Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Neuropharmacology 146 (2019) 1–11

Contents lists available at ScienceDirect

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

Neuroprotection by cannabidiol and hypothermia in a piglet model of T


newborn hypoxic-ischemic brain damage
Lorena Barataa,b, Luis Arruzac, Maria-José Rodríguezc, Esther Aleoc, Eva Viergec, Enrique Criadoc,
Elena Sobrinob, Carlos Vargasa, María Cepriána,d, Ana Gutiérrez-Rodrígueza, William Hinde,
José Martínez-Orgadoa,c,∗
a
Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
b
Instituto de Investigación Puerta de Hierro Majadahonda, Spain
c
Servicio de Neonatología, Hospital Clínico San Carlos – IdISSC, Madrid, Spain
d
Departamento de Bioquímica y Biología Molecular, CIBERNED, IRICYS. Facultad de Medicina, Universidad Complutense de Madrid, Spain
e
GW Research Ltd, Cambridge, UK

H I GH L IG H T S

• Neither hypothermia nor CBD alone reduced brain damage in asphyxiated piglets.
• Administration of CDB alone led to some functional beneficial effects.
• Combining hypothermia and CBD led to robust neuroprotective effects.
• Combining hypothermia and CBD modulated excitotoxicity and inflammation.

A R T I C LE I N FO A B S T R A C T

Keywords: Objective: Hypothermia, the gold standard after a hypoxic-ischemic insult, is not beneficial in all treated new-
Hypoxia-ischemia borns. Cannabidiol is neuroprotective in animal models of newborn hypoxic-ischemic encephalopathy. This
Brain study compared the relative efficacies of cannabidiol and hypothermia in newborn hypoxic-ischemic piglets and
Hypothermia assessed whether addition of cannabidiol augments hypothermic neuroprotection.
Cannabidiol
Methods: One day-old HI (carotid clamp and FiO2 10% for 20 min) piglets were randomized to vehicle or
Neuroprotection
cannabidiol 1 mg/kg i.v. u.i.d. for three doses after being submitted to normothermia or 48 h-long hypothermia
Piglets
with a subsequent rewarming period of 6 h. Non-manipulated piglets (naïve) served as controls. Hemodynamic
or respiratory parameters as well as brain activity (aEEG amplitude) were monitored throughout the experiment.
Following termination, brains were obtained for histological (TUNEL staining, apoptosis; immunohistochemistry
for Iba-1, microglia), biochemical (protein carbonylation, oxidative stress; and TNFα concentration, neuroin-
flammation) or proton magnetic resonance spectroscopy (Lac/NAA: metabolic derangement; Glu/NAA: ex-
citotoxicity).
Results: HI led to sustained depressed brain activity and increased microglial activation, which was significantly
improved by cannabidiol alone or with hypothermia but not by hypothermia alone. Hypoxic-ischemic-induced
increases in Lac/NAA, Glu/NAA, TNFα or apoptosis were not reversed by either hypothermia or cannabidiol
alone, but combination of the therapies did. No treatment modified the effects of HI on oxidative stress or
astroglial activation. Cannabidiol treatment was well tolerated.
Conclusions: cannabidiol administration after hypoxia-ischemia in piglets offers some neuroprotective effects but
the combination of cannabidiol and hypothermia shows some additive effect leading to more complete neuro-
protection than cannabidiol or hypothermia alone.

Abbreviations: aEEG, amplitude-integrated EEG; HI, hypoxic-ischemic; MABP, mean arterial blood pressure; CO, cardiac output; HT, therapeutic hypothermia; HR,
heart rate; NHIE, newborn hypoxic-ischemic encephalopathy; NT, normothermia

Corresponding author. Division of Neonatology, Institute of Children and Adolescents (INA), Hospital Clínico "San Carlos" - IdISSC, Profesor Martin Lagos s/n,
28040, Madrid, Spain.
E-mail address: jose.martinezo@salud.madrid.org (J. Martínez-Orgado).

https://doi.org/10.1016/j.neuropharm.2018.11.020
Received 11 July 2018; Received in revised form 31 October 2018; Accepted 13 November 2018
Available online 20 November 2018
0028-3908/ © 2018 Published by Elsevier Ltd.
L. Barata et al. Neuropharmacology 146 (2019) 1–11

