Abstract
Microglia are widely known for their role in immune surveillance and for their ability to refine neurocircuitry during development, but a growing body of evidence suggests that microglia may also play a complementary role to neurons in regulating the behavioral aspects of substance use disorders. While many of these efforts have focused on changes in microglial gene expression associated with drug-taking, epigenetic regulation of these changes has yet to be fully understood. This review provides recent evidence supporting the role of microglia in various aspects of substance use disorder, with particular focus on changes to the microglial transcriptome and the potential epigenetic mechanisms driving these changes. Further, this review discusses the latest technical advances in low-input chromatin profiling and highlights the current challenges for studying these novel molecular mechanisms in microglia.
Introduction
Substance use disorder (SUD), or addiction, is a chronic relapsing disease characterized by excessive substance use, despite negative consequences (1). SUD is not only a public health concern, but is also economic burden, as the economic impact of substance misuse and SUDs are ever increasing and is estimated to cost the United State over $400 billion annually in crime, lost productivity, and healthcare (US DHSS Surgeon General, NDTA DEA). Individuals that suffer from SUD will transition from an initial voluntary drug usage to compulsive drug seeking and use (2). Moreover, this compulsory drug use can be interrupted by periods of abstinence; however, these periods are typically short-lived, as individuals often relapse within 6 months of stopping use of the drug (3). Historically, studies have focused on how addictive drugs (i.e. cocaine, amphetamines, opioids) affect neuronal populations of the brain’s reward system, specifically focusing on the dopamine pathways, as these are central to the mechanisms of action of various drugs of abuse (4-6). Several therapeutics exist to treat SUD, consisting of medication-based treatment, various behavioral therapies (i.e. cognitive-behavioral therapy), or a combination of both (7). For example, naltrexone, an opioid receptor antagonist, is approved by the FDA to treat alcohol use disorder (AUD) and opioid use disorder (OUD), as it reduces alcohol and opioid cravings (8). However, these effects are short lived, and most patients treated for AUD show no significant effects relative to placebo after 1-3 months of naltrexone discontinuation (9, 10). In OUD patients, the adherence rate to naltrexone is particularly low since patients are typically required to be abstinent prior to starting treatment (11). Currently, medication-assisted treatment, or the use of partial opioid agonists such as methadone and buprenorphine, is most effective in reducing opioid use and increasing patient adherence to treatment (12, 13). Similarly, nicotine cessation therapeutics such as bupropion, and varenicline (14, 15), while effective, can require adherence of at least one month (16, 17). Therefore, there remains a critical need for treatments with high adherence rates that target specific pathways to induce cessation and prevent relapse.
Microglia, the resident immune cells of the central nervous system (CNS), have recently emerged as novel targets in SUD research (18-23), as they are present during critical periods of brain development and play an important role in shaping neural circuitry (24-36). Moreover, accumulating evidence suggests that microglia also play an active role in the development of SUDs and relapse (37, 38). In fact, the impact of microglia on drug-related behaviors can be seen most notably during adolescence, where they contribute to development of brain circuitry (30, 32, 34, 36, 39-41). This is further supported by evidence showing that early-life stress and/or adolescent exposure to drugs of abuse affects susceptibility to drug use later in life through microglial mechanisms (28, 38, 42-46). Specifically, microglia can respond to drugs of abuse through Toll-like receptor (TLR) signaling (47). Microglia express TLR1-9 (48), which recognize a variety of exogenous pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS) from gram-negative bacteria (49), as well as endogenous damage-associated molecular patterns (DAMPs) like high mobility group box 1 and heat shock proteins (50, 51). Interestingly, several drugs of abuse can bind the TLR4/myeloid differentiation factor (MD-2) complex, resulting in the release of numerous proinflammatory cytokines (52-54). Furthermore, microglia could offer insights into known sex differences in SUD (55-59), as these cells are sexually dimorphic and respond to stress in a sex-dependent manner (33, 60-66). For instance, microglia in adult male and female mice vary in density and size, as well as in their transcriptional profile across different brain regions (62), which can affect a variety of behavioral responses (67). Specifically, glucocorticoid signaling and synaptic remodeling via colony stimulating factor 1 (CSF1) impact behavioral response to chronic unpredictable stress in male mice more so than in females (68). Taken together, this evidence indicates that microglial activity in response to various ligands and conditions, including drugs of abuse and their misuse, must be tightly regulated. Therefore, understanding this regulation will yield further insight into the emerging role that microglia play in SUDs.
Several epigenetic mechanisms regulate gene expression (69). Specifically, histone post translational modifications (PTMs) can change the accessibility of chromatin to transcription factors and polymerases, which directly affects gene transcription (70, 71). Furthermore, substantial evidence supports that these mechanisms contribute to behavioral changes observed in SUDs (72). For example, histone acetylation is increased at the promoters of immediate early genes (cFos, FosB, BDNF, Cdk5) in the striatum following acute cocaine injection and cocaine self-administration in male mice (73). Similarly, inhibiting the repressive histone methylating enzyme G9a increases dendritic spine plasticity in the nucleus accumbens and increases cocaine preference in male mice (74). These examples highlight the importance of studying epigenetic modifications and chromatin remodeling in the context of SUD. However, much of this body of evidence, to date, has focused the role of these mechanisms in neuronal cells. Therefore, this review summarizes the current knowledge on the transcriptional and epigenetic changes that microglia undergo in SUDs, and the technical and conceptual challenges that face the field moving forward.
Alcohol
The innate immune system has long been implicated in AUD and dependence. Alcohol, or ethanol, crosses the blood brain barrier (BBB) and can elicit a neuroinflammatory response (75, 76). For instance, analysis of post-mortem brain tissue revealed increased ionized calcium-binding adaptor protein 1 (Iba1) immunoreactivity in the prefrontal cortex (PFC) of male chronic alcohol-dependent individuals, in addition to significant breakdown of BBB integrity (77). Iba1 is often used as a marker for microglia (78) in various inflammatory states (79), as membrane ruffling occurs during the transition from a ramified to ameboid morphology (80). Furthermore, recent single-nucleus RNA sequencing conducted on post-mortem brains from male individuals who were alcohol-dependent showed increased differentially expressed neuroinflammatory genes in oligodendrocytes, astrocytes, and microglia over neurons (81). Specifically, there were a large amount of differentially expressed microglial genes, most notably TLR2, as well as interferon signaling (81). Additionally, rodent studies have confirmed alcohol-induced neuroinflammation and the importance of microglia. For example, in male mice that underwent bidiurnal 2-bottle choice, microglial genes involved in the innate immune response, TLR signaling, and interferon signaling were upregulated with chronic ethanol consumption (82). Chronic intermittent ethanol (CIE) vapor exposure in male mice also resulted in enrichment of several gene expression networks in microglia relating to transforming growth factor-beta (TGF-β) signaling and impaired inflammatory response (83). Given that TGF-β signaling is neuroprotective and regulates homeostatic microglial function (84), the decrease in these modules following CIE indicates alcohol promotes neuroinflammation and disruption of normal microglial function. Altogether, these data suggest microglia are highly reactive to alcohol and may play a role in alcohol-induced neuroinflammation and dependence.
Alcohol induces lasting effects on microglial morphology and activity state (85). Additionally, changes to histone PTMs can drive altered microglial activity states following alcohol exposure. For instance, neonatal exposure to alcohol resulted in hyper-responsive adult hypothalamic microglia that showed increased histone 3 lysine 9 (H3K9) acetylation, a histone mark associated with active promoters (86), in the promoter regions of interleukin(IL)-6 and tumor necrosis factor-alpha (TNF-α) (38). Additionally, increased expression of microglial marker genes C-C Motif Chemokine Ligand 2 (CCL2) and translocator protein (TSPO) in the amygdala of male and female post-mortem samples from alcohol-dependent individuals could be attributed to increased histone 3 lysine 4 (H3K4) tri-methylation (87). Furthermore, dysregulation of inflammatory signaling by microglia was shown to be epigenetically regulated by histone lysine demethylase 6b (KDM6B) in both alcohol-dependent male rats that underwent alcohol vapor exposure and in post-mortem brains from males individuals who were alcohol-dependent (88).
Several studies have shown the therapeutic potential of microglia for treating AUD and alcohol dependence. Indeed, treatment with the anti-inflammatory drug minocycline has been shown to reduce alcohol intake in adult male and female mice undergoing the drinking in the dark paradigm of voluntary alcohol consumption (89). Most notably, microglial depletion in male mice using CSF1R inhibitor PLX5622 resulted in reduction of withdrawal-induced TNF-α and CCL2 expression (90). Moreover, microglial depletion in male mice reduced dependence-induced escalation of voluntary alcohol taking and alleviated anxiety-like behavior by decreasing neuroimmune signaling, preventing upregulation of glutamatergic-related genes in the medial PFC, and reducing glutamatergic and GABAergic signaling in the central amygdala (91). Taken together, these studies suggest microglia may be viable therapeutic targets for the treatment of alcohol dependence.
Opioids
Opioids bind to μ, κ and δ-opioid receptors (ORs) in the CNS (92, 93) and activate reward signaling primarily through μOR-mediated disinhibition of midbrain dopamine neurons (94, 95). Drugs that engage μORs are often the first line treatment for chronic, or severe post-operative pain (96). However, patients can develop tolerance, which could then lead to opioid misuse. Human post-mortem studies have recently shown evidence of increased neuroinflammation in the dorsolateral PFC and nucleus accumbens (NAc) of male and female individuals with OUD (97). Indeed, several rodent studies have suggested that TLR signaling underlies the development of tolerance to opioids and contributes to the addictive potential of these drugs (98-102). Interestingly, microglia express both ORs and TLRs (103, 104), which positions these immune cells as potential regulators of behavioral phenotypes associated with OUD. Furthermore, removal of MyD88, a critical co-adaptor protein for TLR signaling specifically in microglia, increased opioid addiction-like behaviors in male mice including prolonged extinction and enhanced reinstatement of morphine conditioned place preference (CPP) (105). Additionally, microglia-specific knockout of the μOR in both male and female conditional knockout (cKO) mice reduces analgesic tolerance as well as delays morphine tolerance, as measured by increased withdrawal latencies during hot plate tests over 12 days of treatment (106). The same study also showed attenuated opioid-induced hyperalgesia (OIH) in male cKO mice, and decreased withdrawal behaviors in female cKO mice (106). Lastly, microglia may contribute to neuroinflammation due to opioid exposure and misuse (107). For instance, microglia can respond to morphine in vitro resulting in increased IL-1β expression (108). On the other hand, opioids may also be neuroprotective and mitigate withdrawal by “switching off” microglia through inhibitory G protein-coupled receptor signaling (109).
