Next Article in Journal
Long-Term Treatment with Simvastatin Leads to Reduced Migration Capacity of Prostate Cancer Cells
Next Article in Special Issue
Cannabidiol Decreases Intestinal Inflammation in the Ovariectomized Murine Model of Postmenopause
Previous Article in Journal
Brain-Derived Neurotrophic Factor (BDNF) as an Indicator for Effects of Cognitive Behavioral Therapy (CBT): A Systematic Review
Previous Article in Special Issue
Differential Regulation of MMPs, Apoptosis and Cell Proliferation by the Cannabinoid Receptors CB1 and CB2 in Vascular Smooth Muscle Cells and Cardiac Myocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Endocannabinoid System as a Target for Neuroprotection/Neuroregeneration in Perinatal Hypoxic–Ischemic Brain Injury

1
Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
2
Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(1), 28; https://doi.org/10.3390/biomedicines11010028
Submission received: 18 November 2022 / Accepted: 19 December 2022 / Published: 22 December 2022
(This article belongs to the Special Issue Therapeutic Potential for Cannabis and Cannabinoids)

Abstract

:
The endocannabinoid (EC) system is a complex cell-signaling system that participates in a vast number of biological processes since the prenatal period, including the development of the nervous system, brain plasticity, and circuit repair. This neuromodulatory system is also involved in the response to endogenous and environmental insults, being of special relevance in the prevention and/or treatment of vascular disorders, such as stroke and neuroprotection after neonatal brain injury. Perinatal hypoxia–ischemia leading to neonatal encephalopathy is a devastating condition with no therapeutic approach apart from moderate hypothermia, which is effective only in some cases. This overview, therefore, gives a current description of the main components of the EC system (including cannabinoid receptors, ligands, and related enzymes), to later analyze the EC system as a target for neonatal neuroprotection with a special focus on its neurogenic potential after hypoxic–ischemic brain injury.

1. The Endocannabinoid System

The endocannabinoid (EC) system is a cell-signaling system consisting mainly of at least two cannabinoid (CB) receptors, namely CB1 and CB2, their endogenous ligands, and the enzymes responsible for the synthesis, transport, and degradation of endocannabinoids (ECs) [1]. Changes in the expression or activity of CB receptors, ligands, or enzymes are implicated in many pathological conditions [2]. Neurological disorders such as anxiety, depression, schizophrenia, neurodegenerative (e.g., Parkinson’s and Huntington’s disease), and stroke-related disorders, together with osteoporosis, multiple sclerosis, neuropathic pain, cancer, glaucoma, hypertension, and obesity/metabolic syndrome are just the major diseases associated with perturbations of the EC system [3]. Recently, it has been hypothesized that CB2 receptor activation may be also useful to reduce the inflammatory response induced by SARS-CoV-2 infection, due to its capacity to ameliorate the production of cytokines responsible for the pathological phenomenon [4].
However, changes in EC tone are sometimes transient and likely part of the organism’s compensatory response mainly aimed at reducing symptoms or slowing the progression of pathological conditions. In the nervous system, the activity of the EC system also appears related to neuroprotection, because of its ability to modulate the intensity and extent of a series of dangerous biological events involved in the neurodegenerative process. These include modulation of glutamate excitotoxicity [5] and oxidative stress [6], and a reduction in the inflammatory response [7]. This scenario led to considering the EC system as a potential target for developing new neuroprotective therapies [3]. However, it is not always clear whether an increased activity of the EC system can be consequent to a higher biosynthetic activity or a reduction in the metabolic degradation of the endogenous ligands. Therefore, a better understanding of the role and mechanisms underlying EC tone alterations during the neurodegenerative process represents key factors for developing new therapeutic agents acting through this important modulatory system.
Overall, despite the vast amount of knowledge acquired over time, the exploration of the EC system still represents a stimulating goal. Indeed, the complexity of its structures, the species variability of its characteristics, and the overlapping of pharmacological targets, still leave open many questions and scientific opportunities. This review aims to highlight the potential role of the EC system in the neurodegenerative and neuro-reparative processes resulting from hypoxic–ischemic insults occurring during brain development. A summary of CB receptors, ligands, and related enzymes is also reported.

1.1. Cannabinoid Receptors

The effects associated with the endo and exocannabinoid compounds are primarily related to their interaction with the CB1 and CB2 receptors, discovered some decades ago [8,9,10] and characterized based on their neurobiology signaling [11]. Their involvement in many physiological and pathological events justifies the central role that they play as a possible therapeutic key for many diseases. CB receptors can be stimulated or antagonized by different ligands and can also be modulated through the inhibition of the enzymes responsible for the degradation of their endogenous ligands [12]. Unfortunately, the interaction of exocannabinoids with these receptors, especially with the CB1 subtype, is also associated with the psychotropic effects of many recreational drugs, including Cannabis, the so-called new psychoactive substances [13], and smart drugs (SPICE and K2…) [14], or to other undesirable serious effects of synthetic agonist or antagonist drugs [15,16,17].
CB1 receptors are abundantly expressed in the central nervous system (CNS), particularly in the cerebral cortex, hippocampus, basal ganglia, and cerebellum. CB2 receptors, instead, are mostly expressed in the immune system, particularly in B and natural killer cells. However, CB2 receptors have been also found in some districts of the CNS [18] and the CB1 also peripherally, albeit at low levels [11]. More detailed information on the origin, structural aspects, and signaling processes mediated by CB1 and CB2 receptors are reported in [19,20].
Generally, the activation of CB receptors determines the inhibition of adenylate cyclase, with a consequent decrease in the levels of cyclic adenosine monophosphate (cAMP), a second messenger involved in numerous intracellular signaling and essential for the regulation of many cell functions. There is also evidence that the CB1 receptor, in addition to acting on adenylate cyclase, can be coupled to ion channels [21], confirming the key role of CBs in inducing activation or depression of neurotransmission [11].
Recent studies have also revealed the existence of “atypical” EC receptors, i.e., the transient receptor potential vanilloid (TRPV) channels, involved in the nociceptive signaling; the GRP55, G-protein coupled receptors responsible for some independent CB1 and CB2 responses; the peroxisome proliferator-activated receptor gamma (PPAR-γ) receptors, which are physiologically involved in glucose metabolism and insulin signaling, and also in inflammation and pain; and the dopamine, adenosine, opioid, and 5-HT1A receptors [22].

1.2. Endocannabinoids

Endocannabinoids (ECs) are endogenous lipidic compounds formed by a long-chain polyunsaturated fatty acid tail and a polar head containing functional groups such as amide, ester, ether, or hydroxy one. They bind to CB receptors but, unlike most neurotransmitters that are synthesized and stored in vesicles; their synthesis from membrane phospholipids is on-demand and use-dependent [23,24].
ECs are released from postsynaptic terminals in a Ca2+-dependent manner. After their release, they activate presynaptic CB receptors usually through retrograde signaling, although non-retrograde signaling may occur [24]. The retrograde signaling mechanism is responsible for modulating both short-term and long-term neuroplasticity [25]. The short-term type of modulation (seconds) participates in processes, such as depolarization-induced suppression of inhibition and depolarization-induced suppression of excitation. This may occur through the inhibition of Ca2+ voltage-gated channels and the modulation of the synaptic release of various neurotransmitters, including glutamate and γ-aminobutyric acid (GABA) [26,27]. In addition, ECs are also involved in long-term synaptic plasticity (in the order of minutes) through a CB1 repeated stimulation of these brain circuits [24]. This process leads to the long-term depression phenomenon, with the final decrease in the glutamatergic and GABAergic synaptic activity [28]. Thus, ECs may function as a polymodal signal integrator to allow the diversification of synaptic plasticity in a single neuron [29]. EC receptors, in particular those in the CNS, can, therefore, be potential drug targets for the prevention and treatment of neurologic disorders, such as brain ischemia [30].
The best-studied ECs are N-arachidonoylethanolamine (anandamide and AEA, as seen in Figure 1) [31] and 2-arachidonoyl-sn-glycerol (2-AG, as seen in Figure 1) [32,33], but other arachidonic acid derivatives (e.g., noladin ether, virodhamine, and N-arachidonoyldopamine) can bind CB1 and/or CB2 receptors, although their physiological role is not yet clear.
AEA is a full or partial agonist of the CB1 receptor but also shows low activity towards the CB2 receptor [34,35,36,37,38], whereas 2-AG is a full agonist of both CB1 and CB2 receptors [39].
Differences between AEA and 2-AG occur also in the biosynthetic pathways responsible for their formation and degradation. With reference to the synthetic step, N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD) [40,41] or diacylglycerol lipase (DGL) [42,43] are the enzymes directly involved, whereas fatty acid amide hydrolase (FAAH) [44,45,46,47] or monoglyceride lipase (MGL) [48,49,50] are the main enzymes responsible for their metabolism, leading to the formation of arachidonic acid and ethanolamine [51] or glycerol [52] following a cellular internalization process carried out by specific transporters [53,54,55].

1.3. Cannabis, Phytocannabinoids, and Synthetic Cannabinoids

Cannabis contains more than 500 compounds, of which at least 100 are known to be phytocannabinoids [56] owing to pharmacological properties [57]; they are described in detail in [58]. Paleobotanical studies attest that Cannabis was already present during the Holocene epoch about 11,700 years ago, more likely in the territories of Central Asia near the Altai Mountains [59]. The first written testimony on the use of Cannabis for therapeutic purposes dates back to 2700 BC, when the Chinese emperor Shen-Nung reported a detailed description of it in a book that later became the Chinese compendium of drugs [60]. Chinese people used this plant for diseases such as rheumatic pain, malaria, constipation, etc. [61]. Despite their long history, the phytocannabinoids contained in Cannabis were identified only a few decades ago and progressively studied both as single molecules and as their derivatives, and also based on structure–activity relationship studies [62].
The most studied and characterized phytocannabinoids have been Δ9-tetrahydrocannabinol (Δ9-THC, Figure 2) [63], which constitutes the main psychoactive compound of Cannabis, and the non-psychotropic cannabidiol (CBD, Figure 2) [64]. Despite similar structures, their pharmacodynamic properties deeply differ.
Δ9-THC acts as a partial agonist of the CB1 receptor, which explains its strong psychoactive outcomes inducing the tetrad effects (hypothermia, catalepsy, hypolocomotion, and analgesia). These unwanted effects make the medical use of the Δ9-THC strongly restricted despite its beneficial action (neuroprotective, anti-inflammatory, and antispasmodic), which depends on the activation of CB2 and PPAR-γ receptors [56]. In addition, the activity of the molecule is dependent on cell type, receptor expression, and the presence of ECs or other agonists [56,65].
For its part, CBD has a high affinity on a series of targets, including CB1 and CB2, GPR55, TRPV, PPAR-γ, 5-HT1A, dopamine, and opioid receptors, and also on ion channels, which contributes to the beneficial effects of Cannabis in diseases related to a wide range of pathologies (neurological, ischemic stroke, inflammatory, pain, etc.) [22,66,67].
Although Δ9-THC and CBD are the best-known and studied phytocannabinoids, other compounds have been relieved in Cannabis owing to their therapeutic potential. Among these, the main ones are reported below. (1) Cannabigerol (CBG), which is a non-psychotropic derivative with a low affinity for the CB1 and CB2, but it is able to interact with other receptors, such as α2-adrenergic, TRPs superfamily, and 5-HT1A, and to possess various properties (antiproliferative, antibacterial, antioxidant, etc.) [56]. (2) Cannabichromene (CBC), which acts weakly with CB1 and CB2 receptors and is able to inhibit AEA uptake, is the most potent agonist of TRPA1 channels and possesses antinociceptive and anti-inflammatory properties in vitro and in vivo [56]. (3) Δ9-Tetrahydrocannabivarine (Δ9-THCV), which is the n-propyl analogue of Δ9-THC and acts weakly with CB1 receptors and more potently with the CB2 ones. However, it is also able to act with other targets such as TRP, GPR6, GPR55, and D2 receptors and to exert related pharmacological actions [56]. (4) Cannabinol (CBN), which is the first phytocannabinoid structurally characterized; its derivatives are considered as the oxidative by-product of the degradation process of Δ9-THC and CBD [62,68]. It acts as a partial agonist of CB1 and CB2 and exerts neuroprotective, antiepileptic, and analgesic properties [68,69].
Synthetic cannabinoids are ligands that bind to CB receptors and modulate their activity. Their design and the studies aimed to acquire information on the structural requirements to establish interactions with CB receptors. Moreover, the goal was also related to the understanding of the role played by molecules that bind with these targets, the role of the targets themselves, and, more generally, the role of the EC system, since synthetic cannabinoids can be considered tools to increase knowledge in the field. As for natural ligands, the role of synthetic cannabinoids must always be contextualized within the situation of a risk/benefit ratio that would derive from their use. In spite of the large effort in synthesizing and characterizing the pharmacological profile of these molecules, apart from nabilone, there are currently no drugs on the market containing a synthetic cannabinoid, but many of them are used for recreational purposes and are included in the list of substances of abuse [15,70,71,72,73].
The structural requirements that allow synthetic cannabinoids to interact with CB receptors are highly variable, a situation that strongly influences their pharmacological activity, in particular for what concern agonism, antagonism, and, more rarely, inverse agonism. It is interesting to consider, however, that their activity sometimes depends on the experimental model used for their characterization (e.g., antagonism vs. inverse agonism). An overview of these features and the therapeutic potential of CB ligands are presented in Refs. [74,75,76,77,78,79].
The therapeutic interest of the drugs that bind to CB receptors is also proved by the marketed drugs mentioned above. Cesamet® (nabilone—the synthetic dibenzopyran-9-one analog of Δ9-THC), administered for the improvement of chemotherapy-induced nausea and vomiting (CINV) states in patients not responding to conventional antiemetic therapies. Marinol® (dronabinol—the synthetic pure isomer (–)-trans9-THC) is prescribed for the same purposes as the former and also for appetite stimulation in patients with AIDS (acquired immune deficiency syndrome). Sativex®9-THC and cannabidiol in an approximate 1:1 fixed ratio) is used for the symptomatic relief of the pain and/or the management of neuropathic pain and spasticity in adults with multiple sclerosis and is not responsive to other antispasticity therapies. More recently, a new drug containing > 98% CBD and less than 0.15% Δ9-THC (Epidiolex®) has been approved for the treatment of seizures associated with two rare and severe forms of epilepsy (Lennox–Gastaut and Dravet syndromes) in patients two years of age and older [56,66,80,81].
An intriguing and interesting feature of CB receptor ligands is that they are able to exert a neuroprotective role after ischemic injuries [82,83,84,85]. The CB1 and CB2 agonists (–)-CP-55,940 [86] and (R)-(+)-WIN-55212-2 [87], the CB1 inverse agonists SR141716A [88] and AM 251 [89], the CB1 antagonists LY320135 [90], and the CB2 agonist/CB1 antagonist URB447 [91] (Figure 3) are the related tools studied.
Despite the therapeutic use of drugs containing Cannabis and its derivatives or of synthetic ligands of CB receptors, problems related to their abuse remain open. For this reason, molecules inhibiting the degradation of endogenous ligands may represent an interesting alternative for modulating the EC system [92].

