Next Article in Journal
Adenosine-Mimicking Derivatives of 3-Aminopyrazine-2-Carboxamide: Towards Inhibitors of Prolyl-tRNA Synthetase with Antimycobacterial Activity
Next Article in Special Issue
Antitumor Effects of Cannabis sativa Bioactive Compounds on Colorectal Carcinogenesis
Previous Article in Journal
The Pharmacorank Search Tool for the Retrieval of Prioritized Protein Drug Targets and Drug Repositioning Candidates According to Selected Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Insights into Epigenetics and Cannabinoid Receptors

by
Balapal S. Basavarajappa
1,2,3,4,* and
Shivakumar Subbanna
1
1
Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
2
Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY 10032, USA
3
Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
4
Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2022, 12(11), 1560; https://doi.org/10.3390/biom12111560
Submission received: 12 September 2022 / Revised: 29 September 2022 / Accepted: 22 October 2022 / Published: 26 October 2022
(This article belongs to the Special Issue New Advances of Cannabinoid Receptors in Health and Disease)

Abstract

:
The actions of cannabis are mediated by G protein-coupled receptors that are part of an endogenous cannabinoid system (ECS). ECS consists of the naturally occurring ligands N-arachidonylethanolamine (anandamide) and 2-arachidonoylglycerol (2-AG), their biosynthetic and degradative enzymes, and the CB1 and CB2 cannabinoid receptors. Epigenetics are heritable changes that affect gene expression without changing the DNA sequence, transducing external stimuli in stable alterations of the DNA or chromatin structure. Cannabinoid receptors are crucial candidates for exploring their functions through epigenetic approaches due to their significant roles in health and diseases. Epigenetic changes usually promote alterations in the expression of genes and proteins that can be evaluated by various transcriptomic and proteomic analyses. Despite the exponential growth of new evidence on the critical functions of cannabinoid receptors, much is still unknown regarding the contribution of various genetic and epigenetic factors that regulate cannabinoid receptor gene expression. Recent studies have identified several immediate and long-lasting epigenetic changes, such as DNA methylation, DNA-associated histone proteins, and RNA regulatory networks, in cannabinoid receptor function. Thus, they can offer solutions to many cellular, molecular, and behavioral impairments found after modulation of cannabinoid receptor activities. In this review, we discuss the significant research advances in different epigenetic factors contributing to the regulation of cannabinoid receptors and their functions under both physiological and pathological conditions. Increasing our understanding of the epigenetics of cannabinoid receptors will significantly advance our knowledge and could lead to the identification of novel therapeutic targets and innovative treatment strategies for diseases associated with altered cannabinoid receptor functions.

1. Introduction

1.1. Endocannabinoid System: A Brief Overview

Endocannabinoids (eCBs) are bioactive lipids implicated in many physiological mechanisms in the central nervous system (CNS) and peripheral tissues. The eCBs N-arachidonylethanolamine (anandamide, AEA) and 2-arachidonoylglycerol (2-AG) are primarily synthesized in all cell types because they are derived from phospholipids containing arachidonic acid (AA) [1,2,3,4]. The anandamide-synthesizing enzyme is N-acylphosphatidylethanolamine (NAPE)-phospholipase D hydrolase (NAPE-PLD), which catalyzes the synthesis of AEA [5]. Diacylglycerol lipase (DAGL) catalyzes the biosynthesis of 2-AG [6]. In addition to AEA and 2-AG synthesis, other N-acylethanolamines, such as oleoylethanolamide (OEA), linoleoylethanolamide (LEA), palmitoylethanolamide (PEA), and docosahexaenoylethanolamine (DHEA), and other 2-acylglycerols, such as 2-oleoylglycerol and 2-linoleoylglycerol, are synthesized through alternative biochemical routes [7]. These bioactive lipids exhibit different affinities at the cannabinoid receptors CB1 and CB2 and other receptors (transient receptor potential vanilloid 1 (TRPV1), peroxisome proliferator-activated nuclear receptor-α (PPARα) and PPARγ, and the orphan G protein-coupled receptors GPR55 and GPR119 [8]). Upon their action at the CB receptors, eCBs undergo rapid degradation by fatty acid amide hydrolase (FAAH) [9] and monoacylglycerol lipase (MAGL) [10] to ethanolamine, glycerol, AA, and other fatty acids [11,12]. Details on eCB, biosynthetic, and metabolism pathways have been extensively reviewed in recent publications [7,13].
CB1 and CB2 are the extensively studied receptor targets of eCBs, which bind to and activate them with different affinities. CB1 is the brain’s most abundant G protein-coupled receptor [9]. It is responsible for mediating most of the neurobehavioral effects of Δ9 -tetrahydrocannabinol (THC) [14,15], the psychoactive constituent of marijuana [16,17]. Consistent with the well-established functions of eCB, CB1 is enriched in brain areas implicated in memory (e.g., hippocampus, HP), motor coordination (e.g., basal ganglia, cerebellum), and emotional processes (e.g., prefrontal cortex, PFC; amygdala, Amy) [18,19]. CB1 is preferentially restricted to the presynaptic region, and eCBs released from postsynaptic neurons act retrogradely on presynaptic CB1, resulting in short- and long-term suppression of neurotransmitter release [20,21] and the modulation of neuronal activity and network function. This intricate circuit influences various pathophysiological functions, such as emotion, cognition, energy balance, pain sensation, and neuroinflammation [7]. CB1 is also expressed in peripheral tissues, including adipose tissue, liver, and skeletal muscle [22]. CB2 is predominantly expressed in immune cells [23,24], where it seems to facilitate the immunosuppressive effects of eCBs. Interestingly, further discoveries emphasize that CB2 is expressed at low levels in some areas of the brain [24,25,26], where it is activated during injury and inflammation [25,26]. CB2 is mostly located in postsynaptic terminals [27]. However, CB2 is also expressed in some presynaptic terminals [26,28,29].
CB1 is encoded by the CNR1 gene and comprises 472 amino acids in humans and 473 amino acids in rodents (rats and mice), with 97–99% amino acid sequence similarity among them [30]. The CNR1 gene is localized to human chromosome 6q14–15 and mouse chromosome 4. Human CNR1 has four exons, with exon 4 containing the entire protein-coding region [31]. In mice and rats, the coding region of CNR1 is contained within a single exon. Although the 5′ untranslated regions (5′-UTRs) and promoter structures differ between mice and humans [32,33], these structures are not well described in rats [34]. The longest 5′ UTR in human CNR1 is approximately 500 nucleotides and has approximately 600 potential transcription factor-binding regions for 153 distinct transcription factors [35,36]. A few of these binding sites are unique and bind signal transducer and activator of transcription proteins and eventually may regulate many critical aspects of cell growth, survival, and differentiation [36]. The complexity of 5′ UTR of the CNR1 gene emphasizes that multiple transcription factors regulate CNR1 gene expression in a basic manner, which may be necessary for brain development and function [36]. A polymorphic enhancer sequence (ECR1) was identified within intron 2 of the CNR1 locus [37], and disruption of ECR1 using CRISPR genome editing in mice indicated that ECR1 is essential for maintaining normal levels of CNR1 expression within the hippocampus [38]. The CB1 mRNA distribution also paralleled that of the CB1 protein in certain brain areas [39,40,41].
CB2 is encoded by the CNR2 gene and is located on human chromosome 1p36 and mouse chromosome 4 [42,43,44,45]. CB2 displays less homology between species than CB1; for example, human and mouse CB2 share an 82% amino acid homology [46], and mouse and rat CB2 share a 93% amino acid homology. The human, rat, and mouse sequences differ at the C-terminus [47]. The mouse sequence is 13 amino acids shorter, whereas the rat clone is 50 amino acids longer than human CB2 [47]. The CNR2 mRNA distribution has been detected in multiple brain regions, including the PFC, hippocampus, midbrain, and cerebellum [48,49,50,51]. The human CNR2 gene has three exons with three separate promoters [52,53]. However, the current evidence indicates significant species differences in CNR2s in humans, mice, and rats regarding mRNA sizes and gene structure [49,52,53]. Although the functional implication of multiple transcription start sites (TSSs) and core promoters is unknown, this heterogeneity may have significance for the cell type and activation function [54,55]. Additionally, the promoter region of human CNR2 has several transcription factor-binding sites [56,57], which can regulate the expression of CB2 [57]. The promoter of CNR2 has cytosine-phosphate-guanine (CpG) islands and many CCAAT boxes with binding sites for transcription factors associated with the stress response, such as activator protein-1 (AP1), heat shock factor (HSF) and stress response element, GATA-binding factor-1 (erythroid transcription factor), tinman homolog Ntx2.5 (homeodomain factor), and AP4 [58]. The epigenetic regulation of CNR2 loci via DNA methylation might play a decisive function in receptor regulation due to the CpG islands found in the promoter regions. These gene regulatory binding sites are significant, as recent studies have indicated that cannabinoid receptor gene expression could be controlled by chemical modification of DNA and histone tails (epigenetics), resulting in alterations in the chromatin structure and access to transcription factors.

1.2. Epigenetic Mechanisms: A Brief Overview

In general, the primary epigenetic mechanisms that are well recognized to control the expression of genes in the CNS are (1) the main chemical modification of DNA through methylation (-CH3) of cytosine residues in promoter-rich CpG islands; (2) the acetylation (ac), mono- (me1), di- (me2), and tri-methylation (me3) at lysine (K) residues, and other covalent post-translational modification (PTM) of DNA-associated histone protein tails; (3) chromatin remodeling factors that affect gene transcription; (4) the editing and splicing of pre-mRNA by noncoding, small nucleolar RNAs (snoRNAs); e) microRNAs (miRNAs), mRNA processing, the translation and stability of binding proteins, and long noncoding RNAs (lncRNAs); and (5) cellular signaling molecules controlling mRNA translation. Recent publications have reviewed the fundamental features of these epigenetic processes in detail [59,60,61,62,63]. These epigenetic factors/regulators can selectively respond to adverse environmental conditions, causing alterations in the brain’s physiological function and pathological processes. An increasing number of adverse conditions, including exposure to cannabinoids, which activate cannabinoid receptors, have undoubtedly been shown to alter various epigenetic factors. Nevertheless, the mechanism by which altered epigenetic events cause cannabinoid receptor- or cannabinoid-mediated gene expression is poorly defined. In this review, we provide the current understanding of epigenetic changes in cannabinoid receptor gene regulatory regions and cannabinoid-mediated events.

2. Role of DNA Methylation: Cannabinoid Receptors

DNA methylation and other remodeling factors are considered significant epigenetic markers and are known to control gene expression (for reference, see [64,65]). DNA de novo methylation, which occurs in distinct cellular contexts in germ cells and during maturation, is catalyzed by DNA methyltransferases 3A (DNMT3A) and 3B (DNMT3B) in partnership with DNMT3L, a DNMT devoid of catalytic activity. However, it facilitates de novo methylation by promoting the ability of DNMTs to bind to the methyl group donor S-adenosyl-L-methionine (SAM). Additionally, DNA methylation is stabilized by DNMT1. Studies have suggested at least two routes through which the DNA demethylation process occurs: (1) deaminase activity catalyzes the conversion of methylcytosine (mC) to thymidine [66] and (2) the action of the ten-eleven translocation (TET) family (α-ketoglutarate-dependent dioxygenases). TET proteins oxidize 5-mC to 5-hydroxymethylcytosine (5-hmC) using oxygen- and α-ketoglutarate-dependent pathways [67]. DNA demethylation processes through 5-hmC were revealed to function in both developing and adult brains [68], thereby offering the basis for a valuable epigenetic regulator of gene expression [69].
In addition, a different group of proteins that work together with methylated DNA to control gene expression in CNS is the family of methyl CpG-binding proteins (MeCPs). Methyl CpG-binding proteins often function as gene suppressors by binding to methylated cytosines [70,71] in DNA. The MeCP2 protein recognizes and binds to single methylated cytosine (5mC) sites in DNA. Additionally, the binding of MeCP2 to DNA further facilitates the recruitment of transcriptional corepressor complexes [70]. Moreover, phosphorylation of MeCP2 affects its capacity to bind to DNA and regulate gene expression [72,73]. The activity-dependent phosphorylation of MeCP2 promotes its dissociation from promoters, thereby facilitating the DNA demethylation process. Thus, DNA methylation followed by the binding of MeCP2 appears to have a central role in gene expression.