Repeated opioid use results in numerous and persistent transcriptional and epigenetic changes to the CNS (97, 110). For instance, several RNA-sequencing studies of post-mortem tissue from opioid-dependent male and female individuals show upregulation of modules enriched for genes involved in immune and inflammatory processes, regulation of transcription, extracellular matrix, while also highlighting modules enriched for genes involved in synaptic transmission, neurotransmitter secretion, and nervous system development and differentiation (97, 111). Furthermore, the upregulation of these biological processes was the result of de-repression of gene transcription, exhibiting decreased repressive histone marks such as histone 3 lysine 27 (H3K27) tri-methylation (97). Interestingly, preclinical opioid studies in rodents confirm what is seen in OUD patient samples. For example, morphine self-administration in male mice results in upregulation of biological pathways related to immune response and oxidative stress, cell-to-cell signaling, and cell differentiation in the ventral striatum and midbrain (112). Additionally, morphine self-administration has been shown to increase numerous microRNAs, whose potential targets are involved in several immune-related pathways such as mitogen-activated protein kinase (MAPK) and TGF-β. Additionally, acetylation of H3K9 is implicated in OIH, tolerance, and physical dependence, and increases BDNF expression in the mouse spinal cord (113). However, increased histone deacetylase (HDAC) 1 expression can also restore morphine’s antinociceptive effect in a tail-flick assay and decrease TNF-α expression in morphine-tolerant male rats (114). Another mouse study also showed significant upregulation of inflammatory genes including IL-1β and interferon regulatory factor 1 in both the ventral and dorsal striatum following oxycodone self-administration (115). Moreover, exposure to opioids such as morphine during adolescence primes microglia and increases TLR4 signaling specifically in the NAc, predisposing male rats to drug-induced reinstatement of morphine CPP (44).
Reducing neuroinflammation has been shown to attenuate opioid related behaviors. For example in human clinical trials, ibudilast (AV-411), a phosphodiesterase inhibitor, reduced heroin craving and subjective ratings of pain in opioid dependent subjects (116). Additionally, ibudilast treatment during adolescent morphine exposure in male rats prevents morphine-induced reinstatement of CPP and prevents the increase of TLR4 protein levels in microglia compared to controls in adulthood (44). As well, inhibiting microglial activity using minocycline can decrease somatic symptoms of opioid withdrawal such as “wet dog” shakes and expression of several proteins of the mTOR and CREB signaling pathways, which are associated with morphine tolerance, dependence, and drug memory (117).
Opioid-induced neuroinflammation and drug-related behaviors can also be mitigated with early-life interventions. In fact, opioid-related behaviors are affected by early-life experience in a microglia-specific manner. For instance, neonatal handling of male rats results in demethylation of the IL-10 gene and reduces microglial activity, as well as attenuates reinstatement of morphine CPP following drug re-exposure (118). Additionally, inhibition of epigenetic enzymes can ameliorate opioid-induced neuroinflammation. Indeed, morphine has been shown to increase HDAC1, TNFR1, and TNF-α protein expression in the spinal cord of male rats (114). Intrathecal injection of the HDAC inhibitor resveratrol prevents these increases, as well as restores the analgesic properties of morphine, indicated by decreased tail-flick latency (114). Overall, these studies suggest microglia and their epigenome may be novel targets to treat OUD and dependence.
Stimulants
Although stimulants have not been the principal focus of many studies on neuroinflammation and SUD, there is a growing body of evidence suggesting these drugs do alter microglial activity states (23, 119). Overall, studying the effect of stimulants on the immune system in the human CNS has yielded variable outcomes, with much of the post-mortem data demonstrating inconsistent results when measuring glial numbers and expression of cell markers (18, 120). However, most results suggest a connection between stimulant abuse and neuroinflammation. For example, stimulants such as nicotine, cocaine, and amphetamines can increase expression of cytokine receptors, extracellular matrix proteins, and TLRs over the course of several hours in male mice (121). Stimulants act on the reward system by increasing dopamine through various means including inhibition of dopamine reuptake (cocaine, amphetamines, MDMA) (122-124), and through direct activation of midbrain dopamine neurons (nicotine) (125, 126). Interestingly, while nicotine can act as a cognitive stimulant, unlike cocaine and methamphetamine, nicotine decreases locomotion at higher doses (127-129). Nicotine and stimulant use are associated with production of reactive oxygen species (ROS), which can exacerbate neuroinflammatory states and disease (46, 130). Similar to alcohol and opioids, microglia can respond to stimulants such as cocaine and methamphetamine directly through the TLR4/MD-2 complex, which may increase activation of midbrain dopamine neurons and enhance dopamine release in the NAc (131, 132). To this point, human positron emission topography studies show nicotine smokers have lower TSPO expression as measured by radiotracer [11C]DAA1106 binding, suggesting suppressed microglial activity in these individuals (133). In fact, microglia express α7 nicotinic acetylcholine receptor, which may serve as a biological substrate for any direct actions of nicotine on microglial activation (134). Indeed, this could explain the functional deficits seen in microglia after treatment with the drug. Beyond nicotine, stimulants like cocaine and methamphetamine induce a proinflammatory microglial state, where these cells increase expression of cytokines including TNF-α, IL-1β, and IL-6 (131, 135-137), and increase phagocytosis and migration in vitro (138).
While microglia differentially respond to various stimulants, withdrawal from these drugs show microglia alter their gene expression in a similar manner (139-141). For instance, altered microglial morphological states and increased production of ROS can be seen during nicotine withdrawal in the NAc and striatum in male mice (139). These changes coincide with increases in anxiety-associated behaviors (139). Additionally, increased microglial reactivity correlates with impaired neurogenesis in the hippocampus of male mice (142), as well as increased hyperalgesia in male rats during nicotine withdrawal (143). This response is also seen in abstinent male rats following extended-access methamphetamine self-administration (144).
Stimulants have long lasting effects on the innate immune system and on microglia. Several microarray and RNA-sequencing studies demonstrate differential expression of immune-related genes. For example, there is significant overlap between upregulated genes associated with immune/inflammatory response, specifically those regulated by nuclear factor kappa B (NF-κB), in cocaine use disorder (CUD) or OUD (111). Additionally, methamphetamine administration results in delayed upregulation of immune-related genes in male mice, including NF-κB, interleukins and their receptors (145). Consistent with delayed effects following methamphetamine exposure, self-administered methamphetamine increased IL-6 production during abstinence and was associated with activation of nuclear factor of activated T-cell-mediated immune response, which is involved in regulating microglial response and cytokine expression (146).
Early-life stress and exposure to addictive drugs during adolescence increases susceptibility to misuse of stimulants later in life (23, 28). For example, prenatal exposure to nicotine results in downregulation of genes associated with TNF signaling and cytokine-cytokine receptor pathways, as well as increases microglial numbers and changes microglial morphology in the hippocampus of young male and female mice (postnatal day 20) (147). Another study showed adolescent exposure to nicotine increases cocaine self-administration in adult male and female mice in a microglia-specific manner through CXC3 motif chemokine receptor 1 - CXC3 chemokine ligand 1 signaling (43). Additionally, childhood trauma is correlated with increased plasma inflammatory markers in cocaine-dependent human subjects (46). The same study showed that exposing male and female mice to social stress during early life (postnatal day 14-22) sensitizes microglia and peripheral immune cells, and gene ontology analysis of microarray from blood RNA revealed enrichment of “inflammation mediated by chemokine and cytokine signaling” pathway during acute withdrawal from cocaine (46). Furthermore, neonatal exposure to an immune challenge increases methamphetamine-induced reinstated behavioral sensitization including locomotor activity, rearing, and stereotypy in adult male and female rats (148). Despite evidence that altered microglial dynamics early in life can influence later stimulant misuse, suggesting involvement of epigenetic regulation, most studies focus on the epigenetic landscape in neuronal populations. Therefore, there remains a significant need to characterize these changes in non-neuronal populations, particularly in microglia.
Nonetheless, numerous studies demonstrate therapeutic targeting of microglial function can normalize various behaviors related to stimulant use disorder. In human studies, ibudilast reduced reward-related subjective effects of methamphetamine including how “high” the subject felt and if the subject felt the “effect” of the drug (149). This was confirmed and further investigated in male rats using methamphetamine self-administration, where the authors showed that AV1013 (an analog of ibudilast) and minocycline significantly reduced drug intake (150). Minocycline also attenuates nicotine-induced increases in the number of microglia, as well as cocaine-seeking in male and female rats (43). Additionally, minocycline facilitates extinction of methamphetamine CPP and attenuates drug-primed reinstatement of CPP (151). Depletion of microglia is also therapeutic in stimulant-related addictive behaviors. For example, treatment with PLX5622 during nicotine treatment and withdrawal mitigated nicotine-induced increased NADPH oxidase 2 expression and ROS production and reduced anxiogenic behavior in male mice (139). Additionally, microglial depletion using PLX3397, another CSF1R inhibitor, blocked increased cocaine responding induced by nicotine (43). In support of the importance of TLR4 signaling in stimulant use disorders, pretreatment with TLR4 antagonist, (+)-Naloxone, prevented increased cocaine-induced dopamine release and expression of IL-1β in the NAc of male rats (131). Interestingly, in a separate study, the use of bromodomain and extra-terminal motif inhibitor JQ1, which can alter microglial activity (152), also reduced cocaine-induced increased expression of IL-1β in the NAc of male mice (153). Moreover, treatment with GSK-J4, a small molecule inhibitor of KDM6B known to increase macrophage activity (154), reduced reconsolidation of cocaine-conditioned memory and cocaine-primed reinstatement of CPP in male mice (155). Thus, in the context of stimulant use disorders, microglial function may be regulated by epigenetic mechanisms, and may therefore serve as a cellular target for curbing behavioral aspects of stimulant use disorders.
Cannabinoids
With the recent increased use and legalization of medical and recreational marijuana and cannabidiol (CBD) products (SAMHSA), there has been a renewed interest in studying the effects that exogenous cannabinoids have on the CNS. The primary mechanism of action for cannabinoids is modulated through cannabinoid receptor type 1 (CB1R) and cannabinoid receptor type 2 (CB2R) (156). While CB1R is primarily expressed on neurons, CB2R is expressed on immune cells (157), including microglia (158). Indeed, cannabinoids have anti-inflammatory properties (157). For example, cannabinoids can increase release of endogenous IL-1 receptor antagonist, which has neuroprotective effects against excitotoxicity (159). In fact, a recent study demonstrated CB2R is necessary for TLR signaling in mice, dampening microglial transcriptional and morphological response to TLR ligands such as LPS/Interferon-γ, polyinosinic-polycytidylic acid, and unmethylated CpG DNA, and that this occurs through phosphatidylinositol 3-kinase/protein kinase B and MAPK signaling pathways (160). Additionally, the endocannabinoid 2-arachidonylglycerol (2-AG) can increase microglial migration in vitro, and cannabinol and CBD, two nonpsychotropic chemicals in marijuana, inhibit cell migration by antagonizing CB2R (161). Furthermore, 2-AG and anandamide, another CB1R and CB2R ligand, contribute to endocannabinoid tone, which plays a role establishing sex differences in the developing brain by increasing the phagocytic activity of microglia (162).
Several studies have shown that Δ9-tetrahydrocannabinol (THC), the psychoactive component in marijuana, has long-lasting effects on the brain that are age- and sex-dependent (163, 164). Most notably, adolescent exposure to cannabinoids increases vulnerability to substance misuse later in life, suggesting underlying epigenetic regulation. For example, adolescent THC exposure increases heroin self-administration in male rats through decreased repressive trimethylation of H3K9, resulting in increased proenkephalin expression in the shell of the NAc (165). Interestingly, adolescent exposure to cannabinoid receptor agonist WIN55,212-2 increased nicotine self-administration in adult male mice, while nicotine self-administration was decreased in adult female mice (166). However, another study showed treatment with cannabinoid agonist CP 55,940 during adolescence increased acquisition of cocaine-self administration, as well as altered basal brain metabolic activity in adult female rats, but had no effect on males (167). Paradoxically, cannabinoids are also therapeutic in the treatment of SUD and can reduce drug-related behaviors and neuroinflammation. For instance, acute CBD reduced cue-induced drug-craving and anxiety in abstinent heroin users (168). Additionally, treatment with CB2R agonists reduced morphine-induced expression of inflammatory cytokines such as IL-1β, TNF-α, and IL-6 and altered microglial activity in vitro (169) and reduced hyperalgesia and allodynia in male rats (170). Furthermore, microinjection of CBD into the lateral cerebral ventricle prevented acquisition and expression of methamphetamine CPP in male rats (171). Finally, CBD attenuated cognitive deficits, altered microglial morphology, and expression of inflammatory cytokines TNF-α and IL-1β in male mice undergoing nicotine withdrawal (142). Since there is limited access to cannabis and THC due to its scheduling classification, there is a significant need for additional studies to better understand the epigenetic consequences of exogenous cannabinoid exposure, especially in microglia, that may underlie these therapeutic effects.
Current challenges and future directions
Microglia stand as a prime target for studying the behavioral consequences of SUDs. Through their dynamic regulation of neurodevelopment and role in neuroinflammation and psychiatric disease (Fig. 1), microglia stand as a key cell type to be further characterized. However, there are currently a number of theoretical and technical limitations when studying microglia, both as an individual cell type as well as in the context of SUDs.
Figure 1. Microglia regulate drug-related behaviors as a result of transcriptional changes throughout Substance Use Disorders.
Proposed mechanisms for microglial activation following exposure to various drugs of abuse. Microglia may be activated through both direct and indirect means. Following activation, microglia maintain an altered state, with possible changes to morphology, gene expression and epigenetic modifications that lead to functional deficits such as aberrant pruning of synapses and hyperexcitability. These changes are underscored by altered behavior across numerous paradigms. However, treatment with known anti-inflammatory drugs like AV-411 and the tetracycline antibiotic minocycline, as well as complete microglial depletion can mitigate these drug-induced changes both molecularly and behaviorally.
While there is a certain need to better characterize SUD-related gene expression and epigenetic changes in microglia, it is also necessary to further understand microglial activation dynamics from a broader perspective. Microglia have previously been classified through binary terms “M1” (pro-inflammatory) or “M2” (anti-inflammatory) as well as “resting” or “active”; however, recent advances in the field suggest a deeper complexity that extends beyond a binary nomenclature (172, 173). Developing a more comprehensive classification of microglia is essential to understanding the role they play in SUDs and other neurological disorders. In doing so, examining the transcriptional and epigenetic states of microglia can aid in these classifications and therefore broaden the pool of potential therapeutics to correct their altered state in SUDs that may contribute to adverse behavioral outcomes, such as increased drug craving and propensity for relapse.
From an in vitro perspective, the use of microglial cell lines offers a more cost-effective method to study and characterize microglia, as opposed to in vivo models, by directly measuring the effects of a drug. Additionally, in vitro studies have a faster turnaround, are very adaptable (e.g. transfection with siRNA, CRISPR, treatment with small molecules, etc.), and are useful for basic validation of molecular targets. Studies that utilize pharmacological interventions can be particularly modeled using established microglial cell lines such as the murine BV-2 (ATCC; EOC-20, CRL-2469) (174) or the human microglial clone 3 (ATCC; HMC3 CRL-3304) (175). However, while these cell lines display a similar cytokine profile of primary microglial cells (176, 177), they often have reduced expression of key microglial markers, and also display decreased functional activity (175). A recent advancement for studying microglia in vitro involves induced pluripotent stem cell-derived microglia, which are shown to be more transcriptionally and functionally similar to microglia in vivo (178-180); meaning they may offer additional insight into how microglia respond to drugs of abuse. Yet, a major limitation inherent to cell culture models in studying drug addiction is that these cannot account for changes that may be induced by the motivational or affective aspects of drug-seeking and craving. In fact, no in vitro models are currently available to replicate these behavioral phenotypes; therefore, the most reliable method is the use of in vivo behavioral paradigms. One such behavioral technique is intravenous self-administration (IVSA), which is considered the “gold standard” for preclinical studies, as it most closely reflects the motivational aspects and patterns of drug-taking and seeking in humans (181-183). Given how microglia rely on and communicate with other neural cells in both healthy and disease conditions (184, 185), in vivo models of SUDs are currently the most comprehensive for studying the contribution of microglia toward shaping the behavioral course of addiction. Additionally, as previous research has mainly focused on the role of neuronal populations in SUDs (186-188), microglia represent a novel cell population to examine within the context of these conditions.
Another major challenge to studying microglia is the ability to obtain a pure population of these cells from the brain for downstream analyses (Fig. 2). This is key to producing the transcriptomic and epigenetic profiles needed to better understand the role of microglia in SUDs. While the most common method of microglial purification utilizes Florescence Assisted Cellular Sorting (FACS), affinity purification with magnetic beads also yields highly pure populations, as it also targets microglia-specific markers with antibodies (189). Importantly, the multitude of receptors shared between microglia and macrophages (190) has been difficult to overcome, which inherently impacts purity in sorted populations. However, several of these issues have been resolved with the identification of microglia-specific markers such as TMEM119, which has resulted in the generation of several transgenic mouse lines (188). This recent advancement has also allowed for improved resolution when performing cell phenotyping studies using single-cell sequencing (191-193). Along similar lines, RiboTag-seq has been shown to reveal gene networks involved in morphine withdrawal (194), by analyzing the active translatome in microglia (195). While this method offers a number of advantages including compatibility with single-cell sequencing (196) and analyzing cell type specific profiles (197), care must be taken due to the inherent loss of RNA transcripts in this methodology. The discovery of new markers in combination with single-cell sequencing technologies has allowed for better characterization of microglial heterogeneity (198) as well as profiling chromatin accessibility (199). While recent studies have started to profile the epigenome using methods such as Assay for Transposase-Accessible Chromatin (ATAC) sequencing (200, 201) and chromatin immunoprecipitation (ChIP) sequencing (202, 203), the advent of lower input methodologies will enable microglia to be studied on a brain-region specific scale, rather than on a global scale, furthering our understanding of microglial transcriptomics and epigenetics in various disease models (204-206). For example, novel methods like CUT&RUN (Cleavage Under Targets & Release Using Nuclease) (207, 208) and CUT&Tag (Cleavage Under Targets & Tagmentation) (209, 210) allow for higher resolution using lower cell numbers than previous techniques. This new technology facilitates interrogation of transcription factors and histone modifications including various forms of histone methylation and acetylation to identify promoters and enhancers. Given the scarcity of data regarding the transcriptomic and epigenetic landscapes of microglia, these novel low-input, high-resolution methods hold promise to improve our understanding of not only SUDs, but also neurological disorders in general.
Figure 2. Methodologies for studying microglial genomics.
A number of novel technologies have enhanced the ability to sequence the genome and epigenome of microglia. As an initial barrier to profiling microglia, isolation of pure populations has recently been made easier with developments in both FACS and magnetic bead based affinity purification, with both methods achieving similar results. For RNA sequencing, the advent of low input and single cell, kits capable of creating complex libraries for sequencing has enabled investigators to reduce the number of microglia needed to understand their response to certain stimuli and delineate sub-populations within different brain regions. To study chromatin dynamics, methods such as CUT&RUN and CUT&Tag allow for regional microglial populations to be analyzed for various histone marks and transcription factors with very low cellular input, including single cells, at a fraction of the cost of ChIP-sequencing assays. These more recent advances can generate robust datasets comparable to those generated by ChIP-sequencing. ATAC-seq is a convenient method for profiling chromatin accessibility, however its utility is hampered by its limited ability to recognize gene enhancer loci. (FACS: Fluorescence Assisted Cellular Sorting, AFP: Affinity purification, scRNA: Single-cell RNA, CUT&RUN: Cleavage Under Targets & Release Using Nuclease, CUT&Tag: Cleavage Under Targets & Tagmentation, ATAC: Assay for Transposase-Accessible Chromatin) Created with BioRender.