1.4. FAAH and MGL Inhibitors

These compounds increase AEA and 2-AG levels by inhibiting their intracellular degradation. The hypothesis leading to the design and development of EC metabolism inhibitors is based on the fact that, by blocking the degradation of these endogenous mediators, we can increase their concentrations in the physiological districts where they are formed. By using this approach, it may be possible to obtain pharmacological agents characterized by the absence of the psychotropic side effects typical of CB1 exogenous ligands. Indeed, ECs are synthesized and released on-demand in a tissue-specific and time-dependent manner and inhibitors of their metabolic enzymes will cause an increase in EC levels only where and when it is physiologically required. In this way, the activation of CB receptors is obtained through endogenous ligands, but it is prolonged over time. Several pieces of evidence support this approach. For example, it has been reported that FAAH knockdown mice show increased levels of AEA in the brain and other tissues leading to CB1 receptor-mediated analgesia [93,94], or a reduction in anxiety symptoms without the appearance of catalepsy [95].
The possibility of targeting the FAAH and MGL enzymes, therefore, may represent an important therapeutic approach for different pathologies [2,96,97,98,99] and, even if there are no drugs on the market yet, studies carried out in this regard are promising. Disorders related to anxiety, pain, and cigarette and cannabis smoking are the main pathological states in which EC metabolism inhibitors have been studied [2], and clinical studies are in progress. FAAH and MGL inhibitors have also been considered pharmacological tools to increase information on the role of the EC system in neurodegeneration/neuroprotection after ischemic injuries [100,101,102,103,104,105]. The FAAH inhibitor URB597 [95,106,107,108,109,110,111,112,113] and MGL inhibitors URB602 [101,114,115,116], JZL184 [117], KML29 [118], and MJN110 [119] (Figure 4) are the main experimental molecules assessed in these studies.

2. The Endocannabinoid System in Prenatal and Postnatal Development

During prenatal and postnatal brain development, the EC system may play an active role in the control of the cell cycle, proliferation, survival, and differentiation of neural stem cells [120], as well as in the maturation of the nervous system and its functions. The modulation of some of these processes appears regulated by the CB1 receptor, which is expressed in the very early stages of neural development. Indeed, the expression of members of the EC system has been described during early developmental and postnatal stages [121,122,123], and in the embryonic rat brain, its presence was found around day 11 of gestation [124]. In 1998, Berrendero et al. [121] not only demonstrated the existence of CB1 receptors, but they also showed that these receptors were already functional in embryonic stages. In humans, the presence of CB1 receptors has been documented as soon as at week 14 of gestation in the embryo [125].
The regions in which CB1 receptors are expressed in these early stages, i.e., the corpus callosum, stria terminalis, stria medullaris, fasciculus retroflexum, or anterior commissure, are related to processes such as cell proliferation, migration, axonal elongation and synaptogenesis [121,122,123,126]. The later modifications in CB1 receptors’ location during neural development (becoming different in the adult brain), suggest that their expression in the brain changes once their contribution to neural development finishes [121,123].
In murine cell cultures, CB1 receptors appear in several cell types, including stem-like cells, astrocytes, and immature neurons [127]. It has also been observed that the agonist (R)-(+)-methanandamide promoted self-renewal, multipotency, and neuronal differentiation via CB1 activation. When ECs are produced or exogenously administered with bind CB1 receptors, the αi subunit linked to the protein inhibits the activity of adenylyl cyclase and the synthesis of cAMP. Low levels of cAMP reduce the activity of the protein kinase-A and, consequently, the type-A potassium channels are activated and lead to membrane hyperpolarization. The α0 subunit of the G protein associated with the CB1 receptor, instead, inhibits voltage-dependent Ca2+ channels causing cell depolarization. The β and γ subunits, moreover, interact with pathways, such as PI3K or PKB/Akt, that have been shown to induce the expression of transcription factors associated with cell proliferation (CREB, STAT-3, PAX-6, and β-catenin). CB receptors are also closely related to neutral sphingomyelinase, which generates ceramide from sphingomyelin located in the plasma membrane, thus activating the synthesis of transcription factors, such as ERK or p38, that control cell fate and survival [128,129]. The involvement of factors, such as ERK or PI3K, in neurogenesis associated with CB1 activation was also observed by Xapelli et al. [127].
The processes of migration and path-finding during neurogenesis appear also partially regulated by the CB1 receptor. Their blockage with a selective antagonist caused a decrease of 50% in migration in a scratch wound assay in mouse fetal cortex-derived cells [130]. The same authors also labeled the rostral migratory stream explants embedded in Matrigel using the migrating neuroblast markers PSA-NCAM and DCX, and observed a significant reduction (30%) in the migratory distance after the treatment with a CB1 receptor antagonist. The role of the EC system in the path-finding function also became evident when EC signals were proven to be behind axon direction cues, helping neurons find their path [131].
Together with CB receptors, the ECs AEA and 2-AG also make their appearance in the prenatal period. Despite the presence of AEA levels having been detected from the early stages of the embryo [132], 2-AG seems to be predominant in the fetal period, as this molecule has been found in higher concentrations than AEA [133]. Conversely, AEA levels increase gradually during brain development until an adult level is reached, while the concentration of 2-AG remains more or less stable than in the fetal, young, and adult brains [122].

3. The Endocannabinoid System as a Target for Neuroprotection in Hypoxic–Ischemic Encephalopathy

Perinatal HI leading to neonatal encephalopathy (NE) represents a major cause of death and long-term disability in neonates [134]. Each year, up to 20,000 infants are affected by NE in Europe and even more in regions with a lower level of perinatal care [135]. Whereas the incidence of NE in Western Europe and North America is around 1.6/1000 term births [136], neonatal mortality is 6 times higher in developing or low-resourced countries compared with developed or middle-to-high-resourced countries.
Current treatment options for HI are extremely limited, making the management of long-term outcomes or its prevention difficult. Actually, the only approved therapy is therapeutic hypothermia, consisting in lowering the body temperature of patients to 33.5 °C for 72 h through cooling of either the whole body or just the head [137]. Therapeutic hypothermia is routinely implemented in the majority of first-world hospitals to treat term infants with moderate to severe NE; however, cooling is only partially effective as a neuroprotective therapy (>45% of infants have adverse neurodevelopmental outcomes despite treatment) [138]. At the same time, hypothermia can develop some potential side effects due to the slowing of the mechanisms of clearance and metabolism, the induced immunosuppressive activity, and the increase in energy expenditure resulting from the thermoregulatory response [139]. As the current cooling therapy protocols appear to be optimal [140], there is an urgent need to improve neonatal neuroprotection by developing additional safe and effective neuroprotective treatments [141,142].
The EC system is able to limit the deleterious effects caused by multiple toxic stimuli such as glutamate excitotoxicity, oxidative stress, and inflammation, thus providing neuroprotection in different paradigms of brain injury [84,143]. Therefore, compounds that modulate the EC system could be promising neuroprotective and/or neurogenic agents for the treatment of CNS pathologies, including NE.
The first cannabinoid tested in cerebral ischemic models was the synthetic CB1/CB2 agonist (R)-(+)-WIN-55,212-2 [144]. The authors showed that the exogenous administration of this CB agonist significantly reduced the infarct volume and the loss of hippocampal neurons. They also studied the neuroprotective effect of cannabinoids during brain development and showed that exogenous administration of the ECs AEA and 2-AG reduced brain infarction in newborn rats subjected to HI [145]. Later, some of the co-authors described the neuroprotective and long-lasting beneficial effect of URB602, an inhibitor of the degradation of 2-AG [101] in the same murine model. The neuroprotective effect of cannabinoids was also confirmed in an experimental model closer to the human condition, i.e., in the fetal lamb. In this model, the synthetic cannabinoid agonist (R)-(+)-WIN-55,212-2 protected the neonatal brain at very low doses to maintain mitochondrial integrity and functionality [146], to reduce apoptotic cell death [147], and to ameliorate the inflammatory response [148].
The classical way to modulate the EC system is through the activation or blockade of CB1 and CB2 receptors, as described in the first part of this review. However, CB1 receptors seem to play a dual role in post-ischemic neuronal damage, as the decrease in glutamate release due to CB1 activation is accompanied by a parallel decrease in GABA release, resulting in neurotoxicity instead of neuroprotection [149]. Moreover, CB1 overactivation in the perinatal period could be harmful [150] and this can limit the translational interest of CB1 agonists. In addition, CB1-mediated psychoactive effects [151], which are unwanted in clinical treatments, should also be considered.
Activation of the other CB receptor, the CB2, results in potent anti-inflammatory effects [143], and the CB2 antagonism has no described beneficial effect. A therapeutic approach with drugs interacting with CB2 receptors can be developed using either indirect (e.g., cannabidiol) or selective (e.g., GW405833) CB2 agonists. Nevertheless, cannabidiol can induce severe hypotension [152] despite being neuroprotective in different experimental paradigms [153], whereas GW405833 showed no protection after HI [154]. This evidence together with the finding that the CB1 antagonist/inverse agonist rimonabant also exerts a neuroprotective effect, which adds further complexity to the effect of cannabinoid-interacting compounds in neurodegeneration.
Recently, some of the co-authors evaluated the neuroprotective potential of the synthetic cannabinoid URB447 [85]. URB447 is the first mixed CB1 antagonist and CB2 agonist that binds to both CB1 and CB2 receptors with submicromolar affinity and good stereoselectivity [91]. URB447 strongly reduced brain injury when administered before HI in neonatal rats, but more interestingly, the compound was effective also when administered 30 min or 3 h after the initial insult. URB447 reduced cerebral infarction by 95.7% (30 min) and 88% (at 3 h) in the whole ipsilateral (damaged) hemisphere.
Since a pharmacological intervention within 3 h after the injury is considered a clinically feasible therapeutic window to treat perinatal brain injury in humans [155], we characterized the effect of URB447 administered at this time point, focusing on the consequences of HI and URB447 administration on the activation of glial cells and white matter injury. Together with a reduction in astrogliosis and microglial activation, URB447 decreased white matter damage restoring myelin basic protein levels 7 days after HI, confirming the important role played by the EC system in the neurodegenerative and neuroreparative processes after HI.
As commented above, nowadays, the only clinical therapy against HI-induced NE is moderate hypothermia, which exerts a number of neuroprotective responses through the reduction in excitotoxicity, free radical exposure, blood–brain barrier dysfunction, and delayed cell death [156]. Leker et al. [157] observed that a single injection of the CB1 synthetic agonist HU-210 significantly reduced body temperature, conferring a strong neuroprotective effect to the hypoxic–ischemic rats, a beneficial effect that was lost when animals were treated with the selective CB1 antagonist SR141716. The enhancement of hypothermia by stimulating the EC system or by the combined therapy EC system plus hypothermia may have beneficial outcomes in neonates, so these responses are currently under investigation in preclinical models [158,159].