2.1. DNA Methylation on CB1 Receptor Gene (Cnr1) Expression

In the past decade, evidence has accumulated suggesting that CB1 gene expression is under the control of epigenetic mechanisms. This is partly because CB1 gene expression is altered in response to different pathological conditions and upon exposure to adverse insults, including exposure to different drugs [74]. Additionally, many transcription factors implicated in DNA methylation and histone post-translational modifications interact with cannabinoid receptor genes [36,56,57]. The first study demonstrating the association of DNA hypermethylation of the CNR1 gene promoter contributing to the downregulation of CNR1 gene transcription was observed in colon cancer specimens [75]. A similar observation was found in another study in which exposure to prostaglandin E2 suppressed CNR1 gene expression by increasing DNA methylation in the CNR1 promoter region in the human epithelial colon cell line LS-174T, causing tumor growth [76]. Enhanced DNA methylation in the Cnr1 gene promoter region was also found in rodents after maternal separation from postnatal day (PD) 1 to 14 in the first-generation germline [77]. This outcome supports the previously well-established function of CB1 in emotional behavior [78]. In another study, although the mechanisms are less clear, the expression of CB1 by an inhibitor of DNA methyltransferases (5-aza-2′-deoxycytidine, 5-Aza-dC) was found only in those cells (Jurkat cells) in which the expression of CB1 was constitutively inactive [79]. In another study, it was found that DNA hypermethylation of the CNR1 gene promoter was associated with reduced CNR1 mRNA levels in peripheral blood cells of subjects with THC dependence [80]. Selective and transient upregulation of CNR1 gene expression was observed in human colon cancer cells (Caco-2) and rats exposed to short- and long-term dietary extra-virgin olive oil (EVOO) and its phenolic extracts (OPE) or authentic hydroxytyrosol (HT) [81]. Additionally, this treatment caused a reduction in DNA methylation at the Cnr1 gene promoter [81]. Chronic stress-induced visceral pain in the peripheral nervous systems of rats was associated with enhanced DNMT1-mediated DNA hypermethylation at the Cnr1 gene promoter [82]. Furthermore, it decreased Cnr1 gene expression in L6-S2 that transmit pain (nociceptive) signals but not L4-L5 dorsal root ganglia (DRG) [82].
Similarly, enhanced Cnr1 gene expression in PFC was associated with reduced DNA methylation at the Cnr1 gene promoter in a well-validated animal model of schizophrenia (prenatal methylazoxymethanol acetate exposure in rats) and in schizophrenic patients [83]. A significant and selective increase in Cnr1 gene expression in the hypothalamus (HTM) was observed in the initial stages of obesity onset (5 weeks on a high-fat diet) and after 21 weeks of high-fat diet consumption. In addition, there was a significant reduction in DNA methylation at specific CpG sites at Cnr1 gene promoters [84]. Similar observations were found in blood mononuclear cells from younger (<30 years old) human obese subjects [84]. Exposure to THC or alcohol is significantly associated with increased expression of CNR1 in PFC of patients with affective disorder [36]. Additionally, enhanced CNR1 expression was observed in PFC of schizophrenia patients who had committed suicide [36]. It was found that DNA methylation (cg02498983 allele, associated with CNR1 expression) is inversely associated with CNR1 expression [36]. In an activity-based anorexia rat model, Cnr1 gene expression was associated with significant increases in DNA methylation at the Cnr1 gene promoter in the HTM and nucleus accumbens (NAc) brain regions [85].
In an animal model of eating addictive-like behavior, a significant loss of DNA methylation at the Cnr1 gene promoter was observed in PFC. This loss was associated with enhanced CB1 protein expression in the same brain area [86]. Additionally, the pharmacological blockade of CB1 activity during the late training period significantly impaired addictive behavior in mice [86]. This latter observation agreed with the impaired performance of CB1-null mice in this operant training [86]. These findings suggest that DNA methylation-mediated CB1 expression could influence addictive behavior. In another study, selective reduction of DNA methylation at the promoter of CNR1 and enhanced CNR1 gene expression were observed in schizophrenic patients, with no changes in any other disorder [83]. These results from different experimental models indicate that DNA methylation events regulate Cnr1 gene expression (Table 1).

2.2. DNA Methylation on CB2 Gene (Cnr2) Expression

Compared to Cnr1, Cnr2 gene regulation by DNA methylation mechanisms has been less studied. However, THC consumption has been shown to enhance CB2 expression in human blood lymphocytes via changes in DNMT and TET mRNAs [89]. Although no direct link between these events was established, these observations may suggest that increased DNMT-methylating enzymes are associated with some of the pathophysiological processes in schizophrenia and, therefore, should be one of the potential mechanisms linking cannabis use as a trigger for schizophrenia in vulnerable individuals. In another study, CB2-selective agonist (JWH-133)-treated male mice crossed with untreated females exhibited embryonic and placental defects. Additional analysis indicated significantly reduced Tet3 expression in sperm. In addition, significantly increased enrichment of 5mC and reduced 5hmC at paternally expressed genes (Peg10 and Plagl1) in the sperm of JWH-133-treated males was found [90]. In another study, the expression of CB2 by an inhibitor of DNA methyltransferases, 5-Aza-dC, was found only in cells where the expression of the CB2 receptor was silenced. Thus, CB2 was induced by 5-Aza-dC only in SH SY5Y cells but not in Jurkat cells. Although the mechanism is unclear, these findings suggest that already constitutively expressed genes were not regulated in these cells. Altogether, these limited studies suggest that CB2 expression could be regulated by DNA methylation of its promoter and warrant future studies, especially during inflammation [91,92,93], fear memory [94], nerve injury [52], and compulsive drug abuse [95] conditions in which CB2 expression was found to be heightened.

2.3. Cannabinoid Receptor Stimulation on DNA Methylation

Several studies using eCBs or agonists or antagonists acting specifically through CB1 have also demonstrated the participation of DNA methylation in several biological functions. For example, in human keratinocytes (HaCaT cells), AEA reduced keratin 1, keratin 10, involucrin, and transglutaminase-5 gene expression by DNA hypermethylation. Treatment of HaCaT cells with 5-azacytidine ameliorated AEA-inhibited keratin gene expression, indicating that AEA itself was also able to suppress gene transcription by altering both specific and global DNA methylation [96]. Furthermore, it was found that AEA-induced DNMT activity in differentiated keratinocytes was CB1 dependent via p38 MAPK signaling [96]. In THC-treated SIV-infected macaques, it was found that hypermethylation of DNA of several genes was critical for the replication and pathogenesis of human (HIV) and simian (SIV) immunodeficiency viruses [97]. These findings indicated that eCBs could function as transcriptional repressors via less defined DNA methylation mechanisms. In another study, exposure to the CB1 agonist WIN55,212-2 during adolescence increased DNMT3a expression and inhibited cocaine-induced conditioned place preference in mice [98]. THC administration via oral gavage in rats caused significant hypermethylation at Lrrtm4 and significant hypomethylation at Shank1, Syt3, Nrxn1, Nrxn3, Dlg4, and Grid1 of neurodevelopmental genes in rat sperm [99]. THC consumption in patients who have schizophrenic psychosis caused high DNA methylation at the NEUREXIN (NRXN1) promoter, a schizophrenia candidate gene, compared to controls and non-THC consumer patients [100]. Administration of WIN55,212-2 to adolescent rats induced DNA hypermethylation at the intragenic region of the Rgs7 gene, which was associated with a lower rate of mRNA transcription of the Rgs7 gene [101]. Rgs7 acts as an intracellular antagonist of GPCR signaling [101]. It was shown that reduced expression of cannabinoid receptor-interacting protein 1 (CNRIP1) was associated with enhanced DNA methylation of a CpG island site named CNRIP1 MS-2 (CNRIP1 methylation site-2) in intrahepatic cholangiocarcinoma (ICC) cells [102].
Alcohol exposure during development can affect brain development and cause persistent behavioral problems. For example, exposure of PD-7 mice to alcohol heightened CB1 activity (enhanced CB1 expression and anandamide levels) and caused neurodegeneration as measured by active caspase-3 levels [87]. In the same animal model, PD-7 alcohol exposure reduced global DNA methylation by promoting the loss of DNMT1 and DNMT3A in the neonatal brain, and these losses were not observed in CB1-null mice [88]. Additionally, blockade of CB1 with antagonist (SR141716A) prior to PD-7 alcohol exposure in wild-type mice also prevented the loss of DNA methylation [88]. These findings suggest the potential of CB1 in regulating the DNA methylation process. In addition, reduced MeCP2, a protein essential for synaptogenesis and neuronal maturation, was observed in these conditions [103]. Interestingly, the genetic deletion of CB1 prevented the loss of the MeCP2 protein in alcohol-exposed PD-7 mice, and administration of a CB1 antagonist (SR141716A) before PD-7 alcohol exposure precluded this loss [103]. These observations suggested that CB1-mediated instability of MeCP2 and reduced DNA methylation during active synaptic maturation may disrupt synaptic circuit maturation and cause neurobehavioral abnormalities, as found in animal models of fetal alcohol spectrum disorders (FASDs) [59].
Cannabidiol (CBD), a nonpsychotomimetic component of the Cannabis sativa plant, exhibits therapeutic potential in several psychiatric disorders, including schizophrenia. In the prepulse inhibition animal model, CBD-attenuated MK-801, an uncompetitive antagonist of the N-Methyl-D-aspartate (NMDA) receptor, enhanced DNA methylation [104]. These findings indicate that the antipsychotic effects of CBD involve DNA methylation mechanisms in the ventral striatum. The exposure of mice to CBD orally for 2 weeks caused global DNA hypomethylation, including hypomethylation of the de novo methyltransferase DNMT3A and >3000 additional differentially methylated loci enriched for genes [105] involved in the neuronal function and synaptic structure [106]. The effect of CBD hypomethylation on DNMT3A is significant, as the expression of this de novo methyltransferase in PFC has been shown to cause anxiety-like behaviors in adult mice [106]. Together, these findings may suggest that activation of cannabinoid receptors through cannabinoid abuse, specifically during a stage at which the brain is most vulnerable, alters gene expression via DNA methylation.

3. Role of Post-Translational Modification of DNA-Associated Histone Proteins: Cannabinoid Receptors

Histones are proteins that have an essential structural and functional significance in the transition between active and inactive states in chromatin and are responsible for gene regulation and epigenetic silencing [107,108]. The chromatin organization involves two copies of each of the histone H2A, H2B, H3, and H4 proteins, forming a central structured globular domain with a close connection with the DNA [109,110] and a less well-structured amino-terminal tail domain [111,112]. Furthermore, due to the histone fold domain and N-terminal tails, histones are vulnerable to PTMs, such as acetylation, methylation, phosphorylation, and sumoylation [113]. The primary enzymes involved in these PTMs are histone acetyltransferases (HATs), histone lysine deacetylases (HDACs; for example, HDAC-1, HDAC-2, HDAC-3), histone methyltransferase (HMT; for example, G9a, Suv39h1), and histone demethylase (HMD). These PTM-dependent chromatin changes promote the recruitment of DNA-binding proteins, causing a loose or compact chromatin structure at particular genetic loci, which leads to the expression or suppression of a particular gene [113]. Fundamental aspects of different histone modifications have been described in recent reviews [59,62,114].
For the first time, Börner and collaborators demonstrated that exposure to trichostatin A, an HDAC inhibitor, in human Jurkat T cells could regulate CNR1 expression, where the expression of CB1 protein was absent [79]. Reduced expression of Cnr1 in the cingulate cortex of mice with chronic unpredictable stress was associated with decreased levels of histone H3K9 acetylation (H3K9ac) but not H4K8ac with the Cnr1 gene [115]. In the FASD study, it was demonstrated that transcriptional activation of Cnr1 followed by widespread neurodegeneration in the PD-7 alcohol-exposed neonatal brain was due to increased H4K8 acetylation (associated with active transcription) and reduced H3K9 demethylation (correlated with transcriptional silencing) at the Cnr1 gene promoter region [116]. The epigenetic activation of CB1 by PD-7 alcohol exposure was associated with enhanced HDAC-1, HDAC-2, and HDAC-3 gene expression [117]. These events, in turn, suppress the expression of synaptic plasticity-related genes such as Bdnf, c-fos, Egr1, and Arc [117]. Further studies indicated enhanced enrichment of HDAC-1, HDAC-2, and HDAC-3 at the Egr1 and Arc gene promoter regions. Preadministration of a CB1 receptor antagonist (SR141716A) before PD-7 alcohol exposure prevented enrichment of HDACs at the Egr1 and Arc gene promoters and prevented behavioral abnormalities associated with PD-7 alcohol exposure [117]. These observations strongly support the significance of specific histone PTMs’ influence on CB1- and CB1-mediated functions.

