ArticlePDF Available

Abstract and Figures

The neuroprotective actions of cannabidiol and other cannabinoids were examined in rat cortical neuron cultures exposed to toxic levels of the excitatory neurotransmitter glutamate. Glutamate toxicity was reduced by both cannabidiol, a nonpsychoactive constituent of marijuana, and the psychotropic cannabinoid (-)Delta9-tetrahydrocannabinol (THC). Cannabinoids protected equally well against neurotoxicity mediated by N-methyl-D-aspartate receptors, 2-amino-3-(4-butyl-3-hydroxyisoxazol-5-yl)propionic acid receptors, or kainate receptors. N-methyl-D-aspartate receptor-induced toxicity has been shown to be calcium dependent; this study demonstrates that 2-amino-3-(4-butyl-3-hydroxyisoxazol-5-yl)propionic acid/kainate receptor-type neurotoxicity is also calcium-dependent, partly mediated by voltage sensitive calcium channels. The neuroprotection observed with cannabidiol and THC was unaffected by cannabinoid receptor antagonist, indicating it to be cannabinoid receptor independent. Previous studies have shown that glutamate toxicity may be prevented by antioxidants. Cannabidiol, THC and several synthetic cannabinoids all were demonstrated to be antioxidants by cyclic voltametry. Cannabidiol and THC also were shown to prevent hydroperoxide-induced oxidative damage as well as or better than other antioxidants in a chemical (Fenton reaction) system and neuronal cultures. Cannabidiol was more protective against glutamate neurotoxicity than either ascorbate or alpha-tocopherol, indicating it to be a potent antioxidant. These data also suggest that the naturally occurring, nonpsychotropic cannabinoid, cannabidiol, may be a potentially useful therapeutic agent for the treatment of oxidative neurological disorders such as cerebral ischemia.
No caption available
… 
Content may be subject to copyright.
Proc. Natl. Acad. Sci. USA
Vol. 95, pp. 82688273, July 1998
Medical Sciences
Cannabidiol and (2)D
9
-tetrahydrocannabinol are
neuroprotective antioxidants
A. J. HAMPSON*
,M.GRIMALDI
,J.AXELROD*, AND D. WINK
§
*Laboratory of Cellular and Molecular Regulation, National Institutes of Mental Health, Bethesda, MD 20892;
Laboratory of Adaptive Systems, National
Institute of Neurological Disorders and Stroke, Bethesda, MD 20892; and
§
Radiology and Biology Branch, National Cancer Institute, Bethesda,
MD 20892
Contributed by Julius Axelrod, April 27, 1998
ABSTRACT The neuroprotective actions of cannabidiol
and other cannabinoids were examined in rat cortical neuron
cultures exposed to toxic levels of the excitatory neurotrans-
mitter glutamate. Glutamate toxicity was reduced by both
cannabidiol, a nonpsychoactive constituent of marijuana, and
the psychotropic cannabinoid (2)D
9
-tetrahydrocannabinol
(THC). Cannabinoids protected equally well against neuro-
toxicity mediated by N-methyl-D-aspartate receptors, 2-ami-
no-3-(4-butyl-3-hydroxyisoxazol-5-yl)propionic acid recep-
tors, or kainate receptors. N-methyl-D-aspartate receptor-
induced toxicity has been shown to be calcium dependent; this
study demonstrates that 2-amino-3-(4-butyl-3-hydroxyisox-
azol-5-yl)propionic acidykainate receptor-type neurotoxicity
is also calcium-dependent, partly mediated by voltage sensi-
tive calcium channels. The neuroprotection observed with
cannabidiol and THC was unaffected by cannabinoid receptor
antagonist, indicating it to be cannabinoid receptor indepen-
dent. Previous studies have shown that glutamate toxicity may
be prevented by antioxidants. Cannabidiol, THC and several
synthetic cannabinoids all were demonstrated to be antioxi-
dants by cyclic voltametry. Cannabidiol and THC also were
shown to prevent hydroperoxide-induced oxidative damage as
well as or better than other antioxidants in a chemical (Fenton
reaction) system and neuronal cultures. Cannabidiol was
more protective against glutamate neurotoxicity than either
ascorbate or
a
-tocopherol, indicating it to be a potent anti-
oxidant. These data also suggest that the naturally occurring,
nonpsychotropic cannabinoid, cannabidiol, may be a poten-
tially useful therapeutic agent for the treatment of oxidative
neurological disorders such as cerebral ischemia.
Cannabinoid components of marijuana are known to exert
behavioral and psychotropic effects but also to possess ther-
apeutic properties including analgesia (1), ocular hypotension
(2), and antiemesis (3). This report examines another potential
therapeutic role for cannabinoids as neuroprotectants and
describes their mechanism of action in rat cortical neuronal
cultures.
During an ischemic episode, large quantities of the excita-
tory neurotransmitter glutamate are released. This event
causes neuronal death by over-stimulating N-methyl-
D-
aspartate receptors (NMDAr) and 2-amino-3-(4-butyl-3-
hydroxyisoxazol-5-yl)propionic acid (AMPA) and kainate-
type receptors and results in metabolic stress and accumulation
of toxic levels of intracellular calcium (4). In vitro and in vivo
studies (4, 5, 6) have demonstrated that such neurotoxicity can
be reduced by antioxidants or antagonists to NMDAr and
AMPAykainate receptors. Antioxidants such as
a
-tocopherol
(5, 6) are effective neuroprotectants because of their ability to
reduce the toxic reactive oxygen species (ROS) formed during
ischemic metabolism. Cannabinoids like (2)D
9
-tetrahydrocan-
nabinol (THC) and its psychoactive analogues also have been
reported to be neuroprotective against glutamate toxicity in
vitro (7). Cannabinoids have been suggested to prevent gluta-
mate neurotoxicity by activating cannabinoid receptors (7, 8),
which can reduce calcium influx through voltage sensitive
calcium channels (8, 9). A synthetic cannabinoid (HU-211)
also has been demonstrated to be neuroprotective even though
it does not activate cannabinoid receptors. This compound is
an atypical cannabinoid, however, in that it, unlike other
cannabinoids, directly antagonizes NMDAr (10) and possesses
some antioxidant properties (11). The present study examines
classical cannabinoids as neuroprotectants in vitro but focuses
on the nonpsychoactive cannabinoid cannabidiol. Like THC,
cannabidiol is a natural component of the marijuana plant,
Cannabis sativa, although unlike THC, cannabidiol does not
activate cannabinoid receptors and so is devoid of psychoactive
effects (12). This study reports that cannabidiol and other
cannabinoids such as THC are potent antioxidants that protect
neurons from glutamate-induced death without cannabinoid
receptor activation.
MATERIALS AND METHODS
Materials. Cannabidiol, THC, and reagents other than
those specifically listed below were purchased from Sigma.
Cyclothiazide, glutamatergic ligands, and MK-801 were ob-
tained from Tocris Cookson (Bristol, U.K.). Dihydrorhodam-
ine was supplied by Molecular Probes. Tert-butyl hydroperox-
ide, tetraethylammonium chloride, ferric citrate, and sodium
dithionite were all purchased from Aldrich. Agatoxin and
conotoxin were obtained through Biomol (Plymouth Meeting,
PA). All culture media were GIBCOyBRL products.
Solution Preparation. Solutions of cannabinoids, cyclothia-
zide, and other lipophiles were prepared by evaporating a 10
mM ethanolic solution (under a stream of nitrogen) in a
siliconized microcentrifuge tube. Dimethyl sulfoxide (,0.05%
of final volume) was added to ethanol to prevent the lipophile
from completely drying onto the tube wall. After evaporation,
1 ml of culture media was added, and the drug was dispersed
by using a high power sonic probe. Special attention was used
to ensure the solution did not overheat or generate foam. After
dispersal, all solutions were made to their final volume in
siliconized glass tubes by mixing with an appropriate quantity
of culture media.
Neuronal Cultures. Primary cortical neuron cultures were
prepared according to the method of Ventra et al (13). In brief,
fetuses were extracted by C-section from a 17-day pregnant
The publication costs of this article were defrayed in part by page charge
payment. This article must therefore be hereby marked ‘‘advertisement’’ in
accordance with 18 U.S.C. §1734 solely to indicate this fact.
0027-8424y98y958268-6$0.00y0
PNAS is available online at http:yywww.pnas.org.
Abbreviations: AMPA, 2-amino-3-(4-butyl-3-hydroxyisoxazol-5-
yl)propionic acid; BHT, butylhydroxytoluene; NMDAr, N-methyl-D-
aspartate receptors; ROS, reactive oxygen species; THC, (2)D
9
-
tetrahydrocannabinol.
To whom reprint requests should be addressed. e-mail: aidan@
codon.nih.gov.
8268
Wistar rat, and the fetal brains were placed into phosphate
buffered saline. The cortices then were dissected out, cut into
small pieces, and incubated with papain for 9 min at 37°. After
this time, the tissue was dissociated by passage through a
fire-polished Pasteur pipette, and the resultant cell suspension
was separated by centrifugation over a gradient consisting of
10 mgyml BSA and 10 mgyml ovomucoid (a trypsin inhibitor)
in Earle’s balanced salt solution. The pellet then was resus-
pended in high glucose, phenol red-free DMEM containing
10% fetal bovine serum, 2 mM glutamine, 100 units of
penicillin, and 100
m
gyml streptomycin (DMEM). Cells were
counted, were tested for vitality by using the trypan blue
exclusion test, and were seeded onto poly-
D-lysine coated 24
multiwell plates. After 96 hr, 10
m
M fluorodeoxyuridine and 10
m
M uridine were added to block glial cell growth. This protocol
results in a highly neuron-enriched culture (13).
Preparation of (Type I) Astrocytes and Conditioned Media.
Astrocyte-conditioned DMEM (phenol red-free) was used
throughout the AMPAykainate toxicity procedure and after
glutamate exposure in the NMDAr-mediated toxicity proto-
col. Media were conditioned by 24 hr of treatment over a
confluent layer of type I astrocytes prepared from 2-day-old
Wistar rat pups (14). In brief, cortices were dissected, were cut
into small pieces, were digested enzymatically with 0.25%
trypsin, and then were dissociated mechanically by passage
through a plastic pipette. The cell suspension then was plated
into untreated 75-cm
2
T-flasks, and, after 24 hr, the media
were replaced and unattached cells were removed. Once
astrocytes achieved confluency, cells were divided into four
flasks. Media for experiments were conditioned by a 24-hr
exposure to these astrocytes, after which time they were frozen
at 220°C until use. Astrocyte cultures were used to condition
DMEM for no longer than 2 months.
NMDAr-Mediated Toxicity Procedure. NMDAr-mediated
glutamate toxicity was examined by exposing neurons (cul-
tured for 14–18 days) to 250
m
M glutamate for 10 min in a
phenol red-free and magnesium-free saline. The saline was
composed of 125 mM NaCl, 25 mM Glucose, 10 mM Hepes
(pH 7.4), 5 mM KCl, 1.8 mM calcium chloride, and 5% BSA.
After exposure, cells were washed twice with saline and were
incubated for 18 hr in conditioned DMEM. Toxicity was
prevented completely by addition of the NMDAr antagonist
MK-801 (500 nM) (data not shown).
AMPA and Kainate Receptor-Mediated Toxicity Proce-
dures. Unlike NMDAr, which are regulated by magnesium
ions, AMPAykainate receptors rapidly desensitize after ligand
binding. To examine AMPA and kainate receptor-mediated
toxicity, neurons were cultured for 7–13 days and then were
exposed to 100
m
M glutamate and 50
m
M cyclothiazide (used
to prevent AMPA receptor desensitization). Cells were incu-
bated with glutamate in the presence of 500 nM MK-801 for
18–20 hr before analysis. Specific AMPA and kainate receptor
ligands also were used to separately examine the effects of
cannabinoids on AMPA and kainate receptor-mediated
events. Fluorowillardiine (1.5
m
M) and 4-methyl glutamate (10
m
M) were used to investigate AMPA and kainate (15) recep-
tor-mediated toxicity, respectively. When specifically examin-
ing kainate receptor activity, cyclothiazide was replaced with
0.15 mgyml Concanavalin-A.
Although the neuron preparation technique described
above results in a largely neuronal culture, a minority of
astrocytic cells remain. Astrocytes are highly resistant to
glutamate toxicity (16) because of their lack of functional
NMDAr (17, 18), although glutamate toxicity in astrocytes has
been observed to prevent AMPA receptor desensitization if
cyclothiazide is present (19). To examine whether AMPAy
kainate-type toxicity affects astrocytes in our cultures, astro-
cytes (as prepared above) were exposed to glutamate under the
same conditions used on neuron-enriched cultures. Under
these conditions, astrocytes were resistant to glutamate tox-
icity, with 20 hr of exposure resulting in a lactate dehydroge-
nase release of only 5% above background, compared with
100–200% of the background observed in neuron-enriched
cultures (data not shown). It was concluded, therefore, that
astrocyte contamination does not contribute substantially to
the effects of glutamate in our neuronal cultures.
ROS Toxicity Assay. To examine the effects of cannabinoids
on ROS toxicity, 7- to 13-day-old cultured neurons were
incubated with 300
m
M tert-butyl hydroperoxide (an oxidant)
in conditioned DMEM. Tert-butyl hydroperoxide was used
because its miscibility with both water and lipids allows
oxidation to occur in both cytosolic and membrane-delimited
cellular compartments.
Toxicity Assay. Cell toxicity was assessed 18–20 hrs after
insult by measuring lactate dehydrogenase release into the
(phenol red-free) culture media. Experiments were conducted
with triple or quadruple values at each point, and all plates
contained positive (glutamate alone) and baseline controls.
The assay was validated by comparison with a tetrazolium-
based viability assay (XTT) (20). Results were similar with
either system, although lactate dehydrogenase release was
used in this study because it provided a greater signal to noise
ratio than the XTT assay.
Cyclic Voltametry. Cyclic voltametry was performed with an
EG & G Princeton Applied Research potentiostatygalvanostat
(
MODEL 273yPAR 270 software). The working electrode was a
glassy carbon disk with a platinum counter electrode and
silverysilver chloride reference. Tetraethylammonium chlo-
ride in acetonitrile (0.1 M) was used as an electrolyte. Cyclic
voltametry scans were done from 0 to 11.8 V at scan rate of
100 mV per second.
Iron-Catalysed Dihydrorhodamine Oxidation (Fenton Re-
action). The antioxidant activities of each of the compounds
were evaluated by their ability to prevent oxidation of dihy-
drorhodamine to the fluorescent compound rhodamine. Ox-
idant was generated by ferrous catalysis (diothionite-reduced
ferric citrate) of tert-butyl hydroperoxide in a 50:50 water-to-
acetonitrile (volyvol) solution. Dihydrorhodamine (50
m
M)
was incubated with 300
m
M tert-butyl hydroperoxide and 0.5
m
M iron for 5 min. After this time, oxidation was assessed by
spectrofluorimetry (Excitation 5 500 nm, Emission 5 570
nm). Various concentrations of cannabinoids and butylhy-
droxytoluene (BHT) were included to examine their ability to
prevent dihydrorhodamine oxidation.
Data Analysis. Data are reported as mean values plus and
minus standard error. Significance was examined by using a
Student’s t test, (P # 0.05). Kinetic data was analyzed by using
GraphPad’s
PRISM software package (GraphPad, San Diego)
for PC.
RESULTS
Cannabidiol Blocks NMDAr and AMPA and Kainate Re-
ceptor-Mediated Neurotoxicity. Glutamate neurotoxicity can
be mediated by NMDAr, AMPA receptors, or kainate recep-
tors. To examine NMDAr-mediated toxicity, rat cortical neu-
rons were exposed to glutamate for 10 min in a magnesium-
free medium, and the level of lactate dehydrogenase released
was used as an index of cell injury. To examine AMPAykainate
receptor-mediated toxicity, neurons were incubated for 20 hr
with glutamate or a specific AMPA or kainate receptor ligand
(fluorowillardiine or 4-methyl-glutamate, respectively). An
NMDAr antagonist (MK-801) and an agent to prevent recep-
tor desensitization also were included. Cannabidiol prevented
cell death equally well (EC
50
of 2–4
m
M) in both NMDAr and
AMPAykainate toxicity models (Fig. 1 A and B). Similar data
also was observed when glutamate was replaced with either
AMPA-specific or kainate receptor-specific ligands (data not
shown). These results suggest that cannabidiol protects simi-
Medical Sciences: Hampson et al. Proc. Natl. Acad. Sci. USA 95 (1998) 8269
larly, regardless of whether toxicity is mediated by NMDA,
AMPA, or kainate receptors.
AMPAyKainate Toxicity Is Calcium Dependent. Increased
calcium influx is known to be a key factor in NMDAr-induced
cell death (4), but its role in AMPA and kainate toxicity is less
clear. It has been suggested that AMPAykainate receptors may
not directly allow entry of sufficient calcium to kill cells.
However, AMPAykainate receptors flux large amounts of
sodium, which can depolarize cell membranes. Such depolar-
ization may activate both voltage-sensitive calcium channels
(21) and facilitate NMDAr activation (22, 23). In this way,
AMPAykainate receptor stimulation may lead indirectly to
accumulation of toxic intracellular calcium levels. Addition of
the calcium chelator EDTA reduced toxicity in a concentra-
tion-dependent manner, demonstrating the involvement of
calcium in AMPAykainate-type toxicity (data not shown).
EDTA (2 mM) eliminated '70% of glutamate toxicity (Fig. 2)
even though the presence of MK-801 prevented NMDAr
activation. Toxicity also was reduced by inhibitors to L-, N-,
and PyQ- type calcium channels (nifedipine, agatoxin IVa, and
conotoxin GVIa, respectively; Fig. 2), indicating that voltage-
sensitive calcium channels also are involved in AMPAy
kainate-type toxicity. However, a combination of these calcium
channel inhibitors did not completely block EDTA-
preventable (calcium-dependant) cell death.
Neuroprotection by Tetrahydrocannabinol. Unlike canna-
bidiol, THC is a ligand for the brain cannabinoid receptor (24),
and this action has been proposed to explain the ability of THC
to protect neurons from NMDAr toxicity in vitro (7). However,
in AMPAykainate receptor toxicity assays, THC and canna-
bidiol were similarly protective, suggesting that cannabinoid
neuroprotection may be independent of cannabinoid receptor
activation. This was confirmed by inclusion of a cannabinoid
receptor antagonist, SR-141716A (Fig. 3). Neither THC or
cannabidiol neuroprotection was affected by cannabinoid re-
ceptor antagonist.
Cannabinoids as Antioxidants. Cells use easily oxidizable
compounds such as glutathione, ascorbate, and
a
-tocopherol
as antioxidants that protect important cellular structures (e.g.,
DNA, proteins, and membranes) from ROS damage. Studies
have suggested that ROS damage may be involved in glutamate
neurotoxicity (5, 6). To investigate whether cannabinoids
could protect neurons against glutamate by reacting with ROS,
the antioxidant properties of cannabidiol and other cannabi-
noids were assessed. Cyclic voltametry, a procedure that
measures the ability of a compound to accept or donate
electrons under a variable voltage potential, was used to
measure the oxidation potentials of several natural and syn-
thetic cannabinoids. Cannabidiol, THC, and the synthetic
FIG. 2. The involvement of calcium and calcium channels in
AMPAykainate-mediated toxicity. The effects of 2 mM EDTA and
various combinations of the voltage-sensitive calcium channel inhib-
itors
v
-Agatoxin IVa (Ag) (250 nM),
v
-Conotoxin GVIa (CTx) (500
nM), and Nifedipine (Nif) (1
m
M) were used to probe the role and
source of calcium in AMPAykainate receptor-mediated toxicity. Data
represents mean values 6 SEM from four experiments, each with four
replicates. Cannabinoids were present throughout the glutamate ex-
posure period. See Materials and Methods for further experimental
details. Significant difference between EDTA and other treatments is
indicated with an asterisk.
FIG. 1. Effect of cannabidiol on NMDAr- (A) and AMPAykainate receptor- (B) mediated neurotoxicity. Data shown represents mean values 6
SEM from a single experiment with four replicates. Each experiment was repeated on at least four occasions with essentially the same results.
Cannabinoids were present during (and, in the case of NMDAr mediated toxicity, after) the glutamate exposure periods. See Materials and Methods
for further experimental details.
8270 Medical Sciences: Hampson et al. Proc. Natl. Acad. Sci. USA 95 (1998)
cannabinoid HU-211 all donated electrons at a similar poten-
tial as the antioxidant BHT. Anandamide (arachidonyl-
ethanolamide), which is not a cannabinoid in structure but is
an endogenous ligand for the cannabinoid receptor, did not
undergo oxidation in this assay (Fig. 4A). Three other canna-
binoids, cannabinol, nabilone, and levanantrodol, also were
tested, and they, too, exhibited oxidation profiles similar to
cannabidiol and THC (data not shown).
The ability of cannabinoids to be oxidized readily suggests
that they may possess antioxidant properties comparable to
BHT. These properties were examined further in a Fenton
reaction (iron-catalyzed ROS generation). Tert-butyl hy-
droperoxide was used to generate ROS and oxidize dihydror-
hodamine into the fluorescent compound rhodamine. Canna-
bidiol, THC, and BHT all prevented dihydrorhodamine oxi-
dation in a similar, concentration-dependent manner (Fig. 4B),
indicating cannabinoids to be comparable to BHT in antiox-
idant potency. To confirm that cannabinoids act as antioxi-
dants in the intact cell, neurons were incubated with tert-butyl
hydroperoxide and varying concentrations of cannabidiol (Fig.
5A). The oxidant was chosen for its solubility in both aqueous
and organic solvents, thereby facilitating oxidation in both
cytosolic and membrane cell compartments. As observed in
studies with glutamate, cannabidiol protected neurons against
ROS toxicity in a concentration-related manner. Cannabidiol
also was compared with antioxidants in an AMPAykainate
toxicity protocol. Neurons were exposed to glutamate and
equimolar cannabidiol,
a
-tocopherol, BHT, or ascorbate (Fig.
5B). Although all of the antioxidants attenuated glutamate
toxicity, cannabidiol was significantly more protective than
either
a
-tocopherol or ascorbate.
DISCUSSION
The nonpsychoactive marijuana constituent cannabidiol was
found to prevent both glutamate neurotoxicity and ROS-
induced cell death. The psychoactive principle of Cannabis,
THC, also blocked glutamate neurotoxicity with a similar
potency to cannabidiol. In both cases, neuroprotection was
unaffected by cannabinoid receptor antagonist. This suggests
that cannabinoids may have potentially useful therapeutic
effects that are independent of psychoactivity-inducing can-
nabinoid receptors (12) and so are not necessarily accompa-
nied by psychotropic side effects.
Cannabidiol blocked glutamate toxicity in cortical neurons
with equal potency regardless of whether the insult was
mediated by NMDAr, AMPA receptors, or kainate receptors.
This suggests that either cannabinoids antagonize all three
glutamate receptors with the same affinity, or, more likely,
their site of action is downstream of initial receptor activation
FIG. 3. Effect of THC, cannabidiol, and cannabinoid receptor
antagonist on glutamate induced neurotoxicity. Neurons exposed to
glutamate in an AMPAykainate receptor toxicity model were incu-
bated with 10
m
M cannabidiol or THC in the presence or absence of
SR141716A (500nM). See Materials and Methods for experimental
details. Data represents mean values 6 SEM from four experiments,
each with three replicates.
FIG.4. (A) A comparison of the oxidation potentials of cannabinoids and the antioxidant BHT. The oxidation profiles of (750
m
M) BHT,
cannabinoids, and anandamide were compared by cyclic voltametry. Anandamide, a cannabinoid receptor ligand with a noncannabinoid structure,
was used as a nonresponsive control. Experiments were repeated three times with essentially the same results. See Materials and Methods for
experimental details. (B) Effect of cannabidiol and THC on dihydrorhodamine oxidation. Cannabinoids were compared with BHT for their ability
to prevent tert-butyl hydroperoxide-induced oxidation of dihydrorhodamine. See Materials and Methods for experimental details. Data represent
mean values 6 SEM from a single experiment with three replicates. This experiment was repeated four times with essentially the same results.
Medical Sciences: Hampson et al. Proc. Natl. Acad. Sci. USA 95 (1998) 8271
events. Neurotoxic concentrations of glutamate induce mas-
sive calcium influx through NMDAr (4) that ultimately kills
the cell. This study has demonstrated that the toxic effects of
glutamate are also calcium-dependent when mediated by
AMPAykainate receptors. Both EDTA (a calcium chelator)
and voltage-sensitive calcium channel inhibitors reduced
AMPA-ykainate-type neurotoxicity, indicating that a portion
of calcium influx-associated AMPA-ykainate-receptor activa-
tion is mediated by secondary activation of calcium channels.
However, the mixture of calcium channel inhibitors and
NMDAr antagonist did not eliminate completely glutamate
toxicity or reduce cell death as efficiently as EDTA. This
suggests that although toxicity resulting from AMPAykainate
receptor stimulation may be caused by calcium entering the
cell by several routes, it is not caused exclusively by calcium
channel activity. These studies also demonstrate that NMDAr
activation is not required for AMPA-ykainate-type toxicity [as
suggested (22)].
Accumulation of ROS has been shown to be involved in
NMDAr-mediated cell death (4). The current study has sim-
ilarly demonstrated that AMPAykainate receptor-induced
toxicity also involves ROS formation and may be prevented
with antioxidant treatment. Cannabidiol and THC were found
to be comparable with BHT (antioxidant) in both their ability
to prevent dihydrorhodamine oxidation (Fenton reaction) and
their cyclic voltametric profiles. Synthetic cannabinoids such as
HU-211, nabilone, and levanantradol also exhibited similar
profiles. Anandamide, which is a natural cannabinoid receptor
ligand but is not structurally related to cannabinoids, did not
give an antioxidant-like profile by cyclic voltametry, which
indicates that cannabinoids can act as reducing agents (in a
chemical system). To confirm that cannabinoids also can
function as antioxidants in living cells, a lipid hydroperoxide
was used to generate ROS toxicity in neuronal cultures. As
observed in the Fenton reaction system, cannabidiol attenu-
ated this ROS-induced neurotoxicity. These observations in-
dicate that many cannabinoids exert a considerable protective
antioxidant effect in neuronal cultures. The similarity of the
voltamagrams observed with cannabidiol, HU-211, and several
other cannabinoids also suggests that the reported antioxidant
effect of HU-211 is not a feature unique to this atypical
cannabinoid, (as previously implied; e.g., ref. 11) but, rather,
a common property of classical cannabinoid structures. The
potency of cannabidiol as an antioxidant was examined by
comparing it on an equimolar basis with other commonly used
antioxidants. Cannabidiol protected neurons to a greater
degree than either of the dietary antioxidants,
a
-tocopherol or
ascorbate. As in the Fenton reaction system, cannabidiol
protected neurons with comparable efficacy to the potent
antioxidant BHT. The similar antioxidant abilities of canna-
bidiol and BHT in this chemical system and their comparable
protection in neuronal cultures implies that cannabidiol neu-
roprotection is caused by an antioxidant effect.
The antioxidative properties of cannabinoids suggest a
therapeutic use as neuroprotective agents, and the particular
properties of cannabidiol make it a good candidate for such
development. Although cannabidiol was similar in neuropro-
tective capacity to BHT, cannabidiol has no known tumor-
promoting effects [unlike BHT (25, 26)]. The lack of psycho-
activity associated with cannabidiol allows it to be adminis-
tered in higher doses than would be possible with psychotropic
cannabinoids such as THC. Furthermore, the ability of can-
nabidiol to protect against neuronal injury without inhibiting
NMDAr may reduce the occurrence of toxicity or side effects
associated with NMDAr antagonists (27). Previous studies
have indicated that cannabidiol is not toxic, even when chron-
ically administered to humans (28) or given in large acute doses
[700 mgyday (29)]. In vivo studies to examine the efficacy of
cannabidiol as a treatment for experimentally induced isch-
emic stroke are currently in progress.
1. Welch, S. P. & Stevens, D. L. (1992) J. Pharmacol. Exp.Ther. 262,
8–10.
2. Merritt, J. C., Crawford, W. J., Alexander, P. C., Anduze, A. L.
& Gelbart, S. S. (1980) Ophthalmology 87, 222–228.
3. Abrahamov, A. & Mechoulam R. (1995) Life Sci. 56, 2097–2102.
4. Choi, D. W., Koh, J. Y. & Peters, S. (1988) J. Neurosci. 8, 185–196.
5. Ciani, E., Groneng, L., Voltattorni, M., Rolseth, V., Contest-
abile, A. & Paulsen, R. E. (1996) Brain Res. 728, 1–6.
FIG.5. (A) The effect of cannabidiol on oxidative toxicity in neuronal cultures. Tert-butyl hydroperoxide-induced toxicity was examined in the
presence or absence of cannabidiol. (B) Comparison of antioxidants and cannabidiol for their ability to prevent glutamate toxicity in neurons. The
effects of cannabidiol, BHT, ascorbate, and
a
-tocopherol (10
m
M) were examined in a model of AMPAykainate receptor-dependent toxicity. All
drugs were present throughout the glutamate exposure period. Each experiment represents the mean of four replicates repeated on three occasions.
See Materials and Methods for further experimental details. Significant differences between cannabidiol and other antioxidants are indicated with
an asterisk.
8272 Medical Sciences: Hampson et al. Proc. Natl. Acad. Sci. USA 95 (1998)
6. MacGregor, D. G., Higgins, M. J., Jones, P. A., Maxwell, W. L.,
Watson, M. W., Graham, D. I. & Stone, T. W. (1996) Brain Res.
727, 133–144.
7. Skaper, S. D., Buriani, A., Dal Toso, R., Petrell, I. L., Romanello,
L., Facci, L. & Leon, A (1996) Proc. Natl. Acad. Sci. USA 93,
3984–3989.
8. Hampson, A. J., Bornheim, L. M., Scanziani, M., Yost, C. S, Gray,
A. T., Hansen, B. M, Leonoudakis, D. J. & Bickler, P. E. (1998)
J. Neurochem. 70, 671–676.
9. Twitchell, W., Brown, S. & Mackie, K. (1997) J. Neurophysiol. 78,
43–50.
10. Biegon, A. (1995) Ann. N.Y. Acad. Sci. 765, 314.
11. Eshhar, N., Striem, S., Kohen, R., Tirosh, O. & Biegon, A (1995)
Eur. J. Pharmacol. 283(1–3), 19–29.
12. Mansbach, R. S., Rovetti, C. C., Winston, E. N. & Lowe, J. A.,
III (1996) Psychopharmacol. 124, 315–22.
13. Ventra, C, Porcellini, A., Feliciello, A. R., Gallo, A., Paolillo, M.,
Mele, E., Avedimento, V. E. & Schettini, G. (1996) J. Neurochem.
66, 1752–1761.
14. Grimaldi, M., Pozzoli, G., Navarra, P., Preziosi, P. & Schettini,
G. (1994) J. Neurochem. 63, 344–350.
15. Zhou, L. M., Gu, Z. Q., Costa, A. M., Yamada, K. A., Mansson,
P. E., Giordano, T., Skolnick, P. & Jones, K. A (1997) J. Phar-
macol. Exp. Ther. 280, 422–427.
16. Amin, N. & Pearce, B. (1997) Neurochem. Int. 30, 271–276.
17. Holopainen, I., Saransaari, P. & Oja, S. S. (1994) Neurochem. Res.
19, 111–115.
18. Van Bockstaele, E. J. & Colago, E. E. J. (1996) J. Comp. Neurol.
369, 483–496.
19. David, J. C., Yamada, K. A., Bagwe, M. R. & Goldberg, M. P.
(1996) J. Neurosci. 16, 200–209.
20. Roehm, N. W., Rodgers, G. H., Hatfield, S. M. & Glasebrook,
A. L. (1991) J. Immunol. Methods 142, 257–265.
21. Hack, N. & Balazs, R (1995) J. Neurochem. 65, 1077–1084.
22. Berman, F. W. & Murray, T. F. J. (1996) J. Biochem. Toxicol. 11,
111–119.
23. Deupree, D. L., Tang, X. W., Yarom, M., Dickman, E., Kirch,
R. D., Schloss, J. V. & Wu, J. Y. (1996) Neurochem. Int. 29,
255–261.
24. Devane, W. A., Dysarz, F. A., Johnson, M. R., Melvin, L. S. &
Howlett, A. C. (1988) Mol. Pharmacol. 34, 605–613.
25. Thompson, J. A., Bolton, J. L. & Malkinson, A. M. (1991) Exp.
Lung Res. 17, 439453.
26. Lindenschmidt, R. C., Tryka, A. F., Goad, M. E. & Witschi, H. P.
(1986) Toxicology 38, 151–160.
27. Auer, R. N (1994) Psychopharmacol. Bull. 30, 585–591.
28. Cunha, J. M., Carlini, E. A., Pereira, A. E., Ramos, O. L.,
Pimentel, C., Gagliardi, R., Sanvito, W. L., Lander, N. &
Mechoulam, R. (1980) Pharmacology 21, 175–185.
29. Consroe, P., Laguna, J., Allender, J., Snider, S., Stern, L., Sandyk,
R., Kennedy, K. & Schram, K. (1991) Pharmacol. Biochem.
Behav. 40, 701–708.
Medical Sciences: Hampson et al. Proc. Natl. Acad. Sci. USA 95 (1998) 8273
... CBD products are purported to treat numerous health conditions in the popular media, but in almost every instance lack approval from a regulatory agency. While there are studies reporting the beneficial effects of CBD [1,2,36,40,41], evidence on its adverse effects has also emerged [26,27,42]. Its interaction with other drugs is another concern [43]. ...
... It was reported that high levels of GSH were essential for stem cells [56]. CBD was reported to have antioxidative and neuroprotective effects mediated by various mechanisms [41,[57][58][59]; THC has demonstrated different effects on redox homeostasis in different situations [37,38]. In this study, we measured cellular GSH levels to understand NSC redox status after exposure to CBD, its metabolites, and THC for 7 days. ...
Article
Full-text available
Cannabidiol (CBD) has been used for different purposes by different populations in recent years. When consumed by pregnant women, CBD can pass through the placenta and enter the fetal blood stream. There is concern over adverse effects of fetal exposure to CBD and its major metabolites (7-OH-CBD and 7-COOH-CBD). In the present study, human neural stem cells (NSCs) were treated with CBD and its metabolites at different concentrations for various durations to understand how the drug may affect fetal brain development. NSCs were also treated with delta-9 tetrahydrocannabinol (THC) for comparison purposes. CBD, 7-OH-CBD and 7-COOH-CBD dose-dependently reduced NSC viability. CBD and 7-OH-CBD reduced NSC number at the G1 phase. A 24 h exposure did not cause significant change in NSC proliferation. At concentrations comparable to those detected in human blood, longer exposures to CBD, 7-OH-CBD and 7-COOH-CBD caused more obvious cell death. After NSCs differentiation, CBD treatment reduced GFAP and cannabinoid receptor 2 (CB2) expression. THC treatment reduced the GFAP expression, but the change in CB2 expression did not reach statistical significance. The expression of cannabinoid receptor 1 (CB1) and beta-tubulin III were not significantly altered by drug exposures. The study demonstrated that clinically relevant concentrations of CBD, 7-OH-CBD and 7-COOH-CBD affect basic physiological features of human NSCs. After NSC differentiation, the reduced expression of CB2 receptors and GFAP on differentiated cells further indicated the vulnerability of developing central nervous system to CBD and THC. These data will help to contextualize in vivo neurodevelopmental studies that may not accurately model human metabolite profiles of CBD.
... 3,4) CB1R signaling has been widely recognized for its neuroprotective effects against various forms of neurotoxicity. [5][6][7][8][9][10][11][12] This protective role is evidenced by studies showing that CB1R-deficient mice exhibit increased sensitivity to neurotoxicity caused by ischemia or excitotoxicity. 8,9) Furthermore, CB1R agonists demonstrate neuroprotective effects in both in vitro and in vivo models of neurotoxicity. ...
... 8,9) Furthermore, CB1R agonists demonstrate neuroprotective effects in both in vitro and in vivo models of neurotoxicity. [5][6][7] CB1R activity also plays a critical role in mitigating age-related cognitive decline and in preserving neuronal physiology. [10][11][12] Recent findings indicate that CB1R is not only present on the plasma membrane but also localized on the outer membrane of mitochondria in neuronal cells. ...
Article
Full-text available
Cannabinoid receptor type 1 (CB1R) plays a key role in neuronal homeostasis, synaptic plasticity, and neuroprotection. CB1R antagonists typically protect against CB1R agonists-induced neurotoxicity. However, we previously found that the CB1R antagonists rimonabant and its analog AM251 can also be neurotoxic: under serum-free conditions, these compounds induce apoptosis in human neuroblastoma SH-SY5Y cells through mitochondrial damage and endoplasmic reticulum (ER) stress. To elucidate the mechanisms of this neurotoxicity, we examined the effects of CB1R agonists. We co-treated SH-SY5Y cells with rimonabant or AM251 in combination with either the CB1R agonist arachidonyl 2-chloroethylamide (ACEA) or WIN 55212-2 mesylate (WIN). ACEA, but not WIN, protected cells from rimonabant- and AM251-induced apoptosis. While ACEA had only a limited effect on mitochondrial damage, it significantly reduced phosphorylation of the eukaryotic initiation factor 2 alpha (eIF2α), a key marker of ER stress. Given that ACEA also functions as an agonist of transient receptor potential vanilloid 1 (TRPV1), we investigated its role in ACEA-mediated neuroprotection. The TRPV1 antagonist capsazepine blocked ACEA’s protective effects, suggesting that ACEA acts through TRPV1 rather than CB1R. ACEA also prevented apoptosis induced by camptothecin, a well-established apoptosis inducer, through a similar capsazepine-sensitive mechanism, demonstrating its broader protective effects against apoptosis. These findings indicate that rimonabant and AM251 induce neurotoxicity independently of CB1R under serum-free conditions and that ER stress is likely to be a key target of CB1R-independent neuroprotection by ACEA. Our study highlights the complexity of CB1R ligand-associated neurotoxicity and neuroprotection. Fullsize Image
... Our results show that CBD decreases the formation of ROS species induced by Aß 1-42 , Tau, and pTau and recovers neuronal survival after both Aβ 1-42 peptide and NMDA-induced toxicity in primary cortical neurons. These data agree with those published in primary neuronal cultures that show a strong antioxidant effect of CBD against glutamate toxicity [43] and protection from amyloid β-peptide 25-35 -induced neurotoxicity [44]. In this context, transmission electron microscopy assays have also shown that CBD treatment up-regulated the autophagy pathway in hippocampal neurons of APP/PS1 mice model of AD [45]. ...
Article
Full-text available
Background Cannabidiol (CBD), the second most abundant phytocannabinoid in Cannabis sativa, has garnered significant interest due to its non-psychoactive nature and diverse receptor interactions. Methods This study employs in vitro and in vivo methodologies to validate CBD's potential as a treatment for Alzheimer's disease (AD) by addressing key hallmarks of the condition and promoting neuroprotective effects on spatial memory. Results Our findings demonstrate CBD's ability to decrease pTau and Aβ aggregation and to mitigate their axonal transport between cortical and hippocampal neurons. Moreover, CBD treatment was shown to reduce neuroinflammation, as CBD was able to skew microglia towards a neuroprotective M2 phenotype while attenuating proinflammatory cytokine release in the 5xFAD AD mouse model. Notably, daily CBD injections (10 mg/Kg) for 28 days in 5xFAD mice resulted in significant improvements in both short- and long-term spatial memory. The study also reveals CBD's capacity to partially revert neurite formation loss induced by Aβ, Tau, and pTau proteins, suggesting a potential role in promoting neuronal plasticity. Additionally, CBD treatment led to a reduction in reactive oxygen species (ROS) formation and increased neuronal viability in the presence of AD-associated protein aggregates. Conclusions These multifaceted effects of CBD, ranging from molecular-level modulation to behavioral improvements, underscore its potential as a comprehensive therapeutic approach for AD. The findings not only support CBD's neuroprotective properties but also highlight its ability to target multiple pathological processes simultaneously, offering a promising avenue for future AD treatment strategies.
... Over the last few years, pharmacological and pharmacokinetic investigative studies have confirmed the clinical effectiveness of cannabinoids (CBD) as a safe drug for human use. For instance, CBD has been widely reported as a neuroprotective antioxidant [21,22] to inhibit inflammation [23,24] via the activation of the N-methyl-D-aspartate (NMDA) receptor [25]. A study reported that administration of CBD in collagen-induced arthritis mice could modulate the immune system, decreasing the release of interleukin-1 and TNF-α to exert anti-inflammatory effects [24]. ...
Article
Full-text available
Background/Objectives: Age-related macular degeneration (AMD) is the third leading cause of irreversible blindness in elderly individuals aged over 50 years old. Oxidative stress plays a crucial role in the etiopathogenesis of multifactorial AMD disease. The phospholipid bilayer EVs derived from the culture-conditioned medium of human induced pluripotent stem cell (hiPSC) differentiated retinal organoids aid in cell-to-cell communication, signaling, and extracellular matrix remodeling. The goal of the current study is to establish and evaluate the encapsulation of a hydrophobic compound, cannabidiol (CBD), into retinal organoid-derived extracellular vesicles (EVs) for potential therapeutic use in AMD. Methods: hiPSC-derived retinal organoid EVs were encapsulated with CBD via sonication (CBD-EVs), and structural features were elucidated using atomic force microscopy, nanoparticle tracking analysis, and small/microRNA (miRNA) sequencing. ARPE-19 cells and oxidative-stressed (H2O2) ARPE-19 cells treated with CBD-EVs were assessed for cytotoxicity, apoptosis (MTT assay), reactive oxygen species (DCFDA), and antioxidant proteins (immunohistochemistry and Western blot). Results: Distinct miRNA cargo were identified in early and late retinal organoid-derived EVs, implicating their roles in retinal development, differentiation, and functionality. The therapeutic effects of CBD-loaded EVs on oxidative-stressed ARPE-19 cells showed greater viability, decreased ROS production, downregulated expression of inflammation- and apoptosis-related proteins, and upregulated expression of antioxidants by Western blot and immunocytochemistry. Conclusions: miRNAs are both prognostic and predictive biomarkers and can be a target for developing therapy since they regulate RPE physiology and diseases. Our findings indicate that CBD-EVs could potentially alleviate the course of AMD by activating the targeted proteins linked to the adenosine monophosphate kinase (AMPK) pathway. Implicating the use of CBD-EVs represents a novel frontline to promote long-term abstinence from drugs and pharmacotherapy development in treating AMD.
... Cannabinoids, mainly cannabidiol, have been associated with neuroprotec�ve effects and the studies with rat cor�cal neuron cultures exposed to toxic levels of glutamate support a further inves�ga�on of these compounds in different pathologies, including ischemic stroke [88,89]. Also, cannabidiol is increasingly inves�gated in other disorders, including psychiatric illnesses, due to its anxioly�c, an�depressant, and an�psycho�c proper�es, and the benefit of being administered as an adjuvant to current treatment, thus poten�ally decreasing the rate of treatment resistance [90,91]. ...
Article
A significant number of neurological diseases are pathogenetically related to oxidative stress, including but not limited to cerebrovascular afflictions such as ischemic and hemorrhagic stroke, Parkinson’s disease, Alzheimer’s disease, multiple sclerosis, amyotrophic lateral sclerosis, and so on. Natural nutrients may help limit the impact of oxidative stress and, therefore, delay or prevent the impairment of these diseases. Although these natural components have not entered routine use, many have been studied in preclinical or even clinical settings with promising results. Therefore, the need to find the stage of the research in the field of validating the properties and clinical usefulness of such nutrients represents the main reason this narrative review was conducted. This analysis explored the PubMed database for papers related to the influence of natural nutrients on the onset and evolution of some of the most severe neurological disorders. The results of the review provide an overview of the pathways oxidative stress may undertake and how the use of nutrients may counteract these pathways. In conclusion, natural nutrients may have beneficial effects that can be impactful on clinical outcomes, but more good quality research in this field is needed before formulating any clear recommendation.
... Many preclinical and clinical studies have suggested that CBD has a broad range of therapeutic properties, such as antipsychotic, analgesic, anticonvulsant, antineoplastic, antioxidant properties [20]. Recent studies have shown the anxiolytic effect of CBD and also suggest a decrease in the salivary cortisol level [21,22]. It is said that these anxiolytic properties are mediated by the interaction with different receptors, including CB1, 5-HT 1A , TRPV1 [23], but there are also other receptors that CBD is able to modulate, namely PPARγ, GPR55, 5-HT, GABAA. ...
Article
Full-text available
Numerous preclinical and clinical studies indicate that CBD possesses various therapeutic properties, including antipsychotic, analgesic, anticonvulsant, antineoplastic, and antioxidant effects. Recent research has also highlighted its potential anxiolytic effects. This study aimed to evaluate the impact of CBD treatment in a PTSD induction model. To determine CBD’s efficacy, behavioral tests assessing anxiety and memory were conducted. Additionally, two oxidative stress markers were measured to explore its antioxidant properties. Forty adult male rats were used for PTSD induction. The procedure involved exposure to predator odor on day 10, followed by a second exposure on day 20. A secondary stressor, consisting of daily cage partner changes, was also applied. The animals were randomized into four groups: two non-stressed and two stressed groups. CBD was administered at 10 mg/kg. Behavioral effects were evaluated using the open field (OF), elevated plus maze (EPM), novel object recognition (NOR), and Morris Water Maze (MWM) tests. Malondialdehyde and the GSH/GSSG ratio were assessed using liquid chromatography. CBD treatment did not significantly alter anxiety-like behavior in the EPM, though a trend toward increased vertical exploration was observed in the OF test. In memory-related assessments, no significant differences were found in the NOR test, while performance in the MWM indicated improved spatial memory, with CBD-treated rats spending more time in the target quadrant. In addition, malondialdehyde levels decreased in the CBD groups. Elevated cortisol levels in the stressed CBD group suggest a potential anxiolytic effect, warranting further research.
Article
Cannabidiol (CBD) is a phytocannabinoid without intoxicating properties. While CBD can improve neurophysiological functions and subjective symptoms, its effect on cognitive function remains unclear. We summarized the available randomized controlled trials investigating CBD administration and cognitive function. A review of the literature was conducted using the following keywords on PubMed/Medline: (cannabis OR cannabidiol OR cannabinoid OR CBD OR Δ ⁹ -tetrahydrocannabinol OR tetrahydrocannabinol) AND (neurology OR brain OR psychiatric OR neuroscience OR psychology OR cognition) AND (human) AND (randomized controlled trial OR RCT). The search yielded 1038 articles with 36 total included for this literature review. The articles included healthy participants, neurological disease, psychiatric disease, psychosis, paranoia, schizophrenia, and drug-use disorders. Studies with healthy participants included a variety of dosing strategies, suggesting an effect on cognitive function and sleep quality. In Parkinson’s disease, 75–300 mg CBD resulted in mild improvements in daily life activities. Decreases in subjective anxiety were found in patients with psychiatric disease using CBD doses ranging from 300 to 400 mg. In patients with psychosis and paranoia, 600 mg CBD showed inconsistent results in cognitive function. In patients with schizophrenia, up to 1000 mg CBD per day had minimal effects on cognition. Finally, up to 800 mg CBD had minimal effects on cognitive function in patients with substance use disorders. The findings are limited by utilization of acute dosing, variations in CBD dose, and different routes of administration. Standardized dosing and CBD formulations are needed to assess its efficacy for improving cognition.
Article
Full-text available
The antinociceptive effects of various cannabinoids, alone and in combination with opiates, were evaluated in antinociceptive tests in mice. The cannabinoids tested produce marked antinociceptive effects after i.t. administration to mice. The rank order of potency for the drugs using the tail-flick test was levonantradol greater than CP-55,940 = CP-56,667 greater than 11-hydroxy-delta 9-THC greater than delta 9-THC greater than delta 8-THC; dextronantradol was inactive at a dose of 25 micrograms/mouse. Respective ED50 values in the tail-flick test were 0.4, 12.3, 4.2, 15, 45 and 72 micrograms/mouse. Although pretreatment with morphine somewhat enhanced the effects of delta 9-THC, pretreatment of the mice with naloxone (1 mg/kg s.c. or 1 micrograms/mouse i.t.) failed to block the antinociceptive effects of the cannabinoids, indicating that the cannabinoid-induced antinociception does not occur due to direct interaction with the opiate receptor. Pretreatment of mice with 3.13 micrograms/mouse and 6.25 micrograms/mouse of delta 9-THC shifted the ED50 of morphine to 0.15 and 0.05 micrograms/mouse, respectively (a 4-and a 12-fold shift). The shifts in the dose-response curve of the morphine were parallel. Naloxone administration (1 mg/kg s.c.) completely blocked the antinociceptive effects of the combination of 6.25 micrograms of delta 9-THC with morphine. The AD50 for naloxone blockade of the drug combination was 0.24 (0.06-0.94) mg/kg s.c. and the pA2 was 7.7 (6.7-8.9). The pA2 for naloxone blockade of the dimethylsulfoxide-morphine combination was 6.9 (5.7-8.1).(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Full-text available
The antagonist pharmacology of glutamate neurotoxicity was quantitatively examined in murine cortical cell cultures. Addition of 1-3 mM DL-2-amino-5-phosphonovalerate (APV), or its active isomer D-APV, acutely to the exposure solution selectively blocked the neuroexcitation and neuronal cell selectively blocked the neuroexcitation and neuronal cell loss produced by N-methyl-D-aspartate (NMDA), with relatively little effect on that produced by either kainate or quisqualate. As expected, this selective NMDA receptor blockade only partially reduced the neuroexcitation or acute neuronal swelling produced by the broad-spectrum agonist glutamate; surprisingly, however, this blockade was sufficient to reduce glutamate-induced neuronal cell loss markedly. Lower concentrations of APV or D-APV had much less protective effect, suggesting that the blockade of a large number of NMDA receptors was required to acutely antagonize glutamate neurotoxicity. This requirement may be caused by the amplification of small amounts of acute glutamate-induced injury by subsequent release of endogenous NMDA agonists from injured neurons, as the "late" addition of 10-1000 microM APV or D-APV (after termination of glutamate exposure) also reduced resultant neuronal damage. If APV or D-APV were present both during and after glutamate exposure, a summation dose-protection relationship was obtained, showing substantial protective efficacy at low micromolar antagonist concentrations. Screening of several other excitatory amino acid antagonists confirmed that the ability to antagonize glutamate neurotoxicity might correlate with ability to block NMDA-induced neuroexcitation: The reported NMDA antagonists ketamine and DL-2-amino-7-phosphono-heptanoate, as well as the broad-spectrum antagonist kynurenate, were all found to attenuate glutamate neurotoxicity substantially; whereas gamma-D-glutamylaminomethyl sulfonate and L-glutamate diethyl ester, compounds reported to block predominantly quisqualate or kainate receptors, did not affect glutamate neurotoxicity. The present study suggests that glutamate neurotoxicity may be predominantly mediated by the activation of the NMDA subclass of glutamate receptors--occurring both directly, during exposure to exogenous compound, and indirectly, due to the subsequent release of endogenous NMDA agonists. Given other studies linking NMDA receptors to channels with unusually high calcium permeability, this suggestion is consistent with previous data showing that glutamate neurotoxicity depends heavily on extracellular calcium.
Article
In this study we analyzed the involvement of the cyclic AMP (cAMP)-protein kinase A system in the regulation of interleukin 6 production by cultured cortical astrocytes. Vasoactive intestinal peptide strongly increased, in a dose-dependent manner, interleukin 6 production. This effect was reduced when protein kinase A was blocked by KT-5720; it was not affected by calphostin C, a protein kinase C inhibitor. Forskolin caused a concentration-dependent increase in interleukin 6 release that was also inhibited by KT-5720. Because prostaglandins are believed to play a role in interleukin 6 production, we tried to determine whether the stimulatory effects of vasoactive intestinal peptide and forskolin on cytokine release might be mediated by stimulation of prostaglandin production in cortical astrocytes. Vasoactive intestinal peptide did not increase the production of either prostaglandin E2 or F2 alpha. Conversely, forskolin concentration-dependently stimulated the production of both prostaglandins, an effect that was blocked by indomethacin. Indomethacin did not affect either vasoactive intestinal peptide- or forskolin-stimulated interleukin 6 production. To exclude the possibility that prostaglandins participate in interleukin 6 production induced by forskolin, we tested prostaglandins E2 and F2 alpha. The former was completely ineffective in eliciting the cytokine production, whereas prostaglandin F2 alpha slightly increased interleukin 6 production only at the highest concentrations. 8-Bromo-cAMP and dibutyryl-cAMP stimulated interleukin 6 production to a lesser extent than vasoactive intestinal peptide and forskolin. In conclusion, we provide evidence that vasoactive intestinal peptide increases interleukin 6 production by astrocytes through the stimulation of the cAMP-protein kinase A pathway, an effect that is reproduced by cAMP analogues.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Glutamate kills sensitive neurons through several steps downstream to receptor activation: increased free Ca2+ levels, activation of various enzymes and accumulation of reactive oxygen species (ROS). We have evaluated in a well established model of neuronal cultures the neuroprotective effects of blocking these mechanisms, either singularly or by combining multiple enzyme inhibition and/or ROS scavenging. In vitro cultures of cerebellar granule cells were exposed to a toxic concentration of glutamate (100 μM for 15 min in the absence of Mg2+) combined with several pharmacological treatments. Inhibition of nitric oxide synthase (NOS) and phospholipase A2 (PLA2) were effective in decreasing cell death and the combined treatments showed some degree of additivity. By contrast, inhibition of xanthine oxidase (XO) with allopurinol was uneffective. Antioxidants (in particular vitamin E or vitamin E analogs), protected neurons up to more than 50%. A synergistic effect was demonstrated by the combination of vitamin E and C. On the other hand, antioxidants did not increase the protection granted by enzyme inhibitors, suggesting that they act downstream to NOS and PLA2. In conclusion, NOS and PLA2 activated by Ca2+ influx give rise to reactive oxygen species whose deleterious action can be counteracted either by inhibiting these enzymes or by scavenging the excess of free radicals produced by them. Finally, a moderate protection was obtained by blocking protein synthesis with cycloheximide, suggesting a partial contribution of apoptotic mechanisms to the excitotoxic cell death.
Article
This study examines the ability of (+)-(3S,4S)-7-hydroxy-Δ6-tetrahydrocannabinol-1,1-dimethylheptyl (HU-211), a non-competitive NMDA receptor antagonist to: (1) rescue neurons in culture from injury evoked by sodium nitroprusside, hydrogen peroxide (H2O2) and oxygen glucose deprivation; and (2) scavenge reactive oxygen species in vitro. Qualitative and quantitative assessments of cell survival have indicated that: (1) Neuronal cell injury produced following deprivation of oxygen and glucose was significantly attenuated by 5 μM HU-211. (2) Glial and neuronal cell damage induced by sodium nitroprusside was markedly ameliorated by 10 μM HU-211. (3) HU-211 reduced protein oxidation initiated by gamma irradiation, and scavenged peroxyl radicals. (4) HU-211 carries an oxidation potential of 550 mV. These findings suggest that HU-211 holds a unique position among putative neuroprotectant agents in that it combines NMDA receptor antagonistic activity and free radical scavenging abilities in a single molecule.
Article
Based on encouraging preliminary findings, cannabidiol (CBD), a major nonpsychotropic constituent of Cannabis, was evaluated for symptomatic efficacy and safety in 15 neuroleptic-free patients with Huntington's Disease (HD). The effects of oral CBD (10 mg/kg/day for 6 weeks) and placebo (sesame oil for 6 weeks) were ascertained weekly under a double-blind, randomized cross-over design. A comparison of the effects of CBD and placebo on chorea severity and other therapeutic outcome variables, and on a Cannabis side effect inventory, clinical lab tests and other safety outcome variables, indicated no significant (p greater than 0.05) or clinically important differences. Correspondingly, plasma levels of CBD were assayed by GC/MS, and the weekly levels (mean range of 5.9 to 11.2 ng/ml) did not differ significantly over the 6 weeks of CBD administration. In summary, CBD, at an average daily dose of about 700 mg/day for 6 weeks, was neither symptomatically effective nor toxic, relative to placebo, in neuroleptic-free patients with HD.
Article
A new tetrazolium salt XTT, sodium 3'-[1-[(phenylamino)-carbonyl]-3,4-tetrazolium]-bis(4-methoxy-6- nitro)benzene-sulfonic acid hydrate, was evaluated for use in a colorimetric assay for cell viability and proliferation by normal activated T cells and several cytokine dependent cell lines. Cleavage of XTT by dehydrogenase enzymes of metabolically active cells yields a highly colored formazan product which is water soluble. This feature obviates the need for formazan crystal solubilization prior to absorbance measurements, as required when using other tetrazolium salts such as MTT. Bioreduction of XTT by all the murine cells examined was not particularly efficient, but could be potentiated by addition of electron coupling agents such as phenazine methosulfate (PMS) or menadione (MEN). Optimal concentrations of PMS or MEN were determined for the metabolism of XTT by the T cell lines HT-2 and 11.6, NFS-60 a myeloid leukemia, MC/9 a mast cell line and mitogen activated splenic T cells. When used in combination with PMS, each of these cells generated higher formazan absorbance values with XTT than were observed with MTT. Thus the use of XTT in colorimetric proliferation assays offer significant advantages over MTT, resulting from reduced assay time and sample handling, while offering equivalent sensitivity.
Article
The widely used antioxidant butylated hydroxytoluene (BHT, 2,6-di-tert-butyl-4-methylphenol) produces acute pulmonary toxicity in mice, and also enhances the multiplicity of lung tumors in mice when chronically administered following a single dose of a carcinogen such as urethane. Evidence strongly indicates that the pulmonary effects of BHT are caused by one or more of its reactive metabolites, particularly the hydroperoxide or quinone methide products. The former, BHT-OOH (2,6-di-tert-butyl-4-hydroperoxy-4-methylcyclohexa-2,5-dienone+ ++), is later converted to free radicals by cytochrome P-450, and evidence implicating this pathway in BHT-OOH-induced cytotoxicity has been obtained using isolated rat hepatocytes. Pulmonary microsomes from mice effectively hydroxylate BHT to BHT-BuOH [6-tert-butyl-2-(hydroxy-tert-butyl)-4-methylphenol]; this metabolite was several-fold more effective than BHT as a lung tumor promoter, substantially more pneumotoxic than BHT in vivo, and more toxic to isolated rat hepatocytes and mouse bronchiolar Clara cells in vitro. These effects may be a result of oxidation of BHT-BuOH to the corresponding quinone methide, which is a highly electrophilic. The tumor promoting effects of BHT in mouse lung may be a result of selective cytotoxicity or altered signal transduction caused by radical-generating hydroperoxide and/or electrophilic quinone methide metabolites.
Article
The determination and characterization of a cannabinoid receptor from brain are reported. A biologically active bicyclic cannabinoid analgetic CP-55,940 was tritium-labeled to high specific activity. Conditions for binding to rat brain P2 membranes and synaptosomes were established. The pH optimum was between 7 and 8, and specific binding could be eliminated by heating the membranes to 60 degrees. Binding to the P2 membranes was linear within the range of 10 to 50 micrograms of protein/ml. Specific binding (defined as total binding displaced by 1 microM delta 9-tetrahydrocannabinol (delta 9-THC) or 100 nM desacetyllevonantradol) was saturable. The Kd determined from Scatchard analysis was 133 pM, and the Bmax for rat cortical P2 membranes was 1.85 pmol/mg of protein. The Hill coefficient for [3H]CP-55,940 approximated 1, indicating that, under the conditions of assay, a single class of binding sites was determined that did not exhibit cooperativity. The binding was rapid (kon approximately 2.6 x 10(-4) pM-1 min-1) and reversible (Koff approximately 0.016 min-1) and (koff' greater than 0.06 min-1). The two Kd values estimated from the kinetic constants approximately 55 pM and exceeded 200 pM, respectively. The binding of the agonist ligand [3H]CP-55,940 was decreased by the nonhydrolyzable GTP analog guanylylimidodiphosphate. The guanine nucleotide induced a more rapid dissociation of the ligand from the binding site, consistent with an allosteric regulation of the putative receptor by a G protein. The binding was also sensitive to MgCl2 and CaCl2. Binding of [3H]CP-55,940 was displaced by cannabinoid drugs in the following order of potency: CP-55,940 greater than or equal to desacetyllevonantradol greater than 11-OH-delta 9-THC = delta 9-THC greater than cannabinol. Cannabidiol and cannabigerol displaced [3H]CP-55,940 by less than 50% at 1 microM concentrations. The (-)-isomer of CP-55,940 displaced with 50-fold greater potency than the (+)-isomer. This pharmacology is comparable to both the inhibition of adenylate cyclase in vitro and the analgetic activity of these compounds in vivo. The criteria for a high affinity, stereoselective, pharmacologically distinct cannabinoid receptor in brain tissue have been fulfilled.
Article
Male and female C3H mice were fed a diet containing 0.5% or 0.05% of the antioxidant butylated hydroxytoluene (BHT). After 10 months, male but not female animals had a significantly increased incidence of liver tumors compared to animals kept on a BHT-free control diet. In a second experiment, male BALB/c mice were treated subcutaneously with the carcinogens dimethylhydrazine (DMH) or intrarectally with methylnitrosourea (MNU). A diet containing 0.5% BHT significantly increased the incidence of colon tumors in DMH treated animals but had no effect in mice given MNU. It is concluded that the effect of BHT on tumor development depends on strain and target organ examined and possibly also on the chemical carcinogen used.