4. Can Endocannabinoid System Interacting Drugs Modulate Neurogenesis after HI?

The discovery of stem cells in the postnatal and adult mammalian brain changed the previously believed assertion that the adult brain is unable to replace lost neurons [160,161]. Although still unknown with certainty in other regions of the CNS, two neurogenic areas persist after birth: the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus of the hippocampus [162,163,164].
The ability to generate new neurons and glial cells from these niches may contribute to the plasticity of the newborn brain and tissue remodeling after damage [165,166,167,168,169]. Based on their regenerative potential, cells from the SVZ of the ventricles can be molecularly manipulated in situ to induce their proliferation and migration to damaged sites or stimulated in vitro for later transplantation [162,170,171,172,173]. However, the processes of proliferation, migration, differentiation, and survival will depend on a wide range of factors, including the type, intensity, duration, and/or location of the damage [174]. Thus, it is not yet known whether these newly formed neurons are properly integrated into the existing neural network and if they can represent a fully functional microenvironment after a brain injury [175]. It has been estimated that 85% of the new neurons generated in response to the insult do not survive after reaching maturation [176].
The global damage induced by perinatal HI may also affect the neurogenic niches and their neuro-proliferative capacity. After 24–48 h from moderate/severe hypoxic–ischemic damage, the SVZ may show extensive cell death, primarily affecting neuronal stem cells and also oligodendrocyte progenitors [177,178]. Interestingly, the neurogenic potential of this area can be affected independently from cell death [179]. Indeed, in a preclinical model close to the human condition, i.e., newborn piglets, it has been shown that a decreased cellularity is associated with a reduction in cell proliferation and neurogenesis in the SVZ [179]. These effects occurred without necrotic or apoptotic cell death 48 h after hypoxic–ischemic damage. Whether this discrepancy could be related to differences in the severity/duration of the insult or the experimental model employed (rodent vs. piglet) remains the subject of investigation. It should also be considered that the SVZ can present sub-regional sensitivity, with areas and cell types showing selective vulnerability to the insult. Higher rates of survival were observed in its medial zone [180] and in different responses of pre-oligodendrocytes and neuroblasts to hypoxic–ischemic damage [181].
Whereas several works pointed toward HI leading to decreased cell proliferation in the SVZ (for a review, see [182]), other authors have described that the injured ipsilateral SVZ has the ability to increase its size after a longer recovery interval [176,178,183,184], a phenomenon attributed to increased cell proliferation [176,185]. For its part, the undamaged contralateral SVZ can also suffer an expansion after HI [176], with the most undifferentiated precursors being responsible for this increase in its size [185].
The other neurogenic niche, the SGZ of the dentate gyrus of the hippocampus, also revealed conflicting results. Bartley et al. [186] showed that neuronal (together with microglial and endothelial) cell proliferation was significantly increased in the injured ipsilateral hippocampus. The authors used a postnatal day 7 (P7) neonatal mice model subjected to permanent unilateral carotid ligation plus 8% hypoxia for 75 min. Conversely, early after the publication of that work, Kadam et al. [187] described that total counts of new cells were significantly lower in both ipsilateral and contralateral hippocampi, which in turn correlated with lesion-induced atrophy. They used, however, a neonatal stroke model of unilateral carotid ligation alone to produce infarcts in P12 CD1 mice. In a more recent work by Ziemka-Nalecz et al. [188] using the HI permanent unilateral carotid ligation plus hypoxia (7.6% O2 for 60 min) model of brain injury, the authors showed no signs of increased or decreased cell proliferation in the dentate gyrus of the hippocampi, with no differences between sham (non-operated), ipsilateral, or contralateral hippocampi, in none of the five timepoints evaluated (3, 6, 9, 11, or 14 days after HI). This usage of different experimental models of brain injury may add complexity to unraveling the neurogenic response of the neonatal hippocampal SGZ.
To better understand the modulation in cellular populations after HI, experiments have been carried out using flow cytometry with multi-markers in order to quantify the proportion of each cell type. It seems that neuronal stem cells decrease while multipotential, as well as the glial cell progenitors, increase [189]. The increase in the number of reactive astrocytes can be translated into greater production of components of the extracellular matrix, such as hyaluronic acid and chondroitin sulfate, which can, in turn, inhibit the differentiation of oligodendrocytes and limit myelin synthesis [190,191]. This suggests that hypoxia–ischemia may alter the cellular composition of the neurogenic niches [189].
As described, the effect of neonatal HI on the neurogenic response after brain injury remains far from clear [179]. The modulation of the proliferative capacity of the neurogenic niches might be enhanced by using CBs, as the EC system seems to play an important role in processes, such as cell proliferation and differentiation of neural stem cells, during normal brain development.
Aguado et al. [192] observed that stimulation of the EC system enhanced neurogenesis after kainic acid-induced excitotoxicity in neural progenitor cell cultures. The effect was revealed as increased expression of the progenitor markers nestin, Sox-2, and musashi-1, and also as a higher proliferation rate. They also examined whether neural progenitor cell division may result in effective neurogenesis. By immunostaining dividing cells with the mature neuron marker NeuN, the authors described the presence of newly generated neurons one month after injury, suggesting that long-term neurogenesis can be enhanced by EC system modulation [192].
In a model of HI in rodents, Fernández-López et al. [193] stimulated the CB1 receptor by exogenous administration of the synthetic cannabinoid (R)-(+)-WIN-55,212-2, showing increased cell proliferation and doublecortin expression (a marker of neuroblasts) after HI and cannabinoid administration. However, the long-lasting effect described by Aguado et al. [192] here was lost. (R)-(+)-WIN-55,212-2 was able to promote neurogenesis up to 7 days after HI (P14), but the survival of the new neurons decreased shortly after the withdrawal of the treatment. It remains unclear whether prolonging the administration of cannabinoids could be beneficial regarding neurogenesis [193].
Therapeutic hypothermia (the only clinical therapy against HI-induced neonatal encephalopathy) is also able to modulate and enhance endogenous reparative processes. Bregy et al. [194] showed an increase in doublecortin-positive cells in the hippocampal dentate gyrus of cooled animals treated with therapeutic hypothermia after experimental traumatic brain injury. Works using models of ischemic and hypoxic–ischemic brain injury described similar results [195,196]. Rats treated with hypothermia increased their counts of neurogenesis markers compared to normothermic animals. As the activation of the EC system may decrease body temperature, it seems feasible that exogenous administration of CBs may indirectly modulate the neurogenic response after neonatal brain damage.
In addition to neurogenesis, cannabinoids may be of great benefit in white matter recovery after brain damage. The administration of the CB1 agonist ACEA resulted in increased Olig2 (an oligodendrocyte progenitor marker) expressing cells in the SVZ andmyelination in the subcortical white matter [197]. (R)-(+)-WIN-55,212-2 administration after HI also promoted remyelination of the injured external capsule by increasing the number of early oligodendrocyte progenitors and mature oligodendrocytes [193]. The enhancement of oligodendrogenesis is of great interest when treating the developing brain, as increased remyelination is linked with the improvement of sensorimotor functions after hypoxic–ischemic injury [198].

5. Concluding Remarks and Perspectives

The ubiquitous lipid signaling-based EC system is involved in outstanding regulatory functions throughout the human body, including neural development under physiological conditions and neuroprotection, and repair after pathophysiological processes.
In the context of neonatal brain injury, the administration of endogenous or exogenous CBs, or the blockage of EC degradation, has revealed a strong neuroprotective response in different preclinical models after HI. Similarly, the possibility of tissue repair in the developing brain by enhancing the proliferative potential of the SVZ and SGZ neurogenic niches is currently under active investigation. Selective modulation of the EC system in the sites of damage by targeting the enzymes responsible for EC degradation may represent an important therapeutic approach in order to avoid non-desired widespread effects.
Despite the clinical use of CB-related drugs that must be taken with caution, the modulation of the EC system to ameliorate the neurological consequences after neonatal HI is currently an exciting field of research with enormous possibilities for clinical translation.

Author Contributions

Conceptualization A.D. and D.A.-A.; writing—original draft preparation A.D., G.B., A.Á., M.S., S.C., W.B. and D.A.-A.; writing—review and editing, A.D., W.B. and D.A.-A.; supervision A.D. and D.A.-A. All authors have read and agreed to the published version of the manuscript.