3.1. Histone Modifications That Modulate Cnr1 Gene Expression

CB1 has been shown to be expressed in the dorsal root ganglion (DRG) and to contribute to the analgesic properties of cannabinoids. In an animal model of neuropathic pain, enhanced enrichment of H3K9me2, a G9a (HMT)-catalyzed repressive histone mark, was found in the promoter regions of the Cnr1 genes [118]. Furthermore, G9a inhibition in nerve-injured animals not only enhanced CB1 expression in DRG but also potentiated the analgesic effect of a CB1 agonist on nerve injury-induced pain hypersensitivity [118]. Furthermore, in animals lacking G9a in DRG neurons, nerve injury did not reduce CB1 expression in DRG. Additionally, the CB1 agonist failed to produce analgesic effects. In addition, nerve injury weakened the inhibitory effect of the CB1 agonist on synaptic glutamate release from primary afferent nerves to spinal cord dorsal horn neurons in WT mice but not in DRG neuron-specific G9a-null mice [118]. These observations suggest the function of G9a-mediated histone methylation in the expression of CB1 and the analgesic effect of CB1. Cocaine self-administration significantly increased Cnr1 gene expression in NAc, the dorsal striatum (DS), and HP [119]. However, additional studies indicated no enrichment of H3K4me3 and H3K27ac marks [119], two marks usually found at active promoters [120,121]. Additionally, mice exposed to chronic unpredictable stress have been shown to have impaired emotional and nociceptive behaviors and to exhibit reduced CB1 expression in the cingulate cortex. Epigenetic evaluation indicated enhanced HDAC-2 and reduced levels of H3K9ac at the Cnr1 gene in the cingulate cortex compared to controls [115]. It is conceivable that other marks, such as H3K9me2, H3K8ac, or H3K14ac marks, may be altered as found in other conditions and deserve future investigation. Nevertheless, these observations strongly support the consequence of particular histone acetylation or methylation marks on CB1 expression and CB1-mediated functions.

3.2. Histone Modifications That Modulate Cnr2 Gene Expression

CB2 is largely expressed in immune cells, and CB2 agonists have no analgesic effect [122]. Therefore, inhibition of inflammation by CB2 agonists is believed to contribute to the relief of associated pain [123]. Nevertheless, nerve injury enhances CB2 expression in DRG, and CB2 agonists reduce neuropathic pain [115]. Epigenetic analysis indicated increased enrichment of H3K4me3 and H3K9ac (gene-activating histone marks) and reduced enrichment of H3K9me2 and H3K27me3 (repressive histone marks) at the Cnr2 promoter in DRG [115]. These findings indicate that nerve injury associated with CB2 expression involves specific histone acetylation or methylation mechanisms.

3.3. Modulation of Histone Acetylation and Methylation by CB1 and CB2 Activities

Studies where lymph node cells of mice immunized with a superantigen were exposed to THC showed associations of active histone modification signals with Th2 cytokine genes and suppressive modification signals with Th1 cytokine genes, suggesting that such a mechanism may play a significant role in the THC-mediated switch from Th1 to Th2 responses [124]. These studies suggest that some THC regulation of immune responses involves epigenetic pathways. THC exposure in adolescents transiently enhanced H3K9me3 in PFC by increasing the expression of Suv39H1, a histone lysine methyltransferase, but not G9a, and reduced the expression of the Homer1, Mgll, Abat, and Dlg4 genes, which are closely associated with synaptic plasticity [125]. Further epigenetic analysis indicated increased enrichment of H3K9me3 at the Homer1, Mgll, Abat, and Dlg4 genes but not at Abat. In the same study, simultaneous inhibition of Suv39h1 and G9a significantly rescued THC-increased H3K9me3 levels [125]. These observations suggest that adolescent THC-induced cognitive deficits involve specific histone methylation enzymes such as Suv39h1 and G9a. In another study, chronic THC administration significantly transiently enhanced H3K14ac and H3K9me2 levels in HP and NAc [125]. However, in the amygdala, these histone modifications are differentially altered by THC [125].
Low-dose THC administration in mature (12 months old) and old mice (18 months) improved the expression of synaptic plasticity-related proteins (synapsin I, synaptophysin, PSD95, pCREB, pERK), including the Klotho and Bdnf gene in HP and cognitive performance [126]. In addition, these changes were associated with enhanced global H3K9ac and H4K12ac and reduced H3K9me3 levels in HP. Furthermore, there is enhanced enrichment of H3K9ac at the Klotho and Bdnf promoter regions. HAT inhibitor treatment blocked the effects of THC on cognitive function and H3K9ac levels, synapsin 1, Klotho, and Bdnf expression. Consistent with HAT inhibitor effects, glutamatergic neuron-specific CB1-null mice also prevented THC effects on cognitive function and H3K9ac levels, synapsin 1, Klotho, and Bdnf expression [126]. These findings suggest that histone acetylation changes via CB1 signaling in forebrain glutamatergic neurons mediate the beneficial effects of low-dose THC.
CBD has been shown to modify histone marks in different model systems. For example, CBD (10 mg/kg, i.p.) showed enhanced enrichment of H3K4me3 in the FoxA1 binding motif [127]. In the same study, CBD enhanced H3K4me3 and reduced H3K27me3 at specific genes, such as IL-4, IL-5, and IL-13, in splenic CD4+ T cells [127]. In another study, repeated coadministration of CBD and THC (at a 5:1 CBD/THC ratio, i.e., 50 mg/kg/10 mg/kg, i.p. for 15 days), but not the administration of either compound alone, increased H3K9ac and H3K14ac levels, gene-activating histone acetylation marks in the ventral tegmental area of adult male mice [128]. However, similar to DNA methylation, histone modifications in response to exogenous stimuli can vary from tissue to tissue and different brain regions. Consistent with this notion, a study reported differential modifications of H3K4me3, H3K9ac, H3K9me2, H3K27me3, and H3K36me2 in the cerebral cortex, HTM, and pons following systemic administration of CBD (20 mg/kg, i.p.) to adult rats [129]. In the cerebral cortex, CBD enhanced H3K4me3, H3K9ac, and H3K27me3 levels without having any significant influence on H3K9me2 or H3K36me2 levels [129]. In HTM, CBD reduced H3K9ac levels without having any significant influence on H3K4me3, H3K9me2, H3K27me3, and H3K36me2 levels [129]. Last, in the pons, CBD decreased H3K4me3 levels without altering H3K9ac, H3K9me2, H3K27me3, or H3K36me2 marks [129]. The histone modifications induced by CBD were associated with anxiety-related behavioral changes in distinct preclinical studies. For instance, stress duration impacts H3K4, H3K9, and H3K27 methylation levels in HP [130]. In particular, acute stress increased H3K9me3 and reduced H3K27me3 and H3K9me1 levels in HP [130]. In contrast to these results, a week of restraint stress increased H3K9me3 and reduced H3K27me3 and H3K4me3 levels in the same region [130]. Therefore, it is possible that the observed stress-induced reductions in H3K27me3 and H3K4me3 in HP following a week of restraint stress [130] may be reversed by CBD’s enhancing effects on these histone modifications, as observed in the cerebral cortex [129]. Another intriguing anxiety-related epigenetic marker is H3K9ac, and its levels are enhanced in the cerebral cortex by CBD [130]. Additionally, low H3K9ac levels in the central amygdala were suggested to contribute to the maintenance of chronic anxiety and pain [131]. Low-level maternal care-induced anxiety-related behavior in adulthood was associated with reduced H3K9ac levels at the glucocorticoid receptor gene (Nr3c1) in rats [132]. Therefore, in the future, more systematic studies are warranted to examine the link between the protective function of CBD on histone modifications at key genes in health and disease (Table 2).

4. Regulation of microRNAs and Cannabinoid Receptors

MicroRNAs (miRNAs) are small, noncoding RNAs that function as essential epigenetic regulators of gene expression [134]. MiRNAs accomplish their post-transcriptional regulatory functions by interacting with the 3′ untranslated regions (3′ UTRs), 5′ UTRs, coding sequences, and gene promoters of target mRNAs and cause mRNA degradation, leading to translational repression [134,135]. Furthermore, miRNA interactions with their target genes are dynamic and dependent on their subcellular localization, miRNA and mRNA abundances, and miRNA–mRNA interaction affinity [136]. Thus, they can regulate the expression of networks of genes and entire pathways and are considered master regulators of gene expression [137]. MiRNAs are secreted into extracellular fluids and function as signaling molecules in the form of vesicles, such as exosomes, and mediate cell communication [138,139,140]. Furthermore, abnormal miRNA expression is associated with many human disorders [141,142,143]. Recent articles have simplified the canonical and noncanonical miRNA biogenesis pathways and mechanisms underlying miRNA-mediated gene regulation [136,144]. Moreover, the current knowledge of the miRNA secretion, transfer, and uptake of extracellular miRNAs and their functions has been reviewed elsewhere [145,146]. Additionally, miRNAs are indispensable for the maturation and functioning of the adult brain. Undeniably, several studies have demonstrated the participation of different miRNAs in a wide range of cellular homeostatic processes, including cellular differentiation, development, neural patterning, and synaptic plasticity [147,148,149,150]. Genetic deletion studies have demonstrated that miR-124, miR-125b, miR-132, miR-134, miR-137, and miR-138 control dendritic branching and synaptic maturation (for reference, see [151]). The interference of miRNA biogenesis pathways, such as Dicer, which controls the expression of all miRNAs, indicated the miRNA functions in cell differentiation, neuronal size, dendritic branching, and axonal guidance [152]. These studies ultimately indicated the functions of miRNAs in inhibitory synaptic transmission and cognitive function [153]. Thus, an understanding of the regulatory mechanisms that control the patterns and activity of cannabinoid receptors by miRNA expression has the potential to identify a likely mechanism for cannabinoid receptor expression and mediated function. Therefore, we provide an overview of miRNAs that regulate cannabinoid receptor expression and how the modulation of cannabinoid receptors influences miRNA expression.

4.1. miRNAs That Modulate Cnr1 Gene Expression

As discussed above, the cannabinoid system is regulated by and regulates epigenetic mechanisms, though the interactions between cannabinoid receptor modulation and miRNAs are under investigated. Cannabinoids have been shown to drive anticancer effects through miRNA modulation. However, whether ECS promotes tumor growth and progression through miRNA is still unclear. Furthermore, miRNAs have been shown to regulate ECS, promoting or inhibiting cancer growth and progression. For instance, miRNA-1273g-3p promotes the proliferation, migration, and invasion of human colon cancer cells by targeting CB1 [154]. In addition, an inverse association was found between CB1 and hsa-miRNA-29b-3, indicating that CB1 and hsa-miRNA-29b-3 may crosstalk in pediatric low-grade glioma [155]. These findings suggest that miRNAs may interact with cannabinoid receptors to regulate their function, such as in cancer. However, future studies must establish their interactions in various cancers.
The combined computational and experimental evidence indicates that miR-494 controls CB1 expression in myocardial cells [156]. There was also an association between miRNA let-7d and CB1 expression in several neuronal models [157]. In a spinal cord injury model, miR-338-5p has been shown to target Cnr1, and overexpression of miR-338-5p reduced Cnr1 and provided neuroprotection after spinal cord injury [158]. Because cannabinoids exhibit anti-inflammatory responses, several studies have explored the regulation of miRNAs and their subsequent effects. For example, in obese mice, inhibition of CB1 with the specific antagonist AM251 resulted in increased miR-30e-5p and reduced adipocyte storage [159]. MiRNA-30b has been shown to bind to Cnr1 and reduce its expression in cells transfected with miR-30b mimic [160] (Table 3). However, future studies must identify novel miRNAs that may have a direct role in CB1 expression and establish their interactions in various neurobehavioral functions.

4.2. miRNAs That Modulate Cnr2 Gene Expression

Studies on Cnr2 regulation by miRNA are limited. MiR-187-3p was found to target the 3′ untranslated region (UTR) of the CNR2 gene and inhibit CNR2 expression and differentiation of human osteoblastic precursor cells [161]. The combined computational and experimental evidence suggests that miR-665 controls CB2 expression in myocardial cells [156] (Table 3). These findings support the existence of miRNA-binding regions in the Cnr2 gene and their regulatory role in Cnr2 expression. However, additional studies are warranted to examine whether differential regulation of CB2 expression is controlled by different miRNAs in health and pathological conditions.