1. Introduction conducted in an experimental surgical theatre, starting early in the


morning. Briefly, one-day-old male piglets were intubated under 5%
Neonatal Hypoxic-Ischemic Encephalopathy (NHIE) is a serious sevoflurane anesthesia and then mechanically ventilated (Babylog8000,
condition affecting 1–2/1000 live newborns resulting in high mortality Dräger, Germany) under sedoanalgesia and paralysis by continuous
and long-lasting invalidating sequela rate(McAdams and Juul, 2016). infusion of fentanyl at 3 mcg/kg/h, which is typical for this kind of
Current treatment for NHIE is therapeutic hypothermia (HT), where piglet model (Koehler et al., 2018) and vecuronium at 0.6 mg/kg/h,
newborn body temperature to is reduced to 33.5 °C for 72 h which which were maintained throughout the entire experiment. Both carotid
commences within 6 h of birth (McAdams and Juul, 2016). HT is the arteries were exposed and surrounded by an elastic band, and an in-
only therapy that is proven to reduce death and/or severe sequelae in dwelling catheter was placed in the right jugular vein to infuse dextrose
human NHIE so far, however more than 40% of babies suffering from and saline throughout the experimental period. Cardiac output (CO),
severe or moderate forms of NHIE do not benefit from HT (Jacobs et al., heart rate (HR), mean arterial blood pressure (MABP) and central
2010). Therefore, combination therapies enhancing HT neuroprotective temperature were monitored (PiCCO Plus, Pulsion) with a femoral ar-
efficacy are being actively searched (Cilio and Ferriero, 2010; McAdams tery indwelling catheter (Ominare CMS24, HP, Göblingen, Germany).
and Juul, 2016). Rectal temperature was monitored with a probe inserted to a depth of
In this regard, interest in the role of a non-euphoric component of 6 cm and initially maintained at 37.5–38.5 °C using a water mattress
the Cannabis sativa plant, cannabidiol (CBD), has risen lately because (TecoTherm, Inspiration-Healthcare, West Sussex, UK). Finally, brain
CBD has shown robust neuroprotective effects in different in vitro and in activity was continuously monitored by amplitude-integrated EEG
vivo models of NHIE (Castillo et al., 2010; Lafuente et al., 2016; Pazos (aEEG) (BRM3, BrainZ Instruments). Basal and mean amplitude were
et al., 2012, 2013). CBD is a pleiotropic neuroprotectant, modulating quantified, and raw EEG traces were manually reviewed for electro-
excitotoxicity, neuroinflammation and oxidative stress (Mechoulam graphic seizures (periods of rhythmic activity starting with sudden in-
et al., 2007). In fact, combining HT and CBD in asphyxiated newborn crease in voltage of at least 2 μV, accompanied by a narrowing of the
pigs with a 6 h follow-up results in additive neuroprotective effects, band of aEEG activity, lasting at least 15 s) throughout the entire ex-
regarding the reduction of histological or biochemical brain damage, perimental period. Cerebral and systemic regional oxygen content
and also the modulation of excitotoxicity, neuroinflammation and (crSO2 and srSO2, respectively) were continuously monitored using a
oxidative stress (Lafuente et al., 2016). A longer follow up study is near-infrared spectroscopy (NIRS) device (INVOS oximeter, Covidien,
necessary for two reasons: (i) a secondary energy failure period occurs Mansfield, MA).
from 5 to 24 h after the HI-insult, initiating many pathophysiological
processes that largely determine the eventual severity of the HI brain 2.1.2. HI insult
damage (Johnston et al., 2011; Thornton et al., 2012). Thus, the 6 h After a 30 min period of stabilization, piglets underwent a 25 min-
follow up does not capture this secondary energy failure phase and long cerebral HI insult by interrupting carotid blood flow as a result of
therefore does not provide a complete assessment of the final neuro- pulling out the carotid bands and reducing the fraction of inspired
protective potential. (ii) such a short follow up does not accurately oxygen (FiO2) to 10%. HI was confirmed by suppression of brain ac-
reflect the actual clinical situation, where HT and critical care are ad- tivity on aEEG (flat traces < 4 μV) and the decrease of crSO2
ministered for a longer duration (Jacobs et al., 2010; McAdams and to < 20%. At the end of the period of HI, carotid flow was restored and
Juul, 2016). FiO2 was increased to 21%.
The aim of the present work was to study the neuroprotective effects
of combining HT and CBD in a more translational model closer to the
actual clinical situation, a) using newborn piglets, which result in a 2.1.3. Treatment
scenario more similar to that of human newborns compared to newborn Following the HI insult, piglets were randomized to normothermia
rodent models, and b) administering HT and critical care over a period (NT) or hypothermia (HT) treatment. Using the water mattress, rectal
comparable to that of human newborns. temperature was maintained in the NT group at 37.5–38.5 °C and re-
duced within 20 min to 34–34.5 °C in the HT group. Thirty minutes
2. Methods afterwards, the HI piglets were randomly assigned by closed envelop to
receive i.v. either CBD (HC, 1 mg/kg i.v.) or vehicle only (HV). The
The experimental protocol met European and Spanish regulations administrations were repeated 24 and 48 h after HI. CBD was obtained
for protection of experimental animals (2010/63/EU and RD 53/2013) from a 3 mg/mL formulation provided by GW Research Ltd (Cambridge,
and was approved by the Ethics Committee for Animal Welfare from the UK). The total volume of administration (10 mL) was infused by syringe
University Hospital Puerta de Hierro Majadahonda (Madrid, Spain) and pump over 15 min. Dose was selected based upon previous experiments
Hospital Clínico San Carlos (Madrid, Spain). All experimental proce- (Alvarez et al., 2008; Lafuente et al., 2016; Pazos et al., 2013). Since
dures were designed and carried out by personnel qualified in previous experiments by our group reported the item “aEEG” as the one
Laboratory Animal Science, following FELASA recommendations on with the greater variance, sample size was calculated accordingly and
categories B and C to reduce animal stress and to enhance animal based on those previous results (Lafuente et al., 2011) (α = 0.05,
welfare. All surgery was performed under adequate anesthesia and power 80%, variance 7,5, accuracy 4). Therefore, four experimental
analgesia, and great effort was made to minimize suffering and reduce groups were used this study: HV + NT (n = 8), HC + NT (n = 9),
the number of animals used. Furthermore, all experimental procedures HV + HT (n = 8) and HC + HT (n = 9).
on animal welfare (anesthesia and analgesia, drug and substance ad-
ministration) and euthanasia of the animals were conducted in com- 2.1.4. Follow up
pliance with FELASA recommendations. Landrace-White large one-to- The piglets were kept under the above conditions for 48 h.
two-day-old male piglets were obtained from a qualified farm on the Electrographic seizures, as observed in the aEEG, were not treated by
day of the experiment. anticonvulsant substances unless they progressed into status epilepticus
(more than 5 min of continued seizures) or compromised the hemody-
2.1. Experimental protocol namic condition; in this case phenobarbital 20 mg/kg iv was adminis-
tered, adding phenytoin 20 mg/kg and propofol 14 mg/kg if necessary.
2.1.1. Surgical preparation 48 h after starting the treatment, the HT piglets were rewarmed at
The experimental protocol was based on that extensively reported 0.5 °C/h for 6 h. Control animals were non-manipulated piglets from the
elsewhere (Lafuente et al., 2016; Pazos et al., 2013). Experiments were same age (naïve, n = 6).

2
L. Barata et al. Neuropharmacology 146 (2019) 1–11

2.1.5. Sampling 2.3.3. Histological studies


Following completion of the rewarming period, piglets were main- Immunohistochemistry studies were performed in the three central
tained under sedoanalgesia and their brains were cooled by infusing lobes of the parietal cortex, focusing on 1 mm2 areas in the layers II-III
cold saline (4 °C) through both carotid arteries. Then, piglets were in paraffin-embedded coronal sections (4 μm) as reported elsewhere
killed by KCl infusion, their brains were removed, and the right hemi- (Lafuente et al., 2016; Pazos et al., 2013) to identify microglial cells
sphere sliced and immediately frozen in isopentane (less than 1 min (Iba1, 1:400, Wako). Samples were visualized and photographed using
from asystole) and then stored at −80 °C for subsequent spectroscopy a TCS SP5 confocal microscope (Leica Microsystems, Wetzlar, Ger-
and biochemical studies (right hemisphere). The left hemisphere was many). Using the ImageJ 1.43u software (NIH, Bethesda, USA) neu-
then sliced and sections placed into 4% paraformaldehyde for sub- ronal and glial cell density were calculated; in addition, the areal per-
sequent histological and immunohistochemical studies. centage of Iba1-immunoreactive cell bodies and processes were
calculated to determine cell mean size. Finally, brain sections were
2.2. PK studies stained with TUNEL (ApopTag® In Situ Apoptosis Detection Kit, Milli-
pore, MA, USA) to identify cell death.
Blood samples (1 mL) were obtained from HC + NT and HC + HT
piglets just before (0) and 0.25, 0.5, 1, 3, 6 and 12 h after the first CBD 2.4. Statistical analysis
administration, 1 and 12 h after the second CBD administration and 1 h
after the third one. Samples were immediately centrifuged at 1500 rpm Statistical analysis was done using the StatPlus:mac Pro v6.1.25
for 15 min at 4 °C to obtain plasma, which was immediately frozen and software (AnalystSoft Inc., Walnut, CA). Normality of data was tested
stored at −80 °C, together with frozen brain samples (10 mg weight) using the Kolmogorov-Smirnov test. In the case of aEEG data, no evi-
until use. The brains were homogenized in MeOH:water (10:90 v,v), dence against normality was detected; therefore, three-way ANOVA
which was added in a 3:1 solvent:brain ratio (1 g of brain tissue was was selected for time profile comparisons. In this case, results were
taken to equal 1 mL). CBD was extracted from brain tissue homogenate expressed as mean(SEM). In all other data evidence against normality
using liquid-liquid extraction with 5% IPA (hexane). Then, levels of was detected, so non-parametric tests were used for comparison be-
CBD and two of its minor metabolites, 6-OH-CBD and the active me- tween groups: Mann-Whitney or Kruskall-Wallis test for quantitative
tabolite 7-OH-CBD, were quantitatively determined in plasma and brain data and Fisher's test for qualitative data. Results were expressed as
tissue homogenate using LC-MS/MS at LGC Ltd (Teddington, UK). median (IQR). A p < 0.05 was considered significant.