Funding

EITB Maratoia-BIOEF (BIO18/IC/003), the Spanish Ministry of Science and Innovation: (MINECOR20/P66/AEI/10.13039/501100011033).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lu, H.-C.; Mackie, K. An introduction to the endogenous cannabinoid system. Biol. Psychiatry 2016, 79, 516–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Piomelli, D.; Mabou Tagne, A. Endocannabinoid-based therapies. Annu. Rev. Pharmacol. Toxicol. 2022, 62, 483–507. [Google Scholar] [CrossRef] [PubMed]
  3. Lowe, H.; Toyang, N.; Steele, B.; Bryant, J.; Ngwa, W. The endocannabinoid system: A potential target for the treatment of various diseases. Int. J. Mol. Sci. 2021, 22, 9472. [Google Scholar] [CrossRef]
  4. Rossi, F.; Tortora, C.; Argenziano, M.; Di Paola, A.; Punzo, F. Cannabinoid receptor type 2: A possible target in SARS-CoV-2 (CoV-19) infection? Int. J. Mol. Sci. 2020, 21, 3809. [Google Scholar] [CrossRef] [PubMed]
  5. Kim, S.H.; Won, S.J.; Mao, X.O.; Jin, K.; Greenberg, D.A. Molecular mechanisms of cannabinoid protection from neuronal excitotoxicity. Mol. Pharmacol. 2006, 69, 691–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Waksman, Y.; Olson, J.M.; Carlisle, S.J.; Cabral, G.A. The central cannabinoid receptor (CB1) mediates inhibition of nitric oxide production by rat microglial cells. J. Pharmacol. Exp. Ther. 1999, 288, 1357–1366. [Google Scholar]
  7. Walter, L.; Stella, N. Cannabinoids and neuroinflammation. Br. J. Pharmacol. 2004, 141, 775–785. [Google Scholar] [CrossRef] [Green Version]
  8. Devane, W.A.; Dysarz, F.A.; Johnson, M.R.; Melvin, L.S.; Howlett, A.C. Determination and characterization of a cannabinoid receptor in rat brain. Mol. Pharmacol. 1988, 34, 605–613. [Google Scholar]
  9. Matsuda, L.A.; Lolait, S.J.; Brownstein, M.J.; Young, A.C.; Bonner, T.I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990, 346, 561–564. [Google Scholar] [CrossRef]
  10. Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef]
  11. Lutz, B. Neurobiology of cannabinoid receptor signaling. Dialogues Clin. Neurosci. 2020, 22, 207–222. [Google Scholar] [CrossRef] [PubMed]
  12. Ren, S.; Wang, Z.; Zhang, Y.; Chen, N. Potential application of endocannabinoid system agents in neuropsychiatric and neurodegenerative diseases—Focusing on FAAH/MAGL inhibitors. Acta Pharmacol. Sin. 2020, 41, 1263–1271. [Google Scholar] [CrossRef] [PubMed]
  13. UNODC. Early Warning Advisory on New Psychoactive Substances. What are NPS? December 2021. Available online: https://www.unodc.org/LSS/Page/NPS (accessed on 16 November 2022).
  14. Auwärter, V.; Dresen, S.; Weinmann, W.; Müller, M.; Pütz, M.; Ferreirós, N. ‘Spice’ and other herbal blends: Harmless incense or cannabinoid designer drugs? J. Mass Spectrom. 2009, 44, 832–837. [Google Scholar] [CrossRef] [PubMed]
  15. An, D.; Peigneur, S.; Hendrickx, L.A.; Tytgat, J. Targeting cannabinoid receptors: Current status and prospects of natural products. Int. J. Mol. Sci. 2020, 21, 5064. [Google Scholar] [CrossRef] [PubMed]
  16. Deventer, M.H.; Van Uytfanghe, K.; Vinckier, I.M.J.; Reniero, F.; Guillou, C.; Stove, C.P. Cannabinoid receptor activation potential of the next generation, generic ban evading OXIZID synthetic cannabinoid receptor agonists. Drug Test. Anal. 2022, 14, 1565–1575. [Google Scholar] [CrossRef]
  17. Markham, J.; Sparkes, E.; Boyd, R.; Chen, S.; Manning, J.J.; Finlay, D.; Lai, F.; McGregor, E.; Maloney, C.J.; Gerona, R.R.; et al. Defining steric requirements at CB1 and CB2 cannabinoid receptors using synthetic cannabinoid receptor agonists 5F-AB-PINACA, 5F-ADB-PINACA, PX-1, PX-2, NNL-1, and their analogues. ACS Chem. Neurosci. 2022, 13, 1281–1295. [Google Scholar] [CrossRef]
  18. Van Sickle, M.D.; Duncan, M.; Kingsley, P.J.; Mouihate, A.; Urbani, P.; Mackie, K.; Stella, N.; Makriyannis, A.; Piomelli, D.; Davison, J.S.; et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science 2005, 310, 329–332. [Google Scholar] [CrossRef] [Green Version]
  19. Shahbazi, F.; Grandi, V.; Banerjee, A.; Trant, J.F. Cannabinoids and cannabinoid receptors: The story so far. iScience 2020, 23, 101301. [Google Scholar] [CrossRef]
  20. Tang, X.; Liu, Z.; Li, X.; Wang, J.; Li, L. Cannabinoid receptors in myocardial injury: A brother born to rival. Int. J. Mol. Sci. 2021, 22, 6886. [Google Scholar] [CrossRef]
  21. Felder, C.C.; Joyce, K.E.; Briley, E.M.; Mansouri, J.; Mackie, K.; Blond, O.; Lai, Y.; Ma, A.L.; Mitchell, R.L. Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors. Mol. Pharmacol. 1995, 48, 443–450. [Google Scholar]
  22. de Almeida, D.L.; Devi, L.A. Diversity of molecular targets and signaling pathways for CBD. Pharmacol. Res. Perspect. 2020, 8, e00682. [Google Scholar] [CrossRef]
  23. Piomelli, D. The molecular logic of endocannabinoid signalling. Nat. Rev. Neurosci. 2003, 4, 873–884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Augustin, S.M.; Lovinger, D.M. Functional relevance of endocannabinoid-dependent synaptic plasticity in the central nervous system. ACS Chem. Neurosci. 2018, 9, 2146–2161. [Google Scholar] [CrossRef]
  25. Alger, B.E. Retrograde signaling in the regulation of synaptic transmission: Focus on endocannabinoids. Prog. Neurobiol. 2002, 68, 247–286. [Google Scholar] [CrossRef] [PubMed]
  26. Diana, M.A.; Marty, A. Endocannabinoid-mediated short-term synaptic plasticity: Depolarization-induced suppression of inhibition (DSI) and depolarization-induced suppression of excitation (DSE). Br. J. Pharmacol. 2004, 142, 9–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Hohmann, A.G.; Suplita, R.L.; Bolton, N.M.; Neely, M.H.; Fegley, D.; Mangieri, R.; Krey, J.F.; Walker, J.M.; Holmes, P.V.; Crystal, J.D.; et al. An endocannabinoid mechanism for stress-induced analgesia. Nature 2005, 435, 1108–1112. [Google Scholar] [CrossRef] [Green Version]
  28. Castillo, P.E.; Younts, T.J.; Chávez, A.E.; Hashimotodani, Y. Endocannabinoid signaling and synaptic function. Neuron 2012, 76, 70–81. [Google Scholar] [CrossRef] [Green Version]
  29. Puente, N.; Cui, Y.; Lassalle, O.; Lafourcade, M.; Georges, F.; Venance, L.; Grandes, P.; Manzoni, O.J. Polymodal activation of the endocannabinoid system in the extended amygdala. Nat. Neurosci. 2011, 14, 1542–1547. [Google Scholar] [CrossRef]
  30. Estrada, J.A.; Contreras, I. Endocannabinoid receptors in the CNS: Potential drug targets for the prevention and treatment of neurologic and psychiatric disorders. Curr. Neuropharmacol. 2020, 18, 769–787. [Google Scholar] [CrossRef]
  31. Devane, W.A.; Hanus, L.; Breuer, A.; Pertwee, R.G.; Stevenson, L.A.; Griffin, G.; Gibson, D.; Mandelbaum, A.; Etinger, A.; Mechoulam, R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992, 258, 1946–1949. [Google Scholar] [CrossRef]
  32. Mechoulam, R.; Ben-Shabat, S.; Hanus, L.; Ligumsky, M.; Kaminski, N.E.; Schatz, A.R.; Gopher, A.; Almog, S.; Martin, B.R.; Compton, D.R. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem. Pharmacol. 1995, 50, 83–90. [Google Scholar] [CrossRef] [PubMed]
  33. Sugiura, T.; Kondo, S.; Sukagawa, A.; Nakane, S.; Shinoda, A.; Itoh, K.; Yamashita, A.; Waku, K. 2-Arachidonoylglycerol: A possible endogenous cannabinoid receptor ligand in brain. Biochem. Biophys. Res. Commun. 1995, 215, 89–97. [Google Scholar] [CrossRef]
  34. Hillard, C.J.; Campbell, W.B. Biochemistry and pharmacology of arachidonylethanolamide, a putative endogenous cannabinoid. J. Lipid Res. 1997, 38, 2383–2398. [Google Scholar] [CrossRef] [PubMed]
  35. Vogel, Z.; Barg, J.; Levy, R.; Saya, D.; Heldman, E.; Mechoulam, R. Anandamide, a brain endogenous compound, interacts specifically with cannabinoid receptors and inhibits adenylate cyclase. J. Neurochem. 1993, 61, 352–355. [Google Scholar] [CrossRef]
  36. Mackie, K.; Devane, W.A.; Hille, B. Anandamide, an endogenous cannabinoid, inhibits calcium currents as a partial agonist in N18 neuroblastoma cells. Mol. Pharmacol. 1993, 44, 498–503. [Google Scholar]
  37. Facci, L.; Dal Toso, R.; Romanello, S.; Buriani, A.; Skaper, S.D.; Leon, A. Mast cells express a peripheral cannabinoid receptor with differential sensitivity to anandamide and palmitoylethanolamide. Proc. Natl. Acad. Sci. USA 1995, 92, 3376–3380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Bayewitch, M.; Avidor-Reiss, T.; Levy, R.; Barg, J.; Mechoulam, R.; Vogel, Z. The peripheral cannabinoid receptor: Adenylate cyclase inhibition and G protein coupling. FEBS Lett. 1995, 375, 143–147. [Google Scholar] [CrossRef] [Green Version]
  39. Sugiura, T.; Waku, K. 2-Arachidonoylglycerol and the cannabinoid receptors. Chem. Phys. Lipids 2000, 108, 89–106. [Google Scholar] [CrossRef]
  40. Di Marzo, V.; Fontana, A.; Cadas, H.; Schinelli, S.; Cimino, G.; Schwartz, J.C.; Piomelli, D. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 1994, 372, 686–691. [Google Scholar] [CrossRef] [Green Version]
  41. Cadas, H.; di Tomaso, E.; Piomelli, D. Occurrence and biosynthesis of endogenous cannabinoid precursor, N-arachidonoyl phosphatidylethanolamine, in rat brain. J. Neurosci. 1997, 17, 1226–1242. [Google Scholar] [CrossRef] [Green Version]
  42. Farooqui, A.A.; Rammohan, K.W.; Horrocks, L.A. Isolation, characterization, and regulation of diacylglycerol lipases from the bovine brain. Ann. N. Y. Acad. Sci. 1989, 559, 25–36. [Google Scholar] [CrossRef] [PubMed]
  43. Bisogno, T.; Howell, F.; Williams, G.; Minassi, A.; Cascio, M.G.; Ligresti, A.; Matias, I.; Schiano-Moriello, A.; Paul, P.; Williams, E.-J.; et al. Cloning of the first Sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J. Cell Biol. 2003, 163, 463–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Desarnaud, F.; Cadas, H.; Piomelli, D. Anandamide amidohydrolase activity in rat brain microsomes: Identification and partial characterization. J. Biol. Chem. 1995, 270, 6030–6035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hillard, C.J.; Wilkison, D.M.; Edgemond, W.S.; Campbell, W.B. Characterization of the kinetics and distribution of N-Arachidonylethanolamine (Anandamide) hydrolysis by rat brain. Biochim. Biophys. Acta Lipids Lipid Metab. 1995, 1257, 249–256. [Google Scholar] [CrossRef] [PubMed]
  46. Ueda, N.; Kurahashi, Y.; Yamamoto, S.; Tokunaga, T. Partial purification and characterization of the porcin brain enzyme hydrolyzing and synthesizing anandamide. J. Biol. Chem. 1995, 270, 23823–23827. [Google Scholar] [CrossRef] [Green Version]