4.3. Modulation of miRNAs by CB1 and CB2 Activities

THC, which acts through CB1 and CB2 receptors, has been shown to induce functional myeloid-derived suppressor cells (MDSCs) in vivo. In these studies, cells exhibited several differentially expressed miRNAs. Among these miRNAs, miRNA-690 was found to be highly overexpressed in THC-induced MDSCs. These studies suggested that miR-690-targeting genes are involved in myeloid expansion and differentiation and, therefore, in THC immunosuppression effects [163]. AEA anti-inflammatory action significantly involves interleukin 10 (IL-10) induction in the draining lymph nodes (LNs). This function of AEA was associated with miRNAs that target proinflammatory pathways [164]. THC administration before and after simian immunodeficiency virus (SIV) inoculation ameliorated disease progression. In addition, it reduced inflammation in male rhesus macaques. This neuroprotection likely involves miRNAs that regulate the mRNAs of proteins involved in neurotrophin signaling, MAPK signaling, the cell cycle, and the immune response in the striatum of SIV-infected macaques [165]. The anti-inflammatory property of THC was shown to involve an miRNA cluster, specifically miRNA-18a. miRNA-18a is a target of Pten (phosphatase and tensin homolog, an inhibitor of the PI3K/Akt signaling pathway) and is known to suppress T-regulatory cells [166]. THC treatment inhibited the individual miRNAs in the cluster, reversed the effects of staphylococcal enterotoxin B (SEB) on mortality, and alleviated symptoms of toxic shock [166]. Additionally, THC reduced intestinal inflammation in mouse colitis models and SIV-infected rhesus macaques [167]. This neuroprotective function of THC was associated with selective enhancement of miR-10a, miR-24, miR-99b, miR-145, miR-149, and miR-187 expression, which have been shown to target proinflammatory molecules [167]. The combined administration of THC and CBD augmented murine experimental autoimmune encephalomyelitis (EAE) by reducing neuroinflammation and suppressing Th17 and Th1 cells in a CB1- and CB2-dependent manner. In the same model, studies indicated reduced miR-21a-5p, miR-31-5p, miR-122-5p, miR-146a-5p, miR-150-5p, miR-155-5p, and miR-27b-5p expression while enhancing miR-706-5p and miR-7116 expression [168]. CBD alone was shown to suppress inflammation in an animal model of EAE by modulating several miRNAs [127]. It was shown that prenatal THC exposure enhanced miR-122-5p; reduced the expression of its target, insulin-like growth factor 1 receptor (Igf1r), in adult rat ovary follicular cells; and caused follicular apoptosis [169]. In another study, activation of CB2 by a specific agonist (AM1241) was suggested to protect dopaminergic neurons in Parkinson’s disease animals. This function was associated with increased expression of miR-133b-3p and reduced expression of target genes such as Xist and Pitx3 [162]. These studies suggested that the protective function of cannabinoids involves miRNAs. Future studies establishing insight into the complex interactions between miRNA and mRNA in various cannabinoid actions may aid in untangling the molecular underpinnings of the medicinal value of marijuana.

5. Conclusions

Cannabinoid receptors have been shown to function in health and in a number of neuropsychiatric diseases and pathological conditions in response to various insults. In the current preclinical review, we presented an overview of cannabinoid receptor gene structure that offers potential targets for epigenetic modifications in behavioral and neuropharmacological studies that evaluate cannabinoid receptor expression and epigenetic changes. In addition, we outlined evidence suggesting that the activation and inhibition of cannabinoid receptors and their downstream regulation may involve epigenetic mechanisms that include DNA methylation, histone modifications, and the regulation of miRNA expression. Collectively, these studies support the continued evaluation of cannabinoid receptor regulation by epigenetic modifications and can be potential targets in the treatment of cannabinoid receptor-mediated behavioral and pathological conditions (Figure 1). However, future studies are still warranted to evaluate the direct link between cannabinoid receptor activities and epigenetic mechanisms of action. Thus, epigenetic changes are promising targets for the future development of potential therapeutic agents to treat altered cannabinoid receptors functions.

Author Contributions

B.S.B. and S.S. participated in the collection of literature, writing, and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by NIH/NIAAA grant AA019443 (BSB).

Institutional Review Board Statement

All experiments were performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals (National Institutes of Health Publication. 8th edition, revised 2011), as confirmed by the Nathan Kline Institute Ethical Committee.

Acknowledgments

This study was supported by funds from NIH/NIAAA grant AA019443.

Conflicts of Interest

The authors declare no competing financial interests.

