You are on page 1of 11

Toxicology 234 (2007) 216–226

Low concentrations of ketamine initiate dendritic atrophy of


differentiated GABAergic neurons in culture
Laszlo Vutskits a,∗ , Eduardo Gascon b , Gael Potter b ,
Edomer Tassonyi c , Jozsef Z. Kiss b
aDepartment of Anesthesiology, Pharmacology and Intensive Care, University Hospital of Geneva,
24, rue Micheli-du-Crest, 1211 Geneva 14, Switzerland
b Department of Neuroscience, University of Geneva Medical School, Geneva, Switzerland
c Department of Anesthesiology, Pharmacology and Intensive Care, University Hospital of Geneva, Geneva, Switzerland
Received 15 February 2007; received in revised form 28 February 2007; accepted 1 March 2007
Available online 12 March 2007

Abstract
Administration of subanesthetic concentrations of ketamine, a noncompetitive antagonist of the N-methyl-d-aspartate (NMDA)
type of glutamate receptors, is a widely accepted therapeutic modality in perioperative and chronic pain management. Although
extensive clinical use has demonstrated its safety, recent human histopathological observations as well as laboratory data suggest
that ketamine can exert adverse effects on central nervous system neurons. To further investigate this issue, the present study was
designed to evaluate the effects of ketamine on the survival and dendritic arbor architecture of differentiated ␥-aminobutyric acidergic
(GABAergic) interneurons in vitro. We show that short-term exposure of cultures to ketamine at concentrations of ≥20 ␮g/ml leads
to a significant cell loss of differentiated cells and that non-cell death-inducing concentrations of ketamine (10 ␮g/ml) can still initiate
long-term alterations of dendritic arbor in differentiated neurons, including dendritic retraction and branching point elimination.
Most importantly, we also demonstrate that chronic (>24 h) administration of ketamine at concentrations as low as 0.01 ␮g/ml
can interfere with the maintenance of dendritic arbor architecture. These results raise the possibility that chronic exposure to low,
subanesthetic concentrations of ketamine, while not affecting cell survival, could still impair neuronal morphology and thus might
lead to dysfunctions of neural networks.
© 2007 Elsevier Ireland Ltd. All rights reserved.

Keywords: Dendrite; GABA; Ketamine; Pain; Neurotoxicity

1. Introduction venous anaesthetic alternative to phencyclidine (Reich


and Silvay, 1989), the important psychomimetic side
Ketamine is a phencyclidine derivative acting effects observed following exposure to anaesthetic con-
primarily as a noncompetitive antagonist of N-methyl-d- centrations of ketamine (Freuchen et al., 1976) has
aspartate (NMDA) excitatory glutamate receptors (Oye been importantly curtailed its use as an anesthetics
et al., 1992; Orser et al., 1997). While this drug was in current anaesthesia practice. Since the recogni-
first synthesized in an attempt to provide a safer intra- tion of the important role of NMDA receptors in
nociceptive neurotransmission (Petrenko et al., 2003),
renewed interest, however, has focused on the role
∗ Corresponding author. Tel.: +41 22 3723311; fax: +41 22 3727511. of ketamine in pain management (Himmelseher and
E-mail address: laszlo.vutskits@hcuge.ch (L. Vutskits). Durieux, 2005). In fact, at subanesthetic doses, ketamine

0300-483X/$ – see front matter © 2007 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.tox.2007.03.004
L. Vutskits et al. / Toxicology 234 (2007) 216–226 217