  47. Cravatt, B.F.; Giang, D.K.; Mayfield, S.P.; Boger, D.L.; Lerner, R.A.; Gilula, N.B. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature 1996, 384, 83–87. [Google Scholar] [CrossRef] [PubMed]
  48. Tornqvist, H.; Belfrage, P. Purification and some properties of a monoacylglycerol hydrolyzing enzyme of rat adipose tissue. J. Biol. Chem. 1976, 251, 813–819. [Google Scholar] [CrossRef]
  49. Prescott, S.M.; Majerus, P.W. Characterization of 1,2-diacylglycerol hydrolysis in human platelets. Demonstration of an arachidonoyl-monoacylglycerol intermediate. J. Biol. Chem. 1983, 258, 764–769. [Google Scholar] [CrossRef]
  50. Farooqui, A.A.; Taylor, W.A.; Horrocks, L.A. Separation of bovine brain mono- and diacylglycerol lipases by heparin sepharose affinity chromatography. Biochem. Biophys. Res. Commun. 1984, 122, 1241–1246. [Google Scholar] [CrossRef]
  51. Deutsch, D.G.; Chin, S.A. Enzymatic synthesis and degradation of anandamide, a cannabinoid receptor agonist. Biochem. Pharmacol. 1993, 46, 791–796. [Google Scholar] [CrossRef]
  52. Dinh, T.P.; Carpenter, D.; Leslie, F.M.; Freund, T.F.; Katona, I.; Sensi, S.L.; Kathuria, S.; Piomelli, D. Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc. Natl. Acad. Sci. USA 2002, 99, 10819–10824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Beltramo, M.; Stella, N.; Calignano, A.; Lin, S.Y.; Makriyannis, A.; Piomelli, D. Functional role of high affinity anandamide transport, as revelead by selective inhibition. Science 1997, 277, 1094–1097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hillard, C.J.; Edgemond, W.S.; Jarrahian, A.; Campbell, W.B. Accumulation of N-arachidonylethanolamine (anandamide) into cerebellar granule cells occurs via facilitated diffusion. J. Neurochem. 1997, 69, 631–638. [Google Scholar] [CrossRef] [PubMed]
  55. Beltramo, M.; Piomelli, D. Carrier-mediated Transport and enzymatic hydrolysis of the endogenous cannabinoid 2-arachidonoylglycerol. Neuroreport 2000, 11, 1231–1235. [Google Scholar] [CrossRef]
  56. Pagano, C.; Navarra, G.; Coppola, L.; Avilia, G.; Bifulco, M.; Laezza, C. Cannabinoids: Therapeutic use in clinical practice. Int. J. Mol. Sci. 2022, 23, 3344. [Google Scholar] [CrossRef]
  57. Kumar, P.; Mahato, D.K.; Kamle, M.; Borah, R.; Sharma, B.; Pandhi, S.; Tripathi, V.; Yadav, H.S.; Devi, S.; Patil, U.; et al. Pharmacological properties, therapeutic potential, and legal status of Cannabis sativa L.: An overview. Phytother. Res. 2021, 35, 6010–6029. [Google Scholar] [CrossRef]
  58. Gülck, T.; Møller, B.L. Phytocannabinoids: Origins and biosynthesis. Trends Plant Sci. 2020, 25, 985–1004. [Google Scholar] [CrossRef]
  59. Tarasov, P.; Bezrukova, E.; Karabanov, E.; Nakagawa, T.; Wagner, M.; Kulagina, N.; Letunova, P.; Abzaeva, A.; Granoszewski, W.; Riedel, F. Vegetation and climate dynamics during the holocene and eemian interglacials derived from lake baikal pollen records. Palaeogeogr. Palaeoclimatol. Palaeoecol. 2007, 252, 440–457. [Google Scholar] [CrossRef]
  60. Pisanti, S.; Bifulco, M. Medical cannabis: A plurimillennial history of an evergreen. J. Cell. Physiol. 2019, 234, 8342–8351. [Google Scholar] [CrossRef]
  61. Li, H.-L. The origin and use of cannabis in Eastern Asia linguistic-cultural implications. Econ. Bot. 1974, 28, 293–301. [Google Scholar] [CrossRef]
  62. Prandi, C.; Blangetti, M.; Namdar, D.; Koltai, H. Structure-activity relationship of cannabis derived compounds for the treatment of neuronal activity-related diseases. Molecules 2018, 23, 1526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gaoni, Y.; Mechoulam, R. Isolation, structure, and partial synthesis of an active constituent of hashish. J. Am. Chem. Soc. 1964, 86, 1646–1647. [Google Scholar] [CrossRef]
  64. Adams, R.; Hunt, M.; Clark, J.H. Structure of cannabidiol, a product isolated from the marihuana extract of Minnesota wild hemp. I. J. Am. Chem. Soc. 1940, 62, 196–200. [Google Scholar] [CrossRef]
  65. Morales, P.; Hurst, D.P.; Reggio, P.H. Molecular targets of the phytocannabinoids: A complex picture. Prog. Chem. Org. Nat. Prod. 2017, 103, 103–131. [Google Scholar] [CrossRef]
  66. Peng, J.; Fan, M.; An, C.; Ni, F.; Huang, W.; Luo, J. A narrative review of molecular mechanism and therapeutic effect of cannabidiol (CBD). Basic Clin. Pharmacol. Toxicol. 2022, 130, 439–456. [Google Scholar] [CrossRef]
  67. Hayakawa, K.; Mishima, K.; Fujiwara, M. Therapeutic potential of non-psychotropic cannabidiol in ischemic stroke. Pharmaceuticals 2010, 3, 2197–2212. [Google Scholar] [CrossRef] [Green Version]
  68. Dos Santos, R.G.; Hallak, J.E.C.; Crippa, J.A.S. Neuropharmacological effects of the main phytocannabinoids: A narrative review. Adv. Exp. Med. Biol. 2021, 1264, 29–45. [Google Scholar] [CrossRef]
  69. Stone, N.L.; Murphy, A.J.; England, T.J.; O’Sullivan, S.E. A systematic review of minor phytocannabinoids with promising neuroprotective potential. Br. J. Pharmacol. 2020, 177, 4330–4352. [Google Scholar] [CrossRef]
  70. Alves, V.L.; Gonçalves, J.L.; Aguiar, J.; Teixeira, H.M.; Câmara, J.S. The synthetic cannabinoids phenomenon: From structure to toxicological properties. a review. Crit. Rev. Toxicol. 2020, 50, 359–382. [Google Scholar] [CrossRef]
  71. Shafi, A.; Berry, A.J.; Sumnall, H.; Wood, D.M.; Tracy, D.K. New psychoactive substances: A review and updates. Ther. Adv. Psychopharmacol. 2020, 10, 2045125320967197. [Google Scholar] [CrossRef]
  72. Brown, J.D.; Rivera Rivera, K.J.; Hernandez, L.Y.C.; Doenges, M.R.; Auchey, I.; Pham, T.; Goodin, A.J. Natural and synthetic cannabinoids: Pharmacology, uses, adverse drug events, and drug interactions. J. Clin. Pharmacol. 2021, 61 (Suppl. 2), S37–S52. [Google Scholar] [CrossRef] [PubMed]
  73. Chung, E.Y.; Cha, H.J.; Min, H.K.; Yun, J. Pharmacology and adverse effects of new psychoactive substances: Synthetic cannabinoid receptor agonists. Arch. Pharm. Res. 2021, 44, 402–413. [Google Scholar] [CrossRef] [PubMed]
  74. Cinar, R.; Iyer, M.R.; Kunos, G. The therapeutic potential of second and third generation CB1R antagonists. Pharmacol. Ther. 2020, 208, 107477. [Google Scholar] [CrossRef] [PubMed]
  75. Sholler, D.J.; Huestis, M.A.; Amendolara, B.; Vandrey, R.; Cooper, Z.D. Therapeutic potential and safety considerations for the clinical use of synthetic cannabinoids. Pharmacol. Biochem. Behav. 2020, 199, 173059. [Google Scholar] [CrossRef] [PubMed]
  76. Coronado-Álvarez, A.; Romero-Cordero, K.; Macías-Triana, L.; Tatum-Kuri, A.; Vera-Barrón, A.; Budde, H.; Machado, S.; Yamamoto, T.; Imperatori, C.; Murillo-Rodríguez, E. The synthetic CB1 cannabinoid receptor selective agonists: Putative medical uses and their legalization. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 110, 110301. [Google Scholar] [CrossRef]
  77. Saldaña-Shumaker, S.L.; Grenning, A.J.; Cunningham, C.W. Modern approaches to the development of synthetic cannabinoid receptor probes. Pharmacol. Biochem. Behav. 2021, 203, 173119. [Google Scholar] [CrossRef]
  78. Manning, J.J.; Green, H.M.; Glass, M.; Finlay, D.B. Pharmacological selection of cannabinoid receptor effectors: Signalling, allosteric modulation and bias. Neuropharmacology 2021, 193, 108611. [Google Scholar] [CrossRef]
  79. Leo, L.M.; Abood, M.E. CB1 Cannabinoid receptor signaling and biased signaling. Molecules 2021, 26, 5413. [Google Scholar] [CrossRef]
  80. Manera, C.; Bertini, S. Cannabinoid-based medicines and Multiple Sclerosis. Adv. Exp. Med. Biol. 2021, 1264, 111–129. [Google Scholar] [CrossRef]
  81. Products (Outside US). Jazz Pharmaceuticals. Available online: https://www.jazzpharma.com/medicines/our-medicines/ (accessed on 29 July 2022).
  82. Landucci, E.; Scartabelli, T.; Gerace, E.; Moroni, F.; Pellegrini-Giampietro, D.E. CB1 receptors and post-ischemic brain damage: Studies on the toxic and neuroprotective effects of cannabinoids in rat organotypic hippocampal slices. Neuropharmacology 2011, 60, 674–682. [Google Scholar] [CrossRef]
  83. Benyó, Z.; Ruisanchez, É.; Leszl-Ishiguro, M.; Sándor, P.; Pacher, P. Endocannabinoids in cerebrovascular regulation. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H785–H801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Sagredo, O.; Palazuelos, J.; Gutierrez-Rodriguez, A.; Satta, V.; Galve-Roperh, I.; Martínez-Orgado, J. Cannabinoid signalling in the immature brain: Encephalopathies and neurodevelopmental disorders. Biochem. Pharmacol. 2018, 157, 85–96. [Google Scholar] [CrossRef] [PubMed]
  85. Carloni, S.; Crinelli, R.; Palma, L.; Álvarez, F.J.; Piomelli, D.; Duranti, A.; Balduini, W.; Alonso-Alconada, D. The synthetic cannabinoid URB447 reduces brain injury and the associated white matter demyelination after hypoxia-ischemia in neonatal rats. ACS Chem. Neurosci. 2020, 11, 1291–1299. [Google Scholar] [CrossRef] [PubMed]
  86. Melvin, L.S.; Johnson, M.R. Structure-activity relationships of tricyclic and nonclassical bicyclic cannabinoids. NIDA Res. Monogr. 1987, 79, 31–34. [Google Scholar]
  87. Pacheco, M.; Childers, S.R.; Arnold, R.; Casiano, F.; Ward, S.J. Aminoalkylindoles: Actions on specific g-protein-linked receptors. J. Pharmacol. Exp. Ther. 1991, 257, 170–183. [Google Scholar] [PubMed]
  88. Rinaldi-Carmona, M.; Barth, F.; Héaulme, M.; Shire, D.; Calandra, B.; Congy, C.; Martinez, S.; Maruani, J.; Néliat, G.; Caput, D. SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett. 1994, 350, 240–244. [Google Scholar] [CrossRef] [Green Version]
  89. Lan, R.; Makriyannis, A.; Gatley, S.J. Preparation of iodine-123 labeled AM251: A potential SPECT radioligand for the brain cannabinoid CB1 receptor. J. Label. Compd. Radiopharm. 1996, 38, 875–882. [Google Scholar] [CrossRef]
  90. Felder, C.C.; Joyce, K.E.; Briley, E.M.; Glass, M.; Mackie, K.P.; Fahey, K.J.; Cullinan, G.J.; Hunden, D.C.; Johnson, D.W.; Chaney, M.O.; et al. LY320135, a novel cannabinoid CB1 receptor antagonist, unmasks coupling of the CB1 receptor to stimulation of cAMP accumulation. J. Pharmacol. Exp. Ther. 1998, 284, 291–297. [Google Scholar]