References

  1. 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] [PubMed]
  2. 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] [PubMed] [Green Version]
  3. 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.; et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem. Pharmacol. 1995, 50, 83–90. [Google Scholar] [CrossRef]
  4. 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]
  5. Okamoto, Y.; Morishita, J.; Tsuboi, K.; Tonai, T.; Ueda, N. Molecular Characterization of a Phospholipase D Generating Anandamide and Its Congeners. J. Biol. Chem. 2004, 279, 5298–5305. [Google Scholar] [CrossRef] [Green Version]
  6. 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] [Green Version]
  7. Mechoulam, R. A Delightful Trip Along the Pathway of Cannabinoid and Endocannabinoid Chemistry and Pharmacology. Annu. Rev. Pharmacol. Toxicol. 2022, 63. [Google Scholar] [CrossRef]
  8. Veilleux, A.; Di Marzo, V.; Silvestri, C. The Expanded Endocannabinoid System/Endocannabinoidome as a Potential Target for Treating Diabetes Mellitus. Curr. Diabetes Rep. 2019, 19, 117. [Google Scholar] [CrossRef]
  9. 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]
  10. 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]
  11. Basavarajappa, B.S. Endocannabinoid System and Alcohol Abuse Disorders. In Recent Advances in Cannabinoid Physiology and Pathology; Bukiya, A.N., Ed.; Nature Springer: Cham, Switzerland, 2019; 25p. [Google Scholar]
  12. Basavarajappa, B.S.; Joshi, V.; Shivakumar, M.; Subbanna, S. Distinct functions of endogenous cannabinoid system in alcohol abuse disorders. J. Cereb. Blood Flow Metab. 2019, 176, 3085–3109. [Google Scholar] [CrossRef] [PubMed]
  13. Busquets-García, A.; Bolaños, J.P.; Marsicano, G. Metabolic Messengers: Endocannabinoids. Nat. Metab. 2022, 4, 848–855. [Google Scholar] [CrossRef] [PubMed]
  14. Ledent, C.; Valverde, O.; Cossu, G.; Petitet, F.; Aubert, J.-F.; Beslot, F.; Böhme, G.A.; Imperato, A.; Pedrazzini, T.; Roques, B.P.; et al. Unresponsiveness to Cannabinoids and Reduced Addictive Effects of Opiates in CB 1 Receptor Knockout Mice. Science 1999, 283, 401–404. [Google Scholar] [CrossRef] [PubMed]
  15. Zimmer, A.; Zimmer, A.M.; Hohmann, A.G.; Herkenham, M.; Bonner, T.I. Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout mice. Proc. Natl. Acad. Sci. USA 1999, 96, 5780–5785. [Google Scholar] [CrossRef] [Green Version]
  16. Mackie, K. Cannabinoid receptors as therapeutic targets. Annu. Rev. Pharmacol. Toxicol. 2006, 46, 101–122. [Google Scholar] [CrossRef] [Green Version]
  17. Mechoulam, R. (Ed.) The pharmacohistory of Cannabis sativa. In Cannabinoids as Therapeutic Agents; CRC Press: Boca Raton, FL, USA, 1986. [Google Scholar]
  18. Herkenham, M.; Lynn, A.B.; Little, M.D.; Johnson, M.R.; Melvin, L.S.; de Costa, B.R.; Rice, K.C. Cannabinoid receptor localization in brain. Proc. Natl. Acad. Sci. USA 1990, 87, 1932–1936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Tsou, K.; Brown, S.; Sañudo-Peña, M.; Mackie, K.; Walker, J. Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience 1998, 83, 393–411. [Google Scholar] [CrossRef]
  20. Schlicker, E.; Kathmann, M. Modulation of transmitter release via presynaptic cannabinoid receptors. Trends Pharmacol. Sci. 2001, 22, 565–572. [Google Scholar] [CrossRef]
  21. Wilson, R.I.; Nicoll, R.A. Endocannabinoid Signaling in the Brain. Science 2002, 296, 678–682. [Google Scholar] [CrossRef] [Green Version]
  22. Matias, I.; Di Marzo, V. Endocannabinoid synthesis and degradation, and their regulation in the framework of energy balance. J. Endocrinol. Investig. 2006, 29, 15–26. [Google Scholar]
  23. Lunn, C.A.; Reich, E.P.; Bober, L. Targeting the CB2 receptor for immune modulation. Expert Opin. Ther. Targets 2006, 10, 653–663. [Google Scholar] [CrossRef] [PubMed]
  24. Onaivi, E.S.; Ishiguro, H.; Gong, J.P.; Patel, S.; Perchuk, A.; Meozzi, P.A.; Myers, L.; Mora, Z.; Tagliaferro, P.; Gardner, E.; et al. Discovery of the Presence and Functional Expression of Cannabinoid CB2 Receptors in Brain. Ann. N. Y. Acad. Sci. 2006, 1074, 514–536. [Google Scholar] [CrossRef]
  25. Gong, J.-P.; Onaivi, E.S.; Ishiguro, H.; Liu, Q.-R.; Tagliaferro, P.A.; Brusco, A.; Uhl, G.R. Cannabinoid CB2 receptors: Immunohistochemical localization in rat brain. Brain Res. 2006, 1071, 10–23. [Google Scholar] [CrossRef]
  26. 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 CB 2 Receptors. Science 2005, 310, 329–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Sánchez-Zavaleta, R.; Cortés, H.; Avalos-Fuentes, J.A.; García, U.; Vila, J.S.; Erlij, D.; Florán, B. Presynaptic cannabinoid CB2 receptors modulate [3 H]-Glutamate release at subthalamo-nigral terminals of the rat. Synapse 2018, 72, e22061. [Google Scholar] [CrossRef] [PubMed]
  28. Li, X.; Hua, T.; Vemuri, K.; Ho, J.-H.; Wu, Y.; Wu, L.; Popov, P.; Benchama, O.; Zvonok, N.; Locke, K.; et al. Crystal Structure of the Human Cannabinoid Receptor CB2. Cell 2019, 176, 459–467.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Micale, V.; Stepan, J.; Jurik, A.; Pamplona, F.A.; Marsch, R.; Drago, F.; Eder, M.; Wotjak, C.T. Extinction of avoidance behavior by safety learning depends on endocannabinoid signaling in the hippocampus. J. Psychiatr. Res. 2017, 90, 46–59. [Google Scholar] [CrossRef]
  30. Ruehle, S.; Wager-Miller, J.; Straiker, A.; Farnsworth, J.; Murphy, M.N.; Loch, S.; Monory, K.; Mackie, K.; Lutz, B. Discovery and characterization of two novel CB1 receptor splice variants with modified N-termini in mouse. J. Neurochem. 2017, 142, 521–533. [Google Scholar] [CrossRef] [Green Version]
  31. Hoehe, M.R.; Caenazzo, L.; Martinez, M.M.; Hsieh, W.T.; Modi, W.S.; Gershon, E.S.; Bonner, T.I. Genetic and physical mapping of the human cannabinoid receptor gene to chromosome 6q14-q15. New Biol. 1991, 3, 880–885. [Google Scholar]
  32. McCaw, E.A.; Hu, H.; Gomez, G.T.; Hebb, A.L.; Kelly, M.E.; Denovan-Wright, E.M. Structure, expression and regulation of the cannabinoid receptor gene (CB1) in Huntington’s disease transgenic mice. Eur. J. Biochem. 2004, 271, 4909–4920. [Google Scholar] [CrossRef]
  33. Zhang, P.-W.; Ishiguro, H.; Ohtsuki, T.; Hess, J.; Carillo, F.; Walther, D.; Onaivi, E.S.; Arinami, T.; Uhl, G.R. Human cannabinoid receptor 1: 5′ exons, candidate regulatory regions, polymorphisms, haplotypes and association with polysubstance abuse. Mol. Psychiatry 2004, 9, 916–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Miller, L.K.; Devi, L.A. The Highs and Lows of Cannabinoid Receptor Expression in Disease: Mechanisms and Their Therapeutic Implications. Pharmacol. Rev. 2011, 63, 461–470. [Google Scholar] [CrossRef] [PubMed]
  35. Tsunoda, T.; Takagi, T. Estimating transcription factor bindability on DNA. Bioinformatics 1999, 15, 622–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Tao, R.; Li, C.; Jaffe, A.E.; Shin, J.H.; Deep-Soboslay, A.; Yamin, R.; Weinberger, D.R.; Hyde, T.M.; Kleinman, J.E. Cannabinoid receptor CNR1 expression and DNA methylation in human prefrontal cortex, hippocampus and caudate in brain development and schizophrenia. Transl. Psychiatry 2020, 10, 1–13. [Google Scholar] [CrossRef]
  37. Nicoll, G.; Davidson, S.; Shanley, L.; Hing, B.; Lear, M.; McGuffin, P.; Ross, R.; MacKenzie, A. Allele-specific Differences in Activity of a Novel Cannabinoid Receptor 1 (CNR1) Gene Intronic Enhancer in Hypothalamus, Dorsal Root Ganglia, and Hippocampus. J. Biol. Chem. 2012, 287, 12828–12834. [Google Scholar] [CrossRef] [Green Version]
  38. Hay, E.A.; McEwan, A.; Wilson, D.; Barrett, P.; D’Agostino, G.; Pertwee, R.G.; MacKenzie, A. Disruption of an enhancer associated with addictive behaviour within the cannabinoid receptor-1 gene suggests a possible role in alcohol intake, cannabinoid response and anxiety-related behaviour. Psychoneuroendocrinology 2019, 109, 104407. [Google Scholar] [CrossRef]
  39. Katona, I.; Rancz, E.A.; Acsády, L.; Ledent, C.; Mackie, K.; Hajos, N.; Freund, T.F. Distribution of CB1 Cannabinoid Receptors in the Amygdala and their Role in the Control of GABAergic Transmission. J. Neurosci. 2001, 21, 9506–9518. [Google Scholar] [CrossRef]
  40. Stincic, T.L.; Hyson, R.L. Localization of CB1 cannabinoid receptor mRNA in the brain of the chick (Gallus domesticus). Brain Res. 2008, 1245, 61–73. [Google Scholar] [CrossRef] [Green Version]
  41. Van Waes, V.; Beverley, J.A.; Siman, H.; Tseng, K.Y.; Steiner, H. CB1 Cannabinoid Receptor Expression in the Striatum: Association with Corticostriatal Circuits and Developmental Regulation. Front. Pharmacol. 2012, 3, 21. [Google Scholar] [CrossRef] [Green Version]
  42. Jordan, B.; Devi, L.A. G-protein-coupled receptor heterodimerization modulates receptor function. Nature 1999, 399, 697–700. [Google Scholar] [CrossRef]
  43. Joshi, N.; Onaivi, E.S. Endocannabinoid System Components: Overview and Tissue Distribution. Adv. Exp. Med. Biol. 2019, 1162, 1–12. [Google Scholar] [CrossRef] [PubMed]
  44. Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef] [PubMed]
  45. Onaivi, E.S.; Leonard, C.M.; Ishiguro, H.; Zhang, P.W.; Lin, Z.; Akinshola, B.E.; Uhl, G.R. Endocannabinoids and cannabinoid receptor genetics. Prog. Neurobiol. 2002, 66, 307–344. [Google Scholar] [CrossRef]
  46. Shire, D.; Calandra, B.; Rinaldi-Carmona, M.; Oustric, D.; Pessegue, B.; Bonnin-Cabanne, O.; Le Fur, G.; Caput, D.; Ferrara, P. Molecular cloning, expression and function of the murine CB2 peripheral cannabinoid receptor. Biochim. Biophys. Acta (BBA) Gene Struct. Expr. 1996, 1307, 132–136. [Google Scholar] [CrossRef]
  47. Brown, S.M.; Wager-Miller, J.; Mackie, K. Cloning and molecular characterization of the rat CB2 cannabinoid receptor. Biochim. Biophys. Acta (BBA)-Gene Struct. Expr. 2002, 1576, 255–264. [Google Scholar] [CrossRef]
  48. Chen, D.-J.; Gao, M.; Gao, F.-F.; Su, Q.-X.; Wu, J. Brain cannabinoid receptor 2: Expression, function and modulation. Acta Pharmacol. Sin. 2017, 38, 312–316. [Google Scholar] [CrossRef] [Green Version]
  49. Ishiguro, H.; Kibret, B.G.; Horiuchi, Y.; Onaivi, E.S. Potential Role of Cannabinoid Type 2 Receptors in Neuropsychiatric and Neurodegenerative Disorders. Front. Psychiatry 2022, 13, 828895. [Google Scholar] [CrossRef]
  50. Jordan, C.J.; Xi, Z.-X. Progress in brain cannabinoid CB2 receptor research: From genes to behavior. Neurosci. Biobehav. Rev. 2019, 98, 208–220. [Google Scholar] [CrossRef]
  51. Li, Y.; Kim, J. Neuronal expression of CB2 cannabinoid receptor mRNAs in the mouse hippocampus. Neuroscience 2015, 311, 253–267. [Google Scholar] [CrossRef] [Green Version]
  52. Ghosh, K.; Zhang, G.-F.; Chen, H.; Chen, S.-R.; Pan, H.-L. Cannabinoid CB2 receptors are upregulated via bivalent histone modifications and control primary afferent input to the spinal cord in neuropathic pain. J. Biol. Chem. 2022, 298, 101999. [Google Scholar] [CrossRef]
  53. Liu, Q.R.; Pan, C.H.; Hishimoto, A.; Li, C.Y.; Xi, Z.X.; Llorente-Berzal, A.; Viveros, M.P.; Ishiguro, H.; Arinami, T.; Onaivi, E.S.; et al. Species differences in cannabinoid receptor 2 (CNR2 gene): Identification of novel human and rodent CB2 isoforms, differential tissue expression and regulation by cannabinoid receptor ligands. Genes Brain Behav. 2009, 8, 519–530. [Google Scholar] [CrossRef] [PubMed]
  54. Canseco-Alba, A.; Sanabria, B.; Hammouda, M.; Bernadin, R.; Mina, M.; Liu, Q.-R.; Onaivi, E.S. Cell-Type Specific Deletion of CB2 Cannabinoid Receptors in Dopamine Neurons Induced Hyperactivity Phenotype: Possible Relevance to Attention-Deficit Hyperactivity Disorder. Front. Psychiatry 2022, 12, 803394. [Google Scholar] [CrossRef] [PubMed]
  55. Sherwood, T.A.; Nong, L.; Agudelo, M.; Newton, C.; Widen, R.; Klein, T.W. Identification of Transcription Start Sites and Preferential Expression of Select CB2 Transcripts in Mouse and Human B Lymphocytes. J. Neuroimmune Pharmacol. 2009, 4, 476–488. [Google Scholar] [CrossRef]
  56. Onaivi, E.S.; Ishiguro, H.; Gu, S.; Liu, Q.-R. CNS effects of CB2 cannabinoid receptors: Beyond neuro-immuno-cannabinoid activity. J. Psychopharmacol. 2011, 26, 92–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Galán-Ganga, M.; del Río, R.; Jiménez-Moreno, N.; Díaz-Guerra, M.; Lastres-Becker, I. Cannabinoid CB2 Receptor Modulation by the Transcription Factor NRF2 is Specific in Microglial Cells. Cell. Mol. Neurobiol. 2019, 40, 167–177. [Google Scholar] [CrossRef]
  58. Gomes, T.M.; da Silva, D.D.; Carmo, H.; Carvalho, F.; Silva, J.P. Epigenetics and the endocannabinoid system signaling: An intricate interplay modulating neurodevelopment. Pharmacol. Res. 2020, 162, 105237. [Google Scholar] [CrossRef]
  59. Basavarajappa, B.S.; Subbanna, S. Epigenetic Mechanisms in Developmental Alcohol-Induced Neurobehavioral Deficits. Brain Sci. 2016, 6, 12. [Google Scholar] [CrossRef] [Green Version]
  60. Basavarajappa, B.S.; Subbanna, S. Potential Mechanisms Underlying the Deleterious Effects of Synthetic Cannabinoids Found in Spice/K2 Products. Brain Sci. 2019, 9, 14. [Google Scholar] [CrossRef] [Green Version]
  61. Kukreja, L.; Li, C.J.; Ezhilan, S.; Iyer, V.R.; Kuo, J.S. Emerging Epigenetic Therapies for Brain Tumors. Neuromol. Med. 2021, 24, 41–49. [Google Scholar] [CrossRef]
  62. Murshid, N.M.; Lubis, F.A.; Makpol, S. Epigenetic Changes and Its Intervention in Age-Related Neurodegenerative Diseases. Cell. Mol. Neurobiol. 2020, 42, 577–595. [Google Scholar] [CrossRef]
  63. Morselli, M.; Dieci, G. Epigenetic regulation of human non-coding RNA gene transcription. Biochem. Soc. Trans. 2022, 50, 723–736. [Google Scholar] [CrossRef] [PubMed]
  64. Guo, J.U.; Ma, D.K.; Mo, H.; Ball, M.; Jang, M.-H.; Bonaguidi, M.A.; Balazer, J.A.; Eaves, H.L.; Xie, B.; Ford, E.; et al. Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat. Neurosci. 2011, 14, 1345–1351. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, J.U.; Su, Y.; Zhong, C.; Ming, G.-L.; Song, H. Hydroxylation of 5-Methylcytosine by TET1 Promotes Active DNA Demethylation in the Adult Brain. Cell 2011, 145, 423–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Nabel, C.S.; Kohli, R.M. Demystifying DNA Demethylation. Science 2011, 333, 1229–1230. [Google Scholar] [CrossRef] [PubMed]
  67. Wu, S.C.; Zhang, Y. Active DNA demethylation: Many roads lead to Rome. Nat. Rev. Mol. Cell Biol. 2010, 11, 607–620. [Google Scholar] [CrossRef] [Green Version]
  68. Szulwach, K.E.; Li, X.; Li, Y.; Song, C.X.; Wu, H.; Dai, Q.; Irier, H.; Upadhyay, A.K.; Gearing, M.; Levey, A.I.; et al. 5-hmC-mediated epigenetic dynamics during postnatal neurodevelopment and aging. Nat. Neurosci. 2011, 14, 1607–1616. [Google Scholar] [CrossRef] [Green Version]
  69. Kubiura, M.; Okano, M.; Kimura, H.; Kawamura, F.; Tada, M. Chromosome-wide regulation of euchromatin-specific 5mC to 5hmC conversion in mouse ES cells and female human somatic cells. Chromosom. Res. 2012, 20, 837–848. [Google Scholar] [CrossRef] [Green Version]
  70. Nan, X.; Ng, H.H.; Johnson, C.A.; Laherty, C.D.; Turner, B.M.; Eisenman, R.N.; Bird, A. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 1998, 393, 386–389. [Google Scholar] [CrossRef]
  71. Sarraf, S.A.; Stancheva, I. Methyl-CpG binding protein MBD1 couples histone H3 methylation at lysine 9 by SETDB1 to DNA replication and chromatin assembly. Mol. Cell. 2004, 15, 595–605. [Google Scholar] [CrossRef]
  72. Tao, J.; Hu, K.; Chang, Q.; Wu, H.; Sherman, N.E.; Martinowich, K.; Klose, R.J.; Schanen, C.; Jaenisch, R.; Wang, W.; et al. Phosphorylation of MeCP2 at Serine 80 regulates its chromatin association and neurological function. Proc. Natl. Acad. Sci. USA 2009, 106, 4882–4887. [Google Scholar] [CrossRef] [Green Version]
  73. Zhou, Z.; Hong, E.J.; Cohen, S.; Zhao, W.-N.; Ho, H.-Y.H.; Schmidt, L.; Chen, W.G.; Lin, Y.; Savner, E.; Griffith, E.C.; et al. Brain-Specific Phosphorylation of MeCP2 Regulates Activity-Dependent Bdnf Transcription, Dendritic Growth, and Spine Maturation. Neuron 2006, 52, 255–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Laprairie, R.; Kelly, M.; Denovan-Wright, E. The dynamic nature of type 1 cannabinoid receptor (CB1) gene transcription. J. Cereb. Blood Flow Metab. 2012, 167, 1583–1595. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, D.; Wang, H.; Ning, W.; Backlund, M.G.; Dey, S.K.; DuBois, R.N. Loss of Cannabinoid Receptor 1 Accelerates Intestinal Tumor Growth. Cancer Res. 2008, 68, 6468–6476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Xia, D.; Wang, D.; Kim, S.-H.; Katoh, H.; Dubois, R.N. Prostaglandin E2 promotes intestinal tumor growth via DNA methylation. Nat. Med. 2012, 18, 224–226. [Google Scholar] [CrossRef]
  77. Franklin, T.B.; Russig, H.; Weiss, I.C.; Gräff, J.; Linder, N.; Michalon, A.; Vizi, S.; Mansuy, I.M. Epigenetic Transmission of the Impact of Early Stress Across Generations. Biol. Psychiatry 2010, 68, 408–415. [Google Scholar] [CrossRef]
  78. Viveros, M.-P.; Marco, E.-M.; Llorente, R.; López-Gallardo, M. Endocannabinoid System and Synaptic Plasticity: Implications for Emotional Responses. Neural Plast. 2007, 2007, 1–12. [Google Scholar] [CrossRef] [Green Version]
  79. Börner, C.; Martella, E.; Höllt, V.; Kraus, J. Regulation of Opioid and Cannabinoid Receptor Genes in Human Neuroblastoma and T Cells by the Epigenetic Modifiers Trichostatin A and 5-Aza-2′-Deoxycytidine. Neuroimmunomodulation 2012, 19, 180–186. [Google Scholar] [CrossRef]
  80. Rotter, A.; Bayerlein, K.; Hansbauer, M.; Weiland, J.; Sperling, W.; Kornhuber, J.; Biermann, T. CB1 and CB2 Receptor Expression and Promoter Methylation in Patients with Cannabis Dependence. Eur. Addict. Res. 2012, 19, 13–20. [Google Scholar] [CrossRef] [Green Version]
  81. Di Francesco, A.; Falconi, A.; Di Germanio, C.; Di Bonaventura, M.V.M.; Costa, A.; Caramuta, S.; Del Carlo, M.; Compagnone, D.; Dainese, E.; Cifani, C.; et al. Extravirgin olive oil up-regulates CB1 tumor suppressor gene in human colon cancer cells and in rat colon via epigenetic mechanisms. J. Nutr. Biochem. 2015, 26, 250–258. [Google Scholar] [CrossRef]
  82. Hong, S.; Zheng, G.; Wiley, J.W. Epigenetic Regulation of Genes That Modulate Chronic Stress-Induced Visceral Pain in the Peripheral Nervous System. Gastroenterology 2015, 148, 148–157.e7. [Google Scholar] [CrossRef] [Green Version]
  83. D’Addario, C.; Micale, V.; Di Bartolomeo, M.; Stark, T.; Pucci, M.; Sulcova, A.; Palazzo, M.; Babinska, Z.; Cremaschi, L.; Drago, F.; et al. A preliminary study of endocannabinoid system regulation in psychosis: Distinct alterations of CNR1 promoter DNA methylation in patients with schizophrenia. Schizophr. Res. 2017, 188, 132–140. [Google Scholar] [CrossRef] [PubMed]
  84. Pucci, M.; Di Bonaventura, M.V.M.; Vezzoli, V.; Zaplatic, E.; Massimini, M.; Mai, S.; Sartorio, A.; Scacchi, M.; Persani, L.; Maccarrone, M.; et al. Preclinical and Clinical Evidence for a Distinct Regulation of Mu Opioid and Type 1 Cannabinoid Receptor Genes Expression in Obesity. Front. Genet. 2019, 10, 523. [Google Scholar] [CrossRef] [PubMed]
  85. D’Addario, C.; Ms, E.Z.; Giunti, E.; Pucci, M.; Di Bonaventura, M.V.M.; Scherma, M.; Dainese, E.; Maccarrone, M.; Nilsson, I.A.; Cifani, C.; et al. Epigenetic regulation of the cannabinoid receptor CB1 in an activity-based rat model of anorexia nervosa. Int. J. Eat. Disord. 2020, 53, 702–716. [Google Scholar] [CrossRef] [PubMed]
  86. Mancino, S.; Burokas, A.; Gutiérrez-Cuesta, J.; Gutiérrez-Martos, M.; Martín-García, E.; Pucci, M.; Falconi, A.; D’Addario, C.; Maccarrone, M.; Maldonado, R. Epigenetic and Proteomic Expression Changes Promoted by Eating Addictive-Like Behavior. Neuropsychopharmacology 2015, 40, 2788–2800. [Google Scholar] [CrossRef] [Green Version]
  87. Subbanna, S.; Shivakumar, M.; Psychoyos, D.; Xie, S.; Basavarajappa, B.S. Anandamide-CB1 Receptor Signaling Contributes to Postnatal Ethanol-Induced Neonatal Neurodegeneration, Adult Synaptic, and Memory Deficits. J. Neurosci. 2013, 33, 6350–6366. [Google Scholar] [CrossRef] [Green Version]
  88. Nagre, N.N.; Subbanna, S.; Shivakumar, M.; Psychoyos, D.; Basavarajappa, B.S. CB1-receptor knockout neonatal mice are protected against ethanol-induced impairments of DNMT1, DNMT3A, and DNA methylation. J. Neurochem. 2015, 132, 429–442. [Google Scholar] [CrossRef] [Green Version]
  89. Smith, R.C.; Sershen, H.; Janowsky, D.S.; Lajtha, A.; Grieco, M.; Gangoiti, J.A.; Gertsman, I.; Johnson, W.S.; Marcotte, T.D.; Davis, J.M. Changes in Expression of DNA-Methyltransferase and Cannabinoid Receptor mRNAs in Blood Lymphocytes After Acute Cannabis Smoking. Front. Psychiatry 2022, 13, 887700. [Google Scholar] [CrossRef]
  90. Innocenzi, E.; De Domenico, E.; Ciccarone, F.; Zampieri, M.; Rossi, G.; Cicconi, R.; Bernardini, R.; Mattei, M.; Grimaldi, P. Paternal activation of CB2 cannabinoid receptor impairs placental and embryonic growth via an epigenetic mechanism. Sci. Rep. 2019, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  91. Parastouei, K.; Aarabi, M.H.; Hamidi, G.A.; Nasehi, Z.; Arani, S.K.; Jozi, F.; Shahabodin, M.E. A CB2 Receptor Agonist Reduces the Production of Inflammatory Mediators and Improves Locomotor Activity in Experimental Autoimmune Encephalomyelitis. Rep. Biochem. Mol. Biol. 2022, 11, 1–9. [Google Scholar] [CrossRef]
  92. Strisciuglio, C.; Creoli, M.; Tortora, C.; Martinelli, M.; Miele, E.; Paino, S.; Luongo, L.; Rossi, F. Increased expression of CB2 receptor in the intestinal biopsies of children with inflammatory bowel disease. Pediatr. Res. 2022, 1–6. [Google Scholar] [CrossRef]
  93. Kiran, S.; Rakib, A.; Moore, B.M.; Singh, U.P. Cannabinoid Receptor 2 (CB2) Inverse Agonist SMM-189 Induces Expression of Endogenous CB2 and Protein Kinase A That Differentially Modulates the Immune Response and Suppresses Experimental Colitis. Pharmaceutics 2022, 14, 936. [Google Scholar] [CrossRef]
  94. Ten-Blanco, M.; Flores, Á.; Pereda-Pérez, I.; Piscitelli, F.; Izquierdo-Luengo, C.; Cristino, L.; Romero, J.; Hillard, C.J.; Maldonado, R.; Di Marzo, V.; et al. Amygdalar CB2 cannabinoid receptor mediates fear extinction deficits promoted by orexin-A/hypocretin-1. Biomed. Pharmacother. 2022, 149, 112925. [Google Scholar] [CrossRef]
  95. Jayanthi, S.; Peesapati, R.; McCoy, M.T.; Ladenheim, B.; Cadet, J.L. Footshock-Induced Abstinence from Compulsive Methamphetamine Self-administration in Rat Model Is Accompanied by Increased Hippocampal Expression of Cannabinoid Receptors (CB1 and CB2). Mol. Neurobiol. 2022, 59, 1238–1248. [Google Scholar] [CrossRef]
  96. Paradisi, A.; Pasquariello, N.; Barcaroli, D.; Maccarrone, M. Anandamide Regulates Keratinocyte Differentiation by Inducing DNA Methylation in a CB1 Receptor-dependent Manner. J. Biol. Chem. 2008, 283, 6005–6012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Molina, P.E.; Amedee, A.; LeCapitaine, N.J.; Zabaleta, J.; Mohan, M.; Winsauer, P.; Stouwe, C.V. Cannabinoid Neuroimmune Modulation of SIV Disease. J. Neuroimmune Pharmacol. 2011, 6, 516–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Gobira, P.H.; Roncalho, A.L.; Silva, N.R.; Silote, G.P.; Sales, A.J.; Joca, S.R. Adolescent cannabinoid exposure modulates the vulnerability to cocaine-induced conditioned place preference and DNMT3a expression in the prefrontal cortex in Swiss mice. Psychopharmacology 2021, 238, 3107–3118. [Google Scholar] [CrossRef] [PubMed]