3. Results
2.3. Brain damage assessment
Ten out of 34 piglets died before completing the follow-up period
2.3.1. Proton magnetic resonance spectroscopy (H + -MRS) (29.4%), with the following distribution: two piglets from the HV + NT
Ex vivo 1H spectra were performed on frozen samples from par- group, two from the HV + HT, two from the HC + NT and three from
ietooccipital cortex using a Bruer Advance 11.7 T spectrometer (Bruker the HC + HT group (Χ2 = 0.65, p = 0.88). These piglets died during
BioSpin, Karlsruhe, Germany) equipped with a 4 mm triple channel 1H/ the first 24 h except for two piglets from the HC + NT group who died
13C/31P HR-MAS (High Resolution Magic Angle Spinning) resonance 36 h after the end of the HI insult. All died from cardiac arrest. Average
probe at the MRI Unit of the Instituto de Investigacione Biomédicas weight was (median [IQR]) 2.2 [2.1,2.8], 2.35 [2.07,2.5], 2.42
“Alberto Sols” to calculate several ratios, including: lactate/N-acylas- [2.27,2.9 and 2.6].
partate (Lac/NAA) and glutamate/N-acylaspartate (Glu/NAA) ratios, as
reported elsewhere (Lafuente et al., 2016; Pazos et al., 2013). 3.1. PK studies

2.3.2. Western blot studies Plasma: Time profiles of plasma concentration of CBD, 6-OH-CBD
Levels of oxidized proteins were quantified by Western blot analysis and 7-OH-CBD after CBD administration to HC + NT or HC + HT
to assess protein carbonylation in brain tissue as described elsewhere piglets are shown in Fig. 1. Plasma CBD concentration peaked at the
(Lafuente et al., 2016; Pazos et al., 2013). A detection kit (Oxyblot, end of the infusion 15 min after the first administration (Fig. 1A). No
Millipore Iberica; Madrid, Spain) was used according to the manufac- cumulative effect was observed after repeated doses. HT led to a sig-
turer's protocol in brain samples containing 15 μg of total protein. After nificant increase in CBD plasma concentration one hour after the first
incubating with the primary antibody (Rabbit Anti-DNPH 1:150; Mil- administration (Fig. 1A), and a significant increase in 6-OH-CBD 30 min
lipore Iberica, Madrid, Spain) for 1 h and then the secondary antibody post-administration (Fig. 1C) (p < 0.05). Comparisons of the AUCs for
(Goat anti-rabbit IgG HRP conjugated 1:300; Millipore Iberica, Madrid, NT and HT groups revealed a significant elevation of 6-OH-CBD levels
Spain) for 1 h at room temperature, an enhanced chemiluminescent (p < 0.05) (Fig. 1D), but it should be noted that levels of 6-OH-CBD
substrate detection system (GE Healthcare, Buckinghamshire, UK) was were very low and in the proximity of the lower limit of quantification
used to visualize the blots. Oxidized protein levels were quantified via (LLQ) for 6-OH-CBD (1.0 ng/mL).
measurement of the optical density, using the NIH Image J analysis Brain: The combination of HT with CBD did not cause an elevation
software (Bethesda, MD, USA). Results were normalized by total pro- in terminal CBD brain concentration compared to NT (61.2 [39.8–90.8]
tein loading, quantified using GX Stain-Free FastCast gels (Bio Rad, vs 71.9 [61.3–16.5] ng/g, median [IQR], for HC + NT and HC + HT,
Hercules, CA, USA) and expressed as OxyBlot/Total Lane Protein ratio. respectively, NS). 7-OH-CBD was not detectable in brains from
TNFα assays were performed with brain samples containing 20 μg of HC + NT but it was present at very low concentrations in brains from
total protein, as described elsewhere(Lafuente et al., 2016). After pri- HC + HT (3.2 [1.5–4.7] ng/g, median [IQR], NS. LLQ = 1.5 ng/g). 6-
mary antibody incubation (Rabbit anti-TNFα 1:1000, Abcam Plc., OH-CBD was not detectable in brains from HC + NT nor HC + HT
Cambridge, UK) at 4 °C overnight, the membranes were incubated with piglets.
the secondary antibody (Goat anti-rabbit IgG HRP-Conjugated for
TNFα, 1:2000; Bio-Rad Lab., Hercules, CA, USA) for 1 h. Finally, as 3.2. Temperature, hemodynamic and respiratory parameters
above, the ECL system was used, and the films were scanned and
analyzed using ImageJ software. Protein levels were quantified using Data are summarized in Table 1. Temperature remained stable
densitometric analysis, normalized by β-actin (1:500, Abcam Plc., throughout the experimental period in NT piglets (Kruskall-Wallis test,
Cambridge, UK) loading and expressed as TNF-α/β-actin ratio. H:7.76, p = 0.17). Temperature was different between NT and HT

3
L. Barata et al. Neuropharmacology 146 (2019) 1–11

Fig. 1. Plasma levels of CBD (A,C) and it's 6-OH (B,D) and 7-OH (E,F) metabolites following 1 mg/kg CBD i.v. administration, in normothermic (filled circles) or
hypothermic (filled squares) piglets as determined by LC-MS/MS. Piglets received CBD 0.5, 24 and 48 h after a hypoxic-ischemic insult. Data are presented as median
(IQR) of five piglets for A, C and E. AUC was calculated using the trapezoidal method and displayed as box & whisker plots with the central line indicating the median
value, and error bars representing the min and max values (B, D and F). Time course comparisons at each time point and AUC groups were compared using the Mann-
Whitney test. *p < 0.05.

groups during hypothermia (H: 41.52, p = 0.00005). in HV + HT and HC + HT from 24 h after HI (Table 1). Thus, both time
A progressive decrease in MABP was observed starting 48 h after HI (H = 62.07, p = 0.00002) and treatment group (H = 42.5,
in NT groups and 24 h after HI in HT groups. Thus, by the end of the p = 0.000005) influenced HR. CO remained generally stable over the
experiment MABP was lower than at the beginning (H = 69.7, experimental period (Time effect H = 8.0, p = 0.89), although treat-
p = 0.00001). In all cases, MABP was kept over 30 mmHg, the limit to ment group influenced CO (H = 11.2, p = 0.01). Thus, CO slightly fell
avoid cerebral blood flow impairment (Lafuente et al., 2016; Pazos throughout the experimental period in HV + NT piglets with an op-
et al., 2013). This was achieved by infusing supportive drugs as de- posite effect in HC + HT piglets, so that by the end of the experiment
scribed in Table 2. Treatment group influenced MABP (H = 8.69, CO was higher in HC + HT than in HV + NT piglets (Table 1). NIRS
p = 0.03), mainly because of pairwise differences between HC + NT revealed a fall in both crSO2 and srSO2 during the HI episode (Table 1)
and the other groups from 48 h post-HI. (Time effect H = 103.5, p = 0.0 and H = 60.2, p = 0.0000008 for
Hypothermia was associated with a decrease in HR, which was seen srSO2 and crSO2, respectively). No significant differences were

4
L. Barata et al. Neuropharmacology 146 (2019) 1–11

Table 1
Hemodynamic and respiratory parameters.
Data from naïve animals were obtained non-invasively from awake piglets. Blood samples were obtained from ear puncture. No CO or OI data were obtained since
naïve piglets were neither cannulated nor intubated.
NAÏVE (n = 6) HV + NT (n = 6) HV + HT (n = 6) HC + NT (n = 7) HC + HT (n = 6)

Temperature (ºC) B 38.1 (37.8,38.2) 37.9 (37.6,38.3) 38.0 (37.9,38.0) 37.8 (37.6,38.2) 37.8 (37.6,38.1)
E 38.3 (37.8,38.5) 38.0 (37.9,38.2) 37.8 (37.6,38.2) 37.6 (37.6,38.1)
T 38.6 (38.2,39.0) 38.0 (38.0,38.1) 38.5 (38.1,38.8) 38.0 (37.9,38.1)
24 h 38.1 (37.7,38.5) 34.1(34.0,34.2)b,a 37.9 (37.7,38.4) 34.3(34.3,34.4)b,a
48 h 37.9 (37.4,38.0) 33.9(33.8,34.0)b,a 38.3 (37.9,38.6) 34.4(34.3,34.5)b,a
54 h 38.1 (38.0,38.3) 37.8 (37.5,37.9) 37.9 (37.8,38.2) 37.6 (37.5,37.9)
Haemodynamics B 90.5(85.0,100.2) 90.5 (72.7,95.5) 93.5 (86.5,99.0) 92.0 (85.5,99.5) 93.0 (92.0,101.0)
Mean blood pressure (mmHg) E 78.5 (59.2,86.5) 72.0 (68.2,89.5) 74.0 (68.2,87.5) 85.0 (77.0,87.0)
T 89.5 (82.2105.7) 87.5 (84.0,93.2) 94.0 (86.5100.5) 95.0 (86.5100.5)
24 h 81.5 (64.2,88.2) 45.5 (44.0,71.7)b,a 78.0 (69.5,91.5) 55.0 (44.0,78.0)b,a
48 h 64.0 (64.0,71.0)b,a 56.0 (52.0,58.0)b,a 79.0 (71.0,81.0)c 52.0 (48.0,88.0)b,a
54 h 62.0 (60.0,73.0)b,a 53.0 (50.0,55,0)b,a 82.5 (79.0,98.0)c,d 49.0(47.0,80)b,a,e
Heart rate (bpm) B 220 (215,231) 216 (210,228) 221 (212,235) 210 (196,241) 237 (225,250)
E 231 (230,258) 249 (236,250) 250 (245,272) 221 (206,250)
T 217 (118,229) 196 (191,218) 238 (223,247) 218 (216,240)
24 h 219 (192,229) 181 (178,188)a,c 225 (209,243) 183 (182,197)a
48 h 233 (217,242) 161 (157,191)a,c 190 (174,211) 168 (157,175)a,c
54 h 235 (214,242) 198 (168,214) 193 (167,195) 188 (180,195)a,c
Cardiac output (mL/min/kg) B – .36 (.26,.43) .34 (.29,.43) .40 (.36,.47) .38 (.27,.45)
E .32 (.24,.44) .36 (.33,.39) .40 (.32,.47) .36 (.30,.40)
T .35 (.33,.39) .30 (.26,.35) .35 (.32,38) .35 (.25,.39)
24 h .37 (.36,.38) .32 (.28,.35) .37 (.35,.40) .33 (.32,.40)
48 h .33 (.24,.44) .28 (.24,.38) .36 (.32, .36) .37 (.32,.54)#†
54 h .33 (.25,.36) .30 (.25,.40) .36 (.32,.40) .42 (.39,.44)#†
srSO2 (%) B 58.1(55.8,64.0) 57.5 (56.0,66.5) 61.0 (55.0,68.5) 58.0 (53.0,59.0) 65.0 (61.0,65.0)
E 20.0 (16.0,34.5)b,a 34.5 (27.5,36.7)b,a 25.0 (21.5,29.5)b,a 31.0 (30.5,35.0)b,a
T 60.5 (52.5,62.5) 56.0 (53.5,60.0) 50.0 (49.0,53.0) 53.0 (49.0,58.0)
24 h 67.5 (61.7,70.0) 70.5 (63.5,76,5( 66.0 (63.0,70.5) 70.0 (63.0,80.0)
48 h 69.0 (55.0,71.0) 70.0 (58.0,79.0) 65.0 (62.0,66.0) 69.0 (65.0,70.0)
54 h 66.0 (53.0,71.0) 65.0 (60.0,74.0) 68.0 (67.0,69.0) 67.0 (65.0,69.0)
crSO2 (%) B 53.1(49.0,54.0) 49.5(2.2) 53.4(4.0) 50.4(2.1) 51.4(3.4)
E 19.5(3.7)b,a 22.8(7.1)b,a 19.6(3.8)b,a 22.0(4.8)b,a
T 50.6(5.7) 54.5(5.1) 47.8(3.2) 47.0(2.3)
24 h 52.3(5.6) 50.8(8.1) 47.5(7.5) 55.2(2.6)
48 h 41.2(5.1)b 54.6(6.4) 46.8(2.1) 53.1(4.3)
54 h 39.6(4.7)b 52.8(7.0) 52.3(2.9) 54.6(3.6)
Respiratory pH B 7.34(.02) 7.36(.03) 7.37(.02) 7.35(.01) 7.31(.04)
E 7.16(.01)b,a 7.18(.02)b,a 7.24(.02)b,a 7.21(.03)b,a
T 7,20(.05)b,a 7.22(.02)b,a 7.20(.02)b,a 7.17(.04)b,a
24 h 7.41(.03) 7.22(.05) 7.40(.02) 7.21(.03)
48 h 7.32(.05) 7.30(.04) 7.41(.02) 7.25(.05)
54 h 7.29(.05) 7.33(.01) 7.39(.03) 7.29(.03)
pCO2 (mmHg) B 41.4(3.3) 46.8(5.1) 44.7(2.3) 46.4(3.1) 47.2(4.8)
E 39.2(4.2) 41.7(4.2) 40.6(2.1) 40.0(1.8)
T 45.1(5.5) 51.2(3.0) 46.7(3.7) 51.7(3.1)
24 h 44.9(3.6) 44.2(4.1) 44.1(2.4) 46.2(4.5)
48 h 44.8(2.9) 45.0(3.4) 43.2(1.4) 45.6(3.6)
54 h 42.2(3.6) 46.1(5.0) 46.8(2.2) 47.5(1.7)
OI B – 3.0(0.2) 2.9(0.2) 2.9(0.2) 3.1(0.4)
E 3.6(0.4) 3.6(0.4) 3.4(0.3) 3.4(0.2)
T 3.4(0.3) 3.2(0.3) 3.2(0.2) 3.2(0.5)
24 h 3.2(0.2) 3.4(0.3) 3.4(0.3) 2.8(0.2)
48 h 3.5(0.2) 3.0(0.2) 2.9(0.2) 2.3(0.3)#
54 h 3.5(0.2) 3.4(0.3) 2.4(0.2)c,d 2.5(0.1)c,d

Median (IQR). HV: hypoxia-ischemia + vehicle. HC: hypoxia-ischemia + cannabidiol. NT: normothermia. HT: hypothermia. bpm: beats per minute. crSO2 and srSO2:
cerebral and systemic regional oxygen content. OI: oxygenation index (mean airway pressure x fractional inspiration oxygen/arterial pO2). B: basal. E: end of the
hypoxic-ischemic insult. T: Treatment. 24–54 h: time after E. Mood's median test p < 0.05.
a
vs. B in the same group.
b
vs. SHM.
c
vs. HV + NT.
d
vs. HV + HT.
e
vs. HC + NT.

observed among groups afterwards (H = 7.6, p = 0.08 and H = 7.9, no differences among treatment groups (H = 2.6, p = 0.45).
p = 0.07 for srSO2 and crSO2, respectively). Oxygenation index (OI) remained stable over the experimental time
Time influenced pH (H = 48.8, p = 0.000002) since during the HI (H = 18.0, p = 0.16), but it was influenced by the treatment group
episode there was a fall in pH which recovered by one hour after HI, but (H = 26.3, p = 0.00008), since piglets receiving CBD either in NT or
there was no influence of treatment group (H = 5.7, p = 0.12). pCO2 HT showed a subtle but progressive decrease in oxygen needs, to an
remained stable throughout the experiment (H = 8.6, p = 0.12), with extent where OI was lower than in HV animals by the end of the

5
L. Barata et al. Neuropharmacology 146 (2019) 1–11

Table 2
Vasoactive drugs.
NAÏVE (n = 6) HV + NT (n = 6) HV + HT (n = 6) HC + NT (n = 7) HC + HT (n = 6)

a
Need of supportive drugs N/A 6 6 2* 5
Time for starting b N/A 27 (24,28.2)) 14 (10,22) 30 (22,33) 15 (12,18)
Dose c N/A 20 (20,20) 17.5 (13.7, 22.5) 5 (0,17.5)# 20 (20,20)
Dopamine
Dobutamine N/A 20 (20,20) 15 (7.5,22.5) 0 (0, 5)# 20 (15,20)
Epinephrine N/A 0 (0,0.5) 0.9 (0.2,1.6) 0 (0,0.7) 0.4 (0.1, 1)

* Fisher's test p = 0.01 vs. other groups.


# Mood's median test p < 0.05 vs. other groups (X2 = 16, p = 0.001).
a
Number.
b
Hours after HI.
c
μg/kg/min. (b,c): Median (IQR).

amplitude remained depressed throughout the entire follow-up. CBD


alone led to a sustained recovery of brain activity, so that by 48 h post-
HI aEEG amplitude was higher in HC + NT than in HV + NT or
HV + HT piglets (Fig. 2). Adding CBD to HT not only reversed the
depressant effect of HT but led to a fast recovery of brain activity,
reaching basal amplitude over 5 μV, higher than that of HV + NT or
HV + HT piglets, from 12 h post-H. After rewarming, aEEG amplitude
was similar in both HC + NT and HC + HT piglets.
Electrographic seizures were not detected in naïve animals.
Electrographic seizure incidence in HV + NT (four out of six) and
HV + HT (three out of six) was significantly higher than in naïve an-
imals (p < 0.05 using Fisher's test). Electrographic seizure incidence in
HC + NT (two out of seven) and HC + HT (one out of six) was not
statistically different from that of naïve animals, but they were not
statistically different to HV + NT (HV + NT vs CBD + HT using Fisher’
test p = 0.07). All animals with electrographic seizures required an-
ticonvulsant therapy (phenobarbital). One animal from HV + NT and
one from HC + NT needed two anticonvulsants (phenobarbital and
phenytoin). Two animals from HV + NT and one from HV + HT
needed three anticonvulsants (the aforementioned and propofol).

3.4. H + -MRS studies

Metabolic brain damage: The HI insult led to a dramatic increase in


Lac/NAA at 54 h post-HI in HV + NT piglets (Fig. 3A). Neither HT or
CBD alone reduced Lac/NAA ratio. However, combining CBD and HT
prevented this increase so that 54 h after HI the Lac/NAA ratio in
HC + HT piglets was similar to that of naïve animals and significantly
lower than all other HI-treated groups (Fig. 3A).
Excitotoxicity: Glu/NAA ratio was increased at 54 h post-HI
(Fig. 3B). Treatment with HT or CBD alone caused a non-significant
Fig. 2. Time profile of mean (top) and basal (bottom) aEEG amplitude assessed
reduction in Glu/NAA. By contrast, combining CBD and HT prevented
in 1- to 2-day-old piglets over a 54 h-long follow-up after a hypoxic-ischemic
(HI) insult, treated with normothermia (filled symbols) or hypothermia (open
the HI-induced increase of excitotoxicity, with Glu/NAA in HC + HT
symbols) and administered with either vehicle (circles) or CBD (triangles). The being at similar levels to naïve piglets and lower than in the other HI
arrow indicates the start of rewarming in hypothermic piglets. Symbols re- groups (Fig. 3B).
present the mean (SEM) of 5–7 experiments. Two-way ANOVA p < 0.05: (#)
vs. HI + vehicle (Mean amplitude: Time: F = 28.5, p = 0.000001, Group 3.5. Biochemical studies
F = 8.7, p = 0.0002, Interaction: F = 1.1, p = 0.3; Basal amplitude: Time:
F = 26.6, p = 0.000001, Group F = 7.9, p = 0.0002, Interaction: F = 1.05,
Oxidative stress: HI led to increased oxidative stress in piglet brain
p = 0.4). B: basal conditions; E: end of the HI insult; T: treatment.
with greater levels of protein carbonylation in HI + NT compared to
naïve piglets (Fig. 4A). However, no statistically significant differences
experiment. were observed among any groups.
Neuroinflammation: HI led to increased concentrations of TNFα in
3.3. Brain activity the brain (Fig. 4B). This increase was not reduced by HT or CBD alone,
but the combination of both therapies resulted in a significant reduction
aEEG analysis showed a similar profile for both basal and mean compared to HV + NT.
amplitudes (Fig. 2). The HI insult led to a profound decrease of brain
activity. In HV + NT piglets aEEG amplitude increased after the HI 3.6. Histological studies
insult and dramatically fell 36 h after HI. Post-HI recovery of brain
activity was not observed in piglets receiving HT alone, where aEEG Apoptosis: Density of TUNEL-positive cells in cortex was

6
L. Barata et al. Neuropharmacology 146 (2019) 1–11

Fig. 3. Box and whiskers representation of B) lactate/n-acylaspartate (Lac/NAA) and C) glutamate/n-acylaspartate (Glu/NAA) ratios as assessed by H+- MRS in brain
samples obtained from 1- to 2-day-old piglets 54 h after a hypoxic-ischemic insult, treated with normothermia (NT) or hypothermia (HT) and administered with
either vehicle (HV) or CBD (HC). A) Representative H+-MRS spectra showing glutamate (G), N-acylaspartate (N) and lactate (L) peaks. Results from 5 to 7 ex-
periments. Kruskall-Wallis test p < 0.05: (*) vs. NAÏVE; (#) vs. HV + NT, (†) vs. HV + HT; (§) vs. HC + NT (Lac/NAA: H = 14,7, p = 0.005; Glu/NAA: H = 16.08,
p = 0.003).

7
L. Barata et al. Neuropharmacology 146 (2019) 1–11

Fig. 4. Western blot studies in brain samples obtained from


1- to 2-day-old piglets 54 h after a hypoxic-ischemic insult,
treated with normothermia (NT) or hypothermia (HT) and
administered with either vehicle (HV) or CBD (HC). In A),
box and whiskers representation of protein carbonylation,
reflecting oxidative stress. A representative image of im-
munoblotting is shown indicating protein carbonyl forma-
tion. Relative protein carbonyl levels were quantified by
densitometric analyses of the blots (OxyBlot) using anti-
DNPH antibody. The content of oxidized proteins was nor-
malized by total Red Ponceau-stained protein loading and
expressed as OxyBlot/Red Ponceau ratio. In B), box and
whiskers representation of TNFa concentration, reflecting
inflammation. A representative image of immunoblotting
using anti-TNFα antibody is shown. Results from 5 to 7 ex-
periments. Kruskall-Wallis test p < 0.05: (*) vs. NAÏVE; (#)
vs. HV + NT (Oxyblot: H = 2.98, p = 0.59; TNFα:
H = 14.5, p = 0.006).

dramatically increased 54 h after HI, which was not reduced by HT or neuroprotection in newborn piglets after a HI insult, an effect related to
CBD alone (Fig. 5A). By contrast, combining CBD and HT significantly the modulation of several major components of HI brain damage pa-
reduced TUNEL-positive cell density, but these piglets still had higher thophysiology.
levels of apoptosis than naïve piglets. A strength of the present work is that we maintained HT treatment
Microglial activation: Microglial proliferation was activated by HI for 48 h, a period longer than usually reported for similar studies
as shown by the increased density of Iba-1-positive cells in the cortex (Faulkner et al., 2011; Håvard T Garberg et al., 2016; Lafuente et al.,
54 h after HI (Fig. 5B), a response that was similar for all HI groups. 2016; Pazos et al., 2013; Robertson et al., 2013), which enhances its
When studying microglial phenotype, it was observed that HI led to translational value. Although the HT period in our experiments was still
microglial cells of increased size which corresponds to a more ameboid shorter than the clinical scenario (72 h) due to piglet critical care needs,
phenotype. This effect was not modified by HT alone, but administra- it has been proven to be of sufficient duration to demonstrate HT
tion of CBD either alone or in combination with HT reduced mean neuroprotection (Koehler et al., 2018). In addition, the early and high
microglial cell size, correspondding to a more ramified phenotype mortality in our sample, similar to that described for asphyxiated
(Fig. 5C). human newborns (Jacobs et al., 2010), was more than double that re-
ported in similar studies (Faulkner et al., 2011; Garberg et al., 2016;
4. Discussion Lafuente et al., 2016; Robertson et al., 2013), suggesting that the HI
insult in our experiments was severe. This explains the lack of neuro-
The present work shows that combining CBD and HT led to robust protective effect of HT alone in our experiments, since HT is ineffective

8
L. Barata et al. Neuropharmacology 146 (2019) 1–11

Fig. 5. Representative light microphotographs of immunohistochemical studies carried out in cortex from brain samples obtained from 1- to 2-day-old piglets 54 h
after a hypoxic-ischemic insult, treated with normothermia (NT) or hypothermia (HT) and administered with either vehicle (HV) or CBD (HC). In A), TUNEL staining
(ApopTag), identifying apoptosing cells. In B), Iba-1 staining identifying microglial cells, whose phenotype is represented after magnification in C). On the right, box
and whiskers representation of the density of TUNEL positive (top) or Iba-1 positive (middle) cells and of mean Iba-1 positive cell size (bottom). Results from 5 to 7
experiments. Kruskall-Wallis test p < 0.05: (*) vs. NAÏVE; (#) vs. HV + NT; (†) vs. HV + HT (§) vs. HC + NT. Bars: 200 μm in A); and B), 20 μm in C) (TUNEL:
H = 33.6, p = 0.0001; Iba-1 density: H = 14.4, p = 0.004; Iba-1 size: H = 56.4, p = 0.000001).

in severely hypoxic piglets (Garberg et al., 2016). The HT temperature prevented HI-induced increases in Lac/NAA to an extent superior to
used in our experiments is considered the optimal one to afford neu- that reported after combining HT with Xenon (Faulkner et al., 2011) or
roprotection whilst avoiding HT-related deleterious side effects in pig- melatonin (Robertson et al., 2013).
lets (Alonso-Alconada et al., 2015). HI increased the density of TUNEL-positive cells in the cortex, in-
dicating that many cells had died via apoptosis 54 h after HI. This effect
4.1. Metabolic brain damage and apoptosis was not modified by HT or CBD alone. HT or CBD alone reduces cell
necrosis and caspase expression in the piglet brain when assessed 6 h
Neither HT or CBD alone reduced the HI-induced increase in Lac/ after a HI insult (Lafuente et al., 2016; Pazos et al., 2013) but it is
NAA, an excellent indicator of hypoxic-ischemic brain injury and known that this effect is not observable after 48 h of HT (Faulkner et al.,
prognosis (Faulkner et al., 2011; Rocha-Ferreira et al., 2017). In pre- 2011). However, combining CBD and HT after HI led to a remarkable
vious reports CBD reduced Lac/NAA 6 h after a moderate HI (Lafuente reduction of TUNEL-positive cell density, similarly to the combination
et al., 2016; Pazos et al., 2013) but not after a severe hypoxic insult in of HT with Xenon or melatonin in HI piglets (Faulkner et al., 2011;
piglets (Garberg et al., 2016). Regarding HT, existing data are con- Robertson et al., 2013). All these data support the robust neuropro-
flicting with reduction (Rocha-Ferreira et al., 2017), no modification tective effect of combining CBD and HT.
(Håvard T Garberg et al., 2016) or increase (Faulkner et al., 2011;
Robertson et al., 2013) of Lac/NAA in asphyxiated piglets. Therefore, it
is a promising observation that the combination of CBD and HT

9
L. Barata et al. Neuropharmacology 146 (2019) 1–11

4.2. Excitotoxicity, oxidative stress and neuroinflammation effects. In HI newborn rats CBD restores neurofunctional performance
despite modest beneficial effects in MRI or histological studies (Pazos
The immature brain is particularly sensitive to excitotoxicity et al., 2012). Altogether these data suggest that even when CBD is
(Johnston et al., 2011). The Glu/NAA ratio, a good indicator for ex- unable to reduce early brain damage it might exert its beneficial effects
citotoxicity in the brain (Groenendaal et al., 2001), was elevated in HI on the surviving tissue. CBD-induced recovery of aEEG amplitude was
piglets. Neither HT nor CBD alone reduced Glu/NAA at 54 h post-HI. In milder than in previous studies (Pazos et al., 2013) supporting the more
piglets HT or CBD reduce glutamate release in the first 6–10 h after severe nature of the present model. Remarkably, combining CBD and
moderate HI (Håvard T Garberg et al., 2016; Lafuente et al., 2016; HT led to a quick recovery of brain activity to ∼50% of the pre-HI
Pazos et al., 2013; Thoresen et al., 1997) although CBD alone does not levels, which remained stable and significantly higher than in piglets
reduce Glu/NAA 9 h after a severe hypoxic insult (Håvard T Garberg receiving HT alone throughout the experimental period, an effect which
et al., 2016). It is, then, noteworthy that the combination of CBD and is not obtained when combining melatonin and HT in HI piglets
HT fully prevented the HI-induced increase in Glu/NAA in the current (Robertson et al., 2013).
study. Since CBD and HT are thought to act on different locations of the
excitotoxicity cascade (Lafuente et al., 2016), their combination likely 4.4. Electrographic seizures
results from an additive effect.
Protein carbonylation, a reliable marker of oxidative stress in piglet The HI insult led to increased electrographic seizure activity, as
brain (Lafuente et al., 2016; Mueller-Burke et al., 2008; Pazos et al., previously described (Alvarez et al., 2008). HT alone did not reduce the
2013), was increased following HI but to a non-significant extent. incidence of these seizures during the first hours after the HI insult,
Oxidative stress is thought to play its important role in the brain da- similar to that reported for human newborns (Jacobs et al., 2010).
mage process in the first hours after HI (Johnston et al., 2011) returning Despite its well-known anticonvulsant effects (Mechoulam et al., 2007),
progressively to basal levels thereafter(Drury et al., 2014; Mueller- CBD administration led to a non-significant trend to reducing HI-in-
Burke et al., 2008), so that 9 h after a severe hypoxic insult no differ- duced electrographic seizures (p = 0.3). This was in contrast to the
ences in oxidative stress biomarkers are found in piglets (Håvard T effect reported during a 6 h-long follow up after HI in piglets (Alvarez
Garberg et al., 2016). Hence, although both HT and CBD are able to et al., 2008), which may be due to the more severe nature of the HI
reduced protein carbonylation in piglet brain 6 h after HI showing an insult in the present experiments. The effect of CBD was augmented by
additive effect when in combination (Lafuente et al., 2016), in the its combination with HT, leading to a near significant trend to reduction
present study protein carbonylation has been assessed at such a late (p = 0.07) of HI-induced electrographic seizure activity.
stage post-HI that inter-group differences were no longer appreciable.
Inflammation is another major component of HI-induced brain da- 4.5. Tolerability
mage process (Johnston et al., 2011). Although cytokine production
peaks shortly after the HI insult in piglets, progressively decreasing to HT had a detrimental effect on HI piglet hemodynamics, where HI
basal levels during the subsequent 48 h (Rocha-Ferreira et al., 2017), piglets receiving HT experienced greater reductions in MABP
the insult in our model was severe enough to maintain high levels of throughout the experimental period despite receiving higher doses of
TNFα as well as a remarkable microglial proliferation 54 h after the HI inotrope drugs. CBD administration in repeated doses over 48 h was
insult. In consonance, although HT or CBD alone reduced cytokine safe, with no side effects on respiratory or cardiovascular performance,
production in the piglet brain in the first hours after moderate HI insults as reported in shorter follow-up models with CBD single dose (Lafuente
(Lafuente et al., 2016; Rocha-Ferreira et al., 2017) neither HT nor CBD et al., 2016; Pazos et al., 2013). In fact, piglets receiving CBD alone
alone reduced this increase 54 h after the HI insult in the present study. were the most stable from a hemodynamic perspective, as they required
However, combining CBD and HT reduced the HI-induced increase in inotropic drugs least frequently. The effect of HT was not reversed by
TNFα concentration, confirming the additive effect reported previously CBD and can be accounted for by the particular sensitivity of piglets to
6 h after the HI insult (Lafuente et al., 2016). None of the treatments HT.
modulated microglial proliferative response, similar to that reported for
melatonin in combination with HT (Robertson et al., 2013). However, 4.6. Pharmacokinetics
when studying microglial morphology it became apparent that the
microglial cells in HV animals were larger and had adopted an ameboid CBD-treated piglets that were exposed to HT showed higher ex-
shape, which is characteristic of activated microglia(Pierre et al., 2017). posure of both parent and metabolites at selected timepoints in plasma,
HI piglets treated with CBD, however, presented microglial cells of there were significantly higher levels of 6-OH-CBD (p = 0.032) when
smaller size and more ramified, suggesting reduced activation. This is a considering the time-course as a whole (AUC), but not for CBD
relevant effect since microglial activation plays a vital role in in- (p = 0.056). However, with respect to the parent compound, it should
flammation-mediated brain damage after HI (Pierre et al., 2017). be noted that this exploratory PK study was not designed to detect
differences between HT and NT groups. Any differences in the PK
4.3. aEEG (brain activity) profile for HT-treated animals could be a consequence of the retarda-
tion in kinetics that occurs in cooled animal. Also, HT increases plasma
aEEG background and amplitude correlates with the degree of brain levels of drugs with cytochrome P450-related metabolism (Anderson
damag (Blanco et al., 2016). We observed in HV + NT piglets the usual et al., 2016), such as CBD (Jiang et al., 2011). After repeated dosing on
flattening of aEEG during and shortly after the HI insult (Lafuente et al., the last day of administration there was no evidence for any accumu-
2016; Pazos et al., 2013; Robertson et al., 2013) followed by a partial lation of CBD or its metabolites in plasma or brain when compared to a
recovery of aEEG amplitude with further decrease 36 h post-HI, likely single dose. Thus, terminal concentration in HC + NT piglets was si-
corresponding to the second energetic fall (Blanco et al., 2016). HT milar to that reported in HI piglets 6 h after single dose (Pazos et al.,
alone was associated with a permanent low-voltage aEEG background 2013).
after a modest early post-HI recovery, in consonance with previous
studies in piglets (Robertson et al., 2013) and in disagreement with that 4.7. Limitations
observed in asphyxiated human newborns, indicating a particular sen-
sitivity of newborn piglet brain to the effects of HT. CBD alone re- The main limitation of our work is the model itself. Although ex-
covered mean and basal amplitudes, which was intriguing since CBD tensively used in preclinical experiments on neuroprotective strategies
alone did not demonstrate biochemical and histological beneficial for NHIE, the hypoxia plus bilateral carotid occlusion piglet model is

10
L. Barata et al. Neuropharmacology 146 (2019) 1–11

considered less representative than other ones, such as severe hypoxia Ther. Hypothermia Temp. Manag. 6, 169–179. https://doi.org/10.1089/ther.2016.
or hypoxia plus complete asphyxia piglet models (Koehler et al., 2018). 0003.
Blanco, D., Ochoa, C., Alarcon, A., Arna, J., Rı, R., 2016. Electroencephalogram as a
In addition, the model is carried out several hours postpartum. Al- prognostic tool in neonates with hypoxic-ischemic Encephalopathy : a systematic
though most experimental models on NHIE in rodents or large mam- review. PloS One 11, e0165744. https://doi.org/10.1371/journal.pone.0165744.
mals are carried out hours or days postpartum (Koehler et al., 2018; Castillo, A., Tol??n, M.R., Fern??ndez-Ruiz, J., Romero, J., Martinez-Orgado, J., 2010.
The neuroprotective effect of cannabidiol in an in vitro model of newborn hypoxic-
Pazos et al., 2012), this can limit its translational power for intrapartum ischemic brain damage in mice is mediated by CB2 and adenosine receptors.
asphyxia, an aspect that can be better addressed by other models as late Neurobiol. Dis. 37, 434–440.
fetal lamb cord compression (Koehler et al., 2018). Therefore, it would Cilio, M.R., Ferriero, D.M., 2010. Synergistic neuroprotective therapies with hy-
pothermia. Semin. Fetal Neonatal Med. 15, 293–298. https://doi.org/10.1016/j.siny.
be interesting to test CBD + HT in other large animal models before 2010.02.002.
concluding that the beneficial effects of CBD + HT in NHIE would be Drury, P.P., Gunn, E.R., Bennet, L., Gunn, A.J., 2014. Mechanisms of hypothermic neu-
universal. roprotection. Clin. Perinatol. 41, 161–175. https://doi.org/10.1016/j.clp.2013.10.
005.
Faulkner, S., Bainbridge, A., Kato, T., Chandrasekaran, M., Kapetanakis, A.B., Hristova,
5. Conclusions M., Liu, M., Evans, S., De Vita, E., Kelen, D., Sanders, R.D., Edwards, A.D., Maze, M.,
Cady, E.B., Raivich, G., Robertson, N.J., 2011. Xenon augmented hypothermia re-
In conclusion, in a strongly translational model of severe HI brain duces early lactate/N-acetylaspartate and cell death in perinatal asphyxia. Ann.
Neurol. 70, 133–150. https://doi.org/10.1002/ana.22387.
damage in newborn piglets HT was ineffective and CBD administered Garberg, H.T., Huun, M.U., Escobar, J., Martinez-Orgado, J., Løberg, E.-M., Solberg, R.,
alone after the HI insult led to some functional beneficial effects. Didrik Saugstad, O., 2016. Short-term effects of cannabidiol after global hypoxia-
However, the combination of CBD and HT led to robust neuroprotective ischemia in newborn piglets. Pediatr. Res. 80, 710–718. https://doi.org/10.1038/pr.
2016.149.
effects as assessed by histological, biochemical and functional studies. Groenendaal, F., Roelants-Van Rijn, A.M., van Der Grond, J., Toet, M.C., de Vries, L.S.,
The beneficial effects of CBD and HT combination was related to the 2001. Glutamate in cerebral tissue of asphyxiated neonates during the first week of
modulation of excitotoxicity and inflammation. However, the combi- life demonstrated in vivo using proton magnetic resonance spectroscopy.
Neonatology 79, 254–257. https://doi.org/10.1159/000047101.
nation of CBD and HT led to robust neuroprotective effects as assessed Jacobs, S.E., Hunt, R., Tarnow-Mordi, W.O., Inder, T.E., Davis, P.G., 2010. Cochrane
by histological, biochemical and functional studies. The beneficial ef- review: cooling for newborns with hypoxic ischaemic encephalopathy. Evidence-
fects of CBD and HT combination was related to the modulation of based child heal. A Cochrane Rev. J. 5, 474–531. https://doi.org/10.1002/ebch.527.
Jiang, R., Yamaori, S., Takeda, S., Yamamoto, I., Watanabe, K., 2011. Identification of
excitotoxicity and inflammation. The present experiments support the cytochrome P450 enzymes responsible for metabolism of cannabidiol by human liver
clinical investigation of the utility of this approach in asphyxiated microsomes. Life Sci. 89, 165–170. https://doi.org/10.1016/j.lfs.2011.05.018.
newborns. Johnston, M.V., Fatemi, A., Wilson, M.A., Northington, F., 2011. Treatment advances in
neonatal neuroprotection and neurointensive care. Lancet Neurol. 10, 372–382.
https://doi.org/10.1016/S1474-4422(11)70016-3.
Funding sources Koehler, R.C., Yang, Z.J., Lee, J.K., Martin, L.J., 2018. Perinatal hypoxic-ischemic brain
injury in large animal models: relevance to human neonatal encephalopathy. J.
This work was supported by grants from the Carlos III Research Cerebr. Blood Flow Metabol. https://doi.org/10.1177/0271678X18797328.
Lafuente, H., Alvarez, F.J., Pazos, M.R., Alvarez, A., Rey-Santano, M.C., Mielgo, V.,
Institute (ISCiii) according to the Spanish Plan for R + D + I Murgia-Esteve, X., Hilario, E., Martinez-Orgado, J., 2011. Cannabidiol reduces brain
2008–2011 and the State Plan for Scientific and Technical Research and damage and improves functional recovery after acute hypoxia-ischemia in newborn
Innovation 2013-2016, with co-funding from the European Regional pigs. Pediatr. Res. 70, 272–277.
Lafuente, H., Pazos, M.R., Alvarez, A., Mohammed, N., Santos, M., Arizti, M., Alvarez,
Development Funds (FEDER) (FIS- PS1600629), from the Biomedicine F.J., Martinez-Orgado, J.A., 2016. Effects of cannabidiol and hypothermia on short-
Program, Community of Madrid (S2010/BMD-2308) and from GW term brain damage in new-born piglets after acute hypoxia-ischemia. Front. Neurosci.
Research Ltd (GWCRI09119). 10, 323. https://doi.org/10.3389/fnins.2016.00323.
McAdams, R.M., Juul, S.E., 2016. Neonatal encephalopathy. Clin. Perinatol. 43, 485–500.
https://doi.org/10.1016/j.clp.2016.04.007.
Conflicts of interest Mechoulam, R., Peters, M., Murillo-Rodriguez, E., Hanus, L.O., 2007. Cannabidiol–recent
advances. Chem. Biodivers. 4, 1678–1692. https://doi.org/10.1002/cbdv.
200790147.
José Martinez-Orgado has a Research Agreement with GW Research
Mueller-Burke, D., Koehler, R.C., Martin, L.J., 2008. Rapid NMDA receptor phosphor-
Ltd (Cambridge, UK). ylation and oxidative stress precede striatal neurodegeneration after hypoxic
ischemia in newborn piglets and are attenuated with hypothermia. Int. J. Dev.
Neurosci. 26, 67–76. https://doi.org/10.1016/j.ijdevneu.2007.08.015.
Acknowledgments
Pazos, M.R., Cinquina, V., Gómez, A., Layunta, R., Santos, M., Fernández-Ruiz, J.,
Martínez-Orgado, J., 2012. Cannabidiol administration after hypoxia-ischemia to
We are indebted to Martin Santos, PhD, María Dolores Molina newborn rats reduces long-term brain injury and restores neurobehavioral function.
Corzo, Maria Cruz Rodríguez-Bobada and Pablo González López for Neuropharmacology 63, 776–783. https://doi.org/10.1016/j.neuropharm.2012.05.
034.
their help performing this experiment. Pazos, M.R., Mohammed, N., Lafuente, H., Santos, M., Martínez-Pinilla, E., Moreno, E.,
Valdizan, E., Romero, J., Pazos, A., Franco, R., Hillard, C.J., Alvarez, F.J., Martínez-
Appendix A. Supplementary data Orgado, J., 2013. Mechanisms of cannabidiol neuroprotection in hypoxic-ischemic
newborn pigs: role of 5HT(1A) and CB2 receptors. Neuropharmacology 71, 282–291.
https://doi.org/10.1016/j.neuropharm.2013.03.027.
Supplementary data to this article can be found online at https:// Pierre, W.C., Smith, P.L.P., Londono, I., Chemtob, S., Mallard, C., Lodygensky, G.A., 2017.
doi.org/10.1016/j.neuropharm.2018.11.020. Neonatal microglia: the cornerstone of brain fate. Brain Behav. Immun. https://doi.
org/10.1016/j.bbi.2016.08.018.
Robertson, N.J., Faulkner, S., Fleiss, B., Bainbridge, A., Andorka, C., Price, D., Powell, E.,
References Lecky-Thompson, L., Thei, L., Chandrasekaran, M., Hristova, M., Cady, E.B.,
Gressens, P., Golay, X., Raivich, G., 2013. Melatonin augments hypothermic neuro-
protection in a perinatal asphyxia model. Brain 136, 90–105. https://doi.org/10.
Alonso-Alconada, D., Broad, K.D., Bainbridge, A., Chandrasekaran, M., Faulkner, S.D.,
1093/brain/aws285.
Kerenyi, A., Hassell, J., Rocha-Ferreira, E., Hristova, M., Fleiss, B., Bennett, K., Kelen,
Rocha-Ferreira, E., Kelen, D., Faulkner, S., Broad, K.D., Chandrasekaran, M., Kerenyi, Á.,
D., Cady, E., Gressens, P., Golay, X., Robertson, N.J., 2015. Brain cell death is reduced
Kato, T., Bainbridge, A., Golay, X., Sullivan, M., Kramer, B.W., Robertson, N.J., 2017.
with cooling by 3.5 C to 5 C but increased with cooling by 8.5 C in a piglet asphyxia
Systemic pro-inflammatory cytokine status following therapeutic hypothermia in a
model. Stroke 46, 275–278. https://doi.org/10.1161/STROKEAHA.114.007330.
piglet hypoxia-ischemia model. J. Neuroinflammation 14, 44. https://doi.org/10.
Alvarez, F.J., Lafuente, H., Rey-Santano, M.C., Mielgo, V.E., Gastiasoro, E., Rueda, M.,
1186/s12974-017-0821-x.
Pertwee, R.G., Castillo, A.I., Romero, J., Martínez-Orgado, J., 2008. Neuroprotective
Thoresen, M., Satas, S., Puka-Sundvall, M., Whitelaw, A., Hallström, A., Løberg, E.M.,
effects of the nonpsychoactive cannabinoid cannabidiol in hypoxic-ischemic newborn
Ungerstedt, U., Steen, P.A., Hagberg, H., 1997. Post-hypoxic hypothermia reduces
piglets. Pediatr. Res. 64, 653–658.
cerebrocortical release of NO and excitotoxins. Neuroreport 8, 3359–3362.
Anderson, K.B., Poloyac, S.M., Kochanek, P.M., Empey, P.E., 2016. Effect of hypothermia
Thornton, C., Rousset, C.I., Kichev, A., Miyakuni, Y., Vontell, R., Baburamani, A.A., Fleiss,
and targeted temperature management on drug disposition and response following
B., Gressens, P., Hagberg, H., 2012. Molecular mechanisms of neonatal brain injury.
cardiac arrest: a comprehensive review of preclinical and clinical investigations.
Neurol. Res. Int. https://doi.org/10.1155/2012/506320. 2012.

11

You might also like