  91. LoVerme, J.; Duranti, A.; Tontini, A.; Spadoni, G.; Mor, M.; Rivara, S.; Stella, N.; Xu, C.; Tarzia, G.; Piomelli, D. Synthesis and characterization of a peripherally restricted CB1 cannabinoid antagonist, URB447, that reduces feeding and body-weight gain in mice. Bioorg. Med. Chem. Lett. 2009, 19, 639–643. [Google Scholar] [CrossRef] [Green Version]
  92. De Luca, M.A.; Fattore, L. Therapeutic use of synthetic cannabinoids: Still an open issue? Clin. Ther. 2018, 40, 1457–1466. [Google Scholar] [CrossRef] [Green Version]
  93. Cravatt, B.F.; Demarest, K.; Patricelli, M.P.; Bracey, M.H.; Giang, D.K.; Martin, B.R.; Lichtman, A.H. Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc. Natl. Acad. Sci. USA 2001, 98, 9371–9376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lichtman, A.H.; Shelton, C.C.; Advani, T.; Cravatt, B.F. Mice lacking fatty acid amide hydrolase exhibit a cannabinoid receptor-mediated phenotypic hypoalgesia. Pain 2004, 109, 319–327. [Google Scholar] [CrossRef] [PubMed]
  95. Kathuria, S.; Gaetani, S.; Fegley, D.; Valiño, F.; Duranti, A.; Tontini, A.; Mor, M.; Tarzia, G.; La Rana, G.; Calignano, A.; et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat. Med. 2003, 9, 76–81. [Google Scholar] [CrossRef]
  96. Tuo, W.; Leleu-Chavain, N.; Spencer, J.; Sansook, S.; Millet, R.; Chavatte, P. Therapeutic potential of fatty acid amide hydrolase, monoacylglycerol lipase, and N-acylethanolamine acid amidase inhibitors. J. Med. Chem. 2017, 60, 4–46. [Google Scholar] [CrossRef] [PubMed]
  97. Tripathi, R.K.P. A perspective review on fatty acid amide hydrolase (FAAH) inhibitors as potential therapeutic agents. Eur. J. Med. Chem. 2020, 188, 111953. [Google Scholar] [CrossRef] [PubMed]
  98. Van Egmond, N.; Straub, V.M.; van der Stelt, M. Targeting endocannabinoid signaling: FAAH and MAG lipase inhibitors. Annu. Rev. Pharmacol. Toxicol. 2021, 61, 441–463. [Google Scholar] [CrossRef]
  99. Abhishek, K.; Suresh, K.; Rohit, D. A review on structurally diversified synthesized molecules as monoacyl-glycerol lipase inhibitors and their therapeutic uses. Curr. Drug Res. Rev. 2022, 14, 96–115. [Google Scholar] [CrossRef]
  100. Wang, D.-P.; Jin, K.-Y.; Zhao, P.; Lin, Q.; Kang, K.; Hai, J. Neuroprotective effects of VEGF-A nanofiber membrane and FAAH inhibitor URB597 against oxygen-glucose deprivation-induced ischemic neuronal injury. Int. J. Nanomedicine 2021, 16, 3661–3678. [Google Scholar] [CrossRef]
  101. Carloni, S.; Alonso-Alconada, D.; Girelli, S.; Duranti, A.; Tontini, A.; Piomelli, D.; Hilario, E.; Alvarez, A.; Balduini, W. Pretreatment with the monoacylglycerol lipase inhibitor URB602 protects from the long-term consequences of neonatal hypoxic–ischemic brain injury in rats. Pediatr. Res. 2012, 72, 400–406. [Google Scholar] [CrossRef] [Green Version]
  102. Choi, S.-H.; Arai, A.L.; Mou, Y.; Kang, B.; Yen, C.C.-C.; Hallenbeck, J.; Silva, A.C. Neuroprotective effects of MAGL (Monoacylglycerol Lipase) inhibitors in experimental ischemic stroke. Stroke 2018, 49, 718–726. [Google Scholar] [CrossRef]
  103. Piro, J.R.; Suidan, G.L.; Quan, J.; Pi, Y.; O’Neill, S.M.; Ilardi, M.; Pozdnyakov, N.; Lanz, T.A.; Xi, H.; Bell, R.D.; et al. Inhibition of 2-AG hydrolysis differentially regulates blood brain barrier permeability after injury. J. Neuroinflammation 2018, 15, 142. [Google Scholar] [CrossRef] [PubMed]
  104. Xiong, Y.; Yao, H.; Cheng, Y.; Gong, D.; Liao, X.; Wang, R. Effects of monoacylglycerol lipase inhibitor URB602 on lung ischemia-reperfusion injury in mice. Biochem. Biophys. Res. Commun. 2018, 506, 578–584. [Google Scholar] [CrossRef] [PubMed]
  105. Yamasaki, T.; Hatori, A.; Zhang, Y.; Mori, W.; Kurihara, Y.; Ogawa, M.; Wakizaka, H.; Rong, J.; Wang, L.; Liang, S.; et al. Neuroprotective effects of minocycline and KML29, a potent inhibitor of monoacylglycerol lipase, in an experimental stroke model: A small-animal positron emission tomography study. Theranostics 2021, 11, 9492–9502. [Google Scholar] [CrossRef] [PubMed]
  106. Mor, M.; Rivara, S.; Lodola, A.; Plazzi, P.V.; Tarzia, G.; Duranti, A.; Tontini, A.; Piersanti, G.; Kathuria, S.; Piomelli, D. Cyclohexylcarbamic acid 3′- or 4′-Substituted Biphenyl-3-yl esters as fatty acid amide hydrolase inhibitors: Synthesis, quantitative structure-activity relationships, and molecular modeling studies. J. Med. Chem. 2004, 47, 4998–5008. [Google Scholar] [CrossRef] [Green Version]
  107. Fegley, D.; Gaetani, S.; Duranti, A.; Tontini, A.; Mor, M.; Tarzia, G.; Piomelli, D. Characterization of the fatty acid amide hydrolase inhibitor cyclohexyl carbamic acid 3′-carbamoyl-biphenyl-3-yl ester (URB597): Effects on anandamide and oleoylethanolamide deactivation. J. Pharmacol. Exp. Ther. 2005, 313, 352–358. [Google Scholar] [CrossRef] [Green Version]
  108. Gobbi, G.; Bambico, F.R.; Mangieri, R.; Bortolato, M.; Campolongo, P.; Solinas, M.; Cassano, T.; Morgese, M.G.; Debonnel, G.; Duranti, A.; et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc. Natl. Acad. Sci. USA. 2005, 102, 18620–18625. [Google Scholar] [CrossRef]
  109. Piomelli, D.; Tarzia, G.; Duranti, A.; Tontini, A.; Mor, M.; Compton, T.R.; Dasse, O.; Monaghan, E.P.; Parrott, J.A.; Putman, D. Pharmacological profile of the selective FAAH inhibitor KDS-4103 (URB597). CNS Drug Rev. 2006, 12, 21–38. [Google Scholar] [CrossRef] [Green Version]
  110. Russo, R.; Loverme, J.; La Rana, G.; Compton, T.R.; Parrott, J.; Duranti, A.; Tontini, A.; Mor, M.; Tarzia, G.; Calignano, A.; et al. The fatty acid amide hydrolase inhibitor URB597 (cyclohexylcarbamic acid 3′-carbamoylbiphenyl-3-yl ester) reduces neuropathic pain after oral administration in mice. J. Pharmacol. Exp. Ther. 2007, 322, 236–242. [Google Scholar] [CrossRef]
  111. Bortolato, M.; Mangieri, R.A.; Fu, J.; Kim, J.H.; Arguello, O.; Duranti, A.; Tontini, A.; Mor, M.; Tarzia, G.; Piomelli, D. Antidepressant-like activity of the fatty acid amide hydrolase inhibitor URB597 in a rat model of chronic mild stress. Biol. Psychiatry 2007, 62, 1103–1110. [Google Scholar] [CrossRef] [Green Version]
  112. Vacondio, F.; Silva, C.; Lodola, A.; Fioni, A.; Rivara, S.; Duranti, A.; Tontini, A.; Sanchini, S.; Clapper, J.R.; Piomelli, D.; et al. Structure-property relationships of a class of carbamate-based fatty acid amide hydrolase (FAAH) inhibitors: Chemical and biological stability. ChemMedChem 2009, 4, 1495–1504. [Google Scholar] [CrossRef] [Green Version]
  113. Bambico, F.R.; Duranti, A.; Nobrega, J.N.; Gobbi, G. The fatty acid amide hydrolase inhibitor URB597 modulates serotonin-dependent emotional behaviour, and serotonin1A and serotonin2A/C activity in the hippocampus. Eur. Neuropsychopharmacol. 2016, 26, 578–590. [Google Scholar] [CrossRef] [PubMed]
  114. Tarzia, G.; Duranti, A.; Tontini, A.; Piersanti, G.; Mor, M.; Rivara, S.; Plazzi, P.V.; Park, C.; Kathuria, S.; Piomelli, D. Design, synthesis, and structure-activity relationships of alkylcarbamic acid aryl esters, a new class of fatty acid amide hydrolase inhibitors. J. Med. Chem. 2003, 46, 2352–2360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Makara, J.K.; Mor, M.; Fegley, D.; Szabó, S.I.; Kathuria, S.; Astarita, G.; Duranti, A.; Tontini, A.; Tarzia, G.; Rivara, S.; et al. Selective inhibition of 2-AG hydrolysis enhances endocannabinoid signaling in hippocampus. Nat. Neurosci. 2005, 8, 1139–1141. [Google Scholar] [CrossRef] [PubMed]
  116. King, A.R.; Duranti, A.; Tontini, A.; Rivara, S.; Rosengarth, A.; Clapper, J.R.; Astarita, G.; Geaga, J.A.; Luecke, H.; Mor, M.; et al. URB602 inhibits monoacylglycerol lipase and selectively blocks 2-arachidonoylglycerol degradation in intact brain slices. Chem. Biol. 2007, 14, 1357–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Long, J.Z.; Li, W.; Booker, L.; Burston, J.J.; Kinsey, S.G.; Schlosburg, J.E.; Pavón, F.J.; Serrano, A.M.; Selley, D.E.; Parsons, L.H.; et al. Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nat. Chem. Biol. 2009, 5, 37–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Chang, J.W.; Niphakis, M.J.; Lum, K.M.; Cognetta, A.B., III; Wang, C.; Matthews, M.L.; Niessen, S.; Buczynski, M.W.; Parsons, L.H.; Cravatt, B.F. Highly selective inhibitors of monoacylglycerol lipase bearing a reactive group that is bioisosteric with endocannabinoid substrates. Chem. Biol. 2012, 19, 579–588. [Google Scholar] [CrossRef]
  119. Niphakis, M.J.; Cognetta, A.B.; Chang, J.W.; Buczynski, M.W.; Parsons, L.H.; Byrne, F.; Burston, J.J.; Chapman, V.; Cravatt, B.F. Evaluation of NHS carbamates as a potent and selective class of endocannabinoid hydrolase inhibitors. ACS Chem. Neurosci. 2013, 4, 1322–1332. [Google Scholar] [CrossRef] [Green Version]
  120. Gaffuri, A.-L.; Ladarre, D.; Lenkei, Z. Type-1 cannabinoid receptor signaling in neuronal development. Pharmacology 2012, 90, 19–39. [Google Scholar] [CrossRef]
  121. Berrendero, F.; Garcia-Gil, L.; Hernandez, M.L.; Romero, J.; Cebeira, M.; de Miguel, R.; Ramos, J.A.; Fernández-Ruiz, J.J. Localization of MRNA expression and activation of signal transduction mechanisms for cannabinoid receptor in rat brain during fetal development. Development 1998, 125, 3179–3188. [Google Scholar] [CrossRef]
  122. Berrendero, F.; Sepe, N.; Ramos, J.A.; Di Marzo, V.; Fernández-Ruiz, J.J. Analysis of cannabinoid receptor binding and mRNA expression and endogenous cannabinoid contents in the developing rat brain during late gestation and early postnatal period. Synapse 1999, 33, 181–191. [Google Scholar] [CrossRef]
  123. Romero, J.; Garcia-Palomero, E.; Berrendero, F.; Garcia-Gil, L.; Hernandez, M.L.; Ramos, J.A.; Fernández-Ruiz, J.J. Atypical location of cannabinoid receptors in white matter areas during rat brain development. Synapse 1997, 26, 317–323. [Google Scholar] [CrossRef]
  124. Buckley, N.E.; Hansson, S.; Harta, G.; Mezey, É. Expression of the CB1 and CB2 receptor messenger rnas during embryonic development in the rat. Neuroscience 1997, 82, 1131–1149. [Google Scholar] [CrossRef] [PubMed]
  125. Biegon, A.; Kerman, I.A. Autoradiographic study of pre- and postnatal distribution of cannabinoid receptors in human brain. NeuroImage 2001, 14, 1463–1468. [Google Scholar] [CrossRef] [PubMed]
  126. Mato, S.; Del Olmo, E.; Pazos, A. Ontogenetic development of cannabinoid receptor expression and signal transduction functionality in the human brain: Ontogeny of CB1 receptors in human brain. Eur. J. Neurosci. 2003, 17, 1747–1754. [Google Scholar] [CrossRef]
  127. Xapelli, S.; Agasse, F.; Sardà-Arroyo, L.; Bernardino, L.; Santos, T.; Ribeiro, F.F.; Valero, J.; Bragança, J.; Schitine, C.; de Melo Reis, R.A.; et al. Activation of type 1 cannabinoid receptor (CB1R) promotes neurogenesis in murine subventricular zone cell cultures. PLoS ONE 2013, 8, e63529. [Google Scholar] [CrossRef]
  128. Díaz-Alonso, J.; Guzmán, M.; Galve-Roperh, I. Endocannabinoids via CB1 receptors act as neurogenic niche cues during cortical development. Philos. Trans. R. Soc. B Biol. Sci. 2012, 367, 3229–3241. [Google Scholar] [CrossRef]
  129. Fernández-López, D.; Lizasoain, I.; Moro, M.; Martínez-Orgado, J. Cannabinoids: Well-suited candidates for the treatment of perinatal brain injury. Brain Sci. 2013, 3, 1043–1059. [Google Scholar] [CrossRef] [Green Version]
  130. Oudin, M.J.; Gajendra, S.; Williams, G.; Hobbs, C.; Lalli, G.; Doherty, P. Endocannabinoids regulate the migration of subventricular zone-derived neuroblasts in the postnatal brain. J. Neurosci. 2011, 31, 4000–4011. [Google Scholar] [CrossRef] [Green Version]
  131. Berghuis, P.; Rajnicek, A.M.; Morozov, Y.M.; Ross, R.A.; Mulder, J.; Urbán, G.M.; Monory, K.; Marsicano, G.; Matteoli, M.; Canty, A.; et al. Hardwiring the brain: Endocannabinoids shape neuronal connectivity. Science 2007, 316, 1212–1216. [Google Scholar] [CrossRef] [Green Version]
  132. Paria, B.C.; Dey, S.K. Ligand-receptor signaling with endocannabinoids in preimplantation embryo development and implantation. Chem. Phys. Lipids 2000, 108, 211–220. [Google Scholar] [CrossRef]
  133. Fernández-Ruiz, J.; Berrendero, F.; Hernández, M.L.; Ramos, J.A. The endogenous cannabinoid system and brain development. Trends Neurosci. 2000, 23, 14–20. [Google Scholar] [CrossRef] [PubMed]
  134. Douglas-Escobar, M.; Weiss, M.D. Hypoxic-ischemic encephalopathy: A review for the clinician. JAMA Pediatr. 2015, 169, 397–403. [Google Scholar] [CrossRef]
  135. Maiwald, C.A.; Annink, K.V.; Rüdiger, M.; Benders, M.J.N.L.; van Bel, F.; Allegaert, K.; Naulaers, G.; Bassler, D.; Klebermaß-Schrehof, K.; Vento, M.; et al. Effect of allopurinol in addition to hypothermia treatment in neonates for hypoxic-ischemic brain injury on neurocognitive outcome (ALBINO): Study protocol of a blinded randomized placebo-controlled parallel group multicenter trial for superiority (Phase III). BMC Pediatr. 2019, 19, 210. [Google Scholar] [CrossRef] [Green Version]
  136. Lee, A.C.; Kozuki, N.; Blencowe, H.; Vos, T.; Bahalim, A.; Darmstadt, G.L.; Niermeyer, S.; Ellis, M.; Robertson, N.J.; Cousens, S.; et al. Intrapartum-related neonatal encephalopathy incidence and impairment at regional and global levels for 2010 with trends from 1990. Pediatr. Res. 2013, 74, 50–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Davidson, J.O.; Wassink, G.; van den Heuij, L.G.; Bennet, L.; Gunn, A.J. Therapeutic hypothermia for neonatal hypoxic–ischemic encephalopathy—Where to from here? Front. Neurol. 2015, 6, 198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Edwards, A.D.; Brocklehurst, P.; Gunn, A.J.; Halliday, H.; Juszczak, E.; Levene, M.; Strohm, B.; Thoresen, M.; Whitelaw, A.; Azzopardi, D. Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: Synthesis and meta-analysis of trial data. BMJ 2010, 340, c363. [Google Scholar] [CrossRef]
  139. Tetorou, K.; Sisa, C.; Iqbal, A.; Dhillon, K.; Hristova, M. Current therapies for neonatal hypoxic–ischaemic and infection-sensitised hypoxic–ischaemic brain damage. Front. Synaptic Neurosci. 2021, 13, 709301. [Google Scholar] [CrossRef]
  140. Alonso-Alconada, D.; Broad, K.D.; Bainbridge, A.; Chandrasekaran, M.; Faulkner, S.D.; Kerenyi, Á.; Hassell, J.; Rocha-Ferreira, E.; Hristova, M.; Fleiss, B.; et al. Brain cell death is reduced with cooling by 3.5 °C to 5 °C but increased with cooling by 8.5 °C in a piglet asphyxia model. Stroke 2015, 46, 275–278. [Google Scholar] [CrossRef] [Green Version]
  141. Gonzalez, F.F. Neuroprotection strategies for term encephalopathy. Semin. Pediatr. Neurol. 2019, 32, 100773. [Google Scholar] [CrossRef]
  142. Victor, S.; Rocha-Ferreira, E.; Rahim, A.; Hagberg, H.; Edwards, D. New possibilities for neuroprotection in neonatal hypoxic-ischemic encephalopathy. Eur. J. Pediatr. 2022, 181, 875–887. [Google Scholar] [CrossRef]
  143. Fernández-Ruiz, J.; Moro, M.A.; Martínez-Orgado, J. Cannabinoids in neurodegenerative disorders and stroke/brain trauma: From preclinical models to clinical applications. Neurotherapeutics 2015, 12, 793–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Nagayama, T.; Sinor, A.D.; Simon, R.P.; Chen, J.; Graham, S.H.; Jin, K.; Greenberg, D.A. Cannabinoids and neuroprotection in global and focal cerebral ischemia and in neuronal cultures. J. Neurosci. 1999, 19, 2987–2995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Lara-Celador, I.; Castro-Ortega, L.; Álvarez, A.; Goñi-de-Cerio, F.; Lacalle, J.; Hilario, E. Endocannabinoids reduce cerebral damage after hypoxic–ischemic injury in perinatal rats. Brain Res. 2012, 1474, 91–99. [Google Scholar] [CrossRef] [PubMed]
  146. Alonso-Alconada, D.; Alvarez, F.J.; Alvarez, A.; Mielgo, V.E.; Goñi-de-Cerio, F.; Rey-Santano, M.C.; Caballero, A.; Martinez-Orgado, J.; Hilario, E. The cannabinoid receptor agonist WIN 55,212-2 reduces the initial cerebral damage after hypoxic–ischemic injury in fetal lambs. Brain Res. 2010, 1362, 150–159. [Google Scholar] [CrossRef] [PubMed]
  147. Alonso-Alconada, D.; Álvarez, A.; Álvarez, F.J.; Martínez-Orgado, J.A.; Hilario, E. The cannabinoid WIN 55212-2 mitigates apoptosis and mitochondrial dysfunction after hypoxia ischemia. Neurochem. Res. 2012, 37, 161–170. [Google Scholar] [CrossRef]
  148. Alonso-Alconada, D.; Álvarez, A.; Arteaga, O.; Martínez-Ibargüen, A.; Hilario, E. Neuroprotective effect of melatonin: A novel therapy against perinatal hypoxia-ischemia. Int. J. Mol. Sci. 2013, 14, 9379–9395. [Google Scholar] [CrossRef]
  149. Pellegrini-Giampietro, D.E.; Mannaioni, G.; Bagetta, G. Post-ischemic brain damage: The endocannabinoid system in the mechanisms of neuronal death: The endocannabinoid system in cerebral ischemia. FEBS J. 2009, 276, 2–12. [Google Scholar] [CrossRef]
  150. Lombard, C.; Hegde, V.L.; Nagarkatti, M.; Nagarkatti, P.S. Perinatal exposure to Δ 9 -tetrahydrocannabinol triggers profound defects in T cell differentiation and function in fetal and postnatal stages of life, including decreased responsiveness to HIV antigens. J. Pharmacol. Exp. Ther. 2011, 339, 607–617. [Google Scholar] [CrossRef] [Green Version]
  151. Turcotte, C.; Blanchet, M.-R.; Laviolette, M.; Flamand, N. The CB2 receptor and its role as a regulator of inflammation. Cell. Mol. Life Sci. 2016, 73, 4449–4470. [Google Scholar] [CrossRef] [Green Version]
  152. Garberg, H.T.; Solberg, R.; Barlinn, J.; Martinez-Orgado, J.; Løberg, E.-M.; Saugstad, O.D. High-dose cannabidiol induced hypotension after global hypoxia-ischemia in piglets. Neonatology 2017, 112, 143–149. [Google Scholar] [CrossRef] [Green Version]
  153. Martínez-Orgado, J.; Villa, M.; del Pozo, A. Cannabidiol for the treatment of neonatal hypoxic-ischemic brain injury. Front. Pharmacol. 2021, 11, 584533. [Google Scholar] [CrossRef] [PubMed]
  154. Rivers-Auty, J.R.; Smith, P.F.; Ashton, J.C. The cannabinoid CB2 receptor agonist GW405833 does not ameliorate brain damage induced by hypoxia-ischemia in rats. Neurosci. Lett. 2014, 569, 104–109. [Google Scholar] [CrossRef] [PubMed]
  155. Azzopardi, D.; Strohm, B.; Linsell, L.; Hobson, A.; Juszczak, E.; Kurinczuk, J.J.; Brocklehurst, P.; Edwards, A.D.; UK TOBY Cooling Register. Implementation and conduct of therapeutic hypothermia for perinatal asphyxial encephalopathy in the UK—Analysis of national data. PLoS ONE 2012, 7, e38504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Drury, P.P.; Gunn, E.R.; Bennet, L.; Gunn, A.J. Mechanisms of hypothermic neuroprotection. Clin. Perinatol. 2014, 41, 161–175. [Google Scholar] [CrossRef]
  157. Leker, R.R.; Gai, N.; Mechoulam, R.; Ovadia, H. Drug-induced hypothermia reduces ischemic damage: Effects of the cannabinoid HU-210. Stroke 2003, 34, 2000–2006. [Google Scholar] [CrossRef] [Green Version]
  158. Barata, L.; Arruza, L.; Rodríguez, M.-J.; Aleo, E.; Vierge, E.; Criado, E.; Sobrino, E.; Vargas, C.; Ceprián, M.; Gutiérrez-Rodríguez, A.; et al. Neuroprotection by cannabidiol and hypothermia in a piglet model of newborn hypoxic-ischemic brain damage. Neuropharmacology 2019, 146, 1–11. [Google Scholar] [CrossRef]
  159. Lafuente, H.; Pazos, M.R.; Alvarez, A.; Mohammed, N.; Santos, M.; Arizti, M.; Alvarez, F.J.; Martinez-Orgado, J.A. Effects of cannabidiol and hypothermia on short-term brain damage in new-born piglets after acute hypoxia-ischemia. Front. Neurosci. 2016, 10, 323. [Google Scholar] [CrossRef] [Green Version]
  160. Ihrie, R.A.; Álvarez-Buylla, A. Lake-front property: A unique germinal niche by the lateral ventricles of the adult brain. Neuron 2011, 70, 674–686. [Google Scholar] [CrossRef] [Green Version]
  161. Spalding, K.L.; Bergmann, O.; Alkass, K.; Bernard, S.; Salehpour, M.; Huttner, H.B.; Boström, E.; Westerlund, I.; Vial, C.; Buchholz, B.A.; et al. Dynamics of hippocampal neurogenesis in adult humans. Cell 2013, 153, 1219–1227. [Google Scholar] [CrossRef] [Green Version]
  162. Lois, C.; Alvarez-Buylla, A. Proliferating subventricular zone cells in the adult mammalian forebrain can differentiate into neurons and glia. Proc. Natl. Acad. Sci. USA 1993, 90, 2074–2077. [Google Scholar] [CrossRef] [Green Version]
  163. Eriksson, P.S.; Perfilieva, E.; Björk-Eriksson, T.; Alborn, A.-M.; Nordborg, C.; Peterson, D.A.; Gage, F.H. Neurogenesis in the adult human hippocampus. Nat. Med. 1998, 4, 1313–1317. [Google Scholar] [CrossRef] [PubMed]
  164. Kornack, D.R.; Rakic, P. Cell proliferation without neurogenesis in adult primate neocortex. Science 2001, 294, 2127–2130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Asrican, B.; Paez-Gonzalez, P.; Erb, J.; Kuo, C.T. Cholinergic circuit control of postnatal neurogenesis. Neurogenesis 2016, 3, e1127310. [Google Scholar] [CrossRef] [PubMed]
  166. Benner, E.J.; Luciano, D.; Jo, R.; Abdi, K.; Paez-Gonzalez, P.; Sheng, H.; Warner, D.S.; Liu, C.; Eroglu, C.; Kuo, C.T. Protective astrogenesis from the SVZ niche after injury is controlled by notch modulator Thbs4. Nature 2013, 497, 369–373. [Google Scholar] [CrossRef] [Green Version]