  99. Schrott, R.; Rajavel, M.; Acharya, K.; Huang, Z.; Acharya, C.; Hawkey, A.; Pippen, E.; Lyerly, H.K.; Levin, E.D.; Murphy, S.K. Sperm DNA methylation altered by THC and nicotine: Vulnerability of neurodevelopmental genes with bivalent chromatin. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef] [PubMed]
  100. Jahn, K.; Heese, A.; Kebir, O.; Groh, A.; Bleich, S.; Krebs, M.O.; Frieling, H. Differential Methylation Pattern of Schizophrenia Candidate Genes in Tetrahydrocannabinol-Consuming Treatment-Resistant Schizophrenic Patients Compared to Non-Consumer Patients and Healthy Controls. Neuropsychobiology 2020, 80, 36–44. [Google Scholar] [CrossRef]
  101. Tomas-Roig, J.; Benito, E.; Agis-Balboa, R.; Piscitelli, F.; Hoyer-Fender, S.; Di Marzo, V.; Havemann-Reinecke, U. Chronic exposure to cannabinoids during adolescence causes long-lasting behavioral deficits in adult mice. Addict. Biol. 2016, 22, 1778–1789. [Google Scholar] [CrossRef] [PubMed]
  102. Chen, D.; Wu, H.; Feng, X.; Chen, Y.; Lv, Z.; Kota, V.G.; Chen, J.; Wu, W.; Lu, Y.; Liu, H.; et al. DNA Methylation of Cannabinoid Receptor Interacting Protein 1 Promotes Pathogenesis of Intrahepatic Cholangiocarcinoma Through Suppressing Parkin-Dependent Pyruvate Kinase M2 Ubiquitination. Hepatology 2020, 73, 1816–1835. [Google Scholar] [CrossRef]
  103. Subbanna, S.; Nagre, N.N.; Shivakumar, M.; Joshi, V.; Psychoyos, D.; Kutlar, A.; Umapathy, N.S.; Basavarajappa, B.S. CB1R-Mediated Activation of Caspase-3 Causes Epigenetic and Neurobehavioral Abnormalities in Postnatal Ethanol-Exposed Mice. Front. Mol. Neurosci. 2018, 11, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Pedrazzi, J.F.; Sales, A.J.; Guimarães, F.S.; Joca, S.R.; Crippa, J.A.; Del Bel, E. Cannabidiol prevents disruptions in sensorimotor gating induced by psychotomimetic drugs that last for 24-h with probable involvement of epigenetic changes in the ventral striatum. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 111, 110352. [Google Scholar] [CrossRef] [PubMed]
  105. Wanner, N.M.; Colwell, M.; Drown, C.; Faulk, C. Subacute cannabidiol alters genome-wide DNA methylation in adult mouse hippocampus. Environ. Mol. Mutagen. 2020, 61, 890–900. [Google Scholar] [CrossRef]
  106. Elliott, E.; Manashirov, S.; Zwang, R.; Gil, S.; Tsoory, M.; Shemesh, Y.; Chen, A. Dnmt3a in the Medial Prefrontal Cortex Regulates Anxiety-Like Behavior in Adult Mice. J. Neurosci. 2016, 36, 730–740. [Google Scholar] [CrossRef] [Green Version]
  107. Henikoff, S.; Smith, M.M. Histone Variants and Epigenetics. Cold Spring Harb. Perspect. Biol. 2015, 7, a019364. [Google Scholar] [CrossRef] [Green Version]
  108. Park, J.; Lee, K.; Kim, K.; Yi, S.-J. The role of histone modifications: From neurodevelopment to neurodiseases. Signal Transduct. Target. Ther. 2022, 7, 1–23. [Google Scholar] [CrossRef]
  109. Kornberg, R.D.; Lorch, Y. Twenty-Five Years of the Nucleosome, Fundamental Particle of the Eukaryote Chromosome. Cell 1999, 98, 285–294. [Google Scholar] [CrossRef] [Green Version]
  110. Lorch, Y.; Zhang, M.; Kornberg, R.D. Histone Octamer Transfer by a Chromatin-Remodeling Complex. Cell 1999, 96, 389–392. [Google Scholar] [CrossRef] [Green Version]
  111. Goll, M.G.; Bestor, T.H. Histone modification and replacement in chromatin activation: Figure 1. Genes Dev. 2002, 16, 1739–1742. [Google Scholar] [CrossRef] [Green Version]
  112. Grant, P.A. A tale of histone modifications. Genome Biol. 2001, 2, 1–6. [Google Scholar] [CrossRef]
  113. Smolle, M.; Workman, J.L. Transcription-associated histone modifications and cryptic transcription. Biochim. Biophys. Acta 2012, 1829, 84–97. [Google Scholar] [CrossRef] [PubMed]
  114. Joseph, F.M.; Young, N.L. Histone variant-specific post-translational modifications. Semin. Cell Dev. Biol. 2022. [Google Scholar] [CrossRef] [PubMed]
  115. Lomazzo, E.; König, F.; Abassi, L.; Jelinek, R.; Lutz, B. Chronic stress leads to epigenetic dysregulation in the neuropeptide-Y and cannabinoid CB1 receptor genes in the mouse cingulate cortex. Neuropharmacology 2017, 113, 301–313. [Google Scholar] [CrossRef] [PubMed]
  116. Subbanna, S.; Nagre, N.N.; Umapathy, N.S.; Pace, B.; Basavarajappa, B.S. Ethanol Exposure Induces Neonatal Neurodegeneration by Enhancing CB1R Exon1 Histone H4K8 Acetylation and Up-regulating CB1R Function causing Neurobehavioral Abnormalities in Adult Mice. Int. J. Neuropsychopharmacol. 2014, 18, pyu028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Shivakumar, M.; Subbanna, S.; Joshi, V.; Basavarajappa, B.S. Postnatal Ethanol Exposure Activates HDAC-Mediated Histone Deacetylation, Impairs Synaptic Plasticity Gene Expression and Behavior in Mice. Int. J. Neuropsychopharmacol. 2020, 23, 324–338. [Google Scholar] [CrossRef]
  118. Luo, Y.; Zhang, J.; Chen, L.; Chen, S.-R.; Chen, H.; Zhang, G.; Pan, H.-L. Histone methyltransferase G9a diminishes expression of cannabinoid CB1 receptors in primary sensory neurons in neuropathic pain. J. Biol. Chem. 2020, 295, 3553–3562. [Google Scholar] [CrossRef]
  119. Nogueira, D.D.S.; Bourdy, R.; Alcala-Vida, R.; Filliol, D.; Andry, V.; Goumon, Y.; Zwiller, J.; Romieu, P.; Merienne, K.; Olmstead, M.C.; et al. Hippocampal Cannabinoid 1 Receptors Are Modulated Following Cocaine Self-administration in Male Rats. Mol. Neurobiol. 2022, 59, 1896–1911. [Google Scholar] [CrossRef]
  120. De Sa Nogueira, D.; Merienne, K.; Befort, K. Neuroepigenetics and addictive behaviors: Where do we stand? Neurosci. Biobehav. Rev. 2019, 106, 58–72. [Google Scholar] [CrossRef]
  121. Renthal, W.; Nestler, E.J. Epigenetic mechanisms in drug addiction. Trends Mol. Med. 2008, 14, 341–350. [Google Scholar] [CrossRef] [Green Version]
  122. Dhopeshwarkar, A.; Mackie, K. CB2 Cannabinoid receptors as a therapeutic target-what does the future hold? Mol. Pharmacol. 2014, 86, 430–437. [Google Scholar] [CrossRef] [Green Version]
  123. Tang, Y.; Wolk, B.; Britch, S.C.; Craft, R.M.; Kendall, D.A. Anti-inflammatory and antinociceptive effects of the selective cannabinoid CB2 receptor agonist ABK5. J. Pharmacol. Sci. 2020, 145, 319–326. [Google Scholar] [CrossRef] [PubMed]
  124. Yang, X.; VHegde, L.; Rao, R.; Zhang, J.; Nagarkatti, P.S.; Nagarkatti, M. Histone modifications are associated with Delta9-tetrahydrocannabinol-mediated alterations in antigen-specific T cell responses. J. Biol. Chem. 2014, 289, 18707–18718. [Google Scholar] [CrossRef] [PubMed]
  125. Prini, P.; Rusconi, F.; Zamberletti, E.; Gabaglio, M.; Penna, F.; Fasano, M.; Battaglioli, E.; Parolaro, D.; Rubino, T. Adolescent THC exposure in female rats leads to cognitive deficits through a mechanism involving chromatin modifications in the prefrontal cortex. J. Psychiatry Neurosci. 2018, 43, 87–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Bilkei-Gorzo, A.; Albayram, O.; Draffehn, A.; Michel, K.; Piyanova, A.; Oppenheimer, H.; Dvir-Ginzberg, M.; Rácz, I.; Ulas, T.; Imbeault, S.; et al. A chronic low dose of Delta(9)-tetrahydrocannabinol (THC) restores cognitive function in old mice. Nat. Med. 2017, 23, 782–787. [Google Scholar] [CrossRef]
  127. Yang, X.; Bam, M.; Nagarkatti, P.S.; Nagarkatti, M. Cannabidiol Regulates Gene Expression in Encephalitogenic T cells Using Histone Methylation and noncoding RNA during Experimental Autoimmune Encephalomyelitis. Sci. Rep. 2019, 9, 15780. [Google Scholar] [CrossRef] [Green Version]
  128. Todd, S.M.; Zhou, C.; Clarke, D.J.; Chohan, T.W.; Bahceci, D.; Arnold, J.C. Interactions between cannabidiol and Delta(9)-THC following acute and repeated dosing: Rebound hyperactivity, sensorimotor gating and epigenetic and neuroadaptive changes in the mesolimbic pathway. Eur. Neuropsychopharmacol. 2017, 27, 132–145. [Google Scholar] [CrossRef]
  129. Pastrana-Trejo, J.C.; Duarte-Aké, F.; Us-Camas, R.; De-La-Peña, C.; Parker, L.; Pertwee, R.G.; Murillo-Rodríguez, E. Effects on the Post-translational Modification of H3K4Me3, H3K9ac, H3K9Me2, H3K27Me3, and H3K36Me2 Levels in Cerebral Cortex, Hypothalamus and Pons of Rats after a Systemic Administration of Cannabidiol: A Preliminary Study. Central Nerv. Syst. Agents Med. Chem. 2021, 21, 142–147. [Google Scholar] [CrossRef]
  130. Hunter, R.G.; McCarthy, K.J.; Milne, T.A.; Pfaff, D.W.; McEwen, B.S. Regulation of hippocampal H3 histone methylation by acute and chronic stress. Proc. Natl. Acad. Sci. USA 2009, 106, 20912–20917. [Google Scholar] [CrossRef] [Green Version]
  131. Tran, L.; Schulkin, J.; Ligon, C.O.; Meerveld, B.G.-V. Epigenetic modulation of chronic anxiety and pain by histone deacetylation. Mol. Psychiatry 2014, 20, 1219–1231. [Google Scholar] [CrossRef]
  132. Weaver, I.C.G.; Cervoni, N.; Champagne, F.A.; D’Alessio, A.C.; Sharma, S.; Seckl, J.R.; Dymov, S.; Szyf, M.; Meaney, M.J. Epigenetic programming by maternal behavior. Nat. Neurosci. 2004, 7, 847–854. [Google Scholar] [CrossRef]
  133. Prini, P.; Penna, F.; Sciuccati, E.; Alberio, T.; Rubino, T. Chronic Delta(8)-THC Exposure Differently Affects Histone Modifications in the Adolescent and Adult Rat Brain. Int. J. Mol. Sci. 2017, 18, 2094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  135. Vasudevan, S. Posttranscriptional Upregulation by MicroRNAs. Wiley Interdiscip. Rev. RNA 2011, 3, 311–330. [Google Scholar] [CrossRef] [PubMed]
  136. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Martinez, N.J.; Gregory, R.I. MicroRNA Gene Regulatory Pathways in the Establishment and Maintenance of ESC Identity. Cell Stem Cell 2010, 7, 31–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Hayes, J.; Peruzzi, P.P.; Lawler, S. MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol. Med. 2014, 20, 460–469. [Google Scholar] [CrossRef]
  139. Huang, W. MicroRNAs: Biomarkers, Diagnostics, and Therapeutics. Methods Mol. Biol. 2017, 1617, 57–67. [Google Scholar]
  140. Wang, J.; Chen, J.; Sen, S. MicroRNA as Biomarkers and Diagnostics. J. Cell. Physiol. 2015, 231, 25–30. [Google Scholar] [CrossRef]
  141. Paul, P.; Chakraborty, A.; Sarkar, D.; Langthasa, M.; Rahman, M.; Bari, M.; Singha, R.S.; Malakar, A.K.; Chakraborty, S. Interplay between miRNAs and human diseases. J. Cell. Physiol. 2017, 233, 2007–2018. [Google Scholar] [CrossRef]
  142. Tüfekci, K.U.; Öner, M.G.; Meuwissen, R.L.J.; Genç, Ş. The Role of MicroRNAs in Human Diseases. In miRNomics: MicroRNA Biology and Computational Analysis; Humana Press: Totowa, NJ, USA, 2013; pp. 33–50. [Google Scholar] [CrossRef]
  143. Roy, B.; Lee, E.; Li, T.; Rampersaud, M. Role of miRNAs in Neurodegeneration: From Disease Cause to Tools of Biomarker Discovery and Therapeutics. Genes 2022, 13, 425. [Google Scholar] [CrossRef]
  144. Leitão, A.L.; Enguita, F.J. A Structural View of miRNA Biogenesis and Function. Non-Coding RNA 2022, 8, 10. [Google Scholar] [CrossRef] [PubMed]
  145. Lee, C.; Han, J.; Jung, Y. Pathological Contribution of Extracellular Vesicles and Their MicroRNAs to Progression of Chronic Liver Disease. Biology 2022, 11, 637. [Google Scholar] [CrossRef] [PubMed]
  146. Xie, S.; Zhang, Q.; Jiang, L. Current Knowledge on Exosome Biogenesis, Cargo-Sorting Mechanism and Therapeutic Implications. Membranes 2022, 12, 498. [Google Scholar] [CrossRef] [PubMed]