has been shown to reduce pain perception without ate this possibility using our previously described cell
provoking deranging psychomimetic reactions (Oye et culture model of subventricular zone-derived GABAer-
al., 1992; Arendt-Nielsen et al., 1996) and adminis- gic interneurons (Gascon et al., 2005; Vutskits et al.,
tration of low concentrations of ketamine has now 2005,2006).
gained widespread clinical use in perioperative pain
management (Bell et al., 2006). Importantly, long- 2. Methods
term administration of subanesthetic doses of ketamine
has been shown to be effective in the pharmaco- 2.1. Cell cultures and reagents
logical treatment of complex regional pain syndrome
(Rowbotham, 2006), intractable cancer pain (Lossignol After obtaining approval from the Animal Care Committee
of the University Medical Center, cell cultures were prepared
et al., 2005) and central post-stroke pain (Vick and
from newborn (postnatal day 0) Sprague-Dawley rats. Animals
Lamer, 2001).
were sacrificed by decapitation and the brain was carefully
Recent neuropathological findings following contin- removed and transferred into an ice-cold Hank’s magnesium-
uous intrathecal administration of ketamine to relieve and calcium-free solution. Two coronal cuts were then made to
neuropathic cancer pain demonstrate severe histologi- expose the anterior horn of the lateral ventricles and the sub-
cal abnormalities in the spinal cord, including neuronal ventricular zone was microdissected. The small tissue pieces
shrinkage and death as well as reactive gliosis (Vranken obtained were dissociated mechanically and digested with
et al., 2005). In line with these human observations, trypsin (Invitrogen, Life technologies, Paisley, UK) for 15 min
repeated exposure to phencyclidine, ketamine or to at 37 ◦ C. The trypsin reaction was stopped with 1 ml of cold
the noncompetitive NMDA antagonist MK801 led to fetal calf serum and cells recovered after 10 min centrifu-
quite similar histopathological alterations in the cere- gation at 300 × g. To eliminate cell debris, the pellet was
resuspended into 1 ml of PBS and layered onto a 22% Per-
bral cortex of adult rats (Olney et al., 1989), raising
coll gradient (Amershman Pharmacia, Little Chalfont, UK)
concern about the potential neurotoxic effects of these
in phosphate-based saline (PBS) and centrifuged 10 min at
drugs. Indeed, although highly debated (Anand and 500 × g. Cells were washed three times with culture medium
Soriano, 2004; Olney et al., 2004), extensive experi- before plating onto poly-ornithin (Sigma, Saint Louis, MO,
mental evidence indicate that even short-term exposure USA) coated coverslips in 35 mm Petri dishes (Falcon, Ply-
to ketamine and other NMDA antagonists can induce mouth, UK). Seeding density was 5000 cells/cm2 . Cells were
widespread apoptosis in the developing central nervous cultured in Neurobasal medium (Invitrogen, Life technologies,
system (CNS) during the brain growth spurt period Paisley, UK) supplemented with 2% B27 (Invitrogen, Life tech-
(Ikonomidou et al., 1999; Young et al., 2005). While nologies, Paisley, UK), 200 mM l-Glutamine (Invitrogen, Life
the extent of acute ketamine administration-induced cell technologies, Paisley, UK) and 1 mM Na pyruvate (Sigma,
death is significantly less in the adult CNS (Ikonomidou Saint Louis, MO, USA). Under these conditions, cells read-
ily survived and developed as GABAergic neurons for up to 12
et al., 1999; Olney, 2002), little is known about how
days in culture.
chronic exposure to subanesthetic concentrations of
To test the effects of ketamine on neuronal survival and den-
this agent can affect the adult brain. In this con- dritic architecture, cultures were exposed to ketamine (Ketalar,
text, it is important to note that neuronal apoptosis is Parke-Davis, Berlin, Germany) and the NMDA receptor antag-
not the only parameter to be considered in evaluat- onist MK 801 [C16 H15 N·C4 H4 O4 ] (100 ␮M; Tocris, Bristol,
ing potential adverse effects of drug exposure on the UK). The amount and the duration of ketamine treatment are
nervous system. It is now well established that inter- indicated in detail in each experiment in the Section 3. To
ference with the finely tuned molecular mechanisms remove drugs from the culture medium, cultures (control and
orchestrating the maintenance of dendritic arbors in dif- treated) were washed 3 times with warm (37 ◦ C) Neurobasal
ferentiated neurons can induce important reorganization medium and then continued to be cultured in the presence of
of synaptic connections, leading to persistent changes in Neurobasal medium before analysis according to experimental
protocols.
behaviour and psychological functions (Robinson and
Kolb, 2004).
2.2. Immunocytochemistry
In our previous study, we demonstrated that ketamine
can interfere with dendritic arbor development of imma-
Cells were fixed with cold (4 ◦ C) paraformaldehyde 4%
ture GABAergic neurons (Vutskits et al., 2006). It is, in phosphate buffer (pH 7.4). Then, they were rinsed 3 times
however, currently unknown whether chronic exposure in PBS and incubated overnight at 4 ◦ C with the primary anti-
to low concentrations of ketamine can also alter the main- body diluted in PBS containing bovine serum albumine (0.5%)
tenance of dendritic arbor architecture in differentiated and Triton X-100 (0.3%). The mouse monoclonal Ab directed
neurons. The aim of the present study was to evalu- against ␤-tubulin isotype III (Sigma, Saint Louis, MO, USA
218 L. Vutskits et al. / Toxicology 234 (2007) 216–226

