaxonal ion regulation or other distal axon para-
significantly different from age-matched control (P < 0:05).
meters that are dependent on FAT for supply of critical
components (reviewed in LoPachin, 2002; LoPachin
reducing sulfhydryl group content below detectable et al., 2000, 2002a,b, 2003). If impaired synthesis and/
limits and completely blocking neurotransmitter or fast anterograde transport were pathophysiologi-
release and uptake (e.g. Fig. 3A). cally important, secondary alterations in these para-
meters should have occurred. The absence of observed
changes suggests that ACR does not disrupt perikaryal
DISCUSSION or axonal processes and might, instead, cause presy-
naptic dysfunction by acting directly at nerve terminal
In Vivo Effects of ACR on Synaptosomal sites (see ahead).
Function and Protein Content
Identification of Putative Nerve Terminal
Data from electrophysiological, morphological and Protein Targets
neurochemical studies implicate defective neurotrans-
mission at central and peripheral synapses of ACR- To identify possible nerve terminal targets and che-
intoxicated animals (reviewed in LoPachin et al., mical interactions, we characterized [14 C]-ACR label-
2002a, 2003). Goldstein and Lowndes (1979, 1981) ing of synaptosomal proteins. Results show that certain
have suggested that this defect might be mediated by protein bands were variably labeled by in vitro expo-
changes in presynaptic transmitter uptake, synthesis, sure to [14 C]-ACR. Preincubation of synaptosomes
vesicle storage or release. As an initial investigation we with the sulfhydryl-alkylating agent NEM completely
focused on neurotransmitter uptake and release as prevented radiolabeling of proteins, which suggests
possible ACR targets. Results show that Kþ-evoked, that ACR–protein interactions were mediated by thiol
Ca2þ-dependent glutamate release was significantly adduction. Although the neurotoxicological relevance
decreased in synaptosomes isolated from ACR-intoxi- has been questioned (Hashimoto and Aldridge, 1970;
cated rats, whereas the kinetic parameters of high Martenson et al., 1995a,b), adduction of protein thiol
affinity, Naþ-dependent glutamate uptake were not groups is, nonetheless, consistent with the chemical
consistently affected (see ahead). These findings there- structure of ACR. ACR is an a,b-unsaturated carbonyl
fore offer a possible explanation for ACR-induced chemical with electrophilic reactivity at the carbonyl
defective neurotransmission (decreased presynaptic and b-alkene carbon atoms. As such, ACR will interact
release) and suggest an in vitro model system (evoked with nucleophilic centers via a carbonyl condensation
synaptosomal transmitter release) for mechanistic stu- reaction known as the Michael addition (Calleman,
dies (see ahead). It is possible that the observed 1996; Friedman, 1973; Kemp and Vellaccio, 1980). As
reduction in transmitter release was secondary to an electrophile, ACR could interact with biological
decreased perikaryal synthesis and/or kinesin-based nucleophilic centers formed by amines, imidazoles and
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 359

thiol groups. However, the nucleophilic reactivity of for this interaction. However, the ability to produce a
sulfur is significantly greater than that of either carbon, maximum effect (e.g. inhibition of transmitter release)
oxygen or nitrogen (Friedman, 1973; Kemp and equivalent to those of NEM or IAA denotes the full
Vellaccio, 1980) and, consequently, the preferential in efficacy of ACR (Fig. 3A). The low potency/high
vivo target nucleophile for ACR is thiol groups on, for efficacy of ACR is a critical character and indicates
example, protein cysteine residues (see review by Calle- that, although higher in vitro concentrations are needed
man, 1996). The relative affinity of ACR for sulfhydryl to promote protein adduction, the pathophysiological
groups has been demonstrated in previous in vivo and in outcome (i.e. inhibition of release) of the ACR adducts
vitro studies of protein adduct formation (Bergmark is comparable in magnitude to that induced by NEM–
et al., 1991, 1993; Calleman, 1996; Cavins and Fried- or IAA–protein adducts. This has significant implica-
man, 1968; Dixit et al., 1986; Sega et al., 1989) and by tions for in vivo toxicodynamics (see ahead).
ACR-inhibition of zinc iodide–osmium staining of pro- Whereas these in vitro characteristics implicate a
tein thiol groups at the rat neuromuscular junction specific neurotoxic action, it is nonetheless difficult to
(Kemplay and Cavanagh, 1984a,b; Kemplay et al., reconcile the relatively high ACR concentrations
1988). In our studies, preferential thiol adduction was needed to produce such in vitro effects (i.e. the ACR
demonstrated by NEM blockade of [14 C]-ACR protein IC50 for release is 480 mM) with the in vivo exposure
labeling and by ACR reduction of synaptosomal free conditions (i.e. the peak ACR plasma concentration
sulfhydryl groups. In addition, we found that propiona- after 11 days at 50 mg/kg is 422 mM; Barber et al.,
mide did not alter synaptosomal function nor free 2001). This disparity could be related to the relatively
sulfhydryl content. Propionamide is a non-neurotoxic low potency of ACR for sulfhydryl adduction and the
structural analog that lacks the unsubstituted alkene distinctive toxicodynamics of adduct formation during
function of ACR and has little electrophilic reactivity. in vivo and in vitro exposure. Specifically, regardless of
Together, these findings indicate that protein thiol exposure conditions, graded inhibition of transmitter
groups are the principal target nucleophile for ACR. release should occur as the fractional presynaptic
concentration of adducted, functionally impaired pro-
In Vitro Effects of ACR on Synaptosomal tein increases (Goldstein and Lowndes, 1979, 1981;
Neurotransmitter Release and Possible Hinson and Roberts, 1992). However, during acute in
Neurotoxicological Relevance vitro exposure of synaptosomes, high ACR concentra-
tions will be necessary to generate the requisite toxic
Sulfhydryl content and neurotransmitter release levels of adducted protein based on the lower sulfhy-
were determined following in vitro exposure of brain dryl potency of this toxicant and the law of chemical
synaptosomes to ACR or two sulfhydryl alkylating mass action. In contrast, in vivo intoxication is inher-
agents, NEM and IAA. Results show that all three ently more complex. Although ACR has low potency
compounds produced parallel, concentration-depen- for the formation of sulfhydryl adducts, the process is
dent decreases in neurotransmitter release that were an irreversible covalent reaction (see above). Therefore,
highly correlated to respective reductions in synapto- in vivo, detoxification or removal of this adduct will
somal-free sulfhydryl content (Fig. 3A and B). The depend upon the corresponding half-life of the protein.
finding that both in vivo and in vitro exposure to ACR For proteins with long half-lives, nerve terminal adduct
caused inhibition of neurotransmitter release suggests levels would increase during in vivo exposure by a
that the in vivo effect was due to the direct actions of cumulative process. Accordingly, many key regulatory
ACR at nerve terminal sites (see ahead). Furthermore, proteins associated with neurotransmitter release have
this correspondence indicates that in vitro inhibition relatively long half-lives (Calakos and Scheller, 1996)
represents a specific neurotoxicological effect with and are highly sensitive to inhibition by sulfhydryl
possible mechanistic relevance. Specificity is substan- alkylation (see ahead). These proteins would therefore
tiated further by the observation that release was be susceptible to additive, exposure-dependent adduc-
inhibited in a graded, concentration-dependent fashion tion by ACR and subsequent functional inhibition. Low
(Fig. 3A). The sequential and parallel nature of the sulfhydryl potency and cumulative protein adduction are
concentration–response curves for NEM, IAA and consistent with the in vivo neuropathokinetics of ACR;
ACR implies that these compounds interact with a i.e. relatively high mg/kg daily dose-rates and extended
common sulfhydryl-regulated component of the pre- exposure are required for cumulative induction of neu-
synaptic release mechanism. The rank order of these rological deficits in laboratory animals (see Goldstein
curves indicates that ACR has relatively low potency and Lowndes, 1979, 1981; Kuperman, 1958; LoPachin
360 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

et al., 2002b). Resolving the in vivo–in vitro disparity is neurotransmission identified in previous electrophy-
important for validating our in vitro model system and siological studies of intoxicated laboratory animals
for understanding the mechanism of ACR neurotoxicity. (e.g. Goldstein and Lowndes, 1979, 1981; Tsujihata
Accordingly, we are currently using mass spectroscopy et al., 1974) was mediated by a direct effect of ACR at
techniques to assess synaptosomal protein adduct presynaptic sites. Although specific targets have not
formation during in vivo and in vitro ACR exposure. been identified, many proteins involved in neurotrans-
The above scenario is analogous to that proposed for mitter release are thiol-rich and are therefore putative
2,5-hexanedione (HD); i.e. HD produces cumulative candidates for ACR adduction. Indeed, neurotransmit-
neurotoxicity in laboratory animals, which is presumed ter release has been shown to be highly sensitive to
to be mediated by cumulative adduction of long-lived inhibition by thiol alkylating chemicals such as NEM
cytoskeletal proteins, despite demonstrated low in vitro (Lonart and Sudhof, 2000; Nedvetsky et al., 2000; Pan
potency for this molecular effect (reviewed in LoPachin et al., 1995, 1996; Sequeira et al., 1997). The corre-
and Lehning, 1997). sponding molecular mechanism likely involves alkyla-
tion of SNAP-25 (synaptosomal-associated protein of
In Vitro Effects of ACR on Synaptosomal 25 kDa), N-ethylmaleimide-sensitive factor (NSF)
Neurotransmitter Uptake and Possible and/or other presynaptic cysteine-containing proteins
Neurotoxicological Relevance that regulate the exocytotic process (Meffert et al.,
1996; Sollner et al., 1993; Tolar and Pallanck, 1998;
Our results have shown that in vitro exposure of Washbourne et al., 2001). Based on this evidence, we
synaptosomes to ACR, NEM or IAA inhibited Naþ- have proposed that ACR adducts sulfhydryl groups on
dependent L-glutamate uptake and this effect was key regulatory proteins (e.g. NSF) and thereby impairs
correlated to graded reductions in free sulfhydryl neurotransmitter release (LoPachin et al., 2002a,
groups. In brain, glutamate uptake is mediated by 2003). Our results also suggest that in vitro ACR
one neuronal (EAAC1) and two glial (GLT1, GLAST) exposure inhibited synaptosomal neurotransmitter
subtypes (Kanai and Hediger, 1992; Storck et al., 1992). uptake. If this effect is active in vivo, it might play
The Percoll gradient procedure used in the present study a role in promoting synaptic dysfunction. The present
produces a relatively homogeneous synaptosomal pre- studies provide a foundation for ongoing research
paration, although some glial contamination is expected designed to identify molecular targets and decipher
(Dunkley et al., 1988). Regardless, the EAAC1 subtype mechanisms of ACR-induced synaptic dysfunction.
contains two conserved cysteine residues (Bjoras et al., Finally, in addition to the obvious neurotoxicological
1996) that appear to function as sulfhydryl redox-sen- implications, the current findings are consistent with
sing sites and thereby regulate transmitter uptake (Trotti the rapidly developing concept that neurotransmission
et al., 1997a,b). Kinetic analysis revealed that the is regulated by redox modulation of cysteine sulfhydryl
respective IC50’s for ACR, NEM and IAA significantly groups on specific proteins (e.g. NSF; Broillet, 1999;
decreased the uptake Vmax, which is consistent with Gilbert, 1982; Stamler, 1994).
noncompetitive inhibition by irreversible adduction of
cysteine residues (Garret and Grisham, 1999). However,
the neurotoxicological relevance of inhibited in vitro ACKNOWLEDGEMENTS
uptake is unclear, since we did not observe correspond-
ing in vivo changes (see above). This reservation could Research presented in this manuscript was supported
be related to methodological problems such as a low in by a NIH grant from the National Institute of Environ-
vivo signal-to-noise ratio (e.g. a low percentage of mental Health Sciences (RO1 ES03830-17).
affected nerve terminals in a given brain region), which
make detection of subtle but mechanistically important
effects difficult. REFERENCES

Mechanistic Implications Agrawal AK, Seth PK, Squibb RE, Tilson HA, Uphouse LL,
Bondy SC. Neurotransmitter receptors in brain regions of
acrylamide-treated rats. I. Effects of a single exposure to
The present results suggest that ACR impairs synap- acrylamide. Pharmacol Biochem Behav 1981a;14:527–31.
tosomal neurotransmitter release, possibly through Agrawal AK, Squibb RE, Bondy SC. The effects of acrylamide
adduction of thiol groups on nerve terminal proteins. treatment upon the dopamine receptor. Toxicol Appl Pharmacol
These data support the proposal that disruption of 1981b;58:89–99.
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 361

Amaratung A, Leeman SE, Kosik KS, Fine RE. Inhibition of DeRojas TC, Goldstein BD. Primary afferent terminal function
kinesin synthesis in vivo inhibits the rapid transport of following acrylamide: alterations in the dorsal root potential
representative proteins for three transport vesicle classes into and reflex. Toxicol Appl Pharmacol 1987;88:175–82.
the axon. J Neurochem 1995;64:2374–6. Dixit R, Das M, Seth PK, Mukhtar H. Interaction of acrylamide
Ando Y, Steiner M. Sulfhydryl and disulfide groups of platelet with bovine serum albumin. Environ Res 1986;40:365–71.
membranes. I. Determination of sulfhydryl groups. Biochim Dunkley PR, Heath JW, Harrison SM, Jarvie PE, Glenfield PJ,
Biophys Acta 1973;311:26–37. Rostas AP. A rapid Percoll gradient procedure for isolation of
Barber D, Hunt JR, Ehrich M, Lehning E J, LoPachin RM. synaptosomes directly from an S1 fraction: homogeneity and
Metabolism, toxicokinetics and hemoglobin adduct formation morphology of subcellular fractions. Brain Res 1988;441:
in rats following subacute and subchronic acrylamide dosing. 59–71.
Neurotoxicology 2001;22:341–53. Friedman M. Nucleophilic additions. In: The chemistry and
Bergmark E, Calleman CJ, Costa LG. Formation of hemoglobin biochemistry of the sulfhydryl group in amino acids, peptides,
adducts of acrylamide and its epoxide metabolite glycidamide and proteins, chapter 4. New York: Pergamon Press; 1973.
in the rat. Toxicol Appl Pharmacol 1991;111:352–63. pp. 88–134.
Bergmark E, Calleman C, He F, Costa LG. Determination of Garret RH, Grisham CM. Biochemistry. New York: Harcourt
hemoglobin adducts in humans occupationally exposed to College Publishers; 1999. pp. 426–59.
acrylamide. Toxicol Appl Pharmacol 1993;120:45–54. Ghetti B, Wisneiwski HM, Cook RD, Schaumburg HH. Changes
Bjoras M, Gjesdal O, Erickson JD, Torp R, Levy LM, Ottersen OP, in the CNS after acute and chronic acrylamide intoxication. Am
Degree M, Storm-Mathisen J, Seeberg E, Danbolt NC. Cloning J Pathol 1973;70:78A.
and expression of a neuronal rat brain glutamate transporter. Gilbert HF. Biological disulfides: the third messenger? J Biol
Mol Brain Res 1996;36:163–8. Chem 1982;257:12086–112091.
Bondy SC, Tilson HA, Agrawal AK. Neurotransmitter receptors in Gold BG, Schaumburg HH. Acrylamide. In: Spencer PS,
brain regions of acrylamide-treated rats. II. Effects of extended Schaumburg HH, Ludolph AC, editors. Experimental and
exposure to acrylamide. Pharmacol Biochem Behav 1981;14: clinical neurotoxicology. 2nd ed. New York: Oxford University
533–7. Press; 2000. pp. 124–32.
Broillet M-C. S-Nitrosylation of proteins. Cell Mol Life Sci Goldstein BD. Acrylamide neurotoxicity: altered spinal
1999;55:1036–42. monosynaptic response to quipazine, a serotonin agonist in cats.
Calakos N, Scheller RH. Synaptic vesicle biogenesis, docking and Toxicol Appl Pharmacol 1985;78:436–44.
fusion: a molecular description. Physiol Rev 1996;76:1–29. Goldstein BD, Lowndes HE. Spinal cord defect in the peripheral
Calleman CJ. The metabolism and pharmacokinetics of neuropathy resulting from acrylamide. Neurotoxicology
acrylamide: implications for mechanisms of toxicity and human 1979;1:75–87.
risk estimation. Drug Met Rev 1996;28:527–90. Goldstein BD, Lowndes HE. Group Ia primary afferent terminal
Cavanagh JB. The significance of the ‘‘dying-back’’ process in defect in cats with acrylamide neuropathy. Neurotoxicology
experimental and human neurological disease. Int Rev Exp 1981;2:297–312.
Pathol 1964;3:219-s–267. Hashimoto K, Aldridge WN. Biochemical studies of acrylamide, a
Cavanagh JB. The dying back process. Arch Pathol Lab Med neurotoxic agent. Biochem Pharmacol 1970;19:2591–604.
1979;103:659–64. Hinson JA, Roberts DW. Role of covalent and noncovalent
Cavanagh JB. The pathokinetics of acrylamide intoxication: a interactions in cell toxicity: effects on proteins. Annu Rev
reassessment of the problem. Neuropathol Appl Neurobiol Pharmacol Toxicol 1992;32:471–510.
1982a;8:315–36. Kanai Y, Hediger MA. Primary structure and functional
Cavanagh JB. Mechanisms of axon degeneration in three toxic characterization of a high-affinity glutamate transporter. Nature
neuropathies: organophosphorus, acrylamide and hexacarbon 1992;360:467–71.
compared. In: Smith WT, Cavanagh JB, editors. Recent Kemp DS, Vellaccio F. Organic chemistry. New York: Worth
advances in neuropathology. New York: Churchill Livingstone; Publishers; 1980. p. 839–74.
1982b. p. 213–42. Kemplay S, Cavanagh JB. Effects of acrylamide and other
Cavins JF, Friedman M. Specific modification of protein sulfhydryl compound in vivo and in vitro on staining of motor
sulfhydryl groups with a,b-unsaturated compounds. J Biol nerve terminals by the zinc iodide-osmium technique. Muscle
Chem 1968;243:3357–60. Nerve 1984a;7:94–100.
Crofton KM, Padilla S, Tilson HA, Anthony DC, Raymer JH, Kemplay S, Cavanagh JB. Effects of acrylamide and some other
MacPhail RC. The impact of dose rate on the neurotoxicity of sulfhydryl reagents on spontaneous and pathologically induced
acrylamide: the interaction of administered dose, target tissue terminal sprouting from motor end-plates. Muscle Nerve 1984b;
concentrations, tissue damage, and functional effects. Toxicol 7:101–9.
Appl Pharmacol 1996;139:163–76. Kemplay S, Martin P, Wilson S. The effects of thioctic acid on
DeGrandchamp RL, Lowndes HE. Early degeneration and motor nerve terminals in acrylamide-poisoned rats. Neuropathol
sprouting at the rat neuromuscular junction following Appl Neurobiol 1988;14:275–88.
acrylamide administration. Neuropathol Appl Neurobiol 1990; Kuperman AS. Effects of acrylamide on the central nervous
16:239–54. system of the cat. J Pharmacol Exp Ther 1958;123:180–92.
DeGrandchamp RL, Reuhl KR, Lowndes HE. Synaptic terminal Lehning EJ, Persaud A, Dyer KR, Jortner BS, LoPachin RM.
degeneration and remodeling at the rat neuromuscular junction Biochemical and morphologic characterization of acrylamide
resulting from a single exposure to acrylamide. Toxicol Appl peripheral neuropathy. Toxicol Appl Pharmacol 1998;151:
Pharmacol 1990;105:422–33. 211–21.
362 R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363

Lehning EJ, Balaban CD, Ross JF, Reid ML, LoPachin RM. dynein and kinesin to translocate microtubules in vitro. Toxicol
Acrylamide neuropathy. I. Spatiotemporal characteristics of Appl Pharmacol 1995;133:73–81.
nerve cell damage in rat cerebellum. Neurotoxicology 2002a; Meffert MK, Calakos NC, Scheller RH, Schulamn H. Nitric oxide
23:397–414. modulates synaptic vesicle docking/fusion reactions. Neuron
Lehning EJ, Balaban CD, Ross JF, LoPachin RM. Acrylamide 1996;16:1229–36.
neuropathy. II. Spatiotemporal characteristics of nerve cell Miller MS, Spencer PS. The mechanism of acrylamide
damage in rat brainstem and spinal cord. Neurotoxicology axonopathy. Ann Rev Pharmacol Toxicol 1985;25:643–66.
2002b;23:415–29. Nedvetsky PI, Konev SV, Rakovich AA, Petrenko SV, Mongin
Lehning EJ, Balaban CD, Ross JF, LoPachin RM. Acrylamide AA. Effects of nitric oxide donors on Ca2þ-dependent
neuropathy. III. Spatiotemporal characteristics of nerve cell [14 C]GABA release from brain synaptosomes: the role of –SH
damage in rat forebrain. Neurotoxicology 2003;24:125–36. groups. Biochemistry 2000;65:1027–35.
LeQuesne PM. Acrylamide. In: Spencer PS, Schaumburg HH, Pan Z-H, Bahring R, Grantyn R, Lipton SA. Differential
editors, Experimental and clinical neurotoxicology. 1st ed., modulation by sulfhydryl redox agents and glutathione of
Baltimore (MD): Williams & Wilkins; 1980. p. 309–25. GABA- and glycine-evoked currents in rat retinal ganglion
LeQuesne PM. Clinical and morphological findings in acrylamide cells. J Neurosci 1995;15:1384–91.
toxicity. Neurotoxicology 1985;6:17–24. Pan Z-H, Segal MM, Lipton SA. Nitric oxide-related species
Li J-Y, Jahn R, Dahlstrom A. Rab3a, a small GTP-binding protein, inhibit evoked neurotransmission but enhance spontaneous
undergoes fast anterograde transport but not retrograde miniature synaptic currents in central neuronal cultures. Proc
transport in neurons. Eur J Cell Biol 1995;67:137–47. Natl Acad Sci USA 1996;93:15423–8.
Li J-Y, Edelmann L, Jahn R, Dahlstrom A. Axonal transport and Prineas J. The pathogenesis of dying-back polyneuropathies. Part
distribution of synaptobrevin I and II in the rat peripheral II. An ultrastructural study of experimental acrylamide
nervous system. J Neurosci 1996;16:297–307. intoxication in the cat. J Neuropathol Exp Neurol 1969;28:
Lin RC, Scheller RH. Mechanisms of synaptic vesicle exocytosis. 598–621.
Annu Rev Cell Dev Biol 2000;16:19–49. Robinson MB, Hunter-Ensor M, Sinor J. Pharmacologically
Lonart G, Sudhof TC. Assembly of SNARE core complexes prior distinct sodium-dependent L-[3 H]glutamate transport processes
to neurotransmitter release sets the readily releasable pool of in rat brain. Brain Res 1991;544:196–202.
synaptic vesicles. J Biol Chem 2000;275:27703–7. Sabri MI, Spencer PS. Toxic distal axonopathy: biochemical studies
Lowndes HE, Baker T, Michelson LP, Vincent-Ablazey M. and hypothetical mechanisms. In: Spencer PS, Schaumburg HH,
Attenuated dynamic responses of primary endings of muscle editors. Experimental and clinical neurotoxicology. Baltimore:
spindles: a basis for depressed tendon responses in acrylamide Williams & Wilkins; 1980. p. 206–19.
neuropathy. Ann Neurol 1978a;3:433–7. Schaumburg HH, Wisniewski HM, Spencer PS. Ultrastructural
Lowndes HE, Baker T, Cho E-S, Jortner BS. Position sensitivity of studies of the dying-back process. I. Peripheral nerve terminal
de-efferented muscle spindles in experimental acrylamide and axon degeneration in systemic acrylamide intoxication. J
neuropathy. J Pharmacol Exp Ther 1978;205:40–8. Neuropathol Exp Neurol 1974;33:260–84.
LoPachin RM. The role of fast axonal transport in acrylamide Sega GA, Valdivia Alcota RP, Tancongco CP, Brimer PA.
pathophysiology: mechanism or epiphenomenon? Neurotox- Acrylamide binding to the DNA and protamine of spermiogenic
icology 2002;23:253–7. stages in the mouse and its relationship to genetic damage.
LoPachin RM, Lehning EJ. Acrylamide-induced distal axon Mutat Res 1989;216:221–30.
degeneration: a proposed mechanism of action. Neurotoxicology Sequeira SM, Ambrosio AF, Malva JO, Carvalho AP, Carvalho
1994;15:247–60. CM. Modulation of glutamate release from rat hippocampal
LoPachin RM, Lehning EJ. The relevance of axonal swellings and synaptosomes by nitric oxide. Nitric Oxide: Biol Chem 1997;
atrophy to g-diketone neurotoxicity: a forum position paper. 1:315–29.
Neurotoxicology 1997;18:7–22. Shiff G, Morel N. Rapid anterograde axonal transport of the
LoPachin RM, Lehning EJ, Opanashuk LA, Jortner BS. Rate syntaxin-SNAP 25-VAMP complex. J Neurochem 1997;68:
of neurotoxicant exposure determines morphologic mani- 1663–7.
festations of distal axonopathy. Toxicol Appl Pharmacol 2000; Sickles DW, Brady ST, Testino A, Friedman MA, Wrenn RW.
167:75–86. Direct effect of the neurotoxicant acrylamide on kinesin-based
LoPachin RM, Ross JF, Lehning EJ. Nerve terminals as the primary microtubule motility. J Neurosci Res 1996;46:7–17.
site of acrylamide action: a hypothesis. Neurotoxicology 2002a; Sickles DW, Stone D, Friedman MA. Fast axonal transport: a
23:43–60. site of acrylamide neurotoxicity? Neurotoxicology 2002;23:
LoPachin RM, Ross JF, Reid ML, Dasgupta S, Mansukhani S, 223–51.
Lehning EJ. Neurological evaluation of toxic axonopathies in Skene JHP, Willard M. Axonally transported proteins associated
rats: acrylamide and 2-5-hexanedione. Neurotoxicology with axon growth in rabbit central and peripheral nervous
2002;23:95–110. systems. J Cell Biol 1981;89:96–103.
LoPachin RM, Balaban CD, Ross JF. Acrylamide axonopathy Sollner T, Bennett MK, Whiteheart SW, Scheller RH, Rothman
revisited. Toxicol Appl Pharmacol 2003;188:135–53. JE. A protein assembly–disassembly pathway in vitro that may
Martenson CH, Sheetz M, Graham DG. In vitro acrylamide correspond to sequential steps of synaptic vesicle docking,
exposure alters growth cone morphology. Toxicol Appl activation and fusion. Cell 1993;75:409–18.
Pharmacol 1995a;131:119–29. Spencer PS, Schaumburg HH. A review of acrylamide
Martenson CH, Odom A, Sheetz MP, Graham DG. The effect of neurotoxicity. Part I. Properties, uses and human exposure. Can
acrylamide and other sulfhydryl alkylators on the ability of J Neurol Sci 1974a;1:151–69.
R.M. LoPachin et al. / NeuroToxicology 25 (2004) 349–363 363

Spencer PS, Schaumburg HH. A review of acrylamide Tilson HA. The neurotoxicity of acrylamide: an overview.
neurotoxicity. Part II. Experimental animal neurotoxicity and Neurobehav Toxicol Teratol 1981;3:445–61.
pathologic mechanisms. Can J Neurol Sci 1974b;1:170–92. Tolar LA, Pallanck L. NSF function in neurotransmitter
Spencer PS, Schaumburg HH. Central–peripheral distal release involves rearrangement of the SNARE complex
axonopathy—the pathology of dying-back polyneuropathies. downstream of synaptic vesicle docking. J Neurosci 1998;18:
In: Zimmerman H, editors. Progress in neuropathology. 3rd ed. 10250–6.
New York: Grune & Stratton; 1976. p. 253–76. Trotti D, Rizzini BL, Rossi D, Haugeto O, Racagni G, Danbolt
Spencer PS, Schaumburg HH. Ultrastructural studies of the dying- NC, Volterra A. Neuronal and glial glutamate transporters
back process. III. The evolution of experimental peripheral giant possess a SH-based redox regulatory mechanism. Eur J
axonal degeneration. J Neuropathol Exp Neurol 1977a;36: Neurosci 1997a;9:1236–43.
276–99. Trotti D, Nussberger S, Volterra A, Hediger MA. Differential
Spencer PS, Schaumburg HH. Ultrastructural studies of the dying- modulation of the uptake currents by redox interconversion of
back process. IV. Differential vulnerability of PNS and CNS cysteine residues in the human neuronal glutamate transporter
fibers in experimental central–peripheral distal axonopathy. J EAAACI. Eur J Neurosci 1997b;9:2207–12.
Neuropathol Exp Neurol 1977b;36:300–20. Tsujihata M, Engel AG, Lambert EH. Motor end-plate fine
Spencer PS, Schaumburg HH. Classification of neurotoxic disease: structure in acrylamide dying-back neuropathy: a sequential
a morphological approach. In: Spencer PS, Schaumburg HH, morphometric study. Neurology 1974;24:849–56.
editors. Experimental and clinical neurotoxicology. Baltimore: Uphouse L, Russell M. Rapid effects of acrylamide on
Williams & Wilkins; 1980. p. 92–9. spiroperidol and serotonin binding in neural tissue. Neurobehav
Stamler JS. Redox signaling: nitrosylation and related target Toxicol Teratol 1981;3:281–4.
interactions of nitric oxide. Cell 1994;78:931–6. U.S. Environmental Protection Agency, 1988. Preliminary
Stone JD, Peterson AP, Eyer J, Sickles DW. Neurofilaments are non- assessment of health risks from exposure to acrylamide.
essential elements of toxicant-induced reductions in fast axonal Washington (DC): Office of Toxic Substances, US
transport: pulse labeling in CNS neurons. Neurotoxicology Environmental Protection Agency.
2000;21:447–58. Washbourne P, Cansino V, Mathews JR, Graham M, Burgoyne
Storck T, Schulte S, Hofmann K, Stoffel W. Structure expression RD, Wilson MC. Cysteine residues of SNAP-25 are required for
and functional analysis of a Naþ-dependent glutamate/aspartate SNARE disassembly and exocytosis, but not for membrane
transporter from rat brain. Proc Natl Acad Sci USA targeting. Biochem J 2001;357:625–34.
1992;89:10955–9. Zhao C, Tanaka Y, Setou M, Nakagawa T, Takeda S, Yang HW,
Sullivan KF. Structure and utilization of tubulin isotypes. Annu Terada S, Nakata T, Takei Y, Saito M, Tsuji S, Hayashi Y,
Rev Cell Biol 1988;4:687–716. Hirokawa N. Charcot-Marie-Tooth disease type 2A caused
Tareke E, Rydberg P, Karlsson P, Eriksson S, Tornqvist M. by a mutation in a microtubule motor KIPIBb. Cell 2001;
Acrylamide: a cooking carcinogen? Chem Res Toxicol 2000;13: 105:587–97.
517–22.

You might also like