  147. Kuwabara, T.; Hsieh, J.; Nakashima, K.; Taira, K.; Gage, F.H. A Small Modulatory dsRNA Specifies the Fate of Adult Neural Stem Cells. Cell 2004, 116, 779–793. [Google Scholar] [CrossRef] [Green Version]
  148. Poole, R.J.; Hobert, O. Early Embryonic Programming of Neuronal Left/Right Asymmetry in C. elegans. Curr. Biol. 2006, 16, 2279–2292. [Google Scholar] [CrossRef] [Green Version]
  149. Ronshaugen, M.; Biemar, F.; Piel, J.; Levine, M.; Lai, E.C. The Drosophila microRNA iab-4 causes a dominant homeotic transformation of halteres to wings. Genes Dev. 2005, 19, 2947–2952. [Google Scholar] [CrossRef] [Green Version]
  150. Schratt, G.M.; Tuebing, F.; Nigh, E.A.; Kane, C.G.; Sabatini, M.E.; Kiebler, M.; Greenberg, M.E. A brain-specific microRNA regulates dendritic spine development. Nature 2006, 439, 283–289. [Google Scholar] [CrossRef]
  151. Olde Loohuis, N.F.; Kos, A.; Martens, G.J.; Van Bokhoven, H.; Nadif Kasri, N.; Aschrafi, A. MicroRNA networks direct neuronal development and plasticity. Cell. Mol. Life Sci. 2012, 69, 89–102. [Google Scholar] [CrossRef] [Green Version]
  152. Davis, T.H.; Cuellar, T.L.; Koch, S.M.; Barker, A.J.; Harfe, B.D.; McManus, M.T.; Ullian, E.M. Conditional Loss of Dicer Disrupts Cellular and Tissue Morphogenesis in the Cortex and Hippocampus. J. Neurosci. 2008, 28, 4322–4330. [Google Scholar] [CrossRef] [Green Version]
  153. Stark, K.L.; Xu, B.; Bagchi, A.; Lai, W.-S.; Liu, H.; Hsu, R.; Wan, X.; Pavlidis, P.; Mills, A.A.; Karayiorgou, M.; et al. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat. Genet. 2008, 40, 751–760. [Google Scholar] [CrossRef]
  154. Li, M.; Qian, X.; Zhu, M.; Li, A.; Fang, M.; Zhu, Y.; Zhang, J. miR1273g3p promotes proliferation, migration and invasion of LoVo cells via cannabinoid receptor 1 through activation of ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Mol. Med. Rep. 2018, 17, 4619–4626. [Google Scholar] [PubMed] [Green Version]
  155. Sredni, S.T.; Huang, C.-C.; Suzuki, M.; Pundy, T.; Chou, P.; Tomita, T. Spontaneous involution of pediatric low-grade gliomas: High expression of cannabinoid receptor 1 (CNR1) at the time of diagnosis may indicate involvement of the endocannabinoid system. Child’s Nerv. Syst. 2016, 32, 2061–2067. [Google Scholar] [CrossRef] [PubMed]
  156. Möhnle, P.; Schütz, S.V.; Schmidt, M.; Hinske, C.; Hübner, M.; Heyn, J.; Beiras-Fernandez, A.; Kreth, S. MicroRNA-665 is involved in the regulation of the expression of the cardioprotective cannabinoid receptor CB2 in patients with severe heart failure. Biochem. Biophys. Res. Commun. 2014, 451, 516–521. [Google Scholar] [CrossRef] [PubMed]
  157. Chiarlone, A.; Börner, C.; Martín-Gómez, L.; Jiménez-González, A.; García-Concejo, A.; García-Bermejo, M.L.; Lorente, M.; Blázquez, C.; García-Taboada, E.; de Haro, A.; et al. MicroRNA let-7d is a target of cannabinoid CB 1 receptor and controls cannabinoid signaling. Neuropharmacology 2016, 108, 345–352. [Google Scholar] [CrossRef] [PubMed]
  158. Zhang, A.; Bai, Z.; Yi, W.; Hu, Z.; Hao, J. Overexpression of miR-338-5p in exosomes derived from mesenchymal stromal cells provides neuroprotective effects by the Cnr1/Rap1/Akt pathway after spinal cord injury in rats. Neurosci. Lett. 2021, 761, 136124. [Google Scholar] [CrossRef]
  159. Miranda, K.; Mehrpouya-Bahrami, P.; Nagarkatti, P.S.; Nagarkatti, M. Cannabinoid Receptor 1 Blockade Attenuates Obesity and Adipose Tissue Type 1 Inflammation Through miR-30e-5p Regulation of Delta-Like-4 in Macrophages and Consequently Downregulation of Th1 Cells. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef]
  160. Li, L.; Xu, Y.; Zhao, M.; Gao, Z. Neuro-protective roles of long non-coding RNA MALAT1 in Alzheimer’s disease with the involvement of the microRNA-30b/CNR1 network and the following PI3K/AKT activation. Exp. Mol. Pathol. 2020, 117, 104545. [Google Scholar] [CrossRef]
  161. Xu, A.; Yang, Y.; Shao, Y.; Wu, M.; Sun, Y. Inhibiting effect of microRNA-187-3p on osteogenic differentiation of osteoblast precursor cells by suppressing cannabinoid receptor type 2. Differentiation 2019, 109, 9–15. [Google Scholar] [CrossRef]
  162. He, X.; Yang, L.; Huang, R.; Lin, L.; Shen, Y.; Cheng, L.; Jin, L.; Wang, S.; Zhu, R. Activation of CB2R with AM1241 ameliorates neurodegeneration via the Xist/miR-133b-3p/Pitx3 axis. J. Cell. Physiol. 2020, 235, 6032–6042. [Google Scholar] [CrossRef]
  163. Hegde, V.L.; Tomar, S.; Jackson, A.; Rao, R.; Yang, X.; Singh, U.P.; Singh, N.P.; Nagarkatti, P.S.; Nagarkatti, M. Distinct microRNA expression profile and targeted biological pathways in functional myeloid-derived suppressor cells induced by Delta9-tetrahydrocannabinol in vivo: Regulation of CCAAT/enhancer-binding protein alpha by microRNA-690. J. Biol. Chem. 2013, 288, 36810–36826. [Google Scholar] [CrossRef] [Green Version]
  164. Jackson, A.R.; Nagarkatti, P.; Nagarkatti, M. Anandamide Attenuates Th-17 Cell-Mediated Delayed-Type Hypersensitivity Response by Triggering IL-10 Production and Consequent microRNA Induction. PLoS ONE 2014, 9, e93954. [Google Scholar] [CrossRef] [PubMed]
  165. Simon, L.; Song, K.; Stouwe, C.V.; Hollenbach, A.; Amedee, A.; Mohan, M.; Winsauer, P.; Molina, P. Delta9-Tetrahydrocannabinol (Delta9-THC) Promotes Neuroimmune-Modulatory MicroRNA Profile in Striatum of Simian Immunodeficiency Virus (SIV)-Infected Macaques. J. Neuroimmune Pharmacol. 2016, 11, 192–213. [Google Scholar] [CrossRef] [PubMed]
  166. Rao, R.; Nagarkatti, P.S.; Nagarkatti, M. Delta(9) Tetrahydrocannabinol attenuates Staphylococcal enterotoxin B-induced inflammatory lung injury and prevents mortality in mice by modulation of miR-17-92 cluster and induction of T-regulatory cells. Br. J. Pharmacol. 2015, 172, 1792–1806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Chandra, L.C.; Kumar, V.; Torben, W.; Stouwe, C.V.; Winsauer, P.; Amedee, A.; Molina, P.E.; Mohan, M. Chronic administration of Delta9-tetrahydrocannabinol induces intestinal anti-inflammatory microRNA expression during acute simian immunodeficiency virus infection of rhesus macaques. J. Virol. 2015, 89, 1168–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Al-Ghezi, Z.Z.; Miranda, K.; Nagarkatti, M.; Nagarkatti, P.S. Combination of Cannabinoids, Delta9- Tetrahydrocannabinol and Cannabidiol, Ameliorates Experimental Multiple Sclerosis by Suppressing Neuroinflammation Through Regulation of miRNA-Mediated Signaling Pathways. Front. Immunol. 2019, 10, 1921. [Google Scholar] [CrossRef]
  169. Martínez-Peña, A.A.; Lee, K.; Pereira, M.; Ayyash, A.; Petrik, J.J.; Hardy, D.B.; Holloway, A.C. Prenatal Exposure to Delta-9-tetrahydrocannabinol (THC) Alters the Expression of miR-122-5p and Its Target Igf1r in the Adult Rat Ovary. Int. J. Mol. Sci. 2022, 23, 8000. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Epigenetic mechanisms epitomize important gene–environment relation mediators behind many human disorders’ pathogenesis. Various adverse conditions such as drug abuse, including alcohol, cannabinoids, and adverse developmental conditions may potentially affect the expression of genes such as cannabinoid receptors, causing the altered functions, at least through epigenetic modifications.
Figure 1. Epigenetic mechanisms epitomize important gene–environment relation mediators behind many human disorders’ pathogenesis. Various adverse conditions such as drug abuse, including alcohol, cannabinoids, and adverse developmental conditions may potentially affect the expression of genes such as cannabinoid receptors, causing the altered functions, at least through epigenetic modifications.
Biomolecules 12 01560 g001
Table 1. DNA methylation and CB1 receptor gene (Cnr1) expression.
Table 1. DNA methylation and CB1 receptor gene (Cnr1) expression.
ModelTreatment/ExposureDNA Methylation at Cnr1 PromoterCnr1 Gene ExpressionReference
Colon cancer specimens-[75]
Human epithelial colon cell line LS-174TProstaglandin E2[76]
Rodents
(PD 1 to 14)
Maternal separation-[77]
Jurkat cells5-Aza-dC-[79]
HumansTHC[80]
Human colon cancer cells and ratsExtra-virgin olive oil (EVOO).
Phenolic extracts (OPE)
Hydroxytyrosol (HT)
[81]
Rats
L6-S2 (DRG)
Chronic stress[82]
Mice (PD 7)Alcohol[87]
[88]
Schizophrenic patients-[83]
Rat (PFC)Methylazoxymethanol acetate exposure[83]
Human
blood mononuclear cells from younger (<30 years old) human obese subjects
THC/alcohol-[84]
Rat modelAnorexia[85]
Schizophrenia patients-[36]
ND, not determined.
Table 2. Histone modifications at the CB1 receptor gene (Cnr1).
Table 2. Histone modifications at the CB1 receptor gene (Cnr1).
ModelTreatmentHistone ModificationsGene PromoterReference
Mice
(Cingulate cortex)
Chronic unpredictable stress-Increased HDAC-2
-Decreased levels H3K9ac
Cnr1[115]
PD-7 MiceAlcohol-Increased H4K8ac
-Decreased H3K9me2
Cnr1[116]
PD-7 MiceAlcohol-Increased HDAC-1, HDAC-2, and HDAC-3Egr1, Arc[117]
Ratsneuropathic pain-Increased H3K9me2Cnr1[118]
Male RatsCocaine self-administration-Increased H3K4me3, H3K27acCnr1[119,125]
Female adolescent ratsTHC-Increased H3K9me3
-Increased Suv39H1, histone lysine methyltransferase,
ND[125]
Female adolescent ratsTHC-Increased H3K14ac and H3K9me2ND[125,133]
MiceTHCIncreased global H3K9ac, H4K12ac and reduced H3K9me3Klotho and Bdnf[126]
THC and HAT inhibitorNDDecreased H3K9ac, synapsin 1, Klotho, and Bdnf expression
CD4+ T cellsCBDIncreased H3K4me3 and reduced H3K27me3IL-4, IL-5, and IL-13[127]
Adult male miceCBD
and
THC
Increased H3K9ac and H3K14ac levelsND[128]
Adult rats
Cerebral cortex
Hypothalamus
Pons
CBD-Increased H3K4me3, H3K9ac, and H3K27me3 levels
-Decreased H3K9ac levels
-Decreased H3K4me3 levels
ND[129]
RatsAcute stress-Increased H3K9me3
-Decreased H3K27me3 and H3K9me1 levels
ND[130]
Chronic stress-Increased H3K9me3 and
decreased H3K27me3 and H3K4me3 levels
CBD-Increased H3K9ac levels
Prolonged exposure to corticosteroidsChronic anxiety and pain-H3K9ac levels in the central amygdalaND[131]
RatsMaternal care-induced anxiety-related behavior-Decreased H3K9ac levelsglucocorticoid receptor gene (Nr3c1)[132]
ND, not determined.
Table 3. miRNA changes related to cannabinoid receptor gene expression.
Table 3. miRNA changes related to cannabinoid receptor gene expression.
miRNAExperimental ModelTarget/Gene ExpressionReference
miRNA-1273g-3pHuman colon cancer cellsCB1[154]
hsa-miRNA-29b-3Low-grade glioma[155]
miR-494Myocardial cells[156]
miRNA let-7dSH-SY5Y neuroblastoma cells
Zebrafish, mice (cortex, striatum, and hippocampus) primary striatal neurons
[157]
miR-30e-5pMice (obese model)[159]
miR-338-5pRats (spinal cord injury)Cnr1[158]
MiRNA-30bRat models/cell models[160]
MiR-187-3pHuman osteoblastic precursor cellsCnr2[161]
miR-133b-3pPD mouse modelXist and Pitx3[162]
miR-665Human (myocardial cells)CB2 and Cnr2[156]
miRNA-690Mouse (MDSCs)CB1 and CB2[163]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Basavarajappa, B.S.; Subbanna, S. Molecular Insights into Epigenetics and Cannabinoid Receptors. Biomolecules 2022, 12, 1560. https://doi.org/10.3390/biom12111560

AMA Style

Basavarajappa BS, Subbanna S. Molecular Insights into Epigenetics and Cannabinoid Receptors. Biomolecules. 2022; 12(11):1560. https://doi.org/10.3390/biom12111560

Chicago/Turabian Style

Basavarajappa, Balapal S., and Shivakumar Subbanna. 2022. "Molecular Insights into Epigenetics and Cannabinoid Receptors" Biomolecules 12, no. 11: 1560. https://doi.org/10.3390/biom12111560

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