  167. Faiz, M.; Sachewsky, N.; Gascón, S.; Bang, K.W.A.; Morshead, C.M.; Nagy, A. Adult neural stem cells from the subventricular zone give rise to reactive astrocytes in the cortex after stroke. Cell Stem Cell 2015, 17, 624–634. [Google Scholar] [CrossRef] [Green Version]
  168. Livneh, Y.; Adam, Y.; Mizrahi, A. Odor processing by adult-born neurons. Neuron 2014, 81, 1097–1110. [Google Scholar] [CrossRef]
  169. Sakamoto, M.; Ieki, N.; Miyoshi, G.; Mochimaru, D.; Miyachi, H.; Imura, T.; Yamaguchi, M.; Fishell, G.; Mori, K.; Kageyama, R.; et al. Continuous postnatal neurogenesis contributes to formation of the olfactory bulb neural circuits and flexible olfactory associative learning. J. Neurosci. 2014, 34, 5788–5799. [Google Scholar] [CrossRef] [Green Version]
  170. Gil-Perotín, S.; Duran-Moreno, M.; Cebrián-Silla, A.; Ramírez, M.; García-Belda, P.; García-Verdugo, J.M. Adult neural stem cells from the subventricular zone: A review of the neurosphere assay: A review of the neurosphere assay. Anat. Rec. 2013, 296, 1435–1452. [Google Scholar] [CrossRef]
  171. Kukekov, V.G.; Laywell, E.D.; Suslov, O.; Davies, K.; Scheffler, B.; Thomas, L.B.; O’Brien, T.F.; Kusakabe, M.; Steindler, D.A. Multipotent stem/progenitor cells with similar properties arise from two neurogenic regions of adult human brain. Exp. Neurol. 1999, 156, 333–344. [Google Scholar] [CrossRef] [Green Version]
  172. Ostenfeld, T.; Tai, Y.-T.; Martin, P.; Déglon, N.; Aebischer, P.; Svendsen, C.N. Neurospheres modified to produce glial cell line-derived neurotrophic factor increase the survival of transplanted dopamine neurons: GDNF-modified ns improve neuron survival. J. Neurosci. Res. 2002, 69, 955–965. [Google Scholar] [CrossRef]
  173. Yu, S.-J.; Tseng, K.-Y.; Shen, H.; Harvey, B.K.; Airavaara, M.; Wang, Y. Local Administration of AAV-BDNF to subventricular zone induces functional recovery in stroke rats. PLoS ONE 2013, 8, e81750. [Google Scholar] [CrossRef] [PubMed]
  174. Chang, E.H.; Adorjan, I.; Mundim, M.V.; Sun, B.; Dizon, M.L.V.; Szele, F.G. Traumatic brain injury activation of the adult subventricular zone neurogenic niche. Front. Neurosci. 2016, 10, 332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Yu, T.-S.; Washington, P.M.; Kernie, S.G. Injury-induced neurogenesis: Mechanisms and relevance. Neuroscientist 2016, 22, 61–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Plane, J.M.; Liu, R.; Wang, T.-W.; Silverstein, F.S.; Parent, J.M. Neonatal hypoxic–ischemic injury increases forebrain subventricular zone neurogenesis in the mouse. Neurobiol. Dis. 2004, 16, 585–595. [Google Scholar] [CrossRef]
  177. Levison, S.W.; Rothstein, R.P.; Romanko, M.J.; Snyder, M.J.; Meyers, R.L.; Vannucci, S.J. Hypoxia/ischemia depletes the rat perinatal subventricular zone of oligodendrocyte progenitors and neural stem cells. Dev. Neurosci. 2001, 23, 234–247. [Google Scholar] [CrossRef]
  178. Niimi, Y.; Levison, S.W. Pediatric brain repair from endogenous neural stem cells of the subventricular zone. Pediatr. Res. 2018, 83, 385–396. [Google Scholar] [CrossRef]
  179. Alonso-Alconada, D.; Gressens, P.; Golay, X.; Robertson, N.J. Neurogenesis is reduced at 48 h in the subventricular zone independent of cell death in a piglet model of perinatal hypoxia-ischemia. Front. Pediatr. 2022, 10, 793189. [Google Scholar] [CrossRef]
  180. Brazel, C.Y.; Rosti III, R.T.; Boyce, S.; Rothstein, R.P.; Levison, S.W. Perinatal hypoxia/ischemia damages and depletes progenitors from the mouse subventricular zone. Dev. Neurosci. 2004, 26, 266–274. [Google Scholar] [CrossRef] [Green Version]
  181. Romanko, M.J.; Rothstein, R.P.; Levison, S.W. Neural stem cells in the subventricular zone are resilient to hypoxia/ischemia whereas progenitors are vulnerable. J. Cereb. Blood Flow Metab. 2004, 24, 814–825. [Google Scholar] [CrossRef] [Green Version]
  182. Visco, D.B.; Toscano, A.E.; Juárez, P.A.R.; Gouveia, H.J.C.B.; Guzman-Quevedo, O.; Torner, L.; Manhães-de-Castro, R. A systematic review of neurogenesis in animal models of early brain damage: Implications for cerebral palsy. Exp. Neurol. 2021, 340, 113643. [Google Scholar] [CrossRef]
  183. Ong, J.; Plane, J.M.; Parent, J.M.; Silverstein, F.S. Hypoxic-ischemic injury stimulates subventricular zone proliferation and neurogenesis in the neonatal rat. Pediatr. Res. 2005, 58, 600–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Yang, Z.; Levison, S.W. Hypoxia/ischemia expands the regenerative capacity of progenitors in the perinatal subventricular zone. Neuroscience 2006, 139, 555–564. [Google Scholar] [CrossRef] [PubMed]
  185. Felling, R.J. Neural stem/progenitor cells participate in the regenerative response to perinatal hypoxia/ischemia. J. Neurosci. 2006, 26, 4359–4369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Bartley, J.; Soltau, T.; Wimborne, H.; Kim, S.; Martin-Studdard, A.; Hess, D.; Hill, W.; Waller, J.; Carroll, J. BrdU-positive cells in the neonatal mouse hippocampus following hypoxic-ischemic brain injury. BMC Neurosci. 2005, 6, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Kadam, S.D.; Mulholland, J.D.; McDonald, J.W.; Comi, A.M. Neurogenesis and neuronal commitment following ischemia in a new mouse model for neonatal stroke. Brain Res. 2008, 1208, 35–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Ziemka-Nalecz, M.; Jaworska, J.; Sypecka, J.; Polowy, R.; Filipkowski, R.K.; Zalewska, T. Sodium butyrate, a histone deacetylase inhibitor, exhibits neuroprotective/neurogenic effects in a rat model of neonatal hypoxia-ischemia. Mol. Neurobiol. 2017, 54, 5300–5318. [Google Scholar] [CrossRef] [PubMed]
  189. Buono, K.D.; Goodus, M.T.; Guardia Clausi, M.; Jiang, Y.; Loporchio, D.; Levison, S.W. Mechanisms of mouse neural precursor expansion after neonatal hypoxia-ischemia. J. Neurosci. 2015, 35, 8855–8865. [Google Scholar] [CrossRef] [Green Version]
  190. Back, S.A.; Tuohy, T.M.F.; Chen, H.; Wallingford, N.; Craig, A.; Struve, J.; Luo, N.L.; Banine, F.; Liu, Y.; Chang, A.; et al. Hyaluronan accumulates in demyelinated lesions and inhibits oligodendrocyte progenitor maturation. Nat. Med. 2005, 11, 966–972. [Google Scholar] [CrossRef]
  191. Pendleton, J.C.; Shamblott, M.J.; Gary, D.S.; Belegu, V.; Hurtado, A.; Malone, M.L.; McDonald, J.W. Chondroitin sulfate proteoglycans inhibit oligodendrocyte myelination through PTPσ. Exp. Neurol. 2013, 247, 113–121. [Google Scholar] [CrossRef]
  192. Aguado, T.; Romero, E.; Monory, K.; Palazuelos, J.; Sendtner, M.; Marsicano, G.; Lutz, B.; Guzmán, M.; Galve-Roperh, I. The CB1 cannabinoid receptor mediates excitotoxicity-induced neural progenitor proliferation and neurogenesis. J. Biol. Chem. 2007, 282, 23892–23898. [Google Scholar] [CrossRef] [Green Version]
  193. Fernández-López, D.; Pradillo, J.M.; García-Yébenes, I.; Martínez-Orgado, J.A.; Moro, M.A.; Lizasoain, I. The cannabinoid WIN55212-2 promotes neural repair after neonatal hypoxia–ischemia. Stroke 2010, 41, 2956–2964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Bregy, A.; Nixon, R.; Lotocki, G.; Alonso, O.F.; Atkins, C.M.; Tsoulfas, P.; Bramlett, H.M.; Dietrich, W.D. Posttraumatic hypothermia increases doublecortin expressing neurons in the dentate gyrus after traumatic brain injury in the rat. Exp. Neurol. 2012, 233, 821–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Silasi, G.; Colbourne, F. Therapeutic hypothermia influences cell genesis and survival in the rat hippocampus following global ischemia. J. Cereb. Blood Flow Metab. 2011, 31, 1725–1735. [Google Scholar] [CrossRef] [PubMed]
  196. Xiong, M.; Cheng, G.-Q.; Ma, S.-M.; Yang, Y.; Shao, X.-M.; Zhou, W.-H. Post-ischemic hypothermia promotes generation of neural cells and reduces apoptosis by Bcl-2 in the striatum of neonatal rat brain. Neurochem. Int. 2011, 58, 625–633. [Google Scholar] [CrossRef] [PubMed]
  197. Arévalo-Martín, Á.; García-Ovejero, D.; Rubio-Araiz, A.; Gómez, O.; Molina-Holgado, F.; Molina-Holgado, E. Cannabinoids modulate olig2 and polysialylated neural cell adhesion molecule expression in the subventricular zone of post-natal rats through cannabinoid receptor 1 and cannabinoid receptor 2: Cannabinoid receptors in post-natal SVZ. Eur. J. Neurosci. 2007, 26, 1548–1559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Iwai, M.; Stetler, R.A.; Xing, J.; Hu, X.; Gao, Y.; Zhang, W.; Chen, J.; Cao, G. Enhanced oligodendrogenesis and recovery of neurological function by erythropoietin after neonatal hypoxic/ischemic brain injury. Stroke 2010, 41, 1032–1037. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of AEA and 2-AG.
Figure 1. Chemical structures of AEA and 2-AG.
Biomedicines 11 00028 g001
Figure 2. Chemical structures of Δ9-THC and CBD.
Figure 2. Chemical structures of Δ9-THC and CBD.
Biomedicines 11 00028 g002
Figure 3. Chemical structures of neuroprotective CB ligands.
Figure 3. Chemical structures of neuroprotective CB ligands.
Biomedicines 11 00028 g003
Figure 4. Chemical structures of neuroprotective FAAH and MGL inhibitors.
Figure 4. Chemical structures of neuroprotective FAAH and MGL inhibitors.
Biomedicines 11 00028 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Duranti, A.; Beldarrain, G.; Álvarez, A.; Sbriscia, M.; Carloni, S.; Balduini, W.; Alonso-Alconada, D. The Endocannabinoid System as a Target for Neuroprotection/Neuroregeneration in Perinatal Hypoxic–Ischemic Brain Injury. Biomedicines 2023, 11, 28. https://doi.org/10.3390/biomedicines11010028

AMA Style

Duranti A, Beldarrain G, Álvarez A, Sbriscia M, Carloni S, Balduini W, Alonso-Alconada D. The Endocannabinoid System as a Target for Neuroprotection/Neuroregeneration in Perinatal Hypoxic–Ischemic Brain Injury. Biomedicines. 2023; 11(1):28. https://doi.org/10.3390/biomedicines11010028

Chicago/Turabian Style

Duranti, Andrea, Gorane Beldarrain, Antonia Álvarez, Matilde Sbriscia, Silvia Carloni, Walter Balduini, and Daniel Alonso-Alconada. 2023. "The Endocannabinoid System as a Target for Neuroprotection/Neuroregeneration in Perinatal Hypoxic–Ischemic Brain Injury" Biomedicines 11, no. 1: 28. https://doi.org/10.3390/biomedicines11010028

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop