Abstract
Objectives
Preclinical and clinical research in neuropsychiatric disorders, particularly mood and substance use disorders, have historically focused on neurons; however, glial cells – astrocytes, microglia, and oligodendrocytes – also play key roles in these disorders.
Methods
Peer-reviewed PubMed/Medline articles published through December 2012 were identified using the following keyword combinations: glia, astrocytes, oligodendrocytes/glia, microglia, substance use, substance abuse, substance dependence, alcohol, opiate, opioid, cocaine, psychostimulants, stimulants, and glutamate.
Results
Depressive and substance use disorders are highly comorbid, suggesting a common or overlapping aetiology and pathophysiology. Reduced astrocyte cell number occurs in both disorders. Altered glutamate neurotransmission and metabolism – specifically changes in the levels/activity of transporters, receptors, and synaptic proteins potentially related to synaptic physiology – appear to be salient features of both disorders. Glial cell pathology may also underlie the pathophysiology of both disorders via impaired astrocytic production of neurotrophic factors. Microglial/neuroinflammatory pathology is also evident in both depressive and substance use disorders. Finally, oligodendrocyte impairment decreases myelination and impairs expression of myelin-related genes in both substance use and depressive disorders.
Conclusions
Glial-mediated glutamatergic dysfunction is a common neuropathological pathway in both substance use and depression. Therefore, glutamatergic neuromodulation is a rational drug target in this comorbidity.
Keywords: Glia, depression, glutamate, alcohol use disorders, drug use disorders
Introduction
Substance use disorders (SUDs) frequently co-occur with other psychiatric illnesses. Conversely, primary psychiatric disorders can be mistaken for, or masked by, substance misuse. Several epidemiological surveys have demonstrated that co-occurring psychiatric disorders and SUDs are associated with a greater burden of illness and poorer longitudinal course (Mueser et al. 1998; Ziedonis 2004; Niciu et al. 2009); furthermore, clinical features alone are insufficient to diagnose depressive episode subtype in substance-abusing populations (Niciu et al. 2009). With the exception of genetic studies of depression and alcohol dependence (Kertes et al. 2011; Su et al. 2011; Edwards et al. 2012), few preclinical or clinical neuroscience investigations of dual disorders have been conducted. A number of hypotheses, however, have been proposed to explain their shared aetiology and pathophysiology, as there is considerable overlap in neural circuitry and neurotransmitter systems. In particular, the glutamatergic system is critically important in both depressive disorders and SUDs, especially impaired glial (astrocytes, microglia, and oligodendrocytes) home ostasis (Kalivas 2009; Valentine et al. 2009). This review discusses glial cell dysfunction in depressive disorders and SUDs, and attempts to synthesize neuronal-glial glutamatergic dysfunction in their comorbidity.
Methods
Peer-reviewed PubMed/Medline articles published through December 2012 were identified using the following keyword combinations: glia, astrocytes, oligodendrocytes/glia, microglia, substance use, substance abuse, substance dependence, alcohol, opiate, opioid, cocaine, psychostimulants, stimulants, and glutamate. Both preclinical and clinical investigations were reviewed. All articles were written in English. Reviewing the titles and abstracts uncovered 187 relevant reports, which were examined in full and cited herein when relevant.
Astrocytes
Alcohol
Historically, astrocyte activation has been defined by glial fibrillary acidic protein (GFAP) upregulation, although this is an oversimplification (Oberheim et al. 2012). GFAP levels increase in response to acute stressors like ischaemia and neurotoxins. Acute and chronic ethanol exposure in rats dose-dependently increases GFAP expression in several brain regions (Vongvatcharanon et al. 2010; Udomuksorn et al. 2011). However, with chronic ethanol exposure, GFAP downregulation and astrocyte cell death have been observed (Khokhrina et al. 1991). Miguel-Hildago and colleagues confirmed decreased astrocyte density in the dorsolateral and orbitofrontal cortices of alcoholics without Wernicke-Korsakoff syndrome (Miguel-Hidalgo et al. 2002, 2006). In contrast, astrocyte density increases with age and in long-standing alcoholism, which may represent gliosis due to chronic neurotoxicity (Miguel-Hidalgo et al. 2006).
Preclinical studies have demonstrated that pre-frontal cortex (PFC) GFAP-immunoreactive cell density is lower in alcohol-naive and alcohol-preferring rats relative to non-alcohol-preferring rats (Miguel-Hidalgo 2005) (see Table I). Glial cell dysfunction also predates alcohol exposure in this susceptible rodent strain. In the adult rat medial PFC, binge-like alcohol administration during adolescence decreases glial density without affecting neuronal density in male but not female rodents and without affecting cell density in the basolateral amygdala (Koss et al. 2012). However, earlier bingelike alcohol exposure increases bromodeoxyuridine-labelled (dividing) cells; 60% of these cells express glial markers into adulthood (Helfer et al. 2009). Moreover, gliotoxin or gap junction blocker infusion into the prelimbic cortex transiently increases alcohol self-administration (Miguel-Hidalgo et al. 2009; Miguel-Hidalgo 2007). In contrast to these preclinical findings, a postmortem study of alcohol-dependent depressed suicide completers revealed increased glial packing density in the anterior cingulate cortex (ACC) relative to non-alcoholic individuals with depression who committed suicide as well as sudden (non-suicide) deaths (Hercher et al. 2009). Taken together, the results suggest that altered astrocyte density may be a cause instead of a consequence of alcoholism.
Table I.
SUDs | Depression |
---|---|
Decreased astrocyte density in PFC and other brain regions (AUDs) |
Decreased astrocyte density in PFC and other brain regions |
Altered glutamate transporter expression e.g., decreased astrocytic EAAT2/ GLT- 1 in DUDs (cocaine, opioids) |
Impaired glutamate transport, e.g., decreased expression of EAAT1/GLAST, EAAT2/GLT-1, EAAT4, and vGluT1 |
Changes in neurotrophic factors, e.g GDNF, BDNF, and FGF-2/bFGF |
Decreased neurotrophic factor production/secretion, e.g., BDNF |
Reactive gliosis (DUDs) | Altered glutamate and glutamine levels in several brain regions |
Astrocyte-mediated tolerance in DUDs (opioids) |
Gliotrophic effects of traditional antidepressants, e.g SSRIs, TCAs |
Gliotrophic effects of experimental antidepressants, e.g riluzole |
SUD, substance use disorder; PFC, prefrontal cortex; AUD, alcohol use disorder; EAAT2, excitatory amino acid transporter-2; GLT-1, glial glutamate transporter-1; GDNF, glial derived neurotrophic factor; BDNF, brain derived neurotrophic factor; FGF-2/bFGF, basic fibroblast growth factor-2; DUD, drug use disorder; SSRI, selective serotonin reuptake inhibitor; TCA, tricyclic antidepressant.
Italicised text denotes potential convergent mechanisms in SUDs and depressive disorders.
Next, alcohol induces glutamatergic neurotransmission abnormalities in astrocytes. Under physiological conditions, synaptic glutamate is removed by specific astrocyte transporters. The astrocytic excitatory amino acid transporter (EAAT)2 [glial glutamate transporter (GLT-1) in rodents] reuptakes glutamate from the extracellular space where it is intracellularly converted to glutamine for synaptic recycling (Niciu et al. 2012). Excessive extrasynaptic glutamate receptor stimulation initiates apoptosis, and the physiological activity of EAAT2/GLT-1 reduces excitotoxicity from synaptic spillover (Hardingham et al. 2010). In a rodent model of alcoholism, alcohol upregulates EAAT2/GLT-1 (Wu et al. 2011). EAAT2/GLT-1 is also upregulated in the amygdala and cingulate cortex of alcohol-dependent rats, and exposure-induced (but not spontaneously-induced) alcohol use can be antagonized with acamprosate, a glutamatergic neuromodulator (Rimondini et al. 2002; Hoffman et al. 2003). Although its expression is increased, alcohol disrupts EAAT2/ GLT-1-mediated functions, e.g., the modulation of potassium channel (Kv2.1)-dependent hyperpolarization of rat hippocampal neurons (Mulholland et al. 2009). In addition, the pharmacological inhibition or genetic deletion of equilibrative nucleoside transporter, a transmembrane glycoprotein responsible for nucleotide reuptake (Griffiths et al. 1997), increases alcohol consumption by decreasing the expression of EAAT2/GLT-1 and aquaporin, a channel involved in regulating brain water homeostasis, blood flow, glucose transport and metabolism, blood-brain barrier integrity, glutamate turnover, and syn-aptic plasticity (Lee et al. 2013). The cephalosporin antibiotic ceftriaxone not only restores expression of these transporters but also curtails drinking, which suggests crosstalk between adenosine and glutamate (Lee et al. 2013). As a potential genetic underpinning, a silent G→A mutation in exon 5 of EAAT2/ GLT-1 increases vulnerability to alcohol dependence and impulsivity (Foley et al. 2004; Sander et al. 2000). In addition to EAAT2/GLT-1, the other major astrocyte-expressed EAAT (EAAT1/GLAST) also affects alcohol consumption. In mice, a circadian period gene (Per2Brdm1) deletion decreases EAAT1/GLAST expression and increases alcohol consumption; similarly, acamprosate reduces glutamate levels and decreases alcohol intake (Spanagel et al. 2005). Human Per2 single nucleotide polymorphisms are also associated with increased alcohol consumption and may also be associated with sleep problems in alcoholism (Comasco et al. 2010).
Taken together, the evidence from preclinical alcoholism models as well as clinical studies suggests that alcohol alters glial cell density and has profound effects on glutamatergic neurotransmission that may be responsible for neurotoxicity and the modulation of treatment response. Excessive alcohol intake also affects glutamate transporter expression, and vice versa, possibly in an attempt to maintain homeostasis. Furthermore, altered transporter expression/function leads to increased vulnerability for alcohol preference/consumption. Finally, genotypic variations also alter glutamate transporter expression/function and alcohol intake in both rodents and humans.
Illicit drugs
Drugs of abuse also adversely affect astrocytes, providing potential pathophysiological mechanisms for initiation, maintenance, and relapse (see Table I). As in alcohol use disorders, astrocytic glutamate transporters play a pivotal role in the development and maintenance of cocaine misuse. In rodents, cocaine self-administration decreases glutamate in the nucleus accumbens (NAcc) core, which temporally coincides with decreased expression of EAAT2/ GLT-1 and xCT, the catalytic subunit of the glutamate antiporter system xC− which exchanges extracellular cysteine for intracellular glutamate (Knackstedt et al. 2010). Ceftriaxone prevents cocaine-induced reductions in the expression of both transporters and normalizes extracellular glutamate levels, which provides a viable mechanism to reverse cocaine-induced synaptic potentiation, e.g., decreased spontaneous excitatory postsynaptic currents, in the NAcc core and reduce cue- and cocaine-mediated reinstatement (Knackstedt et al. 2010; Trantham-Davidson et al. 2012). N-Acetylcysteine, a cystine prodrug that also stimulates xCT and GLT-1 expression (Knackstedt et al. 2010), decreases rodent cocaine seeking and other cocaine-related behaviours and reduces cravings in non-treatment-seeking cocaine-dependent subjects (Baker et al. 2003; Madayag et al. 2007; Amen et al. 2011). Diffuse astrogliosis has also been linked to methamphetamines (Pubill et al. 2003).
On a behavioural level, the infusion of astrocyte-conditioned media into the rodent NAcc incites locomotor sensitization and reward processing with methamphetamines (Narita et al. 2005, 2006b). The evidence, therefore, suggests that glial-mediated dysfunction, especially in glutamatergic neurotransmission, leads to adverse behavioural sequelae associated with initiation, maintenance, and relapse to cocaine and other psychostimulants. In short, cocaine alters the ability to maintain extracellular levels of glutamate and decreases synaptic plasticity in the NAcc. This renders both mice and humans more susceptible to cue-induced reinstatement. Drugs that increase xCT and/or EAAT2/GLT-1 expression restore extracellular glutamate levels, thus preventing reinstatement.
Similar to the effects of methamphetamines, chronic morphine treatment increases GFAP expression in brain regions involved in addictive disorders: ventral tegmental area (VTA), NAcc, and PFC (Beitner-Johnson et al. 1993; Song et al. 2001; Marie-Claire et al. 2004). Furthermore, the gliotoxin fluorocitrate reduces morphine-induced GFAP immunoreactivity and mitigates tolerance (Song et al. 2001). Finally, morphine tolerance and blunted analgesia correlate with glutamate transporter down-regulation in the spinal cord (Mao et al. 2002). The glutamate transporter stimulator MS-153 significantly reduces conditioned place preference (CPP) in mice treated with opioids (and psychostimulants) without altering locomotion (Nakagawa et al. 2005). MS-153 also attenuates morphine tolerance and reduces the signs/symptoms of opioid dependence (Nakagawa et al. 2001). Furthermore, adenovirally mediated intra-NAcc shell EAAT2/GLT-1 transduction reduces morphine-induced CPP (Fujio et al. 2005). Therefore, astrocyte-based glutamatergic dysfunction is important in the initiation and maintenance of opioid misuse, and targeting these deficits reduces some of the core features of dependence, e.g., CPP.
In addition to their critical role in neurotransmitter dynamics, activated astrocytes also secrete neurotrophic factors, e.g., glial-derived neurotrophic factor (GDNF) (Appel et al. 1997; Lee et al. 2006). GDNF is neuroprotective for medial striatal and VTA dopaminergic neurons on exposure to psycho-stimulants (Cass 1996; Boger et al. 2007). Intra-VTA GDNF infusion blocks numerous biochemical and behavioural sequelae of chronic cocaine and morphine misuse; conversely, intra-VTA anti-GDNF antibodies augment cocaine-induced stereotypy (Messer et al. 2000). Finally, infusion of the dipeptide leucine-isoleucine – an identified GDNF and tumour necrosis factor alpha (TNF-α) inducer of expression – inhibits psychostimulant-induced CPP and sensitization (Niwa et al. 2007).
Depression
Astrocytes have also been implicated in the aetiopathogenesis of preclinical models of despair and clinical depression (see Table I). Postmortem studies of major depressive disorder (MDD) demonstrate decreased glial cell density in several brain regions, including orbitofrontal cortex, dorsolateral PFC (dlPFC) (Rajkowska et al. 1999), ACC (Cotter et al. 2001), and amygdala (Bowley et al. 2002). An age-dependent reduction in GFAP-immunoreactive astrocyte density has also been observed in the PFC of younger individuals with MDD (Miguel-Hidalgo et al. 2000), but not in the supragenual ACC in late-life depression relative to age-matched controls (Khundakar et al. 2011). There is also a significant reduction in aquaporin-immunoreactive astrocytic end feet contacting gray matter vessels in the PFC of individuals with MDD, which suggests that blood-brain barrier permeability may be altered in depression (Rajkowska et al. 2013).
Data are mixed regarding diagnostic specificity, e.g., decreased GFAP expression in the amygdala of individuals with MDD, but not those with bipolar disorder (BD) (Altshuler et al. 2010), and in the cerebellum of individuals with MDD but not those with schizophrenia (Fatemi et al. 2004). Another report identified dlPFC layer-specific GFAP reductions in individuals with schizophrenia compared to healthy controls (Rajkowska et al. 2002). In addition, there is increased GFAP expression in subcortical brain structures in schizophrenia and MDD relative to non-psychiatric controls (Barley et al. 2009). Finally, serum levels of glial-derived S100β are increased in MDD, and these levels are not affected by antidepressant treatment (Schroeter et al. 2008). The preponderance of the above evidence, at the very least, suggests astrocytic dysfunction in the MDD brain.
In reverse translational studies, rats subjected to 15 days of chronic unpredictable stress (CUS) – a validated rodent model of depression – had a 35% decrease in cellular proliferation in the cingulate, motor, and prelimbic cortices; the selective serotonin reuptake inhibitor (SSRI) fluoxetine reversed this deficit and increased sucrose preference, a surrogate marker of anhedonia (Banasr et al. 2007). Interestingly, this study reported reduced endothelial and oligodendrocyte progenitor proliferation but no change in astrocytes and microglia (Banasr et al. 2007). Chronic social defeat by an aggressive conspecific decreased medial PFC gliogenesis and hippocampal dentate gyrus neurogenesis, but had minimal trophic effects in non-limbic brain structures; again, SSRIs reversed these impairments (Czeh et al. 2007), further suggesting the importance of serotonergic circuitry in astrocyte-mediated dysfunction.
In addition to morphological deficits, stress-induction paradigms also affect neurotrophin secretion by astrocytes. Social defeat stress reduces GFAP expression (Araya-Callis et al. 2012); this effect is not reversed by citalopram, but is partially rescued by brain-derived neurotrophic factor (Ye et al. 2011). Central inhibition of astrocyte glutamate reuptake by the EAAT2/GLT-1 inhibitor dihydrokainic acid impairs reward processing and hippocampal-dependent spatial working memory, which correlates with anhedonia-like and cognitive-like symptoms, respectively (Bechtholt-Gompf et al. 2010). The anhedonia-like phenotype could be replicated by dihydrokainic acid injection into the PFC alone (John et al. 2012). In addition, PFC astrocyte ablation by the toxin l-alpha-amino-adipic acid results in despair-like behaviours (Banasr et al. 2008). In contrast, the excito-neurotoxin ibotenate has no effect in depressogenic behavioural paradigms, suggesting that gliotoxicity may be more important than neuronal cell death in rodent despair (Banasr et al. 2008).
Astrocytes secrete many neurotrophins in response to antidepressants; amitriptyline increases basic fibroblast growth factor (FGF-2/bFGF) mRNA expression in astrocyte but not neuronal primary cultures. In the learned helplessness model, imipramine’s effects were attenuated by fluorocitrate (a reversible astrocyte inhibitor) into the hippocampal CA3/dentate gyrus (Iwata et al. 2011). Intracere-broventricular and intra-PFC infusion of FGF-2/ bFGF reverses anhedonia-like behaviours, and this effect can be blocked by infusion of a specific FGF receptor antagonist (Elsayed et al. 2012). As a result, glial-derived neurotrophins are both necessary and sufficient for antidepressant efficacy in rodent models of depression.
Astrocyte perturbations have also been studied in suicide. Astrocytic hypertrophy with longer-ramified processes was reported in ACC white matter (Torres-Platas et al. 2011) with downregulation of the dlPFC astroglial cytoskeletal proteins connexins 30 and 43 (Ernst et al. 2011). In addition, decreased expression of truncated TrkB (TrkB. T1), an astrocyte-specific neurotrophin receptor associated with epigenetic promoter methylation (Ernst et al. 2009), was observed in the frontal cortex of a subpopulation of suicide completers. The secreted calcium-binding astroglial extracellular matrix protein SPARC-like 1/hevin expression stimulates astrocyte-mediated excitatory synaptogenesis (Kucukdereli et al. 2011) and neurodevelopment (Eroglu 2009). Like TrkB.T1, SPARC-like 1/hevin expression is decreased in suicide completers (Zhurov et al. 2012).
Preclinical/mechanistic investigations have been conducted into the aetiopathophysiology of impaired gliogenesis, gliotoxicity, and/or altered synaptic function in depressive disorders. As with SUDs, decreased synaptic glutamate reuptake, synaptic spillover, and extrasynaptic glutamate receptor stimulation were the hypothesized substrates of glutamatergic dysfunction. In rat models, expression of EAAT1/GLAST, EAAT2/GLT-1, EAAT4, and vGluT1 (a vesicle-bound glutamate transporter) is suppressed (Zink et al. 2010; Gourley et al. 2012). Decreased expression of these transporters coincides with increased levels of d-serine (a differential partial agonist at the glycineB coagonist site on the N-methyl-d-aspartate (NMDA) receptor (Sheinin et al. 2001)), which attenuates excitotoxic extrasynaptic stimulation (Papouin et al. 2012; Gomez-Galan et al. 2013). Amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl) propanoic acid (AMPA) receptor membrane localization and activation may also be critical in treatment response to mood stabilizers (Du et al. 2007) and the NMDA receptor antagonist ketamine (Maeng et al. 2008). Finally, the glutamate-modulating drug riluzole stimulates astrocytic glutamate reuptake via EAAT2/GLT-1 upregulation and reverses both glial cell metabolic impairments and GFAP mRNA expression in CUS-exposed rats (Banasr et al. 2010; Gourley et al. 2012).
Adding support for a central role for glutamate, glutamate transporter downregulation has been observed in postmortem neocortical MDD micro-arrays (e.g., SLC1A2 , SLC1A3, and L-glutamate-ammonia ligase ) (Choudary et al. 2005). In the locus ceruleus, glial-expressed high-affinity glutamate transporters (SLC1A2, SLC1A3, and GLUL) are downregulated, while neuronal presynaptic vesicular glutamate transporters (SLC17A6/VGLUT2) and postsynaptic glutamate receptors (GRIA1, GRIK1, GRM1, and GRM5) are upregulated in MDD vs. BD and non-depressed controls (Bernard et al. 2011), suggesting both regional and diagnostic specificity.
Microglia
Alcohol
There is a burgeoning literature on microglia in alcohol use disorders (see Table II). In adolescent Sprague–Dawley rats, binge-like alcohol intake stimulates hippocampal microglial activation for up to 30 days after last consumption, suggesting that circumscribed exposure over a relatively brief neurodevelopmental window has long-lasting consequences (McClain et al. 2011). Ten days of intragastric ethanol administration to C57BL6/J mice also activates microglia, as detected by increasing nicotinamide adenine dinucleotide phosphate oxidase expression and increased reactive oxygen species production (Qin et al. 2012). In addition to stimulating neuroinflammatory cascades, in hypothalamic mixed cultures, ethanol’s apoptotic effects are mediated by microglia (Boyadjieva et al. 2010). Microglial activation is also critical in treatment response: minocycline, a tetracycline antibiotic and microglial inhibitor, decreases drinking in C57BL/6J mice (Agrawal et al. 2011). In a rodent model of drinking cessation, microgliosis preceded neurogenesis and volumetric recovery (Nixon et al. 2008).
Table II.
SUDs | Depression |
---|---|
Increased microglial activation | Increased microglial activation |
Increased cytokine production/secretion, e.g MCP1/CCL2, TNF-α |
Neurobehavioral response to cytokines, e.g.,“sickness behaviour”, and increased cytokine production/secretion |
TNF-α receptor antagonists and minocyclin’s therapeutic benefits in AUDs |
TNF-α receptor antagonists’ therapeutic benefits , e.g depression in psoriasis |
TLR activation→ MAPK/NF-κB signal transduction |
Normalized cytokine levels with standard antidepressants, e.g., SSRIs, MAOIs |
Microglial apoptosis in DUD (opioid) |
MCP1, microglia chemoattractant protein-1; CCL2, chemokine (C-C motif) ligand 2; TNF-α, tumor necrosis factor-α ; AUD, alcohol use disorder; TLR, toll-like receptor; MAPK, mitogen activated protein kinase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; DUD, drug use disorder; SSRI, selective serotonin reuptake inhibitor; MAOI, monoamine oxidase inhibitor.
Italicised text denotes potential convergent mechanisms in SUDs and depressive disorders.
In response to alcohol, several microglial proteins are differentially regulated across multiple brain regions. Genetic deletion of monocyte chemoattractant protein (MCP-1/CCL2), a microglial-derived chemokine, decreases drinking and alcohol preference in mice (Blednov et al. 2005). In another preclinical study, overexpression of MCP-1/CCL2 preserved long-term potentiation and fear conditioning with alcohol exposure (Bray et al. 2013). MCP-1/ CCL2 expression is also globally increased in postmortem brain homogenates from alcohol-dependent subjects (He et al. 2008). In the cingulate cortex, ionized calcium binding adaptor protein-1 (Iba-1) and glucose transporter-5 are increased in alcoholics relative to healthy controls (He et al. 2008). In the VTA and thalamus, however, glucose transporter-5 expression is elevated, but Iba-1 expression is unaffected. Finally, in the amygdala, no microglial marker differences were observed in alcoholics relative to healthy volunteers (He et al. 2008). In addition to these activation markers, alcohol affects the expression of microglial cell surface receptors. Acute alcohol administration induces Toll-like receptor (TLR)4/type I interleukin (IL)-1 receptor signaling mediators: nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen activated protein kinase signal transduction cascades (Blanco et al. 2005). In contrast, alcohol impairs microglial activation in TLR4 null mice (Fernandez-Lizarbe et al. 2009). Although the literature on TNF-α expression is varied (Nelson et al. 1990; Nair et al. 1994; Avogaro et al. 2003), the most recent study indicates that alcohol increases brain TNF-α expression (Qin et al. 2008), which may be neuroprotective; indeed, low concentrations of TNF-α (20 ng/ml) in slice cultures synergize neurotoxicity by impairing EAAT2/GLT-1-mediated glutamate reuptake (Zou et al. 2005). Butylated hydroxytoluene, an antioxidant, reverses this inhibition and blocks the nuclear translocation of the NF-κB subunit p65 (Zou et al. 2005). Finally, in abstinent alcoholics, etanercept (a soluble TNF-α receptor antibody with demonstrated efficacy in the treatment of autoimmune disorders) reduces rapid eye movement sleep, a prognostic indicator for future alcohol relapse (Irwin et al. 2009).
Taken together, the evidence suggests that alcohol has complex temporal and brain area-specific effects on neuroinflammatory cascades that culminate in the differential expression of pro- and anti-inflammatory chemo/cytokines and downstream effectors. As a result, neuroinflammatory modulation (including microglial-mediated glutamate receptor downregulation) may be a rational future target for therapeutic intervention in alcoholism.
Illicit drugs
Unlike alcohol, scant evidence links microglia to drug use disorders (DUDs) (see Table II). Microglia express opioid receptors that inhibit chemotaxis (Chao et al. 1997) and induce apoptosis when stimulated (Hu et al. 2002). On a behavioural level, increased rodent maternal handling inhibits reinstatement of morphine-induced CPP in adulthood by increasing microglial expression of the anti-inflammatory IL-10 in the rat NAcc (Schwarz et al. 2011). Furthermore, microglial p38 (a mitogen activated protein kinase/extracellular signal-related kinase pathway intermediate) in the NAcc also mediates the acquisition and maintenance of morphine-induced CPP (Zhang et al. 2012). Microglial TLRs, e.g., TLR2 (Zhang et al. 2011), TLR4 (Watkins et al. 2009), and TLR9 (He et al. 2011), also modulate the neurochemical and behavioural effects of opioids in rodents.
Other substances of abuse have only been tangentially linked to microglial dysfunction. Striatal microglia are activated by psychostimulants, and a temporal correlation exists between microglial activation and methamphetamine-induced neurotoxicity/tolerance (Thomas et al. 2004a,b). Microglia also mediate some of methamphetamine’s behavioral effects; for instance, minocycline attenuates striatal methamphetamine-induced neurotoxicity and reduces hyperlocomotion (Zhang et al. 2006).
Depression
In rodents, repeated restraint stress activates medical PFC microglia, an effect reversible by minocycline (Hinwood et al. 2012) (see Table II). TNF receptor (TNFR1−/− and TNFR2−/−) knockout mice are more resilient when stressed (Simen et al 2006). TNFR1−/− mice also have reduced fear conditioning, and TNFR2 null mice increase their sucrose ingestion after water deprivation stress. However, no difference has been observed between wild-type and knockout littermates on anxiety-related tests such as the open field and elevated plus maze tests (Simen et al. 2006).
As in SUDs, proinflammatory cytokines are implicated in the pathophysiology of depression. Like bacterial lipopolysaccharide, exogenous TNF-α and IL-1 administration to rodents (both centrally and peripherally) produce“sickness behaviour“, a con stellation of physical and neuropsychiatric symptoms resembling depression (Dantzer 2001a; 2001b). In non-human primates, a ligand that recognizes activated microglia found lipopolysaccharide-induced microglial activation within hours, an effect most likely mediated through cytokine secretion (Hannestad et al. 2012). Likewise, proinflammatory cytokine levels are elevated in isolated and co-occurring depressive disorders (Penninx et al. 2003; Kahl et al. 2006; O’Brien et al. 2007) and abate with antidepressant therapy (Lanquillon et al. 2000; Narita et al. 2006a; O’Brien et al. 2007).
In addition, SSRIs and the serotonin norepinephrine reuptake inhibitor venlafaxine are anti-inflammatory as demonstrated by their ability to decrease proinflammatory cytokine expression in vitro (Liu et al. 2011; Tynan et al. 2012). The SSRIs paroxetine and sertraline inhibit nitric oxide and TNF-α expression in interferon-gamma-activated microglia in a calcium-dependent manner. In contrast to other antidepressants, the monoamine oxidase inhibitor phenelzine increases the proinflammatory cytokines TNF-α and IL-6 in an NF-κB-dependent manner, indicating differential effects based on anti-depressant class (Chung et al. 2012). Interestingly, kinin B1 receptor antagonists, important modulators of microglial TNF-α, reduce depressive-like behaviours in mice (Viana et al. 2010).
Experimental TNF-α findings are being translated into humans. In a novel immunomodulatory treatment study, 618 subjects with moderate-to-severe psoriasis were randomized to the anti-TNF-α antibody etanercept or placebo. Etanercept decreased standard clinical depression scores by ~50% – independent of dermatological and/or musculoskeletal improvement (Tyring et al. 2006). Although a recent placebo-controlled trial of infliximab (another TNF-α neutralizing antibody) revealed a lack of global efficacy in treatment-resistant MDD, an antidepressant response was observed in individuals with a baseline C-reactive protein >5 mg/L (Raison et al. 2012).
Taken together, the evidence suggests that microglia have important and overlapping aetiopathological roles in SUDs and depression.
Oligodendrocytes
Alcohol
Numerous studies have demonstrated white matter dysfunction in alcoholics (Shear et al. 1994; Pfefferbaum et al. 2000; Harper et al. 2003), as well as in rodent models of alcohol use disorders (Pons-Vazquez et al. 2011) (see Table III). At the molecular level, microarray studies revealed altered oligodendrocyte/myelin gene expression in heavy drinkers (Lewohl et al. 2000; Mayfield et al. 2002; Liu et al. 2004). Specifically, alcohol decreases the expression of peroxisome proliferator activated receptor-beta, a transcription factor important for lipid metabolism, in oligodendrocyte-like B12 cells (Leisewitz et al. 2003). Molecular dysfunction is also seen in animal models; in rodents, chronic exposure to alcohol decreases oligodendrocyte myelin glycoprotein mRNA in the hippocampus (Okamoto et al. 2006).
Table III.
SUDs | Depression |
---|---|
Enhanced programmed cell death/apoptosis (AUDs) | Decreased cell density |
Decreased expression of myelin-related genes, e.g., MBP, PLP-1 |
Decreased expression of myelin-related genes, e.g., MBP, MOG, PLP-1 |
Decreased immediate-early gene expression, e.g c-Fos |
Decreased RNA binding protein expression |
Increased activation after ECT/ECS |
SUDs, substance use disorders; MBP, myelin basic protein, PLP-1, phospholipid protein-1; MOG, myelin-oligodendrocyte glycoprotein; RNA, ribonucleic acid; ECT, electroconvulsive therapy; ECS, electroconvulsive seizure.
Italicised text denotes potential convergent mechanisms in SUDs and depressive disorders.
The specific mechanisms of oligodendrocyte dysfunction have been explored with mixed results. In cortical cultures, both Th1 (pro-inflammatory) and Th2 (anti-inflammatory) cytokines decrease alcohol-induced oligodendrocyte cell death (Benjamins et al. 2011). Furthermore, alcohol downregulates oligodendroglial c-Fos (an immediate early gene) and upregulates myelin basic protein (MBP) in a protein kinase C-dependent manner (Bichenkov et al. 2009). However, myelin gene expression (including MBP) is less disrupted (with decreased concomitant neurodegeneration) in TLR4 knockout mice after chronic ethanol exposure (Alfonso-Loeches et al. 2012).
Illicit drugs
Extensive white matter tract and oligodendrocyte abnormalities are seen in cocaine misuse (Chang et al. 1997, 1999; Bartzokis et al. 1999) (see Table III). For example, MBP, proteolipid protein-1 (PLP-1), and myelin-associated oligodendrocyte basic protein (MOBP) are downregulated in chronic cocaine abuse (Albertson et al. 2004). In another microarray study, increased dlPFC PLP-1 levels were observed in four cocaine abusers, and decreased levels were observed in three other cocaine abusers (including two alcohol co-abusers); interestingly, increased dlPFC PLP-1 correlated with recent crack cocaine use (Lehrmann et al. 2003).
Oligodendrocyte dysfunction has also been observed in opioid dependence. In rodents, low-level perinatal buprenorphine exposure increases fetal MBP expression and oligodendrocyte maturation; however, excessive perinatal exposure delays oligodendroglial development and decreases MBP expression (Sanchez et al. 2008). These biphasic effects are mediated by µ-opioid and nociception/orphanin FQ receptors, which are also implicated in the pathogenesis of chronic pain disorders (Eschenroeder et al. 2012).
Depression
Oligodendrocytes have been studied in depressive disorders (Edgar et al. 2012) (see Table III). There is white matter attenuation in MDD, especially in geriatric samples (Taylor et al. 2003, 2004), and this has been confirmed in several postmortem MDD studies (Regenold et al. 2007; Hayashi et al. 2011). Oligodendrocyte density is also decreased in the postmortem MDD brain (−19%) in PFC layer VI and surrounding white matter (Brodmann’s area 9) compared to non-psychiatric controls (Uranova et al. 2004). Decreased oligodendrocyte density has also been observed in sublayers IIIa, IΠb, and IΠc of Brodmann’s area 9 (Vostrikov et al. 2007) and in the amygdala of individuals with MDD but not BD (Hamidi et al. 2004). Other white matter corticolimbic circuitry abnormalities have been observed in BD (Mahon et al. 2010).
As with SUDs, numerous myelin-related genes are downregulated in the depressed brain (Aston et al. 2005; Honer et al. 1999). Quaking homolog, KH domain RNA-binding, an oligodendrocyte-specific RNA-binding protein critical for oligodendrocyte progenitor differentiation, is also reduced in the neocortex, hippocampus, and amygdala of depressed suicide victims (Klempan et al. 2009). Oligodendrocytes are transiently activated by electroconvulsive therapy (Ongur et al. 2004) and rodent electroconvulsive seizures (Jansson et al. 2009). As further molecular evidence of electroconvulsive seizure response, neuronglial antigen 2-expressing oligodendrocyte progenitor cells proliferated and selectively differentiated into mature oligodendrocytes in the rat amygdala (Wennstrom et al. 2004). Thus, overlapping white matter dysfunction and oligodendrocyte cytopathology occurs in both SUDs and depression.
Glial cell-mediated glutamatergic dysfunction in co-occurring SUDs and depression
Most of the studies discussed have focused on isolated SUDs or depressive disorders. To date, only one study has directly compared glial cell pathology in subjects with comorbid depressive symptoms of escalating severity, alcohol dependence alone, and an MDD cohort from an earlier study by the same group (Miguel-Hidalgo et al. 2002). In this study, subjects with a history of alcohol dependence and depressive symptoms (n = 8) had decreased dlPFC glial cell density compared to subjects with alcohol dependence and no depressive symptoms (n = 9) and the historical MDD group (n = 12) (Miguel-Hidalgo et al. 2002).This suggests that subjects with comorbid depression and alcohol dependence may have even more severe glial cell dysfunction than in either illness alone.
Although depression and SUD comorbidity has not been extensively studied (Miguel-Hidalgo et al. 2003), glial cell dysfunction may be particularly important. The following hypotheses may be postulated:
Multiple episodes of substance intoxication/ withdrawal kindle substance-induced depressive episodes, which have cumulative deleterious effects on glia;
Depressed subjects attempt to“self-medicate” their neuropsychiatric symptoms with alcohol and illicit substances, which have cumulative deleterious effects on glia; and
A complex interplay exists between depressive and substance use pathologies that has cumulative and potentially synergistic deleterious consequences on glia.
The first and second hypotheses may pertain to some dually diagnosed subjects (Crum et al. 2013), but these disorders rarely develop in isolation and almost never arise linearly. Therefore, the third cumulative/synergistic hypothesis appears most consistent with the clinical phenomenology of comorbidity.
Ketamine as a glutamatergic probe in comorbidity
In addition to shared glial cell dysfunction, several studies conducted with the NMDA receptor antagonist ketamine support shared glutamatergic dysregulation in comorbid depression and SUDs (Miguel-Hidalgo et al. 2010). Ketamine recreates ethanol-like effects in recently detoxified alcoholics in a dose-dependent manner, which more closely resembles alcohol’s sedative rather than euphoric effects (Krystal et al. 1998). Recently detoxified alcoholics also display fewer dissociative-like symptoms, acute dysphoria, or cognitive/executive dysfunction during ketamine administration relative to non-dependent controls, suggesting that chronic alcohol-induced NMDA receptor antagonism attenuates ketamine’s adverse effects (Krystal et al. 2003). Alcohol-dependent patients may also not experience the negative cognitive and psychological signals to stop drinking beyond the point of mild-to-moderate intoxication (Krystal et al. 2003). Even non-dependent individuals with a family history of alcohol dependence display a more blunted response to ketamine than subjects without this genetic loading (Petrakis et al. 2004).
Although acute and transient ketamine-induced dysphoria is an identified adverse effect, subanesthetic ketamine also has rapid and robust antidepressant effects in patients with treatment-resistant MDD (Berman et al. 2000; Zarate et al. 2006) and bipolar depression (Diazgranados et al. 2010a; Zarate et al. 2012).This enhanced glutamatergic responsivity has also been observed in MDD (Phelps et al. 2009) and bipolar depressed (Luckenbaugh et al. 2012) patients with a first degree relative with alcoholism as these individuals have an augmented anti-depressant response to ketamine.
Future directions/conclusions
Although astrocytes are now known to be critical in synaptic glutamate reuptake, microglia and oligodendrocytes also function in glutamatergic homeostasis [see recent reviews by Wong et al. (2011) and Bakiri et al. (2009) on microglia and oligodendrocytes, respectively]. Microglial upregulation of the NMDA receptor complex has been observed in MDD but not BD (Steiner et al. 2011). Nevertheless, additional research is critically needed on non-astrocyte-based amino acid neurotransmitter dysfunction in both SUDs and depression to better comprehend convergent pathways and offer novel treatment approaches.
There are several exciting avenues for future comorbidity research. First, preclinical models of co-occurring disorders, e.g., rodents exposed to both CUS and alcohol/illicit drugs, may expand our neuropathological understanding of this comorbidity. Ketamine’s improved antidepressant response in those with a family history of alcoholism suggests that subjects with comorbid depression and alcohol dependence may have enhanced antidepressant effects relative to non-alcohol dependent depressed patients. If efficacious, alternative non-parenteral NMDA receptor antagonists with lower addictive liability are better long-term strategies in this population (Ibrahim et al. 2012; Zarate et al. 2013). Despite the great wealth of preclinical data on glial-mediated glutamatergic dysregulation in DUDs (Kalivas et al. 2005, 2011), there are presently no pathophysiological or treatment studies in co-occurring MDD/bipolar depression and DUDs.
Depressed subjects with co-morbid SUDs are at increased risk for suicide (Niciu et al. 2009). Interestingly, ketamine decreases suicidality in high-risk populations (Price et al. 2009; DiazGranados et al. 2010b), including in the emergency setting (Larkin et al. 2011), but it remains to be seen if ketamine or other glutamatergic modulators have anti-suicidal properties in this high risk population.
In summary, the evidence reviewed in this paper highlights the critical role of glia in the aetiopathogenesis of isolated depressive disorders and SUDs. We synthesize the evidence and hypothesize that glial cell-mediated glutamatergic dysfunction may be critical in comorbidity.
Acknowledgements
The authors would like to thank the 7SE Inpatient Mood and Anxiety Disorders Research Unit for their clinical support during the writing of this manuscript.
This work was supported in part by the Intramural Research Program of the National Institute of Mental Health, National Institutes of Health (IRP-NIMH-NIH); by the Department of Veterans Affairs, via its funding of the VA National Center for PTSD (GS); by the National Alliance for Research on Schizophrenia and Depression (NARSAD) (GS); by NIMH grants R01 MH071676-05 and R01 MH081211-3 (GS); by the State of Connecticut Department of Mental Health and Addiction Services (through its support of clinical research services at the Connecticut Mental Health Center and Clinical Neuroscience Research Unit) (GS); by salary support from NIMHT32 MH19961 and R25 MH071584 (MJN); and by a NARSAD Independent Investigator Award and Brain and Behavior Foundation Bipolar Research Award (CAZ).
Dr Sanacora has received consulting fees from Astra-Zeneca, Avanier Pharmaceuticals, Bristol-Myers Squibb, Evotec, Eli Lilly & Co., Hoffman La-Roche, Naurex, Novartis and Novum Pharmaceuticals over the last 24 months. He has also received additional grant support from AstraZeneca, Bristol-Myers Squibb, Eli Lilly, Hoffman La-Roche, Merck & Co., Naurex, and Severier over the last 24 months. In addition, he is a co-inventor on a filed patent application by Yale University (PCTWO06108055A1) concerning the use of glutamate-modulating drugs as antidepressants and anxiolytics. Dr Zarate is listed as a co-inventor on a patent application for the use of ketamine and its metabolites in major depression. Dr Zarate has assigned his rights in the patent to the US Government but will share a percentage of any royalties that may be received.
Footnotes
Statement of Interest
Dr Niciu and Ms Henter have no potential conflicts of interest to disclose, financial or otherwise.
References
- Agrawal RG, Hewetson A, George CM, Syapin PJ, Bergeson SE. Minocycline reduces ethanol drinking. Brain Behav Immun. 2011;25(Suppl 1):S165–S169. doi: 10.1016/j.bbi.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Albertson DN, Pruetz B, Schmidt CJ, Kuhn DM, Kapatos G, Bannon MJ. Gene expression profile of the nucleus accumbens of human cocaine abusers: evidence for dysregulation of myelin. J Neurochem. 2004;88:1211–1219. doi: 10.1046/j.1471-4159.2003.02247.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alfonso-Loeches S, Pascual M, Gomez-Pinedo U, Pascual-Lucas M, Renau-Piqueras J, Guerri C. Toll-like receptor 4 participates in the myelin disruptions associated with chronic alcohol abuse. Glia. 2012;60:948–964. doi: 10.1002/glia.22327. [DOI] [PubMed] [Google Scholar]
- Altshuler LL, Abulseoud OA, Foland-Ross L, Bartzokis G, Chang S, Mintz J, et al. Amygdala astrocyte reduction in subjects with major depressive disorder but not bipolar disorder. Bipolar Disord. 2010;12:541–549. doi: 10.1111/j.1399-5618.2010.00838.x. [DOI] [PubMed] [Google Scholar]
- Amen SL, Piacentine LB, Ahmad ME, Li SJ, Mantsch JR, Risinger RC, et al. Repeated N-acetyl cysteine reduces cocaine seeking in rodents and craving in cocaine-dependent humans. Neuropsychopharmacology. 2011;36:871–878. doi: 10.1038/npp.2010.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Appel E, Kolman O, Kazimirsky G, Blumberg PM, Brodie C. Regulation of GDNF expression in cultured astrocytes by inflammatory stimuli. Neuroreport. 1997;8:3309–3312. doi: 10.1097/00001756-199710200-00023. [DOI] [PubMed] [Google Scholar]
- Araya-Callis C, Hiemke C, Abumaria N, Flugge G. Chronic psychosocial stress and citalopram modulate the expression of the glial proteins GFAP and NDRG2 in the hippocampus. Psychopharmacology. 2012;224:209–222. doi: 10.1007/s00213-012-2741-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aston C, Jiang L, Sokolov BP. Transcriptional profiling reveals evidence for signaling and oligodendroglial abnormalities in the temporal cortex from patients with major depressive disorder. Mol Psychiatry. 2005;10:309–322. doi: 10.1038/sj.mp.4001565. [DOI] [PubMed] [Google Scholar]
- Avogaro A, Sambataro M, Marangoni A, Pianta A, Vettor R, Pagano C, et al. Moderate alcohol consumption, glucose metabolism and lipolysis: the effect on adiponectin and tumor necrosis factor alpha. J Endocrinol Invest. 2003;26:1213–1218. doi: 10.1007/BF03349160. [DOI] [PubMed] [Google Scholar]
- Baker DA, McFarland K, Lake RW, Shen H, Tang XC, Toda S, et al. Neuroadaptations in cystine-glutamate exchange underlie cocaine relapse. Nat Neurosci. 2003;6:743–749. doi: 10.1038/nn1069. [DOI] [PubMed] [Google Scholar]
- Bakiri Y, Burzomato V, Frugier G, Hamilton NB, Karadottir R, Attwell D. Glutamatergic signaling in the brain’s white matter. Neuroscience. 2009;158:266–274. doi: 10.1016/j.neuroscience.2008.01.015. [DOI] [PubMed] [Google Scholar]
- Banasr M, Duman RS. Glial loss in the prefrontal cortex is sufficient to induce depressive-like behaviors. Biol Psychiatry. 2008;64:863–870. doi: 10.1016/j.biopsych.2008.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banasr M, Valentine GW, Li XY, Gourley SL, Taylor JR, Duman RS. Chronic unpredictable stress decreases cell proliferation in the cerebral cortex of the adult rat. Biol Psychiatry. 2007;62:496–504. doi: 10.1016/j.biopsych.2007.02.006. [DOI] [PubMed] [Google Scholar]
- Banasr M, Chowdhury GM, Terwilliger R, Newton SS, Duman RS, Behar KL, et al. Glial pathology in an animal model of depression: reversal of stress-induced cellular, metabolic and behavioral deficits by the glutamate-modulating drug riluzole. Mol Psychiatry. 2010;15:501–511. doi: 10.1038/mp.2008.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barley K, Dracheva S, Byne W. Subcortical oligodendrocyte- and astrocyte-associated gene expression in subjects with schizophrenia, major depression and bipolar disorder. Schizophr Res. 2009;112:54–64. doi: 10.1016/j.schres.2009.04.019. [DOI] [PubMed] [Google Scholar]
- Bartzokis G, Beckson M, Hance DB, Lu PH, Foster JA, Mintz J, et al. Magnetic resonance imaging evidence of“silent” cerebrovascular toxicity in cocaine dependence. Biol Psychiatry. 1999;45:1203–1211. doi: 10.1016/s0006-3223(98)00228-5. [DOI] [PubMed] [Google Scholar]
- Bechtholt-Gompf AJ, Walther HV, Adams MA, Carlezon WA, Jr., Ongur D, Cohen BM. Blockade of astrocytic glutamate uptake in rats induces signs of anhedonia and impaired spatial memory. Neuropsychopharmacology. 2010;35:2049–2059. doi: 10.1038/npp.2010.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beitner-Johnson D, Guitart X, Nestler EJ. Glial fibrillary acidic protein and the mesolimbic dopamine system: regulation by chronic morphine and Lewis-Fischer strain differences in the rat ventral tegmental area. J Neurochem. 1993;61:1766–1773. doi: 10.1111/j.1471-4159.1993.tb09814.x. [DOI] [PubMed] [Google Scholar]
- Benjamins JA, Nedelkoska L, Lisak RP, Hannigan JH, Sokol RJ. Cytokines reduce toxic effects of ethanol on oligodendroglia. Neurochem Res. 2011;36:1677–1686. doi: 10.1007/s11064-011-0401-x. [DOI] [PubMed] [Google Scholar]
- Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–354. doi: 10.1016/s0006-3223(99)00230-9. [DOI] [PubMed] [Google Scholar]
- Bernard R, Kerman IA, Thompson RC, Jones EG, Bunney WE, Barchas JD, et al. Altered expression of glutamate signaling, growth factor, and glia genes in the locus coeruleus of patients with major depression. Mol Psychiatry. 2011;16:634–646. doi: 10.1038/mp.2010.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bichenkov E, Ellingson JS. Ethanol alters the expressions of c-Fos and myelin basic protein in differentiating oligodendrocytes. Alcohol. 2009;43:627–634. doi: 10.1016/j.alcohol.2009.09.026. [DOI] [PubMed] [Google Scholar]
- Blanco AM, Valles SL, Pascual M, Guerri C. Involvement of TLR4/type I IL-1 receptor signaling in the induction of inflammatory mediators and cell death induced by ethanol in cultured astrocytes. J Immunol. 2005;175:6893–6899. doi: 10.4049/jimmunol.175.10.6893. [DOI] [PubMed] [Google Scholar]
- Blednov YA, Bergeson SE, Walker D, Ferreira VM, Kuziel WA, Harris RA. Perturbation of chemokine networks by gene deletion alters the reinforcing actions of ethanol. Behav Brain Res. 2005;165:110–125. doi: 10.1016/j.bbr.2005.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boger HA, Middaugh LD, Patrick KS, Ramamoorthy S, Denehy ED, Zhu H, et al. Long-term consequences of methamphetamine exposure in young adults are exacerbated in glial cell line-derived neurotrophic factor heterozygous mice. J Neurosci. 2007;27:8816–8825. doi: 10.1523/JNEUROSCI.1067-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowley MP, Drevets WC, Ongur D, Price JL. Low glial numbers in the amygdala in major depressive disorder. Biol Psychiatry. 2002;52:404–412. doi: 10.1016/s0006-3223(02)01404-x. [DOI] [PubMed] [Google Scholar]
- Boyadjieva NI, Sarkar DK. Role of microglia in ethanol’s apoptotic action on hypothalamic neuronal cells in primary cultures. Alcohol Clin Exp Res. 2010;34:1835–1842. doi: 10.1111/j.1530-0277.2010.01271.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bray JG, Reyes KC, Roberts AJ, Ransohoff RM, Gruol DL. Synaptic plasticity in the hippocampus shows resistance to acute ethanol exposure in transgenic mice with astrocyte-targeted enhanced CCL2 expression. Neuropharmacology. 2013;67:115–125. doi: 10.1016/j.neuropharm.2012.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cass WA. GDNF selectively protects dopamine neurons over serotonin neurons against the neurotoxic effects of methamphetamine. J Neurosci. 1996;16:8132–8139. doi: 10.1523/JNEUROSCI.16-24-08132.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang L, Mehringer CM, Ernst T, Melchor R, Myers H, Forney D, et al. Neurochemical alterations in asymptomatic abstinent cocaine users: a proton magnetic resonance spectroscopy study. Biol Psychiatry. 1997;42:1105–1114. doi: 10.1016/s0006-3223(97)00135-2. [DOI] [PubMed] [Google Scholar]
- Chang L, Ernst T, Strickland T, Mehringer CM. Gender effects on persistent cerebral metabolite changes in the frontal lobes of abstinent cocaine users. Am J Psychiatry. 1999;156:716–722. doi: 10.1176/ajp.156.5.716. [DOI] [PubMed] [Google Scholar]
- Chao CC, Hu S, Shark KB, Sheng WS, Gekker G, Peterson PK. Activation of mu opioid receptors inhibits microglial cell chemotaxis. J Pharmacol Exp Ther. 1997;281:998–1004. [PubMed] [Google Scholar]
- Choudary PV, Molnar M, Evans SJ, Tomita H, Li JZ, Vawter MP, et al. Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression. Proc Natl Acad Sci USA. 2005;102:15653–15658. doi: 10.1073/pnas.0507901102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung HS, Kim H, Bae H. Phenelzine (monoamine oxidase inhibitor) increases production of nitric oxide and proinflammatory cytokines via the NF-kappaB pathway in lipopolysaccharide-activated microglia cells. Neurochemical Research. 2012;37:2117–1124. doi: 10.1007/s11064-012-0833-y. [DOI] [PubMed] [Google Scholar]
- Comasco E, Nordquist N, Gokturk C, Aslund C, Hallman J, Oreland L, et al. The clock gene PER2 and sleep problems: association with alcohol consumption among Swedish adolescents. Ups J Med Sci. 2010;115:41–48. doi: 10.3109/03009731003597127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cotter D, Mackay D, Landau S, Kerwin R, Everall I. Reduced glial cell density and neuronal size in the anterior cingulate cortex in major depressive disorder. Arch Gen Psychiatry. 2001;58:545–553. doi: 10.1001/archpsyc.58.6.545. [DOI] [PubMed] [Google Scholar]
- Cram RM, Mojtabai R, Lazareck S, Bolton JM, Robinson J, Sareen J, et al. A prospective assessment of reports of drinking to self-medicate mood symptoms with the incidence and persistence of alcohol dependence. JAMA Psychiatry. 2013:1–9. doi: 10.1001/jamapsychiatry.2013.1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Czeh B, Muller-Keuker JI, Rygula R, Abumaria N, Hiemke C, Domenici E, et al. Chronic social stress inhibits cell proliferation in the adult medial prefrontal cortex: hemispheric asymmetry and reversal by fluoxetine treatment. Neuropsychopharmacology. 2007;32:1490–1503. doi: 10.1038/sj.npp.1301275. [DOI] [PubMed] [Google Scholar]
- Dantzer R. Cytokine-induced sickness behavior: mechanisms and implications. Ann NY Acad Sci. 2001a;933:222–234. doi: 10.1111/j.1749-6632.2001.tb05827.x. [DOI] [PubMed] [Google Scholar]
- Dantzer R. Cytokine-induced sickness behavior: where do we stand? Brain Behav Immun. 2001b;15:7–24. doi: 10.1006/brbi.2000.0613. [DOI] [PubMed] [Google Scholar]
- Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, et al. A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry. 2010a;67:793–802. doi: 10.1001/archgenpsychiatry.2010.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiazGranados N, Ibrahim LA, Brutsche NE, Ameli R, Henter ID, Luckenbaugh DA, et al. Rapid resolution of suicidal ideation after a single infusion of an N-methyl-D-aspartate antagonist in patients with treatment-resistant major depressive disorder. J Clin Psychiatry. 2010b;71:1605–1611. doi: 10.4088/JCP.09m05327blu. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du J, Suzuki K, Wei Y, Wang Y, Blumenthal R, Chen Z, et al. The anticonvulsants lamotrigine, riluzole, and valproate differentially regulate AMPA receptor membrane localization: relationship to clinical effects in mood disorders. Neuropsychopharmacology. 2007;32:793–802. doi: 10.1038/sj.npp.1301178. [DOI] [PubMed] [Google Scholar]
- Edgar N, Sibille E. A putative functional role for oligodendrocytes in mood regulation. Transl Psychiatry. 2012;2:el09. doi: 10.1038/tp.2012.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edwards AC, Aliev F, Bierut LJ, Bucholz KK, Edenberg H, Hesselbrock V, et al. Genome-wide association study of comorbid depressive syndrome and alcohol dependence. Psychiatr Genet. 2012;22:31–31. doi: 10.1097/YPG.0b013e32834acd07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elsayed M, Banasr M, Duric V, Fournier NM, Licznerski P, Duman RS. Antidepressant effects of fibroblast growth factor-2 in behavioral and cellular models of depression. Biol Psychiatry. 2012;72:258–265. doi: 10.1016/j.biopsych.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ernst C, Deleva V, Deng X, Sequeira A, Pomarenski A, Klempan T, et al. Alternative splicing, methylation state, and expression profile of tropomyosin-related kinase B in the frontal cortex of suicide completers. Arch Gen Psychiatry. 2009;66:22–32. doi: 10.1001/archpsyc.66.1.22. [DOI] [PubMed] [Google Scholar]
- Ernst C, Nagy C, Kim S, Yang JP, Deng X, Hellstrom IC, et al. Dysfunction of astrocyte connexins 30 and 43 in dorsal lateral prefrontal cortex of suicide completers. Biol Psychiatry. 2011;70:312–319. doi: 10.1016/j.biopsych.2011.03.038. [DOI] [PubMed] [Google Scholar]
- Eroglu C. The role of astrocyte-secreted matricellular proteins in central nervous system development and function. J Cell Commun Signal. 2009;3:167–176. doi: 10.1007/s12079-009-0078-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eschenroeder AC, Vestal-Laborde AA, Sanchez ES, Robinson SE, Sato-Bigbee C. Oligodendrocyte responses to buprenorphine uncover novel and opposing roles of mu-opioid- and nociceptin/orphanin FQ receptors in cell development: implications for drug addiction treatment during pregnancy. Glia. 2012;60:125–136. doi: 10.1002/glia.21253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fatemi SH, Laurence JA, Araghi-Niknam M, Stary JM, Schulz SC, Lee S, et al. Glial fibrillary acidic protein is reduced in cerebellum of subjects with major depression, but not schizophrenia. Schizophr Res. 2004;69:317–323. doi: 10.1016/j.schres.2003.08.014. [DOI] [PubMed] [Google Scholar]
- Fernandez-Lizarbe S, Pascual M, Guerri C. Critical role of TLR4 response in the activation of microglia induced by ethanol. J Immunol. 2009;183:4733–4744. doi: 10.4049/jimmunol.0803590. [DOI] [PubMed] [Google Scholar]
- Foley PF, Loh EW, Innes DJ, Williams SM, Tannenberg AE, Harper CG, et al. Association studies of neurotransmitter gene polymorphisms in alcoholic Caucasians. Ann NY Acad Sci. 2004;1025:39–46. doi: 10.1196/annals.1316.005. [DOI] [PubMed] [Google Scholar]
- Fujio M, Nakagawa T, Sekiya Y, Ozawa T, Suzuki Y, Minami M, et al. Gene transfer of GLT-1, a glutamate transporter, into the nucleus accumbens shell attenuates methampheta-mine- and morphine-induced conditioned place preference in rats. Eur J Neurosci. 2005;22:2744–2754. doi: 10.1111/j.1460-9568.2005.04467.x. [DOI] [PubMed] [Google Scholar]
- Gomez-Galan M, De Bundel D, Van Eeckhaut A, Smolders I, Lindskog M. Dysfunctional astrocytic regulation of glutamate transmission in a rat model of depression. Mol Psychiatry. 2013;18:582–594. doi: 10.1038/mp.2012.10. [DOI] [PubMed] [Google Scholar]
- Gourley SL, Espitia JW, Sanacora G, Taylor JR. Antidepressant-like properties of oral riluzole and utility of incentive disengagement models of depression in mice. Psychopharmacology (Berlin) 2012;219:805–814. doi: 10.1007/s00213-011-2403-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griffiths M, Beaumont N, Yao SY, Sundaram M, Boumah CE, Davies A, et al. Cloning of a human nucleoside transporter implicated in the cellular uptake of adenosine and chemotherapeutic drags. Nat Med. 1997;3:89–93. doi: 10.1038/nm0197-89. [DOI] [PubMed] [Google Scholar]
- Hamidi M, Drevets WC, Price JL. Glial reduction in amygdala in major depressive disorder is due to oligodendrocytes. Biol Psychiatry. 2004;55:563–569. doi: 10.1016/j.biopsych.2003.11.006. [DOI] [PubMed] [Google Scholar]
- Hannestad J, Gallezot JD, Schafbauer T, Lim K, Kloczynski T, Morris ED, et al. Endotoxin-induced systemic inflammation activates microglia: [(1)(1)C]PBR28 positron emission tomography in nonhuman primates. Neuroimage. 2012;63:232–239. doi: 10.1016/j.neuroimage.2012.06.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat Rev Neurosci. 2010;11:682–696. doi: 10.1038/nrn2911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harper C, Dixon G, Sheedy D, Garrick T. Neuropathological alterations in alcoholic brains. Studies arising from the New South Wales Tissue Resource Centre. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:951–961. doi: 10.1016/S0278-5846(03)00155-6. [DOI] [PubMed] [Google Scholar]
- Hayashi Y, Nihonmatsu-Kikuchi N, Yu X, Ishimoto K, Hisanaga SI, Tatebayashi Y. A novel, rapid, quantitative cell-counting method reveals oligodendroglial reduction in the frontopolar cortex in major depressive disorder. Mol Psychiatry. 2011;16:1155–1158. doi: 10.1038/mp.2011.84. [DOI] [PubMed] [Google Scholar]
- He J, Crews FT. Increased MCP-1 and microglia in various regions of the human alcoholic brain. Exp Neurol. 2008;210:349–358. doi: 10.1016/j.expneurol.2007.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He L, Li H, Chen L, Miao J, Jiang Y, Zhang Y, et al. Toll-like receptor 9 is required for opioid-induced microglia apoptosis. PLoS One. 2011;6:el8190. doi: 10.1371/journal.pone.0018190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heifer JL, Calizo LH, Dong WK, Goodlett CR, Greenough WT, Klintsova AY. Binge-like postnatal alcohol exposure triggers cortical gliogenesis in adolescent rats. J Comp Neurol. 2009;514:259–271. doi: 10.1002/cne.22018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hercher C, Parent M, Flores C, Canetti L, Turecki G, Mechawar N. Alcohol dependence-related increase of glial cell density in the anterior cingulate cortex of suicide completers. J Psychiatry Neurosci. 2009;34:281–288. [PMC free article] [PubMed] [Google Scholar]
- Hinwood M, Morandini J, Day TA, Walker FR. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb Cortex. 2012;22:1442–1454. doi: 10.1093/cercor/bhr229. [DOI] [PubMed] [Google Scholar]
- Hoffman PL, Miles M, Edenberg HJ, Sommer W, Tabakoff B, Wehner JM, et al. Gene expression in brain: a window on ethanol dependence, neuroadaptation, and preference. Alcohol Clin Exp Res. 2003;27:155–168. doi: 10.1097/01.ALC.0000060101.89334.11. [DOI] [PubMed] [Google Scholar]
- Honer WG, Falkai P, Chen C, Arango V, Mann JJ, Dwork AJ. Synaptic and plasticity-associated proteins in anterior frontal cortex in severe mental illness. Neuroscience. 1999;91:1247–1255. doi: 10.1016/s0306-4522(98)00679-4. [DOI] [PubMed] [Google Scholar]
- Hu S, Sheng WS, Lokensgard JR, Peterson PK. Morphine induces apoptosis of human microglia and neurons. Neuropharmacology. 2002;42:829–836. doi: 10.1016/s0028-3908(02)00030-8. [DOI] [PubMed] [Google Scholar]
- Ibrahim L, Diaz Granados N, Jolkovsky L, Brutsche N, Luckenbaugh DA, Herring WJ, et al. A Randomized, placebo-controlled, crossover pilot trial of the oral selective NR2B antagonist MK-0657 in patients with treatment-resistant major depressive disorder. J Clin Psychopharmacol. 2012;32:551–557. doi: 10.1097/JCP.0b013e31825d70d6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irwin MR, Olmstead R, Valladares EM, Breen EC, Ehlers CL. Tumor necrosis factor antagonism normalizes rapid eye movement sleep in alcohol dependence. Biol Psychiatry. 2009;66:191–195. doi: 10.1016/j.biopsych.2008.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwata M, Shirayama Y, Ishida H, Hazama GI, Nakagome K. Hippocampal astrocytes are necessary for antidepressant treatment of learned helplessness rats. Hippocampus. 2011;21:877–884. doi: 10.1002/hipo.20803. [DOI] [PubMed] [Google Scholar]
- Jansson L, Wennstrom M, Johanson A, Tingstrom A. Glial cell activation in response to electroconvulsive seizures. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:1119–1128. doi: 10.1016/j.pnpbp.2009.06.007. [DOI] [PubMed] [Google Scholar]
- John CS, Smith KL, Van’t Veer A, Gompf HS, Carlezon WA, Jr., Cohen BM, et al. Blockade of astrocytic glutamate uptake in the prefrontal cortex induces anhedonia. Neuropsychopharmacology. 2012;37:2467–2475. doi: 10.1038/npp.2012.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kahl KG, Bens S, Ziegler K, Rudolf S, Dibbelt L, Kordon A, et al. Cortisol, the cortisol-dehydroepiandrosterone ratio, and pro-inflammatory cytokines in patients with current major depressive disorder comorbid with borderline personality disorder. Biol Psychiatry. 2006;59:667–671. doi: 10.1016/j.biopsych.2005.08.001. [DOI] [PubMed] [Google Scholar]
- Kalivas PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci. 2009;10:561–572. doi: 10.1038/nrn2515. [DOI] [PubMed] [Google Scholar]
- Kalivas PW, Volkow ND. New medications for drug addiction hiding in glutamatergic neuroplasticity. Mol Psychiatry. 2011;16:974–986. doi: 10.1038/mp.2011.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalivas PW, Volkow N, Seamans J. Unmanageable motivation in addiction: a pathology in prefrontal-accumbens glutamate transmission. Neuron. 2005;45:647–650. doi: 10.1016/j.neuron.2005.02.005. [DOI] [PubMed] [Google Scholar]
- Kertes DA, Kalsi G, Prescott CA, Kuo PH, Patterson DG, Walsh D, et al. Neurotransmitter and neuromodulator genes associated with a history of depressive symptoms in individuals with alcohol dependence. Alcohol Clin Exp Res. 2011;35:496–505. doi: 10.1111/j.1530-0277.2010.01366.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khokhrina NT, Kazakova PB, Rakhmanova VI. [Morphometric analysis of the changes in the cerebral cortex of rats after long-term administration of alcohol] Zh Nevropatol Psikhiatr Im S S Korsakova. 1991;91:66–67. [PubMed] [Google Scholar]
- Khundakar AA, Morris CM, Oakley AE, Thomas AJ. Cellular pathology within the anterior cingulate cortex of patients with late-life depression: a morphometric study. Psychiatry Res. 2011;194:184–189. doi: 10.1016/j.pscychresns.2011.04.008. [DOI] [PubMed] [Google Scholar]
- Klempan TA, Ernst C, Deleva V, Labonte B, Turecki G. Characterization of QKI gene expression, genetics, and epigenetics in suicide victims with major depressive disorder. Biol Psychiatry. 2009;66:824–831. doi: 10.1016/j.biopsych.2009.05.010. [DOI] [PubMed] [Google Scholar]
- Knackstedt LA, Melendez RI, Kalivas PW. Ceftriaxone restores glutamate homeostasis and prevents relapse to cocaine seeking. Biol Psychiatry. 2010;67:81–84. doi: 10.1016/j.biopsych.2009.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koss WA, Sadowski RN, Sherrill LK, Gulley JM, Juraska JM. Effects of ethanol during adolescence on the number of neurons and glia in the medial prefrontal cortex and basolateral amygdala of adult male and female rats. Brain Res. 2012;1466:24–32. doi: 10.1016/j.brainres.2012.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krystal JH, Petrakis IL, Limoncelli D, Webb E, Gueorgueva R, D’Souza DC, et al. Altered NMDA glutamate receptor antagonist response in recovering ethanol-dependent patients. Neuropsychopharmacology. 2003;28:2020–2028. doi: 10.1038/sj.npp.1300252. [DOI] [PubMed] [Google Scholar]
- Krystal JH, Petrakis IL, Webb E, Cooney NL, Karper LP, Namanworth S, et al. Dose-related ethanol-like effects of the NMDA antagonist, ketamine, in recently detoxified alcoholics. Arch Gen Psychiatry. 1998;55:354–360. doi: 10.1001/archpsyc.55.4.354. [DOI] [PubMed] [Google Scholar]
- Kucukdereli H, Allen NJ, Lee AT, Feng A, Ozlu MI, Conatser LM, et al. Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc Natl Acad Sci USA. 2011;108:E440–E449. doi: 10.1073/pnas.1104977108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lanquillon S, Krieg JC, Bening-Abu-Shach U, Vedder H. Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology. 2000;22:370–379. doi: 10.1016/S0893-133X(99)00134-7. [DOI] [PubMed] [Google Scholar]
- Larkin GL, Beautrais AL. A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int J Neuropsychopharmacology. 2011:1–5. doi: 10.1017/S1461145711000629. [DOI] [PubMed] [Google Scholar]
- Lee MR, Ruby CL, Hinton DJ, Choi S, Adams CA, Young Kang N, et al. Striatal adenosine signaling regulates EAAT2 and astrocytic AQP4 expression and alcohol drinking in mice. Neuropsychopharmacology. 2013;38:437–445. doi: 10.1038/npp.2012.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee YJ, Jin JK, Jeong BH, Carp RI, Kim YS. Increased expression of glial cell line-derived neurotrophic factor (GDNF) in the brains of scrapie-infected mice. Neurosci Lett. 2006;410:178–182. doi: 10.1016/j.neulet.2006.09.090. [DOI] [PubMed] [Google Scholar]
- Lehrmann E, Oyler J, Vawter MP, Hyde TM, Kolachana B, Kleinman JE, et al. Transcriptional profiling in the human prefrontal cortex: evidence for two activational states associated with cocaine abuse. Pharmacogenomics J. 2003;3:27–40. doi: 10.1038/sj.tpj.6500146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leisewitz AV, Jung JE, Perez-Alzola P, Fuenzalida KM, Roth A, Inestrosa NC, et al. Ethanol specifically decreases peroxisome proliferator activated receptor beta in B12 oligodendrocyte-like cells. J Neurochem. 2003;85:135–141. doi: 10.1046/j.1471-4159.2003.01641.x. [DOI] [PubMed] [Google Scholar]
- Lewohl JM, Wang L, Miles MF, Zhang L, Dodd PR, Harris RA. Gene expression in human alcoholism: microarray analysis of frontal cortex. Alcohol Clin Exp Res. 2000;24:1873–1882. [PubMed] [Google Scholar]
- Liu D, Wang Z, Liu S, Wang F, Zhao S, Hao A. Anti-inflammatory effects of fluoxetine in lipopolysaccharide(LPS)-stimulated microglial cells. Neuropharmacology. 2011;61:592–599. doi: 10.1016/j.neuropharm.2011.04.033. [DOI] [PubMed] [Google Scholar]
- Liu J, Lewohl JM, Dodd PR, Randall PK, Harris RA, Mayfield RD. Gene expression profiling of individual cases reveals consistent transcriptional changes in alcoholic human brain. J Neurochem. 2004;90:1050–1058. doi: 10.1111/j.1471-4159.2004.02570.x. [DOI] [PubMed] [Google Scholar]
- Luckenbaugh DA, Ibrahim L, Brutsche N, Franco-Chaves J, Mathews D, Marquardt CA, et al. Family history of alcohol dependence and antidepressant response to an N-methyl-D-aspartate antagonist in bipolar depression. Bipolar Disord. 2012;14:880–887. doi: 10.1111/bdi.12003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madayag A, Lobner D, Kau KS, Mantsch JR, Abdulhameed O, Hearing M, et al. Repeated N-acetylcysteine administration alters plasticity-dependent effects of cocaine. J Neurosci. 2007;27:13968–13976. doi: 10.1523/JNEUROSCI.2808-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maeng S, Zarate CA, Jr., Du J, Schloesser RJ, McCammon J, Chen G, et al. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol Psychiatry. 2008;63:349–352. doi: 10.1016/j.biopsych.2007.05.028. [DOI] [PubMed] [Google Scholar]
- Mahon K, Burdick KE, Szeszko PR. A role for white matter abnormalities in the pathophysiology of bipolar disorder. Neurosci Biobehav Rev. 2010;34:533–554. doi: 10.1016/j.neubiorev.2009.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao J, Sung B, Ji RR, Lim G. Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity. J Neurosci. 2002;22:8312–8323. doi: 10.1523/JNEUROSCI.22-18-08312.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marie-Claire C, Courtin C, Roques BP, Noble F. Cytoskeletal genes regulation by chronic morphine treatment in rat striatum. Neuropsychopharmacology. 2004;29:2208–2215. doi: 10.1038/sj.npp.1300513. [DOI] [PubMed] [Google Scholar]
- Mayfield RD, Lewohl JM, Dodd PR, Herlihy A, Liu J, Harris RA. Patterns of gene expression are altered in the frontal and motor cortices of human alcoholics. J Neurochem. 2002;81:802–813. doi: 10.1046/j.1471-4159.2002.00860.x. [DOI] [PubMed] [Google Scholar]
- 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(Suppl 1):S120–S128. doi: 10.1016/j.bbi.2011.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Messer CJ, Eisch AJ, Carlezon WA, Jr., Whisler K, Shen L, Wolf DH, et al. Role for GDNF in biochemical and behavioral adaptations to drugs of abuse. Neuron. 2000;26:247–257. doi: 10.1016/s0896-6273(00)81154-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mguel-Hidalgo JJ. Lower packing density of glial fibrillary acidic protein-immunoreactive astrocytes in the prelimbic cortex of alcohol-naive and alcohol-drinking alcohol-preferring rats as compared with alcohol-nonpreferring and Wistar rats. Alcohol Clin Exp Res. 2005;29:766–772. doi: 10.1097/01.alc.0000164378.92680.fa. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mguel-Hidalgo JJ. Transient increase of alcohol preference in Wistar rats after infusion of a blocker of astrocytic gap junctions into the prelimbic cortex. Alcohol Clin Exp Res. 2007;31:88A. [Google Scholar]
- Mguel-Hidalgo JJ, Rajkowska G. Comparison of prefrontal cell pathology between depression and alcohol dependence. J Psychiatr Res. 2003;37:411–420. doi: 10.1016/s0022-3956(03)00049-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mguel-Hidalgo JJ, Baucom C, Dilley G, Overholser JC, Meltzer HY, Stockmeier CA, et al. Glial fibrillary acidic protein immunoreactivity in the prefrontal cortex distinguishes younger from older adults in major depressive disorder. Biol Psychiatry. 2000;48:861–873. doi: 10.1016/s0006-3223(00)00999-9. [DOI] [PubMed] [Google Scholar]
- Mguel-Hidalgo JJ, Wei J, Andrew M, Overholser JC, Jurjus G, Stockmeier CA, et al. Glia pathology in the prefrontal cortex in alcohol dependence with and without depressive symptoms. Biol Psychiatry. 2002;52:1121–1133. doi: 10.1016/s0006-3223(02)01439-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mguel-Hidalgo JJ, Overholser JC, Meltzer HY, Stockmeier CA, Rajkowska G. Reduced glial and neuronal packing density in the orbitofrontal cortex in alcohol dependence and its relationship with suicide and duration of alcohol dependence. Alcohol Clin Exp Res. 2006;30:1845–1855. doi: 10.1111/j.1530-0277.2006.00221.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mguel-Hidalgo J, Shoyama Y, Wanzo V. Infusion of gliotoxins or a gap junction blocker in the prelimbic cortex increases alcohol preference in Wistar rats. J Psychopharmacol. 2009;23:550–557. doi: 10.1177/0269881108091074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mguel-Hidalgo JJ, Waltzer R, Whittom AA, Austin MC, Rajkowska G, Stockmeier CA. Glial and glutamatergic markers in depression, alcoholism, and their comorbidity. J Affect Disord. 2010;127:230–240. doi: 10.1016/j.jad.2010.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueser KT, Drake RE, Wallach MA. Dual diagnosis: a review of etiological theories. Addict Behav. 1998;23:717–734. [PubMed] [Google Scholar]
- Mulholland PJ, Carpenter-Hyland EP, Woodward JJ, Chandler LJ. Ethanol disrupts NMDA receptor and astroglial EAAT2 modulation of Kv2.1 potassium channels in hippocampus. Alcohol. 2009;43:45–50. doi: 10.1016/j.alcohol.2008.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair MP, Schwartz SA, Kronfol ZA, Hill EM, Sweet AM, Greden JF. Suppression of tumor necrosis factor production by alcohol in lipopolysaccharide-stimulated culture. Alcohol Clin Exp Res. 1994;18:602–607. doi: 10.1111/j.1530-0277.1994.tb00917.x. [DOI] [PubMed] [Google Scholar]
- Nakagawa T, Ozawa T, Shige K, Yamamoto R, Minami M, Satoh M. Inhibition of morphine tolerance and dependence by MS-153, a glutamate transporter activator. Eur J Pharmacol. 2001;419:39–45. doi: 10.1016/s0014-2999(01)00965-7. [DOI] [PubMed] [Google Scholar]
- Nakagawa T, Fujio M, Ozawa T, Mnami M, Satoh M. Effect of MS-153, a glutamate transporter activator, on the conditioned rewarding effects of morphine, methamphetamine and cocaine in mice. Behav Brain Res. 2005;156:233–239. doi: 10.1016/j.bbr.2004.05.029. [DOI] [PubMed] [Google Scholar]
- Narita M, Miyatake M, Shibasaki M, Tsuda M, Koizumi S, Yajima Y, et al. Long-lasting change in brain dynamics induced by methamphetamine: enhancement of protein kinase C-dependent astrocytic response and behavioral sensitization. J Neurochem. 2005;93:1383–1392. doi: 10.1111/j.1471-4159.2005.03097.x. [DOI] [PubMed] [Google Scholar]
- Narita K, Murata T, Takahashi T, Kosaka H, Omata N, Wada Y. Plasma levels of adiponectin and tumor necrosis factor-alpha in patients with remitted major depression receiving long-term maintenance antidepressant therapy. Prog Neuropsychopharmacol Biol Psychiatry. 2006a;30:1159–1162. doi: 10.1016/j.pnpbp.2006.03.030. [DOI] [PubMed] [Google Scholar]
- Narita M, Miyatake M, Shibasaki M, Shindo K, Nakamura A, Kuzumaki N, et al. Direct evidence of astrocytic modulation in the development of rewarding effects induced by drugs of abuse. Neuropsychopharmacology. 2006b;31:2476–2488. doi: 10.1038/sj.npp.1301007. [DOI] [PubMed] [Google Scholar]
- Nelson S, Bagby GJ, Summer WR. Alcohol-induced suppression of tumor necrosis factor - a potential risk factor for secondary infection in the acquired immunodeficiency syndrome. Prog Clin Biol Res. 1990;325:211–220. [PubMed] [Google Scholar]
- Niciu MJ, Chan G, Gelernter J, Arias AJ, Douglas K, Weiss R, et al. Subtypes of major depression in substance dependence. Addiction. 2009;104:1700–1709. doi: 10.1111/j.1360-0443.2009.02672.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niciu MJ, Kelmendi B, Sanacora G. Overview of glutamatergic neurotransmission in the nervous system. Pharmacol Biochem Behav. 2012;100:656–664. doi: 10.1016/j.pbb.2011.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niwa M, Nitta A, Yamada Y, Nakajima A, Saito K, Seishima M, et al. An inducer for glial cell line-derived neurotrophic factor and tumor necrosis factor-alpha protects against methamphetamine-induced rewarding effects and sensitization. Biol Psychiatry. 2007;61:890–901. doi: 10.1016/j.biopsych.2006.06.016. [DOI] [PubMed] [Google Scholar]
- Nixon K, Kim DH, Potts EN, He J, Crews FT. Distinct cell proliferation events during abstinence after alcohol dependence: microglia proliferation precedes neurogenesis. Neurobiol Dis. 2008;31:218–229. doi: 10.1016/j.nbd.2008.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien SM, Scully P, Fitzgerald P, Scott LV, Dinan TG. Plasma cytokine profiles in depressed patients who fail to respond to selective serotonin reuptake inhibitor therapy. J Psychiatr Res. 2007;41:326–331. doi: 10.1016/j.jpsychires.2006.05.013. [DOI] [PubMed] [Google Scholar]
- Oberheim NA, Goldman SA, Nedergaard M. Heterogeneity of astrocytic form and function. Methods Mol Biol. 2012;814:23–45. doi: 10.1007/978-1-61779-452-0_3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okamoto H, Mki T, Lee KY, Yokoyama T, Kuma H, Wang ZY, et al. Oligodendrocyte myelin glycoprotein (OMgp) in rat hippocampus is depleted by chronic ethanol consumption. Neurosci Lett. 2006;406:76–80. doi: 10.1016/j.neulet.2006.07.023. [DOI] [PubMed] [Google Scholar]
- Ongur D, Heckers S. A role for glia in the action of electroconvulsive therapy. Harv Rev Psychiatry. 2004;12:253–262. doi: 10.1080/10673220490886185. [DOI] [PubMed] [Google Scholar]
- Papouin T, Ladepeche L, Ruel J, Sacchi S, Labasque M, Hanini M, et al. Synaptic and extrasynaptic NMDA receptors are gated by different endogenous coagonists. Cell. 2012;150:633–636. doi: 10.1016/j.cell.2012.06.029. [DOI] [PubMed] [Google Scholar]
- Penninx BW, Kritchevsky SB, Yaffe K, Newman AB, Simonsick EM, Rubin S, et al. Inflammatory markers and depressed mood in older persons: results from the Health, Aging and Body Composition study. Biol Psychiatry. 2003;54:566–572. doi: 10.1016/s0006-3223(02)01811-5. [DOI] [PubMed] [Google Scholar]
- Petrakis IL, Limoncelli D, Gueorguieva R, Jatlow P, Boutros NN, Trevisan L, et al. Altered NMDA glutamate receptor antagonist response in individuals with a family vulnerability to alcoholism. Am J Psychiatry. 2004;161:1776–1782. doi: 10.1176/ajp.161.10.1776. [DOI] [PubMed] [Google Scholar]
- Pfefferbaum A, Sullivan EV, Hedehus M, Adalsteinsson E, Lim KO, Moseley M. In vivo detection and functional correlates of white matter microstructural disruption in chronic alcoholism. Alcohol Clin Exp Res. 2000;24:1214–1221. [PubMed] [Google Scholar]
- Phelps LE, Brutsche N, Moral JR, Luckenbaugh DA, Manji HK, Zarate CA., Jr. Family history of alcohol dependence and initial antidepressant response to an N-methyl-D-aspartate antagonist. Biol Psychiatry. 2009;65:181–184. doi: 10.1016/j.biopsych.2008.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pons-Vazquez S, Gallego-Pinazo R, Galbis-Estrada C, Zanon-Moreno V, Garcia-Medina JJ, Vila-Bou V, et al. Combined pre- and postnatal ethanol exposure in rats disturbs the myelination of optic axons. Alcohol Alcohol. 2011;46:514–522. doi: 10.1093/alcalc/agr063. [DOI] [PubMed] [Google Scholar]
- Price RB, Nock MK, Charney DS, Mathew SJ. Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry. 2009;66:522–526. doi: 10.1016/j.biopsych.2009.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pubill D, Canudas AM, Pallas M, Camins A, Camarasa J, Escubedo E. Different glial response to methamphetamine- and methylenedioxymethamphetamine-induced neurotoxicity. Naunyn Schmiedebergs Arch Pharmacol. 2003;367:490–499. doi: 10.1007/s00210-003-0747-y. [DOI] [PubMed] [Google Scholar]
- Qin L, Crews FT. NADPH oxidase and reactive oxygen species contribute to alcohol-induced microglial activation and neurodegeneration. J Neuroinflammation. 2012;9:5. doi: 10.1186/1742-2094-9-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin L, He J, Hanes RN, Pluzarev O, Hong JS, Crews FT. Increased systemic and brain cytokine production and neuroinflammation by endotoxin following ethanol treatment. J Neuroinflammation. 2008;5:10. doi: 10.1186/1742-2094-5-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, et al. A randomized controlled trial of the Tumor Necrosis Factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. Arch Gen Psychiatry. 2012:1–11. doi: 10.1001/2013.jamapsychiatry.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajkowska G, Mguel-Hidalgo JJ, Wei J, Dilley G, Pittman SD, Meltzer HY, et al. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol Psychiatry. 1999;45:1085–1098. doi: 10.1016/s0006-3223(99)00041-4. [DOI] [PubMed] [Google Scholar]
- Rajkowska G, Mguel-Hidalgo JJ, Makkos Z, Meltzer H, Overholser J, Stockmeier C. Layer-specific reductions in GFAP-reactive astroglia in the dorsolateral prefrontal cortex in schizophrenia. Schizophr Res. 2002;57:127–138. doi: 10.1016/s0920-9964(02)00339-0. [DOI] [PubMed] [Google Scholar]
- Rajkowska G, Hughes J, Stockmeier CA, Javier Miguel-Hidalgo J, Maciag D. Coverage of blood vessels by astrocytic end-feet is reduced in major depressive disorder. Biol Psychiatry. 2013;73:613–621. doi: 10.1016/j.biopsych.2012.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Regenold WT, Phatak P, Marano CM, Gearhart L, Viens CH, Hisley KC. Myelin staining of deep white matter in the dorsolateral prefrontal cortex in schizophrenia, bipolar disorder, and unipolar major depression. Psychiatry Res. 2007;151:179–188. doi: 10.1016/j.psychres.2006.12.019. [DOI] [PubMed] [Google Scholar]
- Rimondini R, Arlinde C, Sommer W, Heilig M. Long-lasting increase in voluntary ethanol consumption and transcriptional regulation in the rat brain after intermittent exposure to alcohol. FASEB J. 2002;16:27–35. doi: 10.1096/fj.01-0593com. [DOI] [PubMed] [Google Scholar]
- Sanchez ES, Bigbee JW, Fobbs W, Robinson SE, Sato-Bigbee C. Opioid addiction and pregnancy: perinatal exposure to buprenorphine affects myelination in the developing brain. Glia. 2008;56:1017–1027. doi: 10.1002/glia.20675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sander T, Ostapowicz A, Samochowiec J, Smolka M, Winterer G, Schmidt LG. Genetic variation of the glutamate trans- porter EAAT2 gene and vulnerability to alcohol dependence. Psychiatr Genet. 2000;10:103–107. doi: 10.1097/00041444-200010030-00001. [DOI] [PubMed] [Google Scholar]
- Schroeter ML, Abdul-Khaliq H, Krebs M, Diefenbacher A, Blasig IE. Serum markers support disease-specific glial pathology in major depression. J Affect Disord. 2008;111:271–280. doi: 10.1016/j.jad.2008.03.005. [DOI] [PubMed] [Google Scholar]
- 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. J Neurosci. 2011;31:17835–17847. doi: 10.1523/JNEUROSCI.3297-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shear PK, Jernigan TL, Butters N. Volumetric magnetic resonance imaging quantification of longitudinal brain changes in abstinent alcoholics. Alcohol Clin Exp Res. 1994;18:172–176. doi: 10.1111/j.1530-0277.1994.tb00899.x. [DOI] [PubMed] [Google Scholar]
- Sheinin A, Shavit S, Benveniste M. Subunit specificity and mechanism of action of NMDA partial agonist D-cycloserine. Neuropharmacology. 2001;41:151–158. doi: 10.1016/s0028-3908(01)00073-9. [DOI] [PubMed] [Google Scholar]
- Simen BB, Duman CH, Simen AA, Duman RS. TNFalpha signaling in depression and anxiety: behavioral consequences of individual receptor targeting. Biol Psychiatry. 2006;59:775–785. doi: 10.1016/j.biopsych.2005.10.013. [DOI] [PubMed] [Google Scholar]
- Song P, Zhao ZQ. The involvement of glial cells in the development of morphine tolerance. Neurosci Res. 2001;39:281–286. doi: 10.1016/s0168-0102(00)00226-1. [DOI] [PubMed] [Google Scholar]
- Spanagel R, Pendyala G, Abarca C, Zghoul T, Sanchis-Segura C, Magnone MC, et al. The clock gene Per2 influences the glutamatergic system and modulates alcohol consumption. Nat Med. 2005;11:35–42. doi: 10.1038/nm1163. [DOI] [PubMed] [Google Scholar]
- Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai Z, et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? J Neuroinflammation. 2011;8:94. doi: 10.1186/1742-2094-8-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su N, Zhang L, Fei F, Hu H, Wang K, Hui H, et al. The brain-derived neurotrophic factor is associated with alcohol dependence-related depression and antidepressant response. Brain Res. 2011;1415:119–126. doi: 10.1016/j.brainres.2011.08.005. [DOI] [PubMed] [Google Scholar]
- Taylor WD, Steffens DC, MacFall JR, McQuoid DR, Payne ME, Provenzale JM, et al. White matter hyperintensity progression and late-life depression outcomes. Arch Gen Psychiatry. 2003;60:1090–1096. doi: 10.1001/archpsyc.60.11.1090. [DOI] [PubMed] [Google Scholar]
- Taylor WD, MacFall JR, Payne ME, McQuoid DR, Provenzale JM, Steffens DC, et al. Late-life depression and microstructural abnormalities in dorsolateral prefrontal cortex white matter. Am J Psychiatry. 2004;161:1293–1296. doi: 10.1176/appi.ajp.161.7.1293. [DOI] [PubMed] [Google Scholar]
- Thomas DM, Dowgiert J, Geddes TJ, Francescutti-Verbeem D, Liu X, