You are on page 1of 11

Neuropharmacology 110 (2016) 519e529

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Stimulation of brain glucose uptake by cannabinoid CB2 receptors and


its therapeutic potential in Alzheimer's disease
€ falvi a, b, Cristina Lemos a, 2, Ana M. Martín-Moreno c, d, 1, Ba
Attila Ko rbara S. Pinheiro a, 2,
^
Luis García-García e, Miguel A. Pozo e, f, Angela rio-Fernandes a, Rui O. Beleza a,
Vale
a, g
Paula Agostinho , Ricardo J. Rodrigues a, b  c, h, Rodrigo A. Cunha a, g,
, Susana J. Pasquare
c, d, *
María L. de Ceballos
a
CNC e Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
b
Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
c
Neurodegeneration Group, Department of Cellular, Molecular and Developmental Neurobiology, Instituto Cajal, CSIC, Doctor Arce, 37, 28002 Madrid, Spain
d
CIBERNED; Centre for Biomedical Research on Neurodegenerative Diseases, Spain
e
CAI de Cartografía Cerebral, Instituto Pluridisciplinar, UCM, Madrid, Spain
f
PET Technology Institute, Madrid, Spain
g
FMUC, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
h
Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET-Bahía Blanca and Universidad Nacional del Sur, Edificio E1, Camino La
Carrindanga km 7, 8000 Bahía Blanca, Argentina

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

Article history: Cannabinoid CB2 receptors (CB2Rs) are emerging as important therapeutic targets in brain disorders that
Received 20 June 2015 typically involve neurometabolic alterations. We here addressed the possible role of CB2Rs in the
Received in revised form regulation of glucose uptake in the mouse brain. To that aim, we have undertaken 1) measurement of 3H-
18 February 2016 deoxyglucose uptake in cultured cortical astrocytes and neurons and in acute hippocampal slices; 2) real-
Accepted 7 March 2016
time visualization of fluorescently labeled deoxyglucose uptake in superfused hippocampal slices; and 3)
Available online 11 March 2016
in vivo PET imaging of cerebral 18F-fluorodeoxyglucose uptake. We now show that both selective
(JWH133 and GP1a) as well as non-selective (WIN55212-2) CB2R agonists, but not the CB1R-selective
Chemical compounds studied in this article: agonist, ACEA, stimulate glucose uptake, in a manner that is sensitive to the CB2R-selective antagonist,
AM630 (PubChem CID
AM630. Glucose uptake is stimulated in astrocytes and neurons in culture, in acute hippocampal slices, in
4302963)
Anandamide (PubChem CID
different brain areas of young adult male C57Bl/6j and CD-1 mice, as well as in middle-aged C57Bl/6j
5281969) mice. Among the endocannabinoid metabolizing enzymes, the selective inhibition of COX-2, rather than
Arachidonyl-2-chloroethylamide (PubChem that of FAAH, MAGL or a,bDH6/12, also stimulates the uptake of glucose in hippocampal slices of middle-
CID aged mice, an effect that was again prevented by AM630. However, we found the levels of the endo-
5311006) cannabinoid, anandamide reduced in the hippocampus of TgAPP-2576 mice (a model of b-amyloidosis),
2-Arachidonoylglycerol (PubChem CID and likely as a consequence, COX-2 inhibition failed to stimulate glucose uptake in these mice. Together,
5282280) these results reveal a novel general glucoregulatory role for CB2Rs in the brain, raising therapeutic in-
2-Deoxy-D-glucose (PubChem CID
terest in CB2R agonists as nootropic agents.
108223)
© 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND
DuP697 (PubChem CID
3177) license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
GP1a (PubChem CID
10252734)
JWH133 (PubChem CID

Abbreviations: 2-AG, 2-arachidonoylglycerol; 3Rs, replacement, refinement and reduction of animals in research; ABDH6/12, a/b-hydrolase domain 6/12; ACEA, arach-
idonyl-20 -chloroethylamide; AEA, anandamide (N-arachidonoylethanolamine); CB1Rs and CB2Rs, cannabinoid CB1 and CB2 receptors; COX-2, cycloxygenase-2; DMEM,
Dulbecco's modified eagle medium; 3HDG, 3H-2-deoxyglucose; DMSO, dimethyl-sulfoxide; FAAH, fatty acid amide hydrolase; 18FDG, 18F-fluoro-deoxyglucose; HEPES, N-(2-
hydroxyethyl)piperazine-N0 -(2-ethanesulfonic acid); MAGL, monoacylglycerol lipase; 2-NBDG, 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose; TgAPP,
transgenic amyloid precursor protein; WT, wild-type.
* Corresponding author. Neurodegeneration Group, Dept. of Cellular, Molecular and Developmental Neurobiology Cajal Institute, CSIC, Doctor Arce, 37, 28002 Madrid,
Spain.
E-mail address: mceballos@cajal.csic.es (M.L. de Ceballos).
1
Present address: MD Anderson Cancer Center, Arturo Soria 270, 28033 Madrid, Spain.
2
Present address: Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Innsbruck Medical University, Austria.

http://dx.doi.org/10.1016/j.neuropharm.2016.03.015
0028-3908/© 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
520 €falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko

6918505)
JZL184 (PubChem CID
25021165)
LY2183240 (PubChem CID
11507802)
Ouabain (PubChem CID
439501)
WIN55212-2 (PubChem CID
5311501)
WWL70 (PubChem CID:17759121)

Keywords:
Anandamide (AEA)
b-amyloid
Cerebral glucose uptake
Cannabinoid CB2 receptor
Cyclooxygenase-2 (COX-2)
Positron emission tomography (PET)

1. Introduction and Kim, 2015). Furthermore, these hippocampal (and medial en-
torhinal) CB2Rs appear to control the synaptic release of GABA
Brain disorders, including Alzheimer's disease (AD), often (Morgan et al., 2009; Ando  et al., 2012), and the deletion of CB2Rs is
involve specific early alterations in the metabolism of glucose in the associated with impaired synaptic plasticity at excitatory synapses,
brain (Mosconi, 2005; Teune et al., 2010). The idea of alleviating too, in the mouse hippocampus (Li and Kim, 2016a). Not surpris-
symptoms of dementia by boosting cerebral energy metabolism ingly, the CB2R KO mice exhibit memory impairments as well (Li
has been toyed with for decades (Branconnier, 1983), yet safe and Kim, 2016b).
pharmacological agents with well characterized mechanism of Besides these indefinite physiological roles, the therapeutic
action are still lacking. In this sense, we have investigated here the potential of central CB2Rs have also been recognized. There is an
local cerebral glucoregulatory potential of the endocannabinoid increasing clinical interest in endorsing novel selective CB2R ago-
system in rodents. nists to treat different illnesses (Atwood and Mackie, 2010;
The endocannabinoid system mainly consists of two established Pertwee, 2012; Han et al., 2013), including AD. Indeed, prolonged
(CB1 and CB2) receptors that are activated by endocannabinoid oral treatment with a CB2R selective agonist conferred anti-
signalling molecules, such as N-arachidonoylethanolamine (anan- inflammatory effects and reduced microglial activation, which
damide, AEA) and 2-arachidonoylglycerol (2-AG) (Di Marzo and De have been demonstrated to mitigate the cognitive deficits of TgAPP
Petrocellis, 2012; Pertwee, 2012). At the cellular level, endocanna- mice (Martín-Moreno et al., 2012). Similarly, subchronic treatment
binoid signalling via CB1 receptors (CB1Rs) regulates peripheral with another CB2R agonist promoted b-amyloid (Ab) clearance, and
energy metabolism and glucose uptake (Matias et al., 2008). In the restored synaptic plasticity, cognition, and memory in Ab injected
brain, acute activation of the psychoactive CB1R inhibits mito- rats (Wu et al., 2013). These results were extended in other reports
chondrial respiration and glucose metabolism in neurons and as- using TgAPP/PS1 mice (Aso and Ferrer, 2014; Cheng et al., 2014).
trocytes, without robust effect on the uptake of glucose (Be nard Additionally, the severity of Ab pathology correlates positively with
et al., 2012; Duarte et al., 2012; Lemos et al., 2012). Similarly, the the density of the CB2R rather than the CB1R (Esposito et al., 2007;
peripheral glucoregulatory role of the non-psychoactive CB2 re- Solas et al., 2013; Savonenko et al., 2015). Therefore, to map the
ceptors (CB2Rs) has also been recognized (Agudo et al., 2010; putative glucoregulatory potential of CB2Rs in healthy mice and in a
Romero-Zerbo et al., 2012), although this remains to be docu- model of b-amyloidosis, we have taken advantage of recently
mented in the brain. optimized in vitro and in vivo protocols (Lemos et al., 2012; Martín-
CB2Rs were first described in peripheral organs and cells Moreno et al., 2012). Here we report that acute CB2R activation
(Galiegue et al., 1995), and subsequently in activated microglial increases glucose uptake in the brain of young and middle-aged
cells (Carlisle et al., 2002) - the resident immune cell of the brain. mice both in vitro and in vivo, which highlights a novel cerebral
However, during the last decade, the presence and function of role for the CB2R with therapeutic potential.
CB2Rs have been described in other cell types in the brain. An early
evidence was provided by Van Sickle et al. (2005) who identified
and functionally characterized CB2Rs in brainstem neurons. Other 2. Materials and methods
studies have reported CB2R involvement in neurogenesis, axon
guidance, and neuronal excitability (Den Boon et al., 2012; Duff 2.1. Chemicals
et al., 2013; Sierra et al., 2014; Zhang et al., 2014). Furthermore,
3
astrocytes (in particular, activated astrocytes) are also endowed H-2-deoxy-D-glucose (3HDG; specific activity: 60 Ci/mmol),
with CB2R (Sheng et al., 2005; Benito et al., 2008; Bari et al., 2011; arachidonylethanolamide [ethanolamide 1e3H] (50 Ci/mmol) and
Rodríguez-Cueto et al., 2014). However, a recent report added that unlabelled AEA were obtained from American Radiolabeled
the vast majority of specific CB2R immunoreactivity in the healthy Chemicals, Inc. (Saint Louis, MO, USA). 2-arachidonoyl-[1,2,3-3H]
mouse neocortex is neuronal (Savonenko et al., 2015). Nonetheless, glycerol (3H-2-AG; 40 Ci/mmol) was bought from PerkinElmer,
conventional CB2R immunstaining techniques may have possible Madrid, Spain). 2-NBDG was purchased from Invitrogen (Carslbad,
limitations (Ashton, 2011). Therefore, it is important that the highly California, USA). Non-organic reagents, HEPES and DMSO were
sensitive and specific RNAscope technique also proved the presence purchased from Sigma-Aldrich Portugal (Sintra, Portugal) or Merck
of CB2R mRNA in excitatory and inhibitory neurons (but hardly in Biosciences (Darmstadt, Germany). ACEA, AM630, DuP697, GP1a,
resting microglia) throughout the hippocampus of adult mice (Li JWH133, JZL184, LY2183240, WIN55212-2 and WWL70 were ob-
tained from Tocris Bioscience (Bristol, U.K.). All non-water soluble
€falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko 521

substances and 2-NBDG were reconstituted in DMSO at 1000 the deoxyglucose (2-NBDG; 30 mM) was applied through the reser-
final concentration, and stored in aliquots at 20  C. voir and 15 min later, JWH133 (1 mM), GP1a (100 nM) or the DMSO
vehicle alone (0.1% v/v) were administered. All measurements were
2.2. Animals obtained in duplicate from each animal (see Supplementary Fig. 1
for additional details).
All studies were carried out in accordance with the EU (2010/63/
EU) and FELASA guidelines, and they were approved by the Spanish 2.4. Cell culture preparation
and Portuguese Ministries of Agriculture, as well as the local
institutional Animal Care Committees (license No. 280279-31-A Because the embryonic hippocampus is too small to provide
and 025781). All studies involving animals are reported in accor- enough cells for our assays, neuronal and astrocyte primary cul-
dance with the ARRIVE guidelines for reporting experiments tures were prepared from the neocortex as before (Lemos et al.,
involving animals (Kilkenny et al., 2010; McGrath et al., 2010). A 2015). The neocortex of E18 male and female C57Bl/6 rat embryos
total of 73 animals were used in the experiments described here. was digested for 15 min with 0.125% trypsin (type II-S, from porcine
For the experiments which are summarized in Figs. 3AB, 4 and 5, pancreas, Sigma-Aldrich Portugal) and 50 mg/mL DNAse (Sigma-
we used 12-month-old (middle-aged) C57Bl/6j mice (i.e. wild type Aldrich) in Hank's balanced salt solution without calcium and
[WT] littermates as controls), and 12-month-old C57Bl/6j trans- magnesium (in mM: NaCl 137, KCl 5.36, KH2PO4 0.44, NaHCO3 4.16,
genic 2576 mice, containing the human APP695 transgene with Na2HPO4 0.34, D-glucose 5, pH 7.2). After dissociation, astrocytes
mutations at KM670/671NL (hereafter, TgAPP mice). TgAPP mice were grown in plastic Petri dishes (p100) for 14 days in DMEM with
have b-amyloid-burden but no evident cell loss (Hsiao et al., 1996). 10% fetal bovine serum, 50 U/mL penicillin and 50 mg/mL strepto-
At 12 months, TgAPP mice did not differ in weight from the WT mycin (Sigma-Aldrich), at 37  C in an atmosphere of 95%/5% air/
C57Bl/6j mice (WT: 39.6 ± 2.4 g; TgAPP: 38.6 ± 2.3 g), yet they CO2, replacing half of the medium at DIV 7. The cells were then
displayed compromised new object recognition compared to WT trypsinized and plated onto poly-D-lysine (100 mg/mL, Sigma-
mice (data not shown). Data shown in Fig. 3C were obtained from Aldrich) and laminin (10 mg/mL, Sigma-Aldrich) coated 24-well
10-week-old male WT CD-1 mice and their CB1R global KO litter- culture plates at a cell density of 150,000/cm2. Uptake assays
mates (Ledent et al., 1999). All these transgenic and WT mice were were performed 24 h later. For neuronal cultures, cells were plated
genotyped from tail tip biopsies. directly onto poly-D-lysine and laminin coated 24-well culture
Data shown in Fig. 1 were obtained from young adult (8-10 plates, at the same cell density, in DMEM plus 10% fetal bovine
week-old) male C57bl/6 mice (Charles-River, Barcelona, Spain), serum, 50 U/mL penicillin and 50 mg/mL streptomycin. Two hours
while to obtain embryos for cell cultures, we mated young adult after seeding, the medium was replaced by Neurobasal medium
C57Bl/6 mice (Charles-River). with 2% B27 supplement (GIBCO, Life technologies), 50 U/mL
The animals were group-housed under controlled temperature penicillin, 50 mg/ml streptomycin and 2 mM glutamine (Sigma-
(23 ± 2  C) and subjected to a fixed 12 h light/dark cycle, with free Aldrich), and cells were grown at 37  C in an atmosphere of 95%/5%
access to food and water. We applied the principles of the ARRIVE air/CO2 until they were used 14 days later. The medium was
guideline in designing and performing the in vivo and in vitro partially (40%) replaced every 4 days. On average, 15% of cells were
pharmacological experiments (see below), as well as for data found positive for glial fibrillar acidic protein immunostaining in
management and interpretation. All efforts were made to reduce randomly chosen wells (Lemos et al., 2015).
the number of animals used and to minimize their stress and
discomfort. The animals used to perform the in vitro studies were 2.5. In vitro 3H-deoxyglucose uptake in neocortical neuron and
deeply anesthetized with halothane before sacrifice (no reaction to astrocyte cultures
handling or tail pinching, while still breathing).
The assays were carried out according to Lemos et al. (2015).
2.3. In vitro real-time fluorescent measurement of deoxyglucose For these assays, we used 7 different astrocyte cultures and 5
uptake in hippocampal slices neuron cultures from 7 and 5 independent preparations (pregnant
dams). After 15 days in culture, the culture medium was replaced
Experiments were performed as before (Lemos et al., 2015). Five with 250 mL Krebs-HEPES assay solution (pH 7.4; 37  C) (see
young C57bl/6 mice were sacrificed, and their brain was quickly above), containing either the vehicle alone (DMSO 0.1%, in half of
removed and placed in ice-cold Krebs-HEPES solution (in mM: NaCl the plate), or the CB2R-selective antagonist, AM630 (1 mM, in the
113, KCl 3, KH2PO4 1.2, MgSO4 1.2, CaCl2 2.5, NaHCO3 25, glucose 5.5, other 12 wells). The cultures were incubated for 60 min, at 37  C,
HEPES 1.5, pH 7.2). Coronal vibratome slices (300 mm) were left to and the CB2R agonists JWH133 (100 nM) (4 wells/plate) or GP1a
recover for 1 h at room temperature in Krebs-HEPES solution (100 nM) (4 wells/plate), or their vehicle (DMSO 4 wells/plate),
gassed with 5% CO2 and 95% O2, and then gently mounted on was gently added to the wells in a volume of 250 mL, together with
coverslips with the hippocampus in the center, and placed in a RC- AM630 (i.e. 3  4 wells/plate) or its vehicle (i.e. 3  4 wells/plate),
20 superfusion chamber on a PH3 platform (Warner Instruments, and the radioactive glucose analogue 3H-2-deoxyglucose (3HDG;
Harvard, UK). The slices were superfused with gassed Krebs-HEPES 16.7 nM, final concentration). After a further 30 min incubation at
solution at a rate of 0.5 mL min-1 in a closed circuit, and they were 37  C, the plates were transferred onto ice and the 3H content was
then photographed with a CoolSNAP digital camera (Roper Scien- measured in an aliquot from each well (to assess the small dif-
tific, Trenton, NJ, USA) every 30 s over the following 30 min (using a ferences in the individual 3H concentration in each well that
5 PlanNeofluar-objective [NA 0.25] on an inverted Axiovert 200 M pipetting and mixing solutions may introduce). The cells were
fluorescence microscope [Carl Zeiss, Germany], coupled to a then washed gently 3 times with 1 mL ice-cold Krebs-HEPES,
Lambda DG-4 integrated 175 Watt light source and wavelength 300 mL NaOH (0.5 M) was added to each well, and the plates were
switching excitation system [Sutter Instrument Company, Novato, left on a shaker for an hour to recover the proteins and 3HDG
CA, USA] to allow real-time video imaging). The data was band-pass taken up by the cells. Tritium was measured in a Tricarb b-counter
filtered for excitation (BP470/40) and emission (BP525/50). (PerkinElmer, USA), and these values were adjusted for protein
After recording 4 images for auto-fluorescence (to establish the quantity and the 3H concentrations in each individual wells, and
baseline), 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2- multiplied by 3.3  105 (the ratio between 3HDG and D-glucose in
522 €falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko

Fig. 1. Time-course and sub-regional variation of the effect of CB2R agonists on the uptake of the fluorescent glucose analogue, 2-NBDG in hippocampal slices of young C57Bl/6j
male mice. For technical details and additional information, see Supplementary Methods, Supplementary Fig. 1 and Supplementary Video. (A,A0 ) Representative images illustrating
the distribution of the fluorescence signal, using the false color scale of blue (low signal), green (moderate signal) and yellow (strong signal). As indicated in the time-course graph
(B), image (A) was taken just before the treatment (minute 15 of baseline), and image (A0 ) was photographed close to the end of the exposure to GP1a (100 nM: see also Sup-
plementary Video). (B) JWH133 (1 mM) and GP1a (100 nM) both increased the uptake in astrocyte-rich zones of the hippocampus. The displayed curves represent the mean þ S.E.M.
of individual net effects (in duplicate) in each animal, which was obtained after the subtraction of the respective DMSO controls (in duplicates per animal) from the raw treatment
data. Therefore, the DMSO control is represented with the dashed line crossing the zero value. (C) The individual points represent the average of 2 different slices from the same
mouse in duplicate (n ¼ 5): *P < 0.05 and **P < 0.01.

specificity of the transport process (Lemos et al., 2015). Addition-


ally, the plasma membrane Naþ/Kþ-ATPase inhibitor, ouabain (10
mM), also inhibited neuronal glucose uptake in this assay by
42.8 ± 2.1% (n ¼ 3, P < 0.01; figure not shown).

2.6. In vitro 3H-deoxyglucose uptake in hippocampal slices

To better characterize the glucoregulator role of CB2R signalling


pharmacologically, we moved to an in vitro glucose uptake protocol
previously optimized in acute hippocampal slices which allows the
effect of various treatments to be compared simultaneously in
different strains in a pairwise arrangement (Lemos et al., 2012,
2015). Animals were anesthetized with halothane, sacrificed
(around 14:00 o'clock each experimental day to reduce potential
circadian hormonal effects), and their brain was immediately
placed in ice-cold Krebs-HEPES assay. The hippocampus was cut
into 450 mm-thick transverse slices with the help of a McIlwain
tissue chopper, and the slices were gently separated in ice-cold
carboxygenated (95% O2 and 5% CO2) assay solution, and main-
tained at 37  C in a multichamber slice incubator with 50 ml of
carboxygenated assay solution until the end of the experiment.
Four slices were used from each animal and in each experimental
condition. The CB2R-selective antagonist, AM630 or its vehicle
(DMSO 0.1%) were bath-applied from min 55 of the preincubation
period. WIN55212-2, JWH133, ACEA or DuP697, LY2183240, as well
as JZL184 and WWL70 combined or their vehicle (DMSO 0.1%) were
added from min 60 of the preincubation period, and the radioactive
glucose analogue, 3H-2-deoxy-D-glucose (3HDG; final cc. 2 nM) was
applied to the bath 5 min later. After 30 min, the slices were washed
Fig. 2. CB2R activation stimulates hippocampal glucose uptake in astrocytes and twice in ice-cold assay solution for 5 min and collected in 1 mL
neurons in vitro. The CB2R-selective agonists JWH133 (30 nM) and GP1a (100 nM) NaOH (0.5 M) to dissolve the slices. An aliquot of 800 mL was then
stimulated glucose uptake in (A) primary cortical astrocyte and (B) neuronal cultures assayed for 3H in a Tricarb b-counter, while the remaining solution
from C57Bl/6 mice, as assessed using the 3HDG uptake assay (30 min). The CB2R se- was used to quantify the protein in a bicinchoninic acid assay
lective antagonist, AM630 (1 mM) prevented the action of the CB2R agonists, while it
had no effect on the uptake per se. D-glucose uptake is expressed as mean þ SEM in
(Merck Biosciences, Germany). Tritium uptake was multiplied by a
nmol/mg protein (n ¼ 7 astrocyte and n ¼ 5 neuron cultures, performed in quadru- factor of 2.75  106 to estimate the total glucose uptake (corre-
plicate, at a density of 15  104 cells/cm in 24-well plates). *P < 0.05 and **P < 0.01 vs. sponding to the concentration difference between D-glucose and
DMSO control; n.s., not significant. 3
HDG in the uptake medium; Lemos et al., 2012).

2.7. Blood glucose measurements


the medium).
In this experimental model, cytochalasin B (10 mM) largely in- Blood glucose from non-fasted WT and TgAPP mice was
hibits glucose uptake in both cell cultures, thus confirming the assessed before and one hour following the acute administration of
JWH133 0.2 mg/kg i.p. Glucose was measured in one drop of blood
€falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko 523

Fig. 3. CB2R activation stimulates glucose uptake in hippocampal slices of both middle-aged and young mice ex vivo. A) The CB2R-selective agonist, JWH133 (1 mM), the mixed CB1R/
CB2R agonist, WIN55212-2 (1 mM), as well as the COX-2 inhibitor, DuP697 (500 nM) all increased glucose uptake in 12-month-old wild-type (WT) C57Bl/6j mice. All these effects
were prevented by a 5 min preincubation with the CB2R-selective antagonist, AM630 (1 mM). Inhibition of other endocannabinoid metabolizing enzymes, namely, FAAH, MAGL or
a,bDH6, with LY2183240, JZL184 and WWL70 (all at 1 mM) respectively, did not affect glucose uptake. B) By contrast, only JWH133 and WIN55212-2 but not DuP697 stimulated
glucose uptake in the TgAPP 2567 mice. Data represent the mean þ SEM of individual measurements from 6 to 12 animals, with the dashed line indicating the DMSO control values:
*P < 0.05 vs. DMSO-treated slices; n.s., not significant. C) JWH133 (1 mM), stimulated the uptake of glucose in acute hippocampal slices of 10 week-old CB1R KO mice and their WT
littermates. Each of the connected pairs of squares and circles represent one animal. *P < 0.05 vs. DMSO-treated slices (n ¼ 6).

standard, the samples were diluted to 70% water and centrifuged.


The pellets were kept for total protein determination. Solid-phase
extraction (Strata-X 33 mm columns, Phenomenex) was per-
formed before the LC/MS analysis on a Thermo Surveyor liquid
chromatographic system coupled to an electrospray ionization
triple quadrupole TSQ Quantum mass spectrometer operated in
positive ionization mode. Chromatographic analysis was performed
on an ACE 5 mm C8 (150  2.1 mm, Hichrom) column and the
analytes were eluted using an isocratic mobile phase of water/
methanol/formic acid (15/85/0.5 v/v/v) at a flow rate of 200 mL/min.
The column was maintained at 30  C and the sample tray at 4  C
during the analysis. The extraction efficiency was >95% and the
limit of quantification was 0.01 pmol for AEA and <25 pmol for 2-
AG. The sample concentrations were determined from calibration
curves and the endocannabinoid levels were expressed per mg
protein (Bradford assay).

2.9. Endocannabinoid hydrolysis assay

Experiments were carried out as before (Mulder et al., 2011),


and adapted to mice (Pascual et al., 2014). Cortices were mechan-
ically homogenized in TriseHCl (20 mM, pH 7.2) containing 0.32 M
sucrose and protease inhibitors, and centrifuged to eliminate nuclei
and cellular debris (1000g, 10 min, 4  C). Protein concentrations
were determined by Bradford's colorimetric method. The 2-AG-
degrading activity was assessed by incubating sample fractions
Fig. 4. AEA levels were reduced in the hippocampus of TgAPP mice. Of the two
(50 mg protein) in a solution containing TriseHCl (10 mM, pH 7.2),
principal endocannabinoids, the levels of AEA (B) but not of 2-AG (A) were significantly
lower (by 38%) in the TgAPP mice (dark bars) than in the WT (light bars), as assessed by 1 mM EDTA, fatty acid-free bovine serum albumin (1.25 mg/mL),
combined liquid chromatography-mass spectrometry. Data represent the mean þ SEM and 3H-2-AG in a final volume of 200 ml for 15 min at 37  C. Identical
of individual measurements from 7 animals. 2-AG hydrolysis (C) was also unaltered in conditions were used to assess AEA degrading activity using 3H-
hippocampal membranes of TgAPP mice in vitro (n ¼ 6), however, AEA metabolism (D)
AEA ([1-H]ethanolamine, 25 mM, 40 Ci/mmol specific activity; ARC
by FAAH into ethanolamine was significantly smaller in the transgenic hippocampi
in vitro (n ¼ 6). *P < 0.05, ***P < 0.001; n.s., not significant.
Inc.). Reactions were stopped by the addition of 400 mL chlor-
oform:methanol (2:1, v/v) and vigorous vortexing, after centrifu-
from the tail using Glucocard Memory meter strips (Menarini, gation (2000g, 10 min, 4  C), ethanolamine or glycerol, the AEA and
Spain), in a range of 40e500 mg/mL. 2-AG hydrolysis products respectively, were obtained in the upper
phase, and they were transferred into scintillation vials and
radioactivity measured by liquid scintillation spectroscopy.
2.8. Endocannabinoid level measurement in mouse hippocampus The experimental conditions were selected from pilot experi-
ments establishing the relationship of substrate and protein con-
Endocannabinoids were measured externally (Division of centrations to identify optimal AEA-degrading enzymatic activity
Applied Medicine, School of Medicine and Dentistry, University of (data not shown), allowing 50% of AEA converted into
Aberdeen, UK). Lipid extraction was performed by homogenizing ethanolamine.
the frozen hippocampi on ice in 600 mL of acetonitrile/methanol Blanks were prepared identically except that the membranes
(50/50) using an Ultra-Turrax. Upon addition of 6 pmol of N-(2- were omitted or inactivated by boiling for 10 min or by the addition
hydroxyethyl-1,1,2,2-d4)-5Z,8Z-,11Z,14Z-eicosatetraenamide of chloroform:methanol (1:1, v/v) before use. No differences were
[deuterated (d4)-AEA; Tocris Bioscience, UK] as an internal observed among the different blanks. These values were subtracted
524 €falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko

Fig. 5. Evidence for the stimulation of brain glucose uptake by a single low-dose injection of JWH133 in middle-aged mice in vivo. Basal (A1,B1) and JWH133-stimulated A2,B2) 18F-
fluoro-deoxy-glucose (18FDG) uptake in the frontal and temporal cortices, and the hippocampus. A1) Representative color-coded PET of basal 18FDG uptake in 12 month old wild-
type (WT) and B1) TgAPP 2567 mice. Panels A10,B10 ) illustrate the same images merged with the respective CT images, seen from three directions. The effect of acute JWH133
injection (0.2 mg/kg, i.p.) is shown in panels A2,A20 ,B1,B20 ). Color code for FDG activity can be seen vertically on the left of the figure. C) Mean basal uptake values in the three
regions of interest (n ¼ 9 mice). D) Acute JWH133 injection does not alter the blood glucose levels in the two strains and hence, JWH133 does not appear to influence glucose uptake
in the brain through systemic mechanisms. E-G0 ) Before-after graphs summarizing the basal uptake, and 4 days later, the JWH133-stimulated (JWH) uptake in the frontal E, E0 ) and
temporal F, F0 ) cortices, as well as in the hippocampus G, G0 ), of 4 wild-type (WT, E-G) and 4 TgAPP 2567 (E0 -G0 ) mice. Uptake values were corrected for the dose injected and for
body weight, *P < 0.05.

from each enzymatic activity. scanner, Oncovision, Spain). PET images were reconstructed using
Enzyme activity was expressed as picomole [1,2,3-3H]glycerol the ordered subset expectation maximization algorithm (OSEM, 3
formed  mg protein  15 min and picomole [3H]ethanolamine iterations), applying corrections for dead time, radioactive decay,
formed  mg protein  15 min respectively. random coincidences and scattering. For the CT images a high
resolution filtered back projection (FBP) algorithm was used.
2.10. 18F-fluorodeoxy-D-glucose positron emission tomography Metabolic activity was quantified by matching the CT image of
(18FDG-PET) the skull of each animal to a common magnetic resonance (MR)
mouse brain template, in which regions of interest were previously
These experiments were carried out as described previously delineated. After saving the spatial transformation, it was applied
(Martín-Moreno et al., 2012) with only slight modifications. In the to the corresponding fused PET image, to allow the correct co-
PET studies the images of basal (no treatment) uptake were ac- registration of the PET image to the MR brain template. Results
quired, and 4 days later the animals were injected with JWH133 were normalized to the actual radioactivity dose injected and the
0.2 mg/kg ip, and after 30 min, the radiotracer was injected and body weight of each animal (Kuntner et al., 2009). All these pro-
new PET images were acquired (see below). cesses were carried out with PMOD software version 3.0 (PMOD
PET images were acquired in fasted mice injected (i.p.) with Technologies, Switzerland).
18
FDG (approx. 11.1 MBq or 300 mCi/200 ml saline, PET Technology
Institute, Madrid). After a 30 min period for radiotracer uptake, the 2.11. Data presentation and statistical analysis
animals were anesthetized by isoflurane (2%) inhalation, to assure
immobilization during the scanning process, and they were placed All data are expressed as the means ± or þ SEM of the number of
on the bed of the tomograph. Images of 18FDG uptake were ac- independent observations (n) indicated. Normalized data were
quired over 30 min, immediately followed by a CT (computed to- tested for normality with the Kolmogorov-Smirnov normality tests
mography) acquisition (Albira ARS small animal PET/CT dual and statistical significance was calculated by one sample t-test
€falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko 525

against the hypothetical value of zero (Figs. 1C and 2). Alternatively, TgAPP hippocampus amounted to 105.4 ± 8.9% of the WT control
a paired t-test (Fig. 3C) or repeated measures of ANOVA followed by uptake (i.e. þ5.4 ± 8.9% over DMSO control) when assessed in a
Bonferroni's post-hoc test were used, and a value of P < 0.05 was pairwise arrangement (n ¼ 19 pairs, P > 0.05: Fig. 3 A,B). Glucose
accepted as significant. uptake was significantly enhanced by the CB2R-selective agonist
JWH133 (1 mM) in both the WT (þ26.3 ± 10.0%, n ¼ 12, P < 0.05:
3. Results Fig. 3A) and the TgAPP slices (þ16.7 ± 6.7%, n ¼ 12, P < 0.05: Fig. 3B).
Due to the limited availability of 12-month old TgAPP mice, we
3.1. CB2R activation rapidly enhances glucose uptake in made a risk-free decision when testing JWH133 at 1 mM, i.e. at the
hippocampal slices maximal concentration where good CB2R-selectivity is still ach-
ieved (Huffman et al., 1999).
First we assessed whether CB2R activation affects glucose up- Importantly, glucose uptake was also stimulated by the non-
take in superfused hippocampal slices, and the rate of fluorescence- selective (“mixed”) CB1R/CB2R agonist, WIN55212-2 (1 mM), in
labeled deoxyglucose (2-NBDG) accumulation was tested at 30 s both WT (þ22.4 ± 9.2%, n ¼ 12, P < 0.05: Fig. 3A) and TgAPP slices
resolution in such slices. The basal accumulation of 2-NBDG was (þ17.5 ± 7.1%, n ¼ 12, P < 0.05: Fig. 3B). While the selective CB2R
most evident in astrocyte-rich areas of transverse hippocampal antagonist, AM630 (1 mM) had no effect on glucose uptake per se in
slices, i.e. in the in the stratum lacunosum, followed by the strata either strain (n ¼ 12), it did prevent JWH133 and WIN55212-2 from
radiatum and moleculare, and the smallest signal was found in the stimulating glucose uptake in both WT and TgAPP slices (n ¼ 6:
strata pyramidale and granulare (Fig. 1AeA0 ), consistent with pre- Fig. 3 A,B).
vious findings (Jakoby et al., 2014). Importantly, both of the two Cannabinoid agonists exert their actions predominantly via
selective CB2R agonists, GP1a (100 nM, P < 0.01) and JWH133 (1 mM, CB1R activation in the mammalian brain (Katona and Freund, 2012).
P < 0.05), rapidly increased the velocity of 2-NBDG accumulation in To check if CB1Rs are also involved in the stimulation of glucose
these hippocampal slices (Fig. 1 B,C, and Supplementary Video). uptake, first we tested the CB1R-selective agonist ACEA at a con-
Supplementary data related to this article can be found online at centration half way between its Kd values at the CB1R and the CB2R,
http://dx.doi.org/10.1016/j.neuropharm.2016.03.015. i.e. at 1 mM (Hillard et al., 1999). ACEA did not affect glucose uptake
in the middle-aged C57Bl/6 mice (þ5.3 ± 7.9%, n ¼ 7, P > 0.05:
3.2. CB2R activation stimulates glucose transport in cultured Fig. 3A). Subsequently, we asked if CB1Rs were involved in the effect
neurons and astrocytes of JWH133, taken that this ligand has some affinity toward the CB1R
at 1 mM (Huffman et al., 1999). Thus, we tested JWH133 (1 mM) in
Since the use of 2-NBDG underrepresents neuronal glucose acute hippocampal slices of young adult CB1R KO mice and their
transport (Jakoby et al., 2014), we measured the effect of acute WT littermates on the CD-1 strain. JWH133 (1 mM) significantly
in vitro CB2R activation on glucose uptake by primary mouse stimulated glucose uptake þ13.4 ± 3.7% (n ¼ 6, P < 0.05) in the WT
cortical astrocytes and neurons using a 3HDG transport assay. In the mice and þ26.7 ± 9.0% (n ¼ 6, P < 0.05) in the CB1R KO, thus
presence of JWH133 (30 nM) over 30 min, glucose uptake virtually, JWH133 was more twice as more effective in the absence
augmented by 35.4 ± 7.5% in astrocytes (n ¼ 7, P < 0.01 vs. DMSO of the CB1R. These data together with the results with ACEA also
control) and by 13.5 ± 4.0% in neurons (n ¼ 5, P < 0.05: 2 A,B). confirm the absence of acute modulation of hippocampal glucose
Notably, JWH133 exhibits an EC50 value close to 1 mM at the CB1R uptake by CB1Rs.
(Huffman et al., 1999). To avoid the possible non-specific co-acti- Next, we assessed whether endogenous activation of CB2R by
vation of CB1R in these cell cultures, we opted for testing JWH133 the major endocannabinoids, AEA and 2-AG also bolstered glucose
here at 30 nM, i.e. 9-times above its EC50 at the CB2R but more than uptake. To that end, we inhibited either fatty acid amide hydrolase
an order of magnitude below its EC50 at the CB1R (Huffman et al., (FAAH) or cyclooxygenase-2 (COX-2), two enzymes known to be
1999). involved in AEA degradation (Egertova  et al., 2003; Fowler et al.,
Similarly, GP1a at a concentration ~2700-times above its EC50 at 2013). The treatment of slices with DuP697 (500 nM), a selective
the CB2R but ~4-fold below its EC50 at the CB1R, i.e. at 100 nM COX-2 inhibitor (IC50 at COX-2, 10 nM; IC50 at COX-1, 800 nM;
(Murineddu et al., 2006) also increased glucose uptake in astrocytes Gierse et al., 1995), augmented glucose uptake by þ16.6 ± 7.7% in
by 22.4 ± 10.7% (n ¼ 7, P < 0.05) and by 13.7 ± 2.4% in neurons the WT mice (n ¼ 6, P < 0.05: Fig. 3A) - an effect that was prevented
(n ¼ 5, P < 0.01: Fig. 2 A,B). Therefore, with the chosen ligand by CB2R blockade (n ¼ 6: Fig. 3A). However, unlike in the WT,
concentrations we achieved good selectivity toward the CB2R while DuP697 failed to modify glucose uptake in the TgAPP mice (n ¼ 6,
still observing maximal effect amplitudes. The CB2R antagonist Fig. 3B). In contrast, LY2183240 (100 nM), a potent dual inhibitor of
AM630 (1 mM) did not alter glucose uptake per se in cultures of FAAH and AEA reuptake (Nicolussi et al., 2014), did not affect
astrocytes (n ¼ 7) or neurons (n ¼ 5). However, in the presence of glucose uptake in WT mice, and therefore, it was not tested in
AM630, neither GP1a nor JWH133 significantly altered glucose TgAPP mice slices. The major enzymes that degrade 2-AG in the
uptake in either of these two cell types (P > 0.05 vs. AM630 control; mammalian brain are monoacylglycerol lipase (MAGL) and a/b-
astrocytes n ¼ 5, neurons n ¼ 4: Fig. 2 A,B). In summary, CB2R hydrolase domain 6/12 (ABDH6/12) (Savinainen et al., 2012;
activation was more effective at increasing glucose uptake in as- Murataeva et al., 2014). The inhibition of 2-AG hydrolysis by the
trocytes compared with neurons. As stated previously, our neuron concurrent application of JZL184 (1 mM), a MAGL inhibitor (Long
cultures contained 15% of astrocytes, which likely contributed to et al., 2009), and WWL70 (1 mM), an a,bDH6/12 inhibitor (Li et al.,
the effect of the CB2R agonists. However, this low astrocytic 2007), also failed to modify glucose uptake in WT mice (Fig. 3 A).
contamination cannot solely account for the effect amplitudes
observed neuronal cultures. 3.4. Lower AEA levels in the hippocampus of TgAPP mice

3.3. b-amyloidosis hinders endogenous CB2R activation The inability of COX-2 inhibition to stimulate glucose uptake in a
CB2R-dependent fashion in the TgAPP hippocampus suggests a loss
A similar pharmacological analysis was then performed in hip- of endocannabinoid function, which prompted us to measure
pocampal slices of 12-month-old TgAPP mice and their age- endocannabinoid levels in these two strains. The hippocampal
matched controls. The normalized control uptake values of the levels of AEA and 2-AG in the WT mice (Fig. 4) were similar to those
526 €falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko

reported in the rat brain (Stella et al., 1997), and while no difference et al., 2012; Zhang et al., 2014). Interestingly, CB2R expression and
in the 2-AG levels were detected between WT and TgAPP mice density in neurons apparently surpasses that in microglia or as-
(n ¼ 7, P > 0.05), AEA levels in the latter were 37.8 ± 11.4% lower trocytes in the mouse neocortex (Savonenko et al., 2015) and hip-
than in WT (n ¼ 7, P < 0.05) (Fig. 4A,B). Accordingly, there was a pocampus (Li and Kim, 2015).
2.4 ± 0.3 fold increase in the 2-AG/AEA ratio in TgAPP mice (n ¼ 7, The therapeutic activation of CB2R is safe and it does not trigger
P < 0.001). The degradation of 2-AG was also not statistically psychoactivity (Atwood and Mackie, 2010; Pertwee, 2012).
different between the WT and TgAPP (n ¼ 6, P > 0.05) (Fig. 4D). Furthermore, CB2R density is known to increase in Alzheimer's
Interestingly, the degradation of AEA into ethanolamine by FAAH disease and Down's syndrome, as well as in b-amyloidosis in vitro
was significantly reduced in TgAPP by 33.5 ± 8.9% (n ¼ 6, P < 0.001) (Esposito et al., 2007; Ruiz-Valdepen ~ as et al., 2010; Solas et al.,
(Fig. 4D). 2013). This probably reflects an adaptive self-defense process
since CB2R activation confers neuroprotection in several experi-
18
3.5. Effect of CB2R activation on FDG uptake e a PET analysis mental models of Alzheimer's disease (Ramírez et al., 2005;
in vivo Esposito et al., 2007; Ruiz-Valdepen ~ as et al., 2010; Martín-
Moreno et al., 2012).
Finally, we tested the feasibility of targeting CB2R to stimulate
brain glucose uptake in middle-aged mice in vivo. We first evalu- 4.2. AEA and b-amyloidosis
ated the basal uptake of 18FDG in the brain regions typically bearing
the hallmarks of the diminished 18FDG signal in the Alzheimer's The first step in common pathomechanisms toward AD pa-
disease-affected human brain, i.e. the frontal and temporal cortices, thology is excessive b-amyloid accumulation (Sperling et al., 2013;
and the hippocampus (Mosconi, 2005). Like the ex vivo data, we Zahs and Ashe, 2013). Remarkably, the antagonistic relationship
observed no difference in the 18FDG signal in the aforementioned between AEA and b-amyloid appears to be bi-directional: b-amy-
areas of WT and TgAPP mice (n ¼ 9: Fig. 5 A,A0 ,B,B0 ,C). Four days loid toxicity is prevented by AEA in human cell lines in vitro (Milton,
later, the same animals were injected with a small dose of JWH133 2002) and conversely, b-amyloid treatment decreases AEA levels in
(0.2 mg/kg, i.p.) and they were re-evaluated for 18FDG uptake. C6 rat astroglioma cells (Esposito et al., 2007). Accordingly, AEA
While this CB2R agonist did not alter blood glucose levels (Fig. 5 D), levels decrease in the AD brain and they are inversely correlated
it did stimulate glucose uptake by an average of 31.5% in each of the with b-amyloid levels (Jung et al., 2012), consistent with the lower
three regions in the WT mice (n ¼ 4, P < 0.05: Fig. 5EeG), as well as hippocampal AEA levels in TgAPP mice. b-amyloidosis may lower
by an average of 21.3% in the TgAPP mice (n ¼ 4, P < 0.05: AEA levels by either bolstering catabolism and/or by impairing its
Fig. 5E0 eG0 ). synthesis. COX-2 is an important enzyme in AEA metabolism
(Glaser and Kaczocha, 2010; Pamplona et al., 2010), furthermore, b-
4. Discussion amyloidosis induces COX-2 expression in astrocytes (Giovannini
et al., 2002) and in AD-affected neurons. Hence, COX-2 inhibition
4.1. A novel role for cannabinoid CB2 receptors: stimulating glucose may help to decrease the incidence or slow the progress of AD (Berk
uptake in the brain et al., 2013). In fact, in a previous work we treated TgAPP 2576 mice
orally for four months with the CB2R-selective agonist JWH133 and
We show here that CB2R activation in vitro, and more impor- we observed an attenuation of the AD phenotype, including re-
tantly, in vivo rapidly enhances cerebral glucose uptake by ~30% covery from memory impairment and neuroinflammation, as well
over the basal levels. These data are in concert with a recent finding as a normalization of the increase in COX-2 protein levels (Martín-
that the intraperitoneal injection of the mixed CB1R/CB2R agonist, Moreno et al., 2012).
HU210 increased cerebral 18FDG PET signal by 21% in the rat Strikingly, COX-2 inhibition by the selective DuP697 antagonist
(Nguyen et al., 2012). Since CB1R activation decreases mitochon- facilitated glucose uptake via CB2R activation in WT but not in the
drial glucose metabolism (Duarte et al., 2012) CB2Rs are likely TgAPP mice, probably due to the rescue of AEA from COX-2-driven
involved in the stimulation of brain metabolism in the above study. tonic metabolism in WT mice (Glaser and Kaczocha, 2010;
These data support the view of the recently described antagonistic Pamplona et al., 2010). In the TgAPP mice, we found a 38%
interaction between CB1Rs and CB2R in neurons (Calle n et al., 2012). decrease of AEA, suggesting that these levels were already too low
Given that brain glucose availability controls cognition and to stimulate the CB2Rs, even following COX-2 blockade. In fact, this
memory in humans (Messier, 2004) and that central metabolic may illustrate a deficit in synthesis, since AEA production requires
boosting alleviates the cognitive symptoms of dementia, these data the concurrent stimulation of both NMDA and acetylcholine re-
suggest that CB2R agonists might represent a novel class of noot- ceptors (Stella and Piomelli, 2001), both of which become hypo-
ropic drugs (Branconnier, 1983). This raises the interesting hy- functional in AD (Pavía et al., 1998; Giovannini et al., 2002). In the
pothesis that by promoting AEA release, neural activity can activate present work FAAH activity was reduced in TgAPP mice, as judged
central CB2Rs and thereby refuel circuits under heavy load. Indeed, by the decrease in AEA hydrolysis, again in agreement with its
it was recently found that JWH133 enhanced aversive memory reduction in AD brain, which we recently reported (Mulder et al.,
consolidation in mice, while the genetic ablation or pharmacolog- 2011; Pascual et al., 2014). This reduction is mimicked by patho-
ical inhibition of CB2R impaired it (García-Gutie rrez et al., 2013). logically relevant Ab1-40 peptide concentrations (Pascual et al.,
2-NBDG uptake in slices suggested a contribution of astrocytes 2014) and may serve as to preserve AEA levels under lower AEA
in the effects of CB2R activation. Consistent with this, our data in production, as a compensatory mechanism. Even though, FAAH
cultures show that CB2R influences both astrocytic and neuronal inhibition did not stimulate glucose uptake, suggesting that AEA
glucoregulation. CB2R are found at low density on most cell types in metabolism by FAAH is not a rate limiting factor for CB2R activation
the brain, including activated microglia and astrocytes controlling in the WT hippocampus.
neuroinflammation (Ramírez et al., 2005; Martín-Moreno et al.,
2012), and inhibiting astrocytoma growth (Sa nchez et al., 2001). 4.3. Cannabinoid receptors and glucose uptake
Moreover, CB2R mRNA or protein has also been found in several
types of neurons, including cortical, hippocampal and midbrain Although the role of cannabinoid receptors in peripheral glu-
neurons (Atwood and Mackie, 2010; Calle n et al., 2012; Den Boon coregulation is extensively studied (Matias et al., 2008), the
€falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko 527

mechanism through which CB2R activation facilitates glucose 105663/2008 and Pest-C/SAU/LA0001/2013e2014) via
transport in the brain awaits further interrogation. Several mech- FCTeFundaç~ ^ncia e a Tecnologia.
ao para a Cie
anisms could be invoked on that respect. For instance, cannabi- We thank Dr M. Delgado for excellent assistance in the PET
noids stimulate AMP-activated protein kinase (Dagon et al., 2007), a studies, and Drs L. M. García-Segura and M. A. Arevalo for com-
major intracellular energy sensor, which leads to enhanced glucose ments on the manuscript. We are grateful to Drs Catherine Ledent
uptake by stimulating glucose transporter (GLUT) membrane and Catherine Vroonen (IRIBHM, Brussels) for providing the CB1R
expression and transport efficiency (Rutter et al., 2003; Weisova  KO mice and their WT littermates.
et al., 2009). More importantly, the effect was absent in CB2R
knockout mice (Dagon et al., 2007). A possible direct effect of CB2R Appendix A. Supplementary data
agonists on glucose transporters remains unexplored. New syn-
thesis of transporters would not parallel the acute pharmacological Supplementary data related to this article can be found at http://
effects reported here. Therefore, we suggest that CB2R agonists may dx.doi.org/10.1016/j.neuropharm.2016.03.015.
favor transporter localization at the plasma membrane. On the
other hand, it is less likely that CB2Rs increased glucose uptake
References
driven by the stimulation of its metabolism, because we found
previously that WIN55212-2 slows rather than facilitates the Agudo, J., Martin, M., Roca, C., Molas, M., Bura, A.S., Zimmer, A., Bosch, F.,
mitochondrial oxidative metabolism of glucose via CB1R activation Maldonado, R., 2010. Deficiency of CB2 cannabinoid receptor in mice improves
both in neurons and astrocytes (Duarte et al., 2012). insulin sensitivity but increases food intake and obesity with age. Diabetologia
53, 2629e2640. http://dx.doi.org/10.1007/s00125-010-1894-6.
Ando, R.D., Bíro, J., Cso
€lle, C., Ledent, C., Sperl
agh, B., 2012. The inhibitory action of
4.4. Conclusions exo- and endocannabinoids on [3H]GABA release are mediated by both CB1 and
CB2 receptors in the mouse hippocampus. Neurochem. Int. 60, 145e152.
Aso, E., Ferrer, I., 2014. Cannabinoids for treatment of Alzheimer's disease: moving
In summary, the present results provide the first direct phar- toward the clinic. Front. Pharmacol. 5, 37. http://dx.doi.org/10.3389/
macological evidence in vitro and in vivo of a role of CB2R in central fphar.2014.00037.
glucoregulation. Additionally, we found that glucoregulation by Ashton, J.C., 2011. Knockout controls and the specificity of cannabinoid CB2 receptor
antibodies. Br. J. Pharmacol. 163, 1113. http://dx.doi.org/10.1111/j.1476-
endogenous CB2R signalling is negatively affected by b-amyloidosis, 5381.2010.01139.x.
thought to be the first pathological step in AD. Therefore, it would Atwood, B.K., Mackie, K., 2010. CB2: a cannabinoid receptor with an identity crisis.
be interesting to perform further studies to define how CB2R Br. J. Pharmacol. 160, 467e479. http://dx.doi.org/10.1111/j.1476-
5381.2010.00729.x.
mediated glucoregulation contributes to the recently discovered
Bari, M., Bonifacino, T., Milanese, M., Spagnuolo, P., Zappettini, S., Battista, N.,
therapeutic potential of CB2R agonists in animal models of AD Giribaldi, F., Usai, C., Bonanno, G., Maccarrone, M., 2011. The endocannabinoid
(Martín-Moreno et al., 2012; Wu et al., 2013; Aso and Ferrer, 2014; system in rat gliosomes and its role in the modulation of glutamate release. Cell.
Cheng et al., 2014). Mol. Life Sci. CMLS 68, 833e845. http://dx.doi.org/10.1007/s00018-010-0494-4.
nard, G., Massa, F., Puente, N., Lourenço, J., Bellocchio, L., Soria-Go
Be  mez, E.,
Matias, I., Delamarre, A., Metna-Laurent, M., Cannich, A., Hebert-Chatelain, E.,
Conflicts of interest Mulle, C., Ortega-Gutie rrez, S., Martín-Fontecha, M., Klugmann, M.,
Guggenhuber, S., Lutz, B., Gertsch, J., Chaouloff, F., Lo  pez-Rodríguez, M.L.,
Grandes, P., Rossignol, R., Marsicano, G., 2012. Mitochondrial CB receptors
The authors have no conflict of interests to report, and received regulate neuronal energy metabolism. Nat. Neurosci. 15, 558e564. http://
no financial support or compensation from any individual or dx.doi.org/10.1038/nn.3053.
corporate entity over the past 3 years for research or professional Benito, C., Tolo  n, R.M., Pazos, M.R., Nún ~ ez, E., Castillo, A.I., Romero, J., 2008.
Cannabinoid CB2 receptors in human brain inflammation. Br. J. Pharmacol. 153,
services, and there are no personal holdings that could be perceived 277e285. http://dx.doi.org/10.1038/sj.bjp.0707505.
as constituting a potential conflict of interest. Berk, M., Dean, O., Drexhage, H., McNeil, J.J., Moylan, S., O'Neil, A., Davey, C.G.,
Sanna, L., Maes, M., 2013. Aspirin: a review of its neurobiological properties and
therapeutic potential for mental illness. BMC Med. 11, 74. http://dx.doi.org/
Author contributions
10.1186/1741-7015-11-74.
Branconnier, R.J., 1983. The efficacy of the cerebral metabolic enhancers in the
AK and MLC conceived and designed the work, performed data treatment of senile dementia. Psychopharmacol. Bull. 19, 212e219.
n, L., Moreno, E., Barroso-Chinea, P., Moreno-Delgado, D., Corte
Calle s, A., Mallol, J.,
analysis, writing and edition of the manuscript; AK, CL, BSP and AV- , V., Lanciego, J.L., Franco, R., Lluis, C., Canela, E.I., McCormick, P.J., 2012.
Casado
F performed glucose uptake in slices and in the cell cultures; ROB, Cannabinoid receptors CB1 and CB2 form functional heteromers in brain. J. Biol.
PA and RJR made the cell cultures; AMM-M assessed glucose in Chem. 287, 20851e20865. http://dx.doi.org/10.1074/jbc.M111.335273.
plasma, genotyped by PCR and performed the respective data Carlisle, S.J., Marciano-Cabral, F., Staab, A., Ludwick, C., Cabral, G.A., 2002. Differ-
ential expression of the CB2 cannabinoid receptor by rodent macrophages and
analysis; LG-G and MAP conducted the PET studies and the macrophage-like cells in relation to cell activation. Int. Immunopharmacol. 2,
respective data analysis; MLC and SJP performed the endocanna- 69e82.
binoid hydrolysis assay; and RAC interpreted results, and contrib- Cheng, Y., Dong, Z., Liu, S., 2014. b-Caryophyllene ameliorates the Alzheimer-like
phenotype in APP/PS1 Mice through CB2 receptor activation and the PPARg
uted to the writing and the edition of the manuscript, as well as to pathway. Pharmacology 94, 1e12. http://dx.doi.org/10.1159/000362689.
the purchase of some supplies. All authors have reviewed and Dagon, Y., Avraham, Y., Ilan, Y., Mechoulam, R., Berry, E.M., 2007. Cannabinoids
commented on the manuscript. ameliorate cerebral dysfunction following liver failure via AMP-activated pro-
tein kinase. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 21, 2431e2441. http://
dx.doi.org/10.1096/fj.06-7705com.
Acknowledgements Den Boon, F.S., Chameau, P., Schaafsma-Zhao, Q., van Aken, W., Bari, M., Oddi, S.,
Kruse, C.G., Maccarrone, M., Wadman, W.J., Werkman, T.R., 2012. Excitability of
prefrontal cortical pyramidal neurons is modulated by activation of intracellular
This work was supported by the Council of Madrid (S-BIO/0170/ type-2 cannabinoid receptors. Proc. Natl. Acad. Sci. U. S. A. 109, 3534e3539.
2006 and P2010/BMD-2349 to MLC and MAP). AMM-M received a http://dx.doi.org/10.1073/pnas.1118167109.
fellowship from the Spanish Ministry of Education and Science. Di Marzo, V., De Petrocellis, L., 2012. Why do cannabinoid receptors have more than
one endogenous ligand? Philos. Trans. R. Soc. Lond. B Biol. Sci. 367, 3216e3228.
Authors from the University of Coimbra acknowledge funding from
http://dx.doi.org/10.1098/rstb.2011.0382.
the following sources: NARSAD, Santa Casa da Miserco rdia, DARPA Duarte, J.M.N., Ferreira, S.G., Carvalho, R.A., Cunha, R.A., Ko € falvi, A., 2012. CB1 re-
(09-68-ESR-FP-010), FEDER (QREN) (CENTRO-07-ST24-FEDER- ceptor activation inhibits neuronal and astrocytic intermediary metabolism in
002006), Programa Mais Centro under project CENTRO-07-ST24- the rat hippocampus. Neurochem. Int. 60, 1e8. http://dx.doi.org/10.1016/
j.neuint.2011.10.019.
FEDER-002006 and Programa Operacional Factores de Com- Duff, G., Argaw, A., Cecyre, B., Cherif, H., Tea, N., Zabouri, N., Casanova, C., Ptito, M.,
petitividade - COMPETE, and national funds (PTDC/SAU-OSM/ Bouchard, J.-F., 2013. Cannabinoid receptor CB2 modulates axon guidance. PloS
528 €falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko

One 8, e70849. http://dx.doi.org/10.1371/journal.pone.0070849. Li, Y., Kim, J., 2016a. Deletion of CB2 cannabinoid receptors reduces synaptic
Egertova , M., Cravatt, B.F., Elphick, M.R., 2003. Comparative analysis of fatty acid transmission and long-term potentiation in the mouse hippocampus. Hippo-
amide hydrolase and cb1 cannabinoid receptor expression in the mouse brain: campus 26, 275e281.
evidence of a widespread role for fatty acid amide hydrolase in regulation of Li, Y., Kim, J., 2016b. CB2 cannabinoid receptor knockout in mice impairs contextual
endocannabinoid signaling. Neuroscience 119, 481e496. long-term memory and enhances spatial working memory. Neural Plast.
Esposito, G., Iuvone, T., Savani, C., Scuderi, C., De Filippis, D., Papa, M., Di Marzo, V., 9817089 http://dx.doi.org/10.1155/2016/9817089.
Steardo, L., 2007. Opposing control of cannabinoid receptor stimulation on Li, W., Blankman, J.L., Cravatt, B.F., 2007. A functional proteomic strategy to discover
amyloid-beta-induced reactive gliosis: in vitro and in vivo evidence. inhibitors for uncharacterized hydrolases. J. Am. Chem. Soc. 129, 9594e9595.
J. Pharmacol. Exp. Ther. 322, 1144e1152. http://dx.doi.org/10.1124/ Long, J.Z., Li, W., Booker, L., Burston, J.J., Kinsey, S.G., Schlosburg, J.E., Pavo n, F.J.,
jpet.107.121566. Serrano, A.M., Selley, D.E., Parsons, L.H., Lichtman, A.H., Cravatt, B.F., 2009. Se-
Fowler, C.J., Bjo €rklund, E., Lichtman, A.H., Naidu, P.S., Congiu, C., Onnis, V., 2013. lective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid
Inhibitory properties of ibuprofen and its amide analogues towards the hy- behavioral effects. Nat. Chem. Biol. 5, 37e44. http://dx.doi.org/10.1038/
drolysis and cyclooxygenation of the endocannabinoid anandamide. J. Enzyme. nchembio.129.
Inhib. Med. Chem. 28, 172e182. http://dx.doi.org/10.3109/ Martín-Moreno, A.M., Brera, B., Spuch, C., Carro, E., García-García, L., Delgado, M.,
14756366.2011.643304. Pozo, M.A., Innamorato, N.G., Cuadrado, A., de Ceballos, M.L., 2012. Prolonged
Galiegue, S., Mary, S., Marchand, J., Dussossoy, D., Carriere, D., Carayon, P., oral cannabinoid administration prevents neuroinflammation, lowers b-amy-
Bouaboula, M., Shire, D., Le Fur, G., Casellas, P., 1995. Expression of central and loid levels and improves cognitive performance in Tg APP 2576 mice.
peripheral cannabinoid receptors in human immune tissues and leukocyte J. Neuroinflammation 9, 8. http://dx.doi.org/10.1186/1742-2094-9-8.
subpopulations. Eur. J. Biochem. 232, 54e61. Matias, I., Marzo, V.D., Ko €falvi, A., 2008. Endocannabinoids in energy homeostasis
García-Gutie 
rrez, M.S., Ortega-Alvaro, A., Busquets-García, A., Pe rez-Ortiz, J.M., and metabolic disorders. In: Ko € falvi, A. (Ed.), Cannabinoids and the Brain.
Caltana, L., Ricatti, M.J., Brusco, A., Maldonado, R., Manzanares, J., 2013. Synaptic Springer, US, pp. 277e316.
plasticity alterations associated with memory impairment induced by deletion McGrath, J.C., Drummond, G.B., McLachlan, E.M., Kilkenny, C., Wainwright, C.L.,
of CB2 cannabinoid receptors. Neuropharmacology 73, 388e396. http:// 2010. Guidelines for reporting experiments involving animals: the ARRIVE
dx.doi.org/10.1016/j.neuropharm.2013.05.034. guidelines. Br. J. Pharmacol. 160, 1573e1576. http://dx.doi.org/10.1111/j.1476-
Gierse, J.K., Hauser, S.D., Creely, D.P., Koboldt, C., Rangwala, S.H., Isakson, P.C., 5381.2010.00873.x.
Seibert, K., 1995. Expression and selective inhibition of the constitutive and Messier, C., 2004. Glucose improvement of memory: a review. Eur. J. Pharmacol.
inducible forms of human cyclo-oxygenase. Biochem. J. 305, 479e484. 490, 33e57. http://dx.doi.org/10.1016/j.ejphar.2004.02.043.
Giovannini, M.G., Scali, C., Prosperi, C., Bellucci, A., Vannucchi, M.G., Rosi, S., Milton, N.G.N., 2002. Anandamide and noladin ether prevent neurotoxicity of the
Pepeu, G., Casamenti, F., 2002. Beta-amyloid-induced inflammation and human amyloid-beta peptide. Neurosci. Lett. 332, 127e130.
cholinergic hypofunction in the rat brain in vivo: involvement of the p38MAPK Morgan, N.H., Stanford, I.M., Woodhall, G.L., 2009. Functional CB2 type cannabinoid
pathway. Neurobiol. Dis. 11, 257e274. receptors at CNS synapses. Neuropharmacology 57, 356e368.
Glaser, S.T., Kaczocha, M., 2010. Cyclooxygenase-2 mediates anandamide meta- Mosconi, L., 2005. Brain glucose metabolism in the early and specific diagnosis of
bolism in the mouse brain. J. Pharmacol. Exp. Ther. 335, 380e388. http:// Alzheimer's disease. FDG-PET studies in MCI and AD. Eur. J. Nucl. Med. Mol.
dx.doi.org/10.1124/jpet.110.168831. Imaging 32, 486e510. http://dx.doi.org/10.1007/s00259-005-1762-7.
Han, S., Thatte, J., Buzard, D.J., Jones, R.M., 2013. Therapeutic utility of cannabinoid Mulder, J., Zilberter, M., Pasquare , S.J., Alpar, A., Schulte, G., Ferreira, S.G., Ko
€falvi, A.,
receptor type 2 (CB2) selective agonists. J. Med. Chem. 56, 8224e8256. http:// Martín-Moreno, A.M., Keimpema, E., Tanila, H., Watanabe, M., Mackie, K.,
dx.doi.org/10.1021/jm4005626. Hortoba gyi, T., de Ceballos, M.L., Harkany, T., 2011. Molecular reorganization of
Hillard, C.J., Manna, S., Greenberg, M.J., DiCamelli, R., Ross, R.A., Stevenson, L.A., endocannabinoid signalling in Alzheimer's disease. Brain J. Neurol. 134,
Murphy, V., Pertwee, R.G., Campbell, W.B., 1999. Synthesis and characterization 1041e1060. http://dx.doi.org/10.1093/brain/awr046.
of potent and selective agonists of the neuronal cannabinoid receptor (CB1). Murataeva, N., Straiker, A., Mackie, K., 2014. Parsing the players: 2-
J. Pharmacol. Exp. Ther. 289, 1427e1433. arachidonoylglycerol synthesis and degradation in the CNS. Br. J. Pharmacol.
Hsiao, K., Chapman, P., Nilsen, S., Eckman, C., Harigaya, Y., Younkin, S., Yang, F., 171 (6), 1379e1391. http://dx.doi.org/10.1111/bph.12411, 2014 Mar.
Cole, G., 1996. Correlative memory deficits, Abeta elevation, and amyloid pla- Murineddu, G., Lazzari, P., Ruiu, S., Sanna, A., Loriga, G., Manca, I., Falzoi, M., Dessì, C.,
ques in transgenic mice. Science 274, 99e102. Curzu, M.M., Chelucci, G., Pani, L., Pinna, G.A., 2006. Tricyclic pyrazoles. 4.
Huffman, J.W., Liddle, J., Yu, S., Aung, M.M., Abood, M.E., Wiley, J.L., Martin, B.R., Synthesis and biological evaluation of analogues of the robust and selective CB2
1999. 3-(10,10 -Dimethylbutyl)-1-deoxy-delta8-THC and related compounds: cannabinoid ligand 1-(20 ,40 -dichlorophenyl)-6-methyl-N-piperidin-1-yl-1,4-
synthesis of selective ligands for the CB2 receptor. Bioorg. Med. Chem. 7, dihydroindeno[1,2-c]pyrazole-3-carboxamide. J. Med. Chem. 49, 7502e7512.
2905e2914. http://dx.doi.org/10.1021/jm060920d.
Jakoby, P., Schmidt, E., Ruminot, I., Gutie rrez, R., Barros, L.F., Deitmer, J.W., 2014. Nicolussi, S., Chicca, A., Rau, M., Rihs, S., Soeberdt, M., Abels, C., Gertsch, J., 2014.
Higher transport and metabolism of glucose in astrocytes compared with Correlating FAAH and anandamide cellular uptake inhibition using N-alkyl-
neurons: a multiphoton study of hippocampal and cerebellar tissue slices. carbamate inhibitors: from ultrapotent to hyperpotent. Biochem. Pharmacol.
Cereb. Cortex N. Y. N. 1991 (24), 222e231. http://dx.doi.org/10.1093/cercor/ 92, 669e689. http://dx.doi.org/10.1016/j.bcp.2014.09.020.
bhs309. Nguyen, V.H., Verdurand, M., Dedeurwaerdere, S., Wang, H., Zahra, D., Gregoire, M.-
Jung, K.-M., Astarita, G., Yasar, S., Vasilevko, V., Cribbs, D.H., Head, E., Cotman, C.W., C., Zavitsanou, K., 2012. Increased brain metabolism after acute administration
Piomelli, D., 2012. An amyloid b42-dependent deficit in anandamide mobili- of the synthetic cannabinoid HU210: a small animal PET imaging study with
zation is associated with cognitive dysfunction in Alzheimer's disease. Neuro- 18F-FDG. Brain Res. Bull. 87, 172e179. http://dx.doi.org/10.1016/
biol. Aging 33, 1522e1532. http://dx.doi.org/10.1016/ j.brainresbull.2011.11.011.
j.neurobiolaging.2011.03.012. Pamplona, F.A., Menezes-de-Lima, O., Takahashi, R.N., 2010. Aspirin-triggered lip-
Katona, I., Freund, T.F., 2012. Multiple functions of endocannabinoid signaling in the oxin induces CB1-dependent catalepsy in mice. Neurosci. Lett. 470, 33e37.
brain. Annu. Rev. Neurosci. 35, 529e558. http://dx.doi.org/10.1016/j.neulet.2009.12.050.
Kilkenny, C., Browne, W., Cuthill, I.C., Emerson, M., Altman, D.G., NC3Rs Reporting Pascual, A.C., Martín-Moreno, A.M., Giusto, N.M., de Ceballos, M.L., Pasquare , S.J.,
Guidelines Working Group, 2010. Animal research: reporting in vivo experi- 2014. Normal aging in rats and pathological aging in human Alzheimer's dis-
ments: the ARRIVE guidelines. Br. J. Pharmacol. 160, 1577e1579. http:// ease decrease FAAH activity: modulation by cannabinoid agonists. Exp. Ger-
dx.doi.org/10.1111/j.1476-5381.2010.00872.x. ontol. 60, 92e99. http://dx.doi.org/10.1016/j.exger.2014.10.011.
Kuntner, C., Kesner, A.L., Bauer, M., Kremslehner, R., Wanek, T., Mandler, M., Pavía, J., de Ceballos, M.L., Sanchez de la Cuesta, F., 1998. Alzheimer's disease:
Karch, R., Stanek, J., Wolf, T., Müller, M., Langer, O., 2009. Limitations of small relationship between muscarinic cholinergic receptors, beta-amyloid and tau
animal PET imaging with [18F]FDDNP and FDG for quantitative studies in a proteins. Fundam. Clin. Pharmacol. 12, 473e481.
transgenic mouse model of Alzheimer's disease. Mol. Imaging Biol. MIB Off. Pertwee, R.G., 2012. Targeting the endocannabinoid system with cannabinoid re-
Publ. Acad. Mol. Imaging 11, 236e240. http://dx.doi.org/10.1007/s11307-009- ceptor agonists: pharmacological strategies and therapeutic possibilities. Philos.
0198-z. Trans. R. Soc. Lond. B Biol. Sci. 367, 3353e3363. http://dx.doi.org/10.1098/
Ledent, C., Valverde, O., Cossu, G., Petitet, F., Aubert, J.F., Beslot, F., Bo € hme, G.A., rstb.2011.0381.
Imperato, A., Pedrazzini, T., Roques, B.P., Vassart, G., Fratta, W., Parmentier, M., Ramírez, B.G., Bl azquez, C., Go  mez del Pulgar, T., Guzma n, M., de Ceballos, M.L.,
1999. Unresponsiveness to cannabinoids and reduced addictive effects of opi- 2005. Prevention of Alzheimer's disease pathology by cannabinoids: neuro-
ates in CB1 receptor knockout mice. Science 283, 401e404. protection mediated by blockade of microglial activation. J. Neurosci. Off. J. Soc.
Lemos, C., Pinheiro, B.S., Beleza, R.O., Marques, J.M., Rodrigues, R.J., Cunha, R.A., Neurosci. 25, 1904e1913. http://dx.doi.org/10.1523/JNEUROSCI.4540-04.2005.
Rial, D., Ko€falvi, A., 2015. Adenosine A2B receptor activation stimulates glucose Rodríguez-Cueto, C., Benito, C., Ferna ndez-Ruiz, J., Romero, J., Hern andez-
uptake in the mouse forebrain. Purinergic Signal 11, 561e569. http://dx.doi.org/ Ga lvez, M., Go mez-Ruiz, M., 2014. Changes in CB(1) and CB(2) receptors in the
10.1007/s11302-015-9474-3. post-mortem cerebellum of humans affected by spinocerebellar ataxias. Br. J.
Lemos, C., Vale rio-Fernandes, A., Ghisleni, G.C., Ferreira, S.G., Ledent, C., de Pharmacol. 171, 1472e1489. http://dx.doi.org/10.1111/bph.12283.
Ceballos, M.L., Ko €falvi, A., 2012. Impaired hippocampal glucoregulation in the Romero-Zerbo, S.Y., Garcia-Gutierrez, M.S., Sua rez, J., Rivera, P., Ruz-Maldonado, I.,
cannabinoid CB1 receptor knockout mice as revealed by an optimized in vitro Vida, M., Rodriguez de Fonseca, F., Manzanares, J., Bermúdez-Silva, F.J., 2012.
experimental approach. J. Neurosci. Methods 204, 366e373. http://dx.doi.org/ Overexpression of cannabinoid CB2 receptor in the brain induces hyper-
10.1016/j.jneumeth.2011.11.028. glycaemia and a lean phenotype in adult mice. J. Neuroendocrinol. 24,
Li, Y., Kim, J., 2015. Neuronal expression of CB2 cannabinoid receptor mRNAs in the 1106e1119. http://dx.doi.org/10.1111/j.1365-2826.2012.02325.x.
mouse hippocampus. Neuroscience 311, 253e267. Ruiz-Valdepen ~ as, L., Benito, C., Tolo n, R.M., Martínez Orgado, J.A., Romero, J., 2010.
€falvi et al. / Neuropharmacology 110 (2016) 519e529
A. Ko 529

The endocannabinoid system and amyloid-related diseases. Exp. Neurol. 224, Sperling, R.A., Karlawish, J., Johnson, K.A., 2013. Preclinical Alzheimer disease-the
66e73. http://dx.doi.org/10.1016/j.expneurol.2010.03.024. challenges ahead. Nat. Rev. Neurol. 9, 54e58. http://dx.doi.org/10.1038/
Rutter, G.A., Da Silva Xavier, G., Leclerc, I., 2003. Roles of 50 -AMP-activated protein nrneurol.2012.241.
kinase (AMPK) in mammalian glucose homoeostasis. Biochem. J. 375, 1e16. Stella, N., Piomelli, D., 2001. Receptor-dependent formation of endogenous canna-
http://dx.doi.org/10.1042/BJ20030048. binoids in cortical neurons. Eur. J. Pharmacol. 425, 189e196. http://dx.doi.org/
S
anchez, C., de Ceballos, M.L., Gomez del Pulgar, T., Rueda, D., Corbacho, C., 10.1523/jneurosci.4540-04.2005.
Velasco, G., Galve-Roperh, I., Huffman, J.W., Ramo  n y Cajal, S., Guzman, M., 2001. Stella, N., Schweitzer, P., Piomelli, D., 1997. A second endogenous cannabinoid that
Inhibition of glioma growth in vivo by selective activation of the CB(2) modulates long-term potentiation. Nature 388, 773e778.
cannabinoid receptor. Cancer Res. 61, 5784e5789. Teune, L.K., Bartels, A.L., de Jong, B.M., Willemsen, A.T.M., Eshuis, S.A., de Vries, J.J.,
Savinainen, J.R., Saario, S.M., Laitinen, J.T., 2012. The serine hydrolases MAGL, van Oostrom, J.C.H., Leenders, K.L., 2010. Typical cerebral metabolic patterns in
ABHD6 and ABHD12 as guardians of 2-arachidonoylglycerol signalling through neurodegenerative brain diseases. Mov. Disord. Off. J. Mov. Disord. Soc. 25,
cannabinoid receptors. Acta Physiol. (Oxf) 204, 267e276. http://dx.doi.org/ 2395e2404. http://dx.doi.org/10.1002/mds.23291.
10.1111/j.1748-1716.2011.02280.x. Van Sickle, M.D., Duncan, M., Kingsley, P.J., Mouihate, A., Urbani, P., Mackie, K.,
Savonenko, A.V., Melnikova, T., Wang, Y., Ravert, H., Gao, Y., Koppel, J., Lee, D., Stella, N., Makriyannis, A., Piomelli, D., Davison, J.S., Marnett, L.J., Di Marzo, V.,
Pletnikova, O., Cho, E., Sayyida, N., Hiatt, A., Troncoso, J., Davies, P., Dannals, R.F., Pittman, Q.J., Patel, K.D., Sharkey, K.A., 2005. Identification and functional
Pomper, M.G., Horti, A.G., 2015. Cannabinoid CB2 receptors in a mouse model of characterization of brainstem cannabinoid CB2 receptors. Science 310, 329e332.
Ab amyloidosis: immunohistochemical analysis and suitability as a PET http://dx.doi.org/10.1126/science.1115740.
biomarker of neuroinflammation. PLoS One 10 (6), e0129618. http://dx.doi.org/ Weisova , P., Concannon, C.G., Devocelle, M., Prehn, J.H.M., Ward, M.W., 2009.
10.1371/journal.pone.0129618. Regulation of glucose transporter 3 surface expression by the AMP-activated
Sheng, W.S., Hu, S., Min, X., Cabral, G.A., Lokensgard, J.R., Peterson, P.K., 2005. protein kinase mediates tolerance to glutamate excitation in neurons.
Synthetic cannabinoid WIN55,212-2 inhibits generation of inflammatory me- J. Neurosci. Off. J. Soc. Neurosci. 29, 2997e3008. http://dx.doi.org/10.1523/
diators by IL-1beta-stimulated human astrocytes. Glia 49, 211e219. http:// JNEUROSCI.0354-09.2009.
dx.doi.org/10.1002/glia.20108. Wu, J., Bie, B., Yang, H., Xu, J.J., Brown, D.L., Naguib, M., 2013. Activation of the CB2
Sierra, S., Luquin, N., Rico, A.J., Go mez-Bautista, V., Roda, E., Dopeso-Reyes, I.G., receptor system reverses amyloid-induced memory deficiency. Neurobiol. Ag-
Vazquez, A., Martínez-Pinilla, E., Labandeira-García, J.L., Franco, R., Lanciego, J.L., ing 34, 791e804. http://dx.doi.org/10.1016/j.neurobiolaging.2012.06.011.
2014. Detection of cannabinoid receptors CB1 and CB2 within basal ganglia Zahs, K.R., Ashe, K.H., 2013. b-Amyloid oligomers in aging and Alzheimer's disease.
output neurons in macaques: changes following experimental parkinsonism. Front. Aging Neurosci. 5, 28. http://dx.doi.org/10.3389/fnagi.2013.00028.
Brain Struct. Funct. 1e18. http://dx.doi.org/10.1007/s00429-014-0823-8. Zhang, H.-Y., Gao, M., Liu, Q.-R., Bi, G.-H., Li, X., Yang, H.-J., Gardner, E.L., Wu, J., Xi, Z.-
Solas, M., Francis, P.T., Franco, R., Ramirez, M.J., 2013. CB2 receptor and amyloid X., 2014. Cannabinoid CB2 receptors modulate midbrain dopamine neuronal
pathology in frontal cortex of Alzheimer's disease patients. Neurobiol. Aging 34, activity and dopamine-related behavior in mice. Proc. Natl. Acad. Sci. U. S. A. 111,
805e808. http://dx.doi.org/10.1016/j.neurobiolaging.2012.06.005. E5007eE5015. http://dx.doi.org/10.1073/pnas.1413210111.

You might also like