From a translational standpoint, the therapeutic value of epigenetic targets remains promising. However, owing to the complex dynamics of epigenetic mechanisms during development and throughout the lifespan, development of treatments targeting specific epigenetic modifications has been challenging. While some studies show promise using HDAC inhibitors such as resveratrol for treatment of OUD (114), bromodomain-containing 4 inhibitor JQ1 for CUD (153), and GSK-J4 for AUD and CUD in vitro and in vivo, respectively (88, 155, 211), the pharmacokinetics, brain bioavailability, and lack of cell-type specificity of these drugs lend themselves to limited translational value. Furthermore, given the recent emergence of microglia in the study of SUDs, the importance of microglial chromatin architecture and the role that it may play in SUDs is not yet fully understood. Finally, epigenetic targets, while promising, are shared across numerous cell types, which may result in off-target effects. Since this review discusses several epigenetic changes noted in microglia for specific SUDs, repurposing of epigenetic drugs could provide a novel therapeutic avenue, but studies into novel epigenetic treatments should focus on cellular specificity to reduce off-target effects and increase efficacy.
While the fields of addiction and neuroscience continue to advance, recent evidence has positioned microglia as important mediators in the development and progression of SUDs. This review highlights recent advances in our understanding of microglia, from the shift in focus to glial involvement in SUD research as well as to setting our sights forward to crucial transcriptomic and epigenetic hypotheses regarding microglial regulation in SUDs. However, important gaps in this understanding still remain. While in vitro and clinically relevant in vivo behavioral models are essential to understanding the importance of microglia to addiction pathophysiology, genomics studies employing cell type-specific RNA or single cell sequencing applications in combination with low-input CUT&RUN/CUT&Tag chromatin profiling methods will provide the molecular depth to understanding the driving factors that may underlie drug-taking, craving and relapse. Further studies that better characterize these changes may shed light on the role of microglia in neuroinflammation, addiction, and may unlock a host of novel therapeutic targets for the treatment of SUDs.
Highlights.
We explore the recent evidence supporting a role for microglia in regulating the behavioral aspects of substance use disorders.
We focus on the transcriptional and proposed epigenetic mechanisms underlying microglial responses to drugs of abuse.
We discuss the latest advances and challenges in low-input epigenetic/molecular profiling of microglia.
Funding:
This work was funded by the National Institutes of Health [DP1DA051858, K01DA045294, and F99NS130871], and by support from the Shipley Foundation.
Footnotes
Competing interests: The Authors have no competing interests to declare.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Literature
- 1.Hyman SE, Malenka RC, Nestler EJ. Neural Mechanisms of Addiction: The Role of Reward-Related Learning and Memory. 2006. [DOI] [PubMed] [Google Scholar]
- 2.Hyman SE, Malenka RC, Nestler EJ. Neural Mechanisms of Addiction: The Role of Reward-Related Learning and Memory. 2006. [DOI] [PubMed] [Google Scholar]
- 3.Crombag HS, Bossert JM, Koya E, Shaham Y. Review. Context-induced relapse to drug seeking: a review. Philos Trans R Soc Lond B Biol Sci. 2008;363(1507):3233–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry. 2016;3(8):760–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Koob George F, Bloom Floyd E. Cellular and Molecular Mechanisms of Drug Dependence. Science. 1988;242(4879):715–23. [DOI] [PubMed] [Google Scholar]
- 6.Koob GF, Le Moal M. Drug Addiction, Dysregulation of Reward, and Allostasis. Neuropsychopharmacology. 2001;24(2):97–129. [DOI] [PubMed] [Google Scholar]
- 7.Abuse NIoD. Principles of Drug Addiction Treatment: A Research-Based Guide (Third Edition). 2018. [Google Scholar]
- 8.Singh D, Saadabadi A. Naltrexone. StatPearls. Treasure Island (FL)2022. [Google Scholar]
- 9.Carter CJ. The natural history and epidemiology of venous thrombosis. Prog Cardiovasc Dis. 1994;36(6):423–38. [DOI] [PubMed] [Google Scholar]
- 10.Anton RF, Moak DH, Latham PK, Waid LR, Malcolm RJ, Dias JK, et al. Posttreatment results of combining naltrexone with cognitive-behavior therapy for the treatment of alcoholism. J Clin Psychopharmacol. 2001;21(1):72–7. [DOI] [PubMed] [Google Scholar]
- 11.Jarvis BP, Holtyn AF, Subramaniam S, Tompkins DA, Oga EA, Bigelow GE, et al. Extended-release injectable naltrexone for opioid use disorder: a systematic review. Addiction. 2018;113(7):1188–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kreek MJ. Methadone-related opioid agonist pharmacotherapy for heroin addiction. History, recent molecular and neurochemical research and future in mainstream medicine. Ann N Y Acad Sci. 2000;909:186–216. [DOI] [PubMed] [Google Scholar]
- 13.Connery HS. Medication-assisted treatment of opioid use disorder: review of the evidence and future directions. Harv Rev Psychiatry. 2015;23(2):63–75. [DOI] [PubMed] [Google Scholar]
- 14.Stead LF, Perera R, Bullen C, Mant D, Hartmann-Boyce J, Cahill K, et al. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146. [DOI] [PubMed] [Google Scholar]
- 15.Mills EJ, Wu P, Lockhart I, Thorlund K, Puhan M, Ebbert JO. Comparisons of high-dose and combination nicotine replacement therapy, varenicline, and bupropion for smoking cessation: a systematic review and multiple treatment meta-analysis. Ann Med. 2012;44(6):588–97. [DOI] [PubMed] [Google Scholar]
- 16.Gilpin EA, Pierce JP, Farkas AJ. Duration of smoking abstinence and success in quitting. J Natl Cancer Inst. 1997;89(8):572–6. [DOI] [PubMed] [Google Scholar]
- 17.Zhang B, Cohen JE, Bondy SJ, Selby P. Duration of nicotine replacement therapy use and smoking cessation: a population-based longitudinal study. Am J Epidemiol. 2015;181(7):513–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Coller JK, Hutchinson MR. Implications of central immune signaling caused by drugs of abuse: Mechanisms, mediators and new therapeutic approaches for prediction and treatment of drug dependence. Pharmacology and Therapeutics: Pergamon; 2012. p. 219–45. [DOI] [PubMed] [Google Scholar]
- 19.Crews FT, Vetreno RP. Addiction, adolescence, and innate immune gene induction. Frontiers in Psychiatry: Frontiers; 2011. p. 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Crews FT, Zou J, Qin L. Induction of innate immune genes in brain create the neurobiology of addiction. Brain, Behavior, and Immunity: Academic Press; 2011. p. S4–S12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. Journal of Neurochemistry: John Wiley & Sons, Ltd; (10.1111); 2016. p. 136–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lacagnina MJ, Rivera PD, Bilbo SD. Glial and Neuroimmune Mechanisms as Critical Modulators of Drug Use and Abuse. Neuropsychopharmacology: Nature Publishing Group; 2017. p. 156–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Clark KH, Wiley CA, Bradberry CW. Psychostimulant Abuse and Neuroinflammation: Emerging Evidence of Their Interconnection. Neurotoxicity Research 2013. p. 174–88. [DOI] [PubMed] [Google Scholar]
- 24.Paolicelli Rosa C, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic Pruning by Microglia Is Necessary for Normal Brain Development. Science. 2011;333(6048):1456–8. [DOI] [PubMed] [Google Scholar]
- 25.Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, et al. The Classical Complement Cascade Mediates CNS Synapse Elimination. Cell: Elsevier; 2007. p. 1164–78. [DOI] [PubMed] [Google Scholar]
- 26.Wilton DK, Dissing-Olesen L, Stevens B. Neuron-Glia Signaling in Synapse Elimination. 10.1146/annurev-neuro-070918-050306: Annual Reviews; 2019. p. 107–27. [DOI] [PubMed] [Google Scholar]
- 27.Arambula SE, McCarthy MM. Neuroendocrine-Immune Crosstalk Shapes Sex-Specific Brain Development. Endocrinology: Oxford Academic; 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Guerri C, Pascual M. Impact of neuroimmune activation induced by alcohol or drug abuse on adolescent brain development. 2019. [DOI] [PubMed] [Google Scholar]
- 29.Hove HV, Martens L, Scheyltjens I, Vlaminck KD, Antunes ARP, Prijck SD, et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nature Neuroscience 2019: Nature Publishing Group; 2019. p. 1. [DOI] [PubMed] [Google Scholar]
- 30.Parkhurst Christopher N, Yang G, Ninan I, Savas Jeffrey N, Yates John R, Lafaille Juan J, et al. Microglia Promote Learning-Dependent Synapse Formation through Brain-Derived Neurotrophic Factor. Cell: Elsevier; 2013. p. 1596–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Li Q, Cheng Z, Zhou L, Darmanis S, Neff NF, Okamoto J, et al. Developmental Heterogeneity of Microglia and Brain Myeloid Cells Revealed by Deep Single-Cell RNA Sequencing. Neuron: Cell Press; 2019. p. 207–23.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Miyamoto A, Wake H, Ishikawa AW, Eto K, Shibata K, Murakoshi H, et al. Microglia contact induces synapse formation in developing somatosensory cortex. Nature Communications: Nature Publishing Group; 2016. p. 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Nelson LH, Saulsbery AI, Lenz KM. Small cells with big implications: Microglia and sex differences in brain development, plasticity and behavioral health. Progress in Neurobiology: Pergamon; 2019. p. 103–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Microglia: Actively surveying and shaping neuronal circuit structure and function, (2013). [DOI] [PubMed] [Google Scholar]
- 35.Ferrini F, De Koninck Y. Microglia control neuronal network excitability via BDNF signalling. Neural plasticity 2013. p. 429815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Schafer Dorothy P, Lehrman Emily K, Kautzman Amanda G, Koyama R, Mardinly Alan R, Yamasaki R, et al. Microglia Sculpt Postnatal Neural Circuits in an Activity and Complement-Dependent Manner. Neuron: Elsevier; 2012. p. 691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Liao K, Guo M, Niu F, Yang L, Callen SE, Buch S. Cocaine-mediated induction of microglial activation involves the ER stress-TLR2 axis. J Neuroinflammation. 2016;13:33-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chastain LG, Franklin T, Gangisetty O, Cabrera MA, Mukherjee S, Shrivastava P, et al. Early life alcohol exposure primes hypothalamic microglia to later-life hypersensitivity to immune stress: possible epigenetic mechanism.Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2019;44(9):1579–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Favuzzi E, Huang S, Saldi GA, Binan L, Ibrahim LA, Fernández-Otero M, et al. GABA-receptive microglia selectively sculpt developing inhibitory circuits. Cell: Cell Press; 2021. p. 4048–63.e32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Badimon A, Strasburger HJ, Ayata P, Chen X, Nair A, Ikegami A, et al. Negative feedback control of neuronal activity by microglia. Nature: Nature Publishing Group; 2020. p. 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cheadle L, Rivera SA, Phelps JS, Ennis KA, Stevens B, Burkly LC, et al. Sensory Experience Engages Microglia to Shape Neural Connectivity through a Non-Phagocytic Mechanism. Neuron: Cell Press; 2020. p. 451–68.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kopec AM, Smith CJ, Ayre NR, Sweat SC, Bilbo SD. Microglial dopamine receptor elimination defines sex-specific nucleus accumbens development and social behavior in adolescent rats. Nature Communications 2018 9:1: Nature Publishing Group; 2018. p. 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Linker KE, Elabd MG, Tawadrous P, Cano M, Green KN, Wood MA, et al. Microglial activation increases cocaine self-administration following adolescent nicotine exposure. Nature Communications: Nature Research; 2020. p. 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Schwarz JM, Bilbo SD. Adolescent morphine exposure affects long-term microglial function and later-life relapse liability in a model of addiction. Journal of Neuroscience: Society for Neuroscience; 2013. p. 961–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Brown KT, Levis SC, O'Neill CE, Northcutt AL, Fabisiak TJ, Watkins LR, et al. Innate immune signaling in the ventral tegmental area contributes to drug-primed reinstatement of cocaine seeking. Brain, Behavior, and Immunity 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lo lacono L, Catale C, Martini A, Valzania A, Viscomi MT, Chiurchiù V, et al. From Traumatic Childhood to Cocaine Abuse: The Critical Function of the Immune System. Biological Psychiatry: Elsevier; 2018. p. 905–16. [DOI] [PubMed] [Google Scholar]
- 47.Bachtell R, Hutchinson MR, Wang X, Rice KC, Maier SF, Watkins LR. Targeting the Toll of Drug Abuse: The Translational Potential of Toll-Like Receptor 4. CNS Neurol Disord Drug Targets. 2015;14(6):692–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Olson JK, Miller SD. Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs. J Immunol. 2004;173(6):3916–24. [DOI] [PubMed] [Google Scholar]
- 49.Medzhitov R, Janeway C Jr. Innate immunity. N Engl J Med. 2000;343(5):338–44. [DOI] [PubMed] [Google Scholar]
- 50.Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm. 2010;2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hanke ML, Kielian T. Toll-like receptors in health and disease in the brain: mechanisms and therapeutic potential. Clin Sci (Lond). 2011;121(9):367–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hutchinson MR, Zhang Y, Shridhar M, Evans JH, Buchanan MM, Zhao TX, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav Immun. 2010;24(1):83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Northcutt AL, Hutchinson MR, Wang X, Baratta MV, Hiranita T, Cochran TA, et al. DAT isn't all that: cocaine reward and reinforcement require Toll-like receptor 4 signaling. Mol Psychiatry. 2015;20(12):1525–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang X, Northcutt AL, Cochran TA, Zhang X, Fabisiak TJ, Haas ME, et al. Methamphetamine Activates Toll-Like Receptor 4 to Induce Central Immune Signaling within the Ventral Tegmental Area and Contributes to Extracellular Dopamine Increase in the Nucleus Accumbens Shell. ACS Chem Neurosci. 2019;10(8):3622–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Brady KT, Randall CL. GENDER DIFFERENCES IN SUBSTANCE USE DISORDERS. Psychiatric Clinics of North America: Elsevier; 1999. p. 241–52. [DOI] [PubMed] [Google Scholar]
- 56.McHugh RK, Votaw VR, Sugarman DE, Greenfield SF. Sex and gender differences in substance use disorders. Clinical Psychology Review: Pergamon; 2018. p. 12–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Brynildsen JK, Sanchez V, Yohn NL, Carpenter MD, Blendy JA. Sex-specific transgenerational effects of morphine exposure on reward and affective behaviors. Behav Brain Res: Elsevier B.V.; 2020. p. 112842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Doncheck EM, Liddiard GT, Konrath CD, Liu X, Yu L, Urbanik LA, et al. Sex, stress, and prefrontal cortex: influence of biological sex on stress-promoted cocaine seeking. Neuropsychopharmacology: Springer Science and Business Media LLC; 2020. p. 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kumar M, Rainville JR, Williams K, Lile JA, Hodes GE, Vassoler FM, et al. Sexually dimorphic neuroimmune response to chronic opioid treatment and withdrawal. Neuropharmacology: Elsevier Ltd; 2021. p. 108469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Osborne BF, Turano A, Schwarz JM. Sex differences in the neuroimmune system. Current Opinion in Behavioral Sciences: Elsevier; 2018. p. 118–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Darling JS, Bayless DW, Dartez LR, Taylor JJ, Mehrotra A, Smith WL, et al. Sex differences in impulsivity in adult rats are mediated by organizational actions of neonatal gonadal hormones and not by hormones acting at puberty or in adulthood. Behav Brain Res: Elsevier B.V.; 2020. p. 112843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Guneykaya D, Ivanov A, Hernandez DP, Haage V, Wojtas B, Meyer N, et al. Transcriptional and Translational Differences of Microglia from Male and Female Brains. Cell Reports 2018. p. 2773–83.e6. [DOI] [PubMed] [Google Scholar]
- 63.McCarthy MM, Nugent BM, Lenz KM. Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nature Reviews Neuroscience: Nature Publishing Group; 2017. p. 471–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Walker DM, Zhou X, Cunningham AM, Lipschultz AP, Ramakrishnan A, Cates HM, et al. Sex-Specific Transcriptional Changes in Response to Adolescent Social Stress in the Brain’s Reward Circuitry. Biological Psychiatry: Elsevier BV; 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bollinger JL, Collins KE, Patel R, Wellman CL. Behavioral stress alters corticolimbic microglia in a sex- and brain region-specific manner. PLoS ONE: Public Library of Science; 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Woodburn SC, Bollinger JL, Wohleb ES. Synaptic and behavioral effects of chronic stress are linked to dynamic and sex-specific changes in microglia function and astrocyte dystrophy. Neurobiol Stress. 2021;14:100312-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Arambula SE, McCarthy MM. Neuroendocrine-Immune Crosstalk Shapes Sex-Specific Brain Development. Endocrinology. 2020;161(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Woodburn SC, Bollinger JL, Wohleb ES. Synaptic and behavioral effects of chronic stress are linked to dynamic and sex-specific changes in microglia function and astrocyte dystrophy. Neurobiol Stress. 2021; 14:100312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity. 2010;105. [DOI] [PubMed] [Google Scholar]
- 70.Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17(8):487–500. [DOI] [PubMed] [Google Scholar]
- 71.Kouzarides T Chromatin modifications and their function. Cell. 2007;128(4):693–705. [DOI] [PubMed] [Google Scholar]
- 72.Renthal W, Nestler EJ. Epigenetic mechanisms in drug addiction. Trends Mol Med. 2008;14(8):341–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kumar A, Choi KH, Renthal W, Tsankova NM, Theobald DE, Truong HT, et al. Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron. 2005;48(2):303–14. [DOI] [PubMed] [Google Scholar]
- 74.Maze I, Covington HE, Dietz DM, LaPlant Q, Renthal W, Russo SJ, et al. Essential Role of the Histone Methyltransferase G9a in Cocaine-Induced Plasticity. Science. 2012;327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wei J, Dai Y, Wen W, Li J, Ye LL, Xu S, et al. Blood-brain barrier integrity is the primary target of alcohol abuse. Chem Biol Interact. 2021. ;337:109400. [DOI] [PubMed] [Google Scholar]
- 76.Vore AS, Deak T. blood-brain barrier function in health and disease across development. International Review of Neurobiology. 2021;161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Rubio-Araiz A, Porcu F, Pérez-hernández M, García-gutiérrez MS, Aracil-fernández MA, Gutierrez-lópez MD, et al. Disruption of blood – brain barrier integrity in postmortem alcoholic brain : preclinical evidence of TLR4 involvement from a binge-like drinking model. Addiction Biology. 2016:1103–16. [DOI] [PubMed] [Google Scholar]
- 78.Ahmed Z, Shaw G, Sharma VP, Yang C, McGowan E, Dickson DW. Actin-binding Proteins Coronin-1a and IBA-1 are Effective Microglial Markers for Immunohistochemistry. Journal of Histochemistry & Cytochemistry. 2007;55. [DOI] [PubMed] [Google Scholar]
- 79.Hovens IB, Nyakas C, Schoemaker RG. A novel method for evaluating microglial activation using ionized calcium-binding adaptor protein-1 staining: cell body to cell size ratio. Neuroimmunol Neuroinflammation. 2014;1(2). [Google Scholar]
- 80.Sasaki Y, Ohsawa K, Kanazawa H, Kohsaka S, Imai Y. Iba1 is an actin-cross-linking protein in macrophages/microglia. Biochemical and Biophysical Research Communications. 2001;286(2). [DOI] [PubMed] [Google Scholar]
- 81.Brenner E, Tiwari GR, Kapoor M, Liu Y, Brock A, Mayfield RD. Single cell transcriptome profiling of the human alcohol-dependent brain. Human Molecular Genetics. 2020;29:1144–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.McCarthy GM, Farris SP, Blednov YA, Harris RA, Mayfield RD. Microglial-specific transcriptome changes following chronic alcohol consumption. Neuropharmacology. 2018;128:416–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Erickson EK, Blednov YA, Harris RA, Mayfield RD. Glial gene networks associated with alcohol dependence. Scientific Reports. 2019;9:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zoller T, Schneider A, Kleimeyer C, Masuda T, Potru PS, Pfeifer D, et al. Silencing of TGFbeta signalling in microglia results in impaired homeostasis. Nat Commun. 2018;9(1):4011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.McClain JA, Morris SA, Deeny MA, Marshall SA, Hayes DM, Kiser ZM, et al. Adolescent binge alcohol exposure induces long-lasting partial activation of microglia. Brain, Behavior, and Immunity. 2011;25:S120–S8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Karmodiya K, Krebs AR, Oulad-Abdelghani M, Kimura H, Tora L. H3K9 and H3K14 acetylation co-occur at many gene regulatory elements, while H3K14ac marks a subset of inactive inducible promoters in mouse embryonic stem cells. BMC Genomics. 2012;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ponomarev I, Wang S, Zhang L, Adron Harris R, Dayne Mayfield R. Gene Coexpression Networks in Human Brain Identify Epigenetic Modifications in Alcohol Dependence. Journal of Neuroscience. 2012;32:1884–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Johnstone AL, Andrade NS, Barbier E, Khomtchouk BB, Rienas CA, Lowe K, et al. Dysregulation of the histone demethylase KDM6B in alcohol dependence is associated with epigenetic regulation of inflammatory signaling pathways. Addiction Biology. 2019:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Agrawal RG, Owen JA, Levin PS, Hewetson A, Berman AE, Franklin R, et al. Bioinformatics Analyses Reveals Age-Specific Neuroimmune Modulation as a Target for Treatment of High Ethanol Drinking. Alcohol Clin Exp Res. 2015;38:428–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Walter TJ, Crews FT. Microglial depletion alters the brain neuroimmune response to acute binge ethanol withdrawal. J Neuroinflammation: BioMed Central; 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Warden AS, Wolfe SA, Khom S, Varodayan FP, Patel RR, Steinman MQ, et al. Microglia Control Escalation of Drinking in Alcohol-Dependent Mice: Genomic and Synaptic Drivers. Biological Psychiatry: Elsevier; 2020. p. 910–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pathan H, Williams J. Basic opioid pharmacology: an update. British Journal of Pain. 2012;6:11–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kopecky EA. Opioid Pharmacology: Developmental Effects on Opioid Metabolism. Clinical Journal of Pain. 2019;35:481–6. [DOI] [PubMed] [Google Scholar]
- 94.Stimulatory effects of opioids on transmitter release and possible cellular mechanisms: Overview and original results, (1996). [DOI] [PubMed] [Google Scholar]
- 95.Johnson SW, North RA. Opioids excite dopamine neurons by hyperpolarization of local interneurons. Journal of Neuroscience. 1992;12:483–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Nafziger AN, Barkin RL. Opioid Therapy in Acute and Chronic Pain. The Journal of Clinical Pharmacology. 2018;58(9):1111–22. [DOI] [PubMed] [Google Scholar]
- 97.Seney ML, Kim SM, Glausier JR, Hildebrand MA, Xue X, Zong W, et al. Transcriptional Alterations in Dorsolateral Prefrontal Cortex and Nucleus Accumbens Implicate Neuroinflammation and Synaptic Remodeling in Opioid Use Disorder. Biological Psychiatry. 2021;90:550–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hutchinson MR, Zhang Y, Shridhar M, Evans JH, Buchanan MM, Zhao TX, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain, Behavior, and Immunity: Academic Press; 2010. p. 83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hutchinson MR, Northcutt AL, Hiranita T, Wang X, Lewis SS, Thomas J, et al. Opioid Activation of Toll-Like Receptor 4 Contributes to Drug Reinforcement. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Theberge FR, Li X, Kambhampati S, Pickens CL, St. Laurent R, Bossert JM, et al. Effect of chronic delivery of the toll-like receptor 4 antagonist (+)-naltrexone on incubation of heroin craving. Biological Psychiatry. 2013;73:729–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Eidson LN, Inoue K, Young LJ, Tansey MG, Murphy AZ. Toll-like Receptor 4 Mediates Morphine-Induced Neuroinflammation and Tolerance via Soluble Tumor Necrosis Factor Signaling. Neuropsychopharmacology. 2017;42:661–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chen J-X, Huang K-M, Liu M, Jiang J-X, Liu J-P, Zhang Y-X, et al. Activation of TLR4/STAT3 signaling in VTA contributes to the acquisition and maintenance of morphine-induced conditioned place preference. Behav Brain Res. 2017;335:151–7. [DOI] [PubMed] [Google Scholar]
- 103.Bsibsi M, Ravid R, Gveric D, Van Noort JM. Broad Expression of Toll-Like Receptors in the Human Central Nervous System. Journal of Neuropathology & Experimental Neurology. 2002;61:1013–21. [DOI] [PubMed] [Google Scholar]
- 104.Pocock J, Kettenmann H. Neurotransmitter receptors on microglia. Trends in Neurosciences 2007. p. 527–35. [DOI] [PubMed] [Google Scholar]
- 105.Rivera PD, Hanamsagar R, Kan MJ, Tran PK, Stewart D, Jo YC, et al. Removal of microglial-specific MyD88 signaling alters dentate gyrus doublecortin and enhances opioid addiction-like behaviors. Brain, Behavior, and Immunity. 2019;76:104–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Reiss D, Maduna T, Maurin H, Audouard E, Gaveriaux-Ruff C. Mu opioid receptor in microglia contributes to morphine analgesic tolerance, hyperalgesia, and withdrawal in mice. Journal of Neuroscience Research. 2022;100:203–19. [DOI] [PubMed] [Google Scholar]
- 107.Zhang H, Largent-Milnes TM, Vanderah TW. Glial neuroimmune signaling in opioid reward. Brain Research Bulletin. 2020;155:102–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liang Y, Chu H, Jiang Y, Yuan L. Morphine enhances IL-1β release through toll-like receptor 4-mediated endocytic pathway in microglia. Purinergic Signaling. 2016;12:637–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Coffey KR, Lesiak AJ, Marx RG, Vo EK, Garden GA, Neumaier JF. A cAMP-Related Gene Network in Microglia Is Inversely Regulated by Morphine Tolerance and Withdrawal. Biological Psychiatry Global Open Science. 2021;0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Browne CJ, Godino A, Salery M, Nestler EJ. Epigenetic Mechanisms of Opioid Addiction. Biological Psychiatry. 2020;87:22–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Saad MH, Rumschlag M, Guerra MH, Savonen CL, Jaster AM, Olson PD, et al. Differentially expressed gene networks, biomarkers, long noncoding RNAs, and shared responses with cocaine identified in the midbrains of human opioid abusers. Scientific Reports 2019 9:1. 2019;9:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Tapocik JD, Luu TV, Mayo CL, Wang BD, Doyle E, Lee AD, et al. Neuroplasticity, axonal guidance and micro-RNA genes are associated with morphine self-administration behavior. Addiction Biology. 2013;18:480–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liang D-Y, Li X, Clark JD. Epigenetic Regulation of Opioid-Induced Hyperalgesia, Dependence, and Tolerance in Mice. The Journal of Pain. 2013;14:36–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Tsai R-Y, Wang J-C, Chou K-Y, Wong C-S, Cherng C-H. Resveratrol reverses morphine-induced neuroinflammation in morphine-tolerant rats by reversal HDAC1 expression. Journal of the Formosan Medical Association. 2016;325:445–54. [DOI] [PubMed] [Google Scholar]
- 115.Zhang Y, Liang Y, Levran O, Randesi M, Yuferov V, Zhao C, et al. Alterations of expression of inflammation/immune-related genes in the dorsal and ventral striatum of adult C57BL/6J mice following chronic oxycodone self-administration: a RNA sequencing study. Psychopharmacology. 2017;234:2259–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Metz VE, Jones JD, Manubay J, Sullivan MA, Mogali S, Segoshi A, et al. Effects of Ibudilast on the Subjective, Reinforcing, and Analgesic Effects of Oxycodone in Recently Detoxified Adults with Opioid Dependence. Neuropsychopharmacology. 2017;42:1825–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Liu Q, Tang S, Du X, Sun Y, Lai J, Wang X. Injection of minocycline into the periaqueductal gray attenuates morphine withdrawal signs. Neuroscience Letters. 2020;736:135283. [DOI] [PubMed] [Google Scholar]
- 118.Schwarz JM, Hutchinson MR, Bilbo SD. Early-Life Experience Decreases Drug-Induced Reinstatement of Morphine CPP in Adulthood via Microglial-Specific Epigenetic Programming of Anti-Inflammatory IL-10 Expression. Journal of Neuroscience. 2011;31:17835–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Neuroimmune mechanisms of psychostimulant and opioid use disorders, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Shaerzadeh F, Streit WJ, Heysieattalab S, Khoshbouei H. Methamphetamine neurotoxicity, microglia, and neuroinflammation. J Neuroinflammation. 2018;15:341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Piechota M, Korostynski M, Solecki W, Gieryk A, Slezak M, Bilecki W, et al. The dissection of transcriptional modules regulated by various drugs of abuse in the mouse striatum. Genome Biol. 2010;11(5):R48–R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sora I, Hall FS, Andrews AM, Itokawa M, Li XF, Wei HB, et al. Molecular mechanisms of cocaine reward: combined dopamine and serotonin transporter knockouts eliminate cocaine place preference. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(9):5300–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kish SJ. Pharmacologic mechanisms of crystal meth. CMAJ : Canadian Medical Association journal = journal de l'Association medicale canadienne. 2008;178:1679–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Fantegrossi WE, Ciullo JR, Wakabayashi KT, De La Garza R, 2nd, Traynor JR, Woods JH. A comparison of the physiological, behavioral, neurochemical and microglial effects of methamphetamine and 3,4-methylenedioxymethamphetamine in the mouse. Neuroscience. 2008;151 (2):533–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Mansvelder HD, McGehee DS. Cellular and synaptic mechanisms of nicotine addiction. Journal of Neurobiology. 2002;53:606–17. [DOI] [PubMed] [Google Scholar]
- 126.Wonnacott S, Barik J. Molecular and cellular mechanisms of action of nicotine in the CNS. Handbook of Experimental Pharmacology. 2009;192:173–207. [DOI] [PubMed] [Google Scholar]
- 127.Marks MJ, Burch JB, Collins AC. Genetics of nicotine response in four inbred strains of mice. J Pharmacol Exp Ther. 1983;226(1):291–302. [PubMed] [Google Scholar]
- 128.Freeman GB, Sherman KA, Gibson GE. Locomotor activity as a predictor of times and dosages for studies of nicotine's neurochemical actions. Pharmacol Biochem Behav. 1987;26(2):305–12. [DOI] [PubMed] [Google Scholar]
- 129.Kita T, Nakashima T, Shirase M, Asahina M, Kurogochi Y. Effects of nicotine on ambulatory activity in mice. Jpn J Pharmacol. 1988;46(2):141–6. [DOI] [PubMed] [Google Scholar]
- 130.Ahmed SH, Lutjens R, van der Stap LD, Lekic D, Romano-Spica V, Morales M, et al. Gene expression evidence for remodeling of lateral hypothalamic circuitry in cocaine addiction. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(32):11533–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Northcutt AL, Hutchinson MR, Wang X, Baratta MV, Hiranita T, Cochran TA, et al. DAT isn't all that: cocaine reward and reinforcement require Toll-like receptor 4 signaling. Mol Psychiatry. 2015;20(12):1525–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wang X, Northcutt AL, Cochran TA, Zhang X, Fabisiak TJ, Haas ME, et al. Methamphetamine Activates Toll-Like Receptor 4 to Induce Central Immune Signaling within the Ventral Tegmental Area and Contributes to Extracellular Dopamine Increase in the Nucleus Accumbens Shell. ACS Chemical Neuroscience: American Chemical Society; 2019. p. 3622–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Brody AL, Hubert R, Enoki R, Garcia LY, Mamoun MS, Okita K, et al. Effect of Cigarette Smoking on a Marker for Neuroinflammation: A [11C]DAA1106 Positron Emission Tomography Study. Neuropsychopharmacology. 2017;42:1630–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Guan YZ, Jin XD, Guan LX, Yan HC, Wang P, Gong Z, et al. Nicotine Inhibits Microglial Proliferation and Is Neuroprotective in Global Ischemia Rats. Molecular Neurobiology. 2015;51:1480–8. [DOI] [PubMed] [Google Scholar]
- 135.Gonçalves J, Martins A, Ferreira R, Milhazes N, Borges F, Ribeiro CF, et al. Methamphetamine-lnduced Early Increase of IL-6 and TNF-α mRNA Expression in the Mouse Brain. Annals of the New York Academy of Sciencies. 2008;1139:103–11. [DOI] [PubMed] [Google Scholar]
- 136.Burkovetskaya ME, Small R, Guo L, Buch S, Guo ML. Cocaine self-administration differentially activates microglia in the mouse brain. Neuroscience Letters. 2020;728:134951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Lewitus GM, Konefal SC, Greenhalgh AD, Pribiag H, Augereau K, Stellwagen D. Microglial TNF-α Suppresses Cocaine-Induced Plasticity and Behavioral Sensitization. Neuron. 2016;90:483–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Fernandes NC, Sriram U, Gofman L, Cenna JM, Ramirez SH, Potula R. Methamphetamine alters microglial immune function through P2X7R signaling. J Neuroinflammation. 2016;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Adeluyi A, Guerin L, Fisher ML, Galloway A, Cole RD, Chan SSL, et al. Microglia morphology and proinflammatory signaling in the nucleus accumbens during nicotine withdrawal. Science Advances. 2019;5:eaax7031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cruickshank CC, Dyer KR. A review of the clinical pharmacology of methamphetamine. Addiction. 2009;104:1085–99. [DOI] [PubMed] [Google Scholar]
- 141.Ru Q, Xiong Q, Zhou M, Chen L, Tian X, Xiao H, et al. Withdrawal from chronic treatment with methamphetamine induces anxiety and depression-like behavior in mice. Psychiatry Research. 2019;271:476–83. [DOI] [PubMed] [Google Scholar]
- 142.Saravia R, Ten-Bianco M, Grande MT, Maldonado R, Berrendero F. Anti-inflammatory agents for smoking cessation? Focus on cognitive deficits associated with nicotine withdrawal in male mice. Brain, Behavior, and Immunity. 2019;75:228–39. [DOI] [PubMed] [Google Scholar]
- 143.Zhang X, Xu P, Li C, Zhu W, Wu S, Yu A, et al. Spinal microglial P2X4 receptor–brain-derived neurotrophic factor signaling regulates nicotine withdrawal-induced hyperalgesia. NeuroReport. 2017;28:339–47. [DOI] [PubMed] [Google Scholar]
- 144.Silva AP, Ambrosio E, Leitão RA, Assis MA, Gonçalves J, Coria SM, et al. Extended-access methamphetamine self-administration elicits neuroinflammatory response along with blood-brain barrier breakdown. Brain, Behavior, and Immunity. 2017;62:306–17. [DOI] [PubMed] [Google Scholar]
- 145.Cadet JL, Jayanthi S, Mccoy MT, Vawter M, Ladenheim B. Temporal profiling of methamphetamine-induced changes in gene expression in the mouse brain: Evidence from cDNA array. Synapse. 2001;41:40–8. [DOI] [PubMed] [Google Scholar]
- 146.Krasnova IN, Justinova Z, Cadet JL. Methamphetamine addiction: involvement of CREB and neuroinflammatory signaling pathways. Psychopharmacology. 2016;233:1945–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Zhou L, Tao X, Pang G, Mu M, Sun Q, Liu F, et al. Maternal Nicotine Exposure Alters Hippocampal Microglia Polarization and Promotes Anti-inflammatory Signaling in Juvenile Offspring in Mice. Front Pharmacol. 2021;12:661304-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Tien L-T, Cai Z, Rhodes PG, Fan L-W. Neonatal exposure to lipopolysaccharide enhances methamphetamine-induced reinstated behavioral sensitization in adult rats. Behav Brain Res. 2011;224:166–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Worley MJ, Heinzerling KG, Roche DJO, Shoptaw S. Ibudilast attenuates subjective effects of methamphetamine in a placebo-controlled inpatient study. Drug and Alcohol Dependence 2016. p. 245–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Snider SE, Hendrick ES, Beardsley PM. Glial cell modulators attenuate methamphetamine self-administration in the rat. European Journal of Pharmacology. 2013;701:124–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Attarzadeh-Yazdi G, Arezoomandan R, Haghparast A. Minocycline, an antibiotic with inhibitory effect on microglial activation, attenuates the maintenance and reinstatement of methamphetamine-seeking behavior in rat. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2014;53:142–8. [DOI] [PubMed] [Google Scholar]
- 152.Wang H, Huang W, Liang M, Shi Y, Zhang C, Li Q, et al. (+)-JQ1 attenuated LPS-induced microglial inflammation via MAPK/NFκB signaling. Cell and Bioscience. 2018;8:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sartor GC, Powell SK, Brothers SP, Wahlestedt C. Epigenetic Readers of Lysine Acetylation Regulate Cocaine- Induced Plasticity. Journal of Neuroscience. 2015;35:15062–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kruidenier L, Chung C-w, Cheng Z, Liddle J, Che K, Joberty G, et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature. 2012;488:404–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zhang Y-X, Akumuo RC, España RA, Yan C-X, Gao W-J, Li Y-C. The histone demethylase KDM6B in the medial prefrontal cortex epigenetically regulates cocaine reward memory. Neuropharmacology. 2018;141:113–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Iversen L. Cannabis and the brain. Brain. 2003;126:1252–70. [DOI] [PubMed] [Google Scholar]
- 157.Turcotte C, Blanchet MR, Laviolette M, Flamand N. The CB2 receptor and its role as a regulator of inflammation. Cellular and Molecular Life Sciences 2016 73:23. 2016;73:4449–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Cabral GA, Marciano-Cabral F. Cannabinoid receptors in microglia of the central nervous system: immune functional relevance. Journal of Leukocyte Biology. 2005;78:1192–7. [DOI] [PubMed] [Google Scholar]
- 159.Molina-Holgado F, Pinteaux E, Moore JD, Molina-Holgado E, Guaza C, Gibson RM, et al. Endogenous Interleukin-1 Receptor Antagonist Mediates Anti-Inflammatory and Neuroprotective Actions of Cannabinoids in Neurons and Glia. Journal of Neuroscience. 2003;23:6470–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Reusch N, Ravichandran KA, Olabiyi BF, Komorowska-Müller JA, Hansen JN, Ulas T, et al. Cannabinoid receptor 2 is necessary to induce toll-like receptor-mediated microglial activation. Glia. 2021;70:71–88. [DOI] [PubMed] [Google Scholar]
- 161.Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, et al. Nonpsychotropic Cannabinoid Receptors Regulate Microglial Cell Migration. Journal of Neuroscience. 2003;23:1398–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.VanRyzin JW, Marquardt AE, Argue KJ, Vecchiarelli HA, Ashton SE, Arambula SE, et al. Microglial Phagocytosis of Newborn Cells Is Induced by Endocannabinoids and Sculpts Sex Differences in Juvenile Rat Social Play. Neuron. 2019;102:435–49.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Filbey FM, Aslan S, Calhoun VD, Spence JS, Damaraju E, Caprihan A, et al. Long-term effects of marijuana use on the brain. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:16913–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Battistella G, Fornari E, Annoni JM, Chtioui H, Dao K, Fabritius M, et al. Long-Term Effects of Cannabis on Brain Structure. Neuropsychopharmacology 2014 39:9. 2014;39:2041–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Tomasiewicz HC, Jacobs MM, Wilkinson MB, Wilson SP, Nestler EJ, Hurd YL. Proenkephalin Mediates the Enduring Effects of Adolescent Cannabis Exposure Associated with Adult Opiate Vulnerability. Biological Psychiatry. 2012;72:803–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Dukes Ba AJ, Fowler JP, Lallai V, Pushkin Bs AN, Fowler CD. Adolescent Cannabinoid and Nicotine Exposure Differentially Alters Adult Nicotine Self-Administration in Males and Females. Nicotine & Tobacco Research.2020:1364–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Higuera-Matas A, Luisa Soto-Montenegro M, Del Olmo N, Miguéns M, Torres I, José Vaquero J, et al. Augmented Acquisition of Cocaine Self-Administration and Altered Brain Glucose Metabolism in Adult Female but not Male Rats Exposed to a Cannabinoid Agonist during Adolescence. Neuropsychopharmacology 2008 33:4. 2007;33:806–13. [DOI] [PubMed] [Google Scholar]
- 168.Hurd YL, Spriggs S, Alishayev J, Winkel G, Gurgov K, Kudrich C, et al. Cannabidiol for the reduction of cue-induced craving and anxiety in drug-abstinent individuals with heroin use disorder: A double-blind randomized placebo-controlled trial. American Journal of Psychiatry. 2019;176:911–22. [DOI] [PubMed] [Google Scholar]
- 169.Merighi S, Gessi S, Varani K, Fazzi D, Mirandola P, Borea PA. Cannabinoid CB2 receptor attenuates morphine-induced inflammatory responses in activated microglial cells. British Journal of Pharmacology. 2012;166:2371–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Tumati S, Largent-Milnes TM, Keresztes A, Ren J, Roeske WR, Vanderah TW, et al. Repeated morphine treatment-mediated hyperalgesia, allodynia and spinal glial activation are blocked by co-administration of a selective cannabinoid receptor type-2 agonist. Journal of Neuroimmunology. 2012;244:23–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Anooshe M, Nouri K, Karimi-Haghighi S, Mousavi Z, Haghparast A. Cannabidiol efficiently suppressed the acquisition and expression of methamphetamine-induced conditioned place preference in the rat. Behav Brain Res. 2021;404:113158. [DOI] [PubMed] [Google Scholar]
- 172.Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. British journal of pharmacology. 2016;173(4):649–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Jurga AM, Paleczna M, Kuter KZ. Overview of General and Discriminating Markers of Differential Microglia Phenotypes. Frontiers in cellular neuroscience. 2020;14:198-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Blasi E, Barluzzi R, Bocchini V, Mazzolla R, Bistoni F. Immortalization of murine microglial cells by a v-raf / v-myc carrying retrovirus. Journal of Neuroimmunology. 1990;27(2):229–37. [DOI] [PubMed] [Google Scholar]
- 175.Dello Russo C, Cappoli N, Coletta I, Mezzogori D, Paciello F, Pozzoli G, et al. The human microglial HMC3 cell line: where do we stand? A systematic literature review. J Neuroinflammation. 2018;15(1):259-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Lindberg C, Hjorth E, Post C, Winblad B, Schultzberg M. Cytokine production by a human microglial cell line: Effects of ßamyloid and α-melanocyte-stimulating hormone. Neurotoxicity Research. 2005;8(3):267–76. [DOI] [PubMed] [Google Scholar]
- 177.Liu J, Hjorth E, Zhu M, Calzarossa C, Samuelsson E-B, Schultzberg M, et al. Interplay between human microglia and neural stem/progenitor cells in an allogeneic co-culture model. J Cell Mol Med. 2013;17(11):1434–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Abud EM, Ramirez RN, Martinez ES, Healy LM, Nguyen CHH, Newman SA, et al. iPSC-Derived Human Microglia-like Cells to Study Neurological Diseases. Neuron. 2017;94(2):278–93 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.McQuade A, Coburn M, Tu CH, Hasselmann J, Davtyan H, Blurton-Jones M. Development and validation of a simplified method to generate human microglia from pluripotent stem cells. Mol Neurodegener. 2018;13(1):67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Drager NM, Sattler SM, Huang CT, Teter OM, Leng K, Hashemi SH, et al. A CRISPRi/a platform in human iPSC-derived microglia uncovers regulators of disease states. Nat Neurosci. 2022;25(9):1149–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Mello NK, Negus SS. Preclinical Evaluation of Pharmacotherapies for Treatment of Cocaine and Opioid Abuse Using Drug Self-Administration Procedures. Neuropsychopharmacology. 1996;14(6):375–424. [DOI] [PubMed] [Google Scholar]
- 182.Thomsen M, Caine SB. Intravenous drug self-administration in mice: Practical considerations. Behavior Genetics. 2007;37:101–18. [DOI] [PubMed] [Google Scholar]
- 183.Dickson PE, Miller MM, Calton MA, Bubier JA, Cook MN, Goldowitz D, et al. Systems genetics of intravenous cocaine self-administration in the BXD recombinant inbred mouse panel. Psychopharmacology. 2016;233(4):701–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Lacagnina MJ, Rivera PD, Bilbo SD. Glial and Neuroimmune Mechanisms as Critical Modulators of Drug Use and Abuse. Neuropsychopharmacology. 2017;42:156–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Szepesi Z, Manouchehrian O, Bachiller S, Deierborg T. Bidirectional Microglia-Neuron Communication in Health and Disease. Frontiers in cellular neuroscience. 2018;12:323-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Easley-Neal C, Foreman O, Sharma N, Zarrin AA, Weimer RM. CSF1R Ligands IL-34 and CSF1 Are Differentially Required for Microglia Development and Maintenance in White and Gray Matter Brain Regions. Frontiers in Immunology. 2019;0:2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Das A, Chai JC, Kim SH, Lee YS, Park KS, Jung KH, et al. Transcriptome sequencing of microglial cells stimulated with TLR3 and TLR4 ligands. BMC Genomics. 2015;16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, et al. New tools for studying microglia in the mouse and human CNS. Proceedings of the National Academy of Sciences. 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Williamson LL, Sholar PW, Mistry RS, Smith SH, Bilbo SD. Microglia and memory: modulation by early-life infection. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2011;31(43):15511–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Amici SA, Dong J, Guerau-de-Arellano M. Molecular Mechanisms Modulating the Phenotype of Macrophages and Microglia. Frontiers in immunology. 2017;8:1520-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Masuda T, Sankowski R, Staszewski O, Böttcher C, Amann L, Scheiwe C, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566:388. [DOI] [PubMed] [Google Scholar]
- 192.Hove HV, Martens L, Scheyltjens I, Vlaminck KD, Antunes ARP, Prijck SD, et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nature Neuroscience 2019. 2019:1. [DOI] [PubMed] [Google Scholar]
- 193.Li Q, Cheng Z, Zhou L, Darmanis S, Neff NF, Okamoto J, et al. Developmental Heterogeneity of Microglia and Brain Myeloid Cells Revealed by Deep Single-Cell RNA Sequencing. Neuron. 2019;101:207–23.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Coffey KR, Lesiak AJ, Marx RG, Vo EK, Garden GA, Neumaier JF. RiboTag-Seq Reveals a Compensatory cAMP Responsive Gene Network in Striatal Microglia Induced by Morphine Withdrawal. bioRxiv. 2020:2020.02.10.942953. [Google Scholar]
- 195.Haimon Z, Volaski A, Orthgiess J, Boura-Halfon S, Varol D, Shemer A, et al. Re-evaluating microglia expression profiles using RiboTag and cell isolation strategies. Nat Immunol. 2018;19(6):636–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Gao Y, Zhao X. sncRiboTag-Seq: cell-type-specific RiboTag-Seq for cells in low abundance in mouse brain tissue. STAR Protoc. 2020;2(1):100231-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Sanz E, Bean JC, Carey DP, Quintana A, McKnight GS. RiboTag: Ribosomal Tagging Strategy to Analyze Cell-Type-Specific mRNA Expression In Vivo. Curr Protoc Neurosci. 2019;88(1):e77–e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A, et al. Single-Cell RNA Sequencing of Microglia throughout the Mouse Lifespan and in the Injured Brain Reveals Complex Cell-State Changes. Immunity. 2019;50:253–71.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Yeh H, Ikezu T. Transcriptional and Epigenetic Regulation of Microglia in Health and Disease. Trends in Molecular Medicine. 2018;25:96–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Fujita Y, Yamashita T. Alterations in Chromatin Structure and Function in the Microglia. Front Cell Dev Biol. 2021;8:626541-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Schmunk G, Kim CN, Soliman SS, Keefe MG, Bogdanoff D, Tejera D, et al. Human microglia upregulate cytokine signatures and accelerate maturation of neural networks. bioRxiv. 2020:2020.03.24.006874. [Google Scholar]
- 202.Guo X, Chen D, An S, Wang Z. ChIP-seq Profiling Identifies Histone Deacetylase 2 Targeting Genes Involved in Immune and Inflammatory Regulation Induced by Calcitonin Gene-Related Peptide in Microglial Cells. J Immunol Res. 2020;2020:4384696-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Satoh J-I, Asahina N, Kitano S, Kino Y. A Comprehensive Profile of ChIP-Seq-Based PU.1/Spi1 Target Genes in Microglia. Gene Regul Syst Bio. 2014;8:127–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh YE, et al. Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci. 2018;21(8):1049–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Grabert K, Michoel T, Karavolos MH, Clohisey S, Baillie JK, Stevens MP, et al. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat Neurosci. 2016;19(3):504–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Bachiller S, Jimenez-Ferrer I, Paulus A, Yang Y, Swanberg M, Deierborg T, et al. Microglia in Neurological Diseases: A Road Map to Brain-Disease Dependent-Inflammatory Response. Front Cell Neurosci. 2018;12:488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Skene PJ, Henikoff JG, Henikoff S. Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nature Protocols. 2018;13(5):1006–19. [DOI] [PubMed] [Google Scholar]
- 208.Skene PJ, Henikoff S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife. 2017;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Kaya-Okur HS, Wu SJ, Codomo CA, Pledger ES, Bryson TD, Henikoff JG, et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nature Communications 2019 10:1.2019;10:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Kaya-Okur HS, Janssens DH, Henikoff JG, Ahmad K, Henikoff S. Efficient low-cost chromatin profiling with CUT&Tag. Nature Protocols. 2020;15:3264–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Zhang YX, Akumuo RC, Espana RA, Yan CX, Gao WJ, Li YC. The histone demethylase KDM6B in the medial prefrontal cortex epigenetically regulates cocaine reward memory. Neuropharmacology. 2018;141:113–25. [DOI] [PMC free article] [PubMed] [Google Scholar]