1:400 dilution) was used to identify neurons. Bound Abs were of arborisation pattern: while dendrites still continue to
revealed with rhodamine- or fluorescein-conjugated sheep anti- discretely sprout on poly-ornithine-covered coverslips,
mouse IgG (Boehringer, Mannheim, Germany; dilution 1:40 no additional increase in the number of primary dendrites
for rhodamine and 1:80 for fluorescein) diluted in PBS con- (PD) and branching points (BP) can be observed by time
taining 0.5% BSA. (Fig. 1E and F). These findings are consistent with our
recent results, indicating functional maturation of these
2.3. Cell counts, statistical analysis, image acquisition cells by the 6th DIV that corresponds to the functional
and processing
shift from the depolarizing to the hyperpolarizing action
Immunostained cultures were examined with a Nikon
of the neurotransmitter GABA on these cells (Gascon et
Eclipse TE2000-U microscope (Nikon Corp, Tokyo, Japan). al., 2006).
For cell counts, a 10× Nikon objective and a digital cam-
era (Retiga EX, Qimaging, Burnaby, Canada) controlled by 3.2. Effect of short-term ketamine exposure on the
the Openlab software (Version 3.1.2. Improvision, Coven- survival of SVZ-derived differentiated GABAergic
try, England) was used. On each coverslip, 30 samples were neurons
randomly taken and then samples were pooled (i.e., total sur-
face measured per coverslip was approximately 5 mm2 ). Cell In our recent study, we showed that short-term expo-
counts were made using the Image J (NIH) software (data are sure of immature SVZ-derived neurons to ketamine
expressed as the number of neurons per mm2 ± standard error induces apoptosis in a dose-dependent manner (Vutskits
of mean, and reflect results obtained form at least 3 independent
et al., 2006). As the effect of ketamine on neuronal sur-
experiments).
For quantitative analysis of dendritic arbours, ␤-tubulin iso-
vival has been reported to be age dependent (Ikonomidou
type III positive cells were photographed using a 40× Nikon et al., 1999), we first assessed whether short-term expo-
objective linked through the digital camera to the Openlab soft- sure to ketamine can induce cell loss in 6 DIV cultures
ware. Before analysis, brightness and contrast were optimized containing differentiated neurons and, if yes, whether
with Adobe Photoshop program (Adobe Systems Incorpo- cell-death-inducing concentrations of ketamine in these
rated, San Jose, California, USA). The following parameters cells are different from those observed in immature
of dendritic shape and extent were then determined: number of neurons (Vutskits et al., 2006). To this end, we first
primary dendrites, length of dendrites and the number of den- exposed 6 DIV cultures to ketamine for 1 h in concen-
dritic branches. Total dendritic length was measured drawing trations ranging from 1 to 100 ␮g/ml and cell survival
all visible processes with Image J. The remaining parameters was evaluated up to 72 h following drug exposure (for
were manually scored on the image. Processes shorter than
experimental setup see Fig. 2A). We found no cell
5 ␮m were excluded from the analysis.
Through the whole study, values were expressed as
loss at any of these concentrations when cell survival
means ± standard error of mean and analyzed for statistical was assessed immediately (time 0) after this treatment
significance. Differences between groups were first discrim- (Fig. 2B). In contrast to immature neurons, where a
inated by one-way ANOVA and then the unpaired t-test was 1-hour-long exposure to ketamine induced cell death
performed, where t was corrected for multiple comparisons at concentrations of 10 ␮g/ml as early as 24 h follow-
against the untreated group using the Bonferroni test. *P < 0.05 ing treatment (Vutskits et al., 2006), this experimental
compared with the untreated control group. paradigm did not influence survival of 6 DIV differ-
entiated neurons at any of the time points examined
3. Results (Fig. 2B). We found, however, a significant cell loss of
differentiated neurons 24 h after ketamine treatment at
3.1. Subventricular zone-derived neurons display a concentrations ≥ 40 ␮g/ml and also at later time points
differentiated phenotype after 6 days in culture using concentrations ≥ 20 ␮g/ml (Fig. 2B). An 8-hour-
long-exposure to ketamine (for experimental setup see
We have previously shown that subventricular Fig. 2A) did not induce apparent cell loss following
zone-derived neuronal precursors differentiate into the end of this treatment (time 0) up to drug concen-
GABAergic neurons under serum-free conditions in vitro trations of 20 ␮g/ml (Fig. 2C). However, a significant
(Gascon et al., 2005). These neurons rapidly elaborate (P < 0.05) cell loss was detected from the 1st day
their dendritic arbor during the first few days in culture post-exposure in cultures receiving ketamine at con-
(Fig. 1), thus providing a useful model system to study centrations of ≥ 10 ␮g/ml (Fig. 2C). Since one major
the effects of anesthetics on dendritic development and action of ketamine is the blockade of the NMDA-type
survival of these cells (Vutskits et al., 2005,2006). By the glutamate receptors, we also examined cell survival in
6th day in vitro (DIV), cells reach a plateau phase in terms the presence of the noncompetitive NMDA receptor
L. Vutskits et al. / Toxicology 234 (2007) 216–226 219

Fig. 1. Dendritic arbor architecture development of subventricular zone-derived GABAergic neurons. (A) 24 h after seeding, isolated neuroblasts
exhibit an immature morphology under serum-free conditions. (B) Representative photomicrograph of a differentiating neuron by the end of the
3rd day in culture. (C) Neurons develop a complex dendritic architecture by the 6th day in vitro. (D) While dendrites still continue to discretely
sprout on poly-ornithine-coated coverslips, no further increase in dendritic complexity can be observed at later time points (9th day in culture).
(E–F) Quantitative assessment of dendritic arbor development in terms of the number of primary dendrites and branching points, respectively. In
photomicrographs (A–D), cells were stained with the neuron specific marker tubulin-␤-III antibody. Correction bar (A–D): 25 ␮m. In figures E–F,
values on the Y-axis are expressed as the number of primary dendrites and branching points per cell. Results are presented as mean ± S.E.M.; n = 3
independent experiments for each time point expressed.

blocker MK801 (100 ␮M). The presence of MK801 up 3.3. Dose-dependent effect of short-term ketamine
to 8 h in the culture medium did not induce cell death exposure on dendritic arbor architecture of
(Fig. 2B and C). Altogether, these results indicate that differentiated GABAergic neurons
sensitivity to ketamine-induced cytotoxicity decreases
along with neuronal maturation and that these effects We next examined whether short-term exposure to
appear to be independent of the blockade of NMDA non-cell-death-inducing concentrations of ketamine can
receptor. still alter dendritic tree architecture in differentiated
220 L. Vutskits et al. / Toxicology 234 (2007) 216–226

total dendritic length (TDL); (2) the number of primary


dendrites (PD), defined as those arising from the cell
body; and (3) the number of branching points (BP). As
seen in Fig. 3, ketamine treatment up to 8 h, at concentra-
tions ≤ 5 ␮g/ml, did not affect any aspects of dendritic
architecture. In contrast, even a 1-hour-long exposure to
this drug at concentrations of 10 ␮g/ml induced a signif-
icant remodelling of dendritic arbor, including retraction
of dendrites as well as elimination of branching points.
These effects could be observed as early as 24 h follow-
ing drug exposure (not shown) and became significant
(P < 0.05) by the 3rd day post exposure (Fig. 3 H). An 8-
hour-long exposure to 10 ␮g/ml of ketamine had an even
more robust effect (Fig. 3I). In contrast, the presence of
MK801 up to 8 h in the culture medium did not influ-
ence dendritic morphology (Fig. 3H and I). These results
thus demonstrate that even a single short episode of
ketamine treatment at non-cell-death-inducing concen-
trations can lead to persistent changes of dendritic arbor
architecture.

3.4. Effect of chronic ketamine exposure on the


survival of differentiated GABAergic neurons

Long-term administration of low, subanesthetic doses


of ketamine has become an accepted therapeutical
Fig. 2. Effect of short-term ketamine treatment on the survival of modality in postoperative sedation and pain control as
subventricular zone-derived differentiated neurons. (A) Experimental
protocol. (B) Quantitative analysis of cell survival following a 1-
well as in chronic pain treatment (Himmelseher and
hour-long treatment to ketamine (5–40 ␮g/ml) and MK801 (100 ␮M). Durieux, 2005; Rowbotham, 2006). To study the effects
(C) Quantitative analysis of cell survival following an 8-hour-long of long-term exposure of differentiated neurons to sub-
treatment to ketamine (2–20 ␮g/ml) and MK801 (100 ␮M). In fig- anesthetic concentrations of ketamine, 6 DIV cultures
ures (B–C), results are presented as mean ± S.E.M.; n = 3 independent were exposed to the continuous presence of this drug
experiments for each time point and each treatment expressed. Values
are expressed as the number of neurons/mm2 . *P < 0.05 compared with
(0.01–5 ␮g/ml) for 72 h and cell loss was assessed in
the untreated control group. corresponding sister cultures every 24 h (for experimen-
tal protocol see Fig. 4A). Using this protocol, we found a
GABAergic neurons. Using the abovementioned exper- significant (P < 0.05) decrease in cell number after 48 h
imental paradigms (Fig. 3A), cultures were exposed to at concentrations ≥ 2 ␮g/ml and cell loss became also
ketamine at concentrations ranging from 1 to 10 ␮g/ml significant by the end of the 3rd day of continuous treat-
and the impact of this treatment on neuronal dendritic ment at concentrations of ≥ 1 ␮g/ml. Similarly, chronic
architecture, was measured using three parameters: (1) application of MK 801 (100 ␮M) led to a significant

Fig. 3. Effects of short-term ketamine treatment on dendritic arbor architecture of subventricular zone-derived differentiated neurons. (A) Exper-
imental protocol. (B) Representative epifluorescent micrograph of a 6-day-old neuron, showing that no effect on dendritic architecture was found
at any doses of ketamine applied (up to 40 ␮g/ml) when morphology was immediately (time 0) assessed following drug exposure. (C) In control,
placebo treated cultures, neuronal arbor architecture remained stable up to 72 h following treatment. Similarly, (D) neither exposure to ketamine
at concentrations of ≤ 5 ␮g/ml nor (E) treatment with MK801 (100 ␮M) up to 8 h had any long-term effect on neuronal morphology. In contrast,
(F) as short as a 1-hour-long treatment with ketamine at concentrations of 10 ␮g/ml induced dendritic pruning of differentiated neurons and (G)
these effects were even more robust when the length of the treatment period reached 8 h. (H–I) Quantitative assessment of dendritic morphology
72 h after a (H) 1-hour-long and an (I) 8-hour-long ketamine (1–20 ␮g/ml) or MK801 (100 ␮M) treatment. In photomicrographs (A–D), cells were
stained with the neuron specific marker tubulin-␤-III antibody. Correction bar (A–D): 20 ␮m. In figures (H–I), values on the Y-axis are expressed as
the number of primary dendrites and branching points per cell. Results are presented as mean ± S.E.M.; n = 3 independent experiments (3 culture
dish per experiment, 30 neurons per culture dish) for each time point and each treatment expressed. *P < 0.05 compared with the untreated control
group.
L. Vutskits et al. / Toxicology 234 (2007) 216–226 221
222 L. Vutskits et al. / Toxicology 234 (2007) 216–226

the end of the 3rd day of drug exposure. Importantly,


following 48 h of treatment, the number of branch points
was also significantly decreased in cultures treated with
ketamine at a concentration of 0.1 ␮g/ml. Finally, elimi-
nation of branch points could also be observed in groups
receiving ketamine at a concentration of 0.01 ␮g/ml,
although it did not reach significance. Taken together,
these data indicate that chronic exposure to low doses
of ketamine, while not affecting cell survival, results
in clear atrophy of dendritic arbors of differentiated
GABAergic neurons.

4. Discussion

The NMDA-type of excitatory glutamate receptors


play an important role in central sensitization following
Fig. 4. Quantitative assessment of cell survival following chronic
painful stimuli (Petrenko et al., 2003) and pharmaco-
application of ketamine (0.01–5 ␮g/ml) and MK801 (100 ␮M). Results
are presented as mean ± S.E.M.; n = 3 independent experiments for logical blockade of these receptors, using subanesthetic
each time point and each treatment expressed. Values are expressed as concentrations of ketamine, has been show to be an
the number of neurons/mm2 . *P < 0.05 compared with the untreated effective treatment modality in both perioperative and
control group. chronic pain management (Himmelseher and Durieux,
2005; Rowbotham, 2006). Based on previous laboratory
decrease in the number of neurons in culture, suggesting and human histopathological observations, suggesting
that NMDA-dependent mechanisms are involved in the that ketamine can exert potential adverse effects on CNS
survival of GABAergic neurons (Fig. 4B). It is impor- neurons (Olney et al., 1989; Vranken et al., 2005), the
tant to note that, while application of ketamine 1 ␮g/ml present study was designed to evaluate the dose- and
as well as MK801 induced death of both immature and exposure-time dependent effects of ketamine on differ-
differentiated neurons, the extent of cell death was more entiated GABAergic neurons. We show that short-term
important in immature cells (52 ± 6% in immature ver- exposure to ketamine induces cell death and dendritic
sus 19 ± 4% in differentiated neurons), further arguing arbor retraction of surviving neurons only when this drug
for the differential sensitivity of young versus adult-like is administered at relatively high doses (≥10 ␮g/ml).
neurons to ketamine. Most importantly, we also demonstrate that chronic
administration of ketamine at concentrations as low as
3.5. Effect of chronic (>24 h) ketamine exposure on 0.01 ␮g/ml can induce an important atrophy of neuronal
dendritic arbor architecture of differentiated dendritic architecture. These results suggest that long-
GABAergic neurons term administration of subanesthetic concentrations of
ketamine, while not affecting survival, may still impair
Studying the effects of long-term ketamine treatment neuronal network functions by interfering with funda-
as well as the impact of NMDA receptor blockade on mental mechanisms governing maintenance of dendritic
dendritic architecture of mature GABAergic neurons arbor in differentiated neurons.
(for experimental protocol see Fig. 4A), we found that In vitro models provide a useful complementary
dendritic morphology was severely impaired following approach to in vivo animal experiments in the evaluation
chronic exposure even to low concentrations of this drug of potential drug-induced adverse effects on an organism
(Fig. 5). During the first 24 h in the presence of ketamine (Spielmann and Liebsch, 2001). In the present study, we
(0.01–1 ␮g/ml) or MK801 (100 ␮M), no significant dif- used our previously validated in vitro model, that allowed
ference could be detected between control and treated us to study the effects of anesthetics on the survival and
cultures in any aspects of dendritic morphology analyzed dendritic arborization of developing GABAergic neu-
(Fig. 5). In contrast, by the end of 2nd day, 1 ␮g/ml rons in culture (Gascon et al., 2005; Vutskits et al.,
of ketamine as well as MK801 induced a significant 2005,2006). Here we show that our model is also a
decrease in total dendritic length and the number of valuable tool to assess the potentially toxic effects of
branch points compared to the beginning of treatment these agents on more differentiated cells, presenting
(time 0) and these perturbations further accentuated by functional characteristics of mature neurons. In these
L. Vutskits et al. / Toxicology 234 (2007) 216–226 223

Fig. 5. Effect of long-term ketamine (0.01–1 ␮g/ml) and MK 801 (100 ␮M) treatment on dendritic architecture of differentiated GABAergic
neurons. 6 days after seeding (time 0), ketamine was applied to the culture medium and left there for up to 72 h. Representative examples of dendritic
arbor expansion in (A) time 0; (B) control; (C) ketamine (0.1 ␮g/ml); (D) ketamine (1 ␮g/ml); and (E) MK801 (100 ␮M)-treated neurons. (F–H)
Quantitative analysis of the effects of continuous exposure to low concentrations of ketamine (0.01–1 ␮g/ml) and MK801 (100 ␮M), on dendritic
arbor as expressed by the total dendritic length (F), the number of primary dendrites (G) and the number of branching points (H). Cells were stained
with the neuron specific marker tubulin-␤-III antibody. Correction bar (A–E): 35 ␮m. In figures F–H, values on the Y-axis are expressed as the
number of primary dendrites and branching points per cell. Results are presented as mean ± S.E.M.; n = 3 independent experiments (3 culture dish
per experiment, 30 neurons per culture dish) for each time point and each treatment expressed. *P < 0.05 compared with the untreated control group.
224 L. Vutskits et al. / Toxicology 234 (2007) 216–226

cultures, we recently demonstrated a GABAA receptor- One of the most intriguing finding of the present
mediated functional shift from the depolarizing toward study was that long-term exposure of differentiated neu-
the hyperpolarizing cellular responses to GABA by the rons to low, non-cell-death-inducing concentrations of
6th DIV and this is correlated with a significant increase ketamine can still induce a significant atrophy of den-
in the expression pattern of K+ /Cl− co-transporter KCC2 drites, including retraction of branches and elimination
(Gascon et al., 2006). Parallel to these functional changes of branching points. Such atrophy and debranching of
in neuronal phenotype, there is an important decrease in dendritic arbors could have important long-term func-
the expression of PSA-NCAM, a cell surface marker of tional consequences, as even minor changes in neuronal
immature neurons, from the end of the 1st week in vitro morphology have been shown to exert a significant
(Gascon et al., 2005). Finally, results presented in this impact on neuronal responses to stimuli (van Ooyen et
study show that differentiating neurons reach steady- al., 2002; Robinson and Kolb, 2004). Little is known
state in terms of dendritic arborization pattern by the about the mechanisms that underlie pruning of dendritic
6th DIV. Altogether, these results thus strongly suggest arbors in the CNS (Williams and Truman, 2005) and,
that neuronal precursors in these preparations undergo to our knowledge, the role of chronic NMDA-mediated
morphofunctional maturation and acquire, by the 6th neurotransmission blockade in this context has not been
DIV, functional characteristics similar to those of mature established. Our data, showing that chronic admin-
neurons. istration of the NMDA receptor antagonist MK801,
Using 6 DIV cultures to evaluate the effect of short- similarly to low doses of ketamine, also results in reduced
term ketamine exposure on cell survival, we demonstrate dendritic arbor suggests that NMDA-dependent mech-
that higher concentrations and exposure times of this anisms are involved in the maintenance of dendritic
drug are needed to induce cell death in differentiated structures.
GABAergic cells, compared to their immature coun- In an attempt to draw parallel between our data and
terparts (Vutskits et al., 2006). These results are in those obtained in previous in vivo animal studies (Olney
agreement with previous in vivo studies (Ikonomidou et al., 1989; Ikonomidou et al., 1999; Scallet et al.,
et al., 1999; Jevtovic-Todorovic et al., 2001), and sug- 2004; Young et al., 2005), an important issue to con-
gest that sensitivity to ketamine-induced cytotoxicity sider is whether ketamine concentrations administered
decreases along with neuronal maturation. Although the in the present study are comparable to those used in ear-
precise molecular mechanisms responsible for the dif- lier in vivo experiments. It has been previously reported
ferential sensitivity of immature versus mature neurons that following a single dose intraperitoneal injection of
to ketamine remains to be determined, one plausi- 20 mg/kg ketamine, known to induce apoptosis in the
ble explanation of these observations could be the cerebral cortex of young mice (Young et al., 2005),
developmental switch in the subunit composition of plasma concentrations of this drug are approximately
NMDA receptors that occurs during neuronal differenti- 5–6 ␮g/ml (Scallet et al., 2004). Importantly, ketamine
ation both in vivo and in vitro (Williams et al., 1993; rapidly penetrates the blood-brain barrier and brain con-
Feldmeyer and Cull-Candy, 1996; Kew et al., 1998). centrations of this drug are up to six times higher than
It is also important to underline that, in contrast to those in the blood (Edwards et al., 2002). It is thus con-
high-dose regimens of ketamine, we did not find any ceivable that the abovementioned treatment paradigm
effect of short-term exposure of cells to the noncom- could lead to a transient accumulation of ketamine in
petitive NMDA receptor antagonist MK801 either on the brain parenchyma at concentrations of ≥ 20 ␮g/ml,
survival or dendritic morphology. These results suggest a scenario quite similar to our “short-term exposure”
that ketamine-induced neurotoxicity following short- protocol where the presence of ketamine for 1-hour in
term exposure to relatively high concentrations of this the culture medium, representing conceptually the brain
drug is, at least partially, independent of NMDA recep- rather than the plasma environment, at concentrations
tor blockade. As ketamine is known to interact with of ≥ 20 ␮g/ml also induced cell loss. To our knowledge,
a multitude of signaling pathways mediating neuro- no in vivo data exists on the effects of chronic low-
transmission in the CNS (Adams, 1998), it is possible dose ketamine exposure on the CNS. It is important to
that, in the presence of important concentrations of emphasize that the present observations were made on
this anesthetics, additive or synergistic effects between isolated neurons without synaptic contacts. It will be,
these signaling mechanisms could rapidly initiate den- therefore, essential to validate these data by determining
dritic remodeling and/or apoptosis. Alternatively, high how neuronal dendritic arbor architecture is influenced
doses of ketamine could exert a nonspecific neurotoxic by ketamine in a more complex and physiological envi-
effect. ronment.
L. Vutskits et al. / Toxicology 234 (2007) 216–226 225

Whereas anaesthesia-inducing plasma concentrations Adler, C.M., Goldberg, T.E., Malhotra, A.K., Pickar, D., Breier, A.,
of ketamine in humans have been reported to be above 1998. Effects of ketamine on thought disorder, working memory,
1 ␮g/ml (Malinovsky et al., 1996; Weber et al., 2004), and semantic memory in healthy volunteers. Biol. Psychiatry 43,
811–816.
adequate pain relief-associated plasma levels of this Anand, K.J., Soriano, S.G., 2004. Anesthetic agents and the immature
anesthetics are several fold lower, situating around brain: are these toxic or therapeutic? Anesthesiology 101, 527–
20 ng/ml (Suzuki et al., 2005). Although it is ethically 530.
impossible to determine blood-brain distribution coef- Arendt-Nielsen, L., Nielsen, J., Petersen-Felix, S., Schnider, T.W.,
ficient of ketamine in humans, based on experimental Zbinden, A.M., 1996. Effect of racemic mixture and the (S+)-
isomer of ketamine on temporal and spatial summation of pain.
studies of ketamine pharmacokinetics (Edwards et al., Br. J. Anaesth. 77, 625–631.
2002), it is thus strongly possible, as discussed above, Bell, R.F., Dahl, J.B., Moore, R.A., Kalso, E., 2006. Perioperative
that plasma concentrations of 20 ng/ml of ketamine result ketamine for acute postoperative pain. Cochrane Database Syst
in several fold higher brain concentrations. In this con- Rev, CD004603.
text, chronic exposure to subanesthetic concentrations of Berde, C., Cairns, B., 2000. Developmental pharmacology across
species: promise and problems. Anesth. Analg. 91, 1–5.
ketamine would potentially result in drug levels between Edwards, S.R., Minto, C.F., Mather, L.E., 2002. Concurrent ketamine
0.01 and 0.1 ␮g/ml in the human brain parenchyma. and alfentanil administration: pharmacokinetic considerations. Br.
This clinical situation could be similar to our “long-term J. Anaesth. 88, 94–100.
treatment” paradigm, where exposure of GABAergic Feldmeyer, D., Cull-Candy, S., 1996. Functional consequences of
neurons to ketamine in this concentration range induced changes in NMDA receptor subunit expression during develop-
ment. J. Neurocytol. 25, 857–867.
dendritic retraction. It is, however, important to note Freuchen, I., Ostergaard, J., Kuhl, J.B., Mikkelsen, B.O., 1976. Reduc-
that extrapolation of these laboratory results to clinical tion of psychotomimetic side effects of Ketalar (ketamine) by
practice requires extreme caution. Indeed, due to inter- Rohypnol (flunitrazepam). A randomized, double-blind trial. Acta
species differences in terms of drug effects (Berde and Anaesthesiol. Scand. 20, 97–103.
Cairns, 2000), animal data provide an imprecise basis Gascon, E., Vutskits, L., Zhang, H., Barral-Moran, M.J., Kiss, P.J.,
Mas, C., Kiss, J.Z., 2005. Sequential activation of p75 and TrkB is
for evaluating human risk. On the other hand, given involved in dendritic development of subventricular zone-derived
that dendrites are important morphofunctional substrates neuronal progenitors in vitro. Eur. J. Neurosci. 21, 69–80.
underlying complex cognitive functions, our results Gascon, E., Dayer, A.G., Sauvain, M.O., Potter, G., Jenny, B., De
demonstrating the effects of ketamine on neuronal den- Roo, M., Zgraggen, E., Demaurex, N., Muller, D., Kiss, J.Z., 2006.
dritic arbour should give further arguments to promote GABA regulates dendritic growth by stabilizing lamellipodia in
newly generated interneurons of the olfactory bulb. J. Neurosci.
clinical research on this topic. While the effect of short- 26, 12956–12966.
term ketamine exposure has been extensively studied in Himmelseher, S., Durieux, M.E., 2005. Ketamine for perioperative
a wide variety of neuropsychological tests (Adler et al., pain management. Anesthesiology 102, 211–220.
1998; Krystal et al., 1994; Rowland et al., 2005), to our Ikonomidou, C., Bosch, F., Miksa, M., Bittigau, P., Vockler, J., Dikra-
knowledge, there are currently no data on detailed neu- nian, K., Tenkova, T.I., Stefovska, V., Turski, L., Olney, J.W., 1999.
Blockade of NMDA receptors and apoptotic neurodegeneration in
ropsychological outcome in patients receiving ketamine the developing brain. Science 283, 70–74.
for extended periods. Results of these future studies Jevtovic-Todorovic, V., Wozniak, D.F., Benshoff, N.D., Olney, J.W.,
would be necessary to initiate discussion about the 2001. A comparative evaluation of the neurotoxic properties of
risk/benefit evaluation of chronic ketamine treatment. ketamine and nitrous oxide. Brain Res. 895, 264–267.
Kew, J.N., Richards, J.G., Mutel, V., Kemp, J.A., 1998. Developmental
changes in NMDA receptor glycine affinity and ifenprodil sen-
Acknowledgements sitivity reveal three distinct populations of NMDA receptors in
individual rat cortical neurons. J. Neurosci. 18, 1935–1943.
This work was supported by Swiss National Foun- Krystal, J.H., Karper, L.P., Seibyl, J.P., Freeman, G.K., Delaney, R.,
dation Grants (Bern, Switzerland) 310000 113555/1 to Bremner, J.D., Heninger, G.R., Bowers Jr., M.B., Charney, D.S.,
1994. Subanesthetic effects of the noncompetitive NMDA antago-
L.V.; 3100A0-104059/1 to J.Z.K. and the Anesthesi- nist, ketamine, in humans. Psychotomimetic, perceptual, cognitive,
ology Department Fund of the University Hospital of and neuroendocrine responses. Arch. Gen. Psychiatry 51, 199–214.
Geneva to L.V. We wish to thank Sylvie Chliate (techni- Lossignol, D.A., Obiols-Portis, M., Body, J.J., 2005. Successful use
cian, Department of Neuroscience, University of Geneva of ketamine for intractable cancer pain. Support. Care Cancer 13,
Medical School, Switzerland) for technical assistance. 188–193.
Malinovsky, J.M., Servin, F., Cozian, A., Lepage, J.Y., Pinaud, M.,
1996. Ketamine and norketamine plasma concentrations after i.v.,
References nasal and rectal administration in children. Br. J. Anaesth. 77,
203–207.
Adams, H.A., 1998. Mechanisms of action of ketamine. Anaesthesiol. Olney, J.W., 2002. New insights and new issues in developmental
Reanim. 23, 60–63. neurotoxicology. Neurotoxicology 23, 659–668.
226 L. Vutskits et al. / Toxicology 234 (2007) 216–226

Olney, J.W., Labruyere, J., Price, M.T., 1989. Pathological changes Suzuki, M., Kinoshita, T., Kikutani, T., Yokoyama, K., Inagi, T.,
induced in cerebrocortical neurons by phencyclidine and related Sugimoto, K., Haraguchi, S., Hisayoshi, T., Shimada, Y., 2005.
drugs. Science 244, 1360–1362. Determining the plasma concentration of ketamine that enhances
Olney, J.W., Young, C., Wozniak, D.F., Ikonomidou, C., Jevtovic- epidural bupivacaine-and-morphine-induced analgesia. Anesth.
Todorovic, V., 2004. Anesthesia-induced developmental neu- Analg. 101, 777–784.
roapoptosis. Does it happen in humans? Anesthesiology 101, van Ooyen, A., Duijnhouwer, J., Remme, M.W., van Pelt, J., 2002. The
273–275. effect of dendritic topology on firing patterns in model neurons.
Orser, B.A., Pennefather, P.S., MacDonald, J.F., 1997. Multiple mech- Network 13, 311–325.
anisms of ketamine blockade of N-methyl-d-aspartate receptors. Vick, P.G., Lamer, T.J., 2001. Treatment of central post-stroke pain
Anesthesiology 86, 903–917. with oral ketamine. Pain 92, 311–313.
Oye, I., Paulsen, O., Maurset, A., 1992. Effects of ketamine on sensory Vranken, J.H., Troost, D., Wegener, J.T., Kruis, M.R., van der
perception: evidence for a role of N-methyl-d-aspartate receptors. Vegt, M.H., 2005. Neuropathological findings after continuous
J. Pharmacol. Exp. Ther. 260, 1209–1213. intrathecal administration of S(+)-ketamine for the management
Petrenko, A.B., Yamakura, T., Baba, H., Shimoji, K., 2003. The role of of neuropathic cancer pain. Pain 117, 231–235.
N-methyl-d-aspartate (NMDA) receptors in pain: a review. Anesth. Vutskits, L., Gascon, E., Tassonyi, E., Kiss, J.Z., 2005. Clinically
Analg. 97, 1108–1116. relevant concentrations of propofol but not midazolam alter in
Reich, D.L., Silvay, G., 1989. Ketamine: an update on the first vitro dendritic development of isolated gamma-aminobutyric acid-
twenty-five years of clinical experience. Can. J. Anaesth. 36, 186– positive interneurons. Anesthesiology 102, 970–976.
197. Vutskits, L., Gascon, E., Tassonyi, E., Kiss, J.Z., 2006. Effect of
Robinson, T.E., Kolb, B., 2004. Structural plasticity associated with ketamine on dendritic arbor development and survival of immature
exposure to drugs of abuse. Neuropharmacology 47 (Suppl 1), GABAergic neurons in vitro. Toxicol. Sci. 91, 540–549.
33–46. Weber, F., Wulf, H., Gruber, M., Biallas, R., 2004. S-ketamine and
Rowbotham, M.C., 2006. Pharmacologic management of complex s-norketamine plasma concentrations after nasal and i.v. adminis-
regional pain syndrome. Clin. J. Pain. 22, 425–429. tration in anesthetized children. Paediatr. Anaesth. 14, 983–988.
Rowland, L.M., Astur, R.S., Jung, R.E., Bustillo, J.R., Lauriello, J., Williams, D.W., Truman, J.W., 2005. Cellular mechanisms of den-
Yeo, R.A., 2005. Selective cognitive impairments associated with drite pruning in Drosophila: insights from in vivo time-lapse of
NMDA receptor blockade in humans. Neuropsychopharmacology remodeling dendritic arborizing sensory neurons. Development
30, 633–639. 132, 3631–3642.
Scallet, A.C., Schmued, L.C., Slikker Jr., W., Grunberg, N., Faustino, Williams, K., Russell, S.L., Shen, Y.M., Molinoff, P.B., 1993. Devel-
P.J., Davis, H., Lester, D., Pine, P.S., Sistare, F., Hanig, J.P., 2004. opmental switch in the expression of NMDA receptors occurs in
Developmental neurotoxicity of ketamine: morphometric confir- vivo and in vitro. Neuron. 10, 267–278.
mation, exposure parameters, and multiple fluorescent labeling of Young, C., Jevtovic-Todorovic, V., Qin, Y.Q., Tenkova, T., Wang,
apoptotic neurons. Toxicol. Sci. 81, 364–370. H., Labruyere, J., Olney, J.W., 2005. Potential of ketamine and
Spielmann, H., Liebsch, M., 2001. Lessons learned from validation midazolam, individually or in combination, to induce apoptotic
of in vitro toxicity test: from failure to acceptance into regulatory neurodegeneration in the infant mouse brain. Br J. Pharmacol. 146,
practice. Toxicol. In Vitro 15, 585–590. 189–197.

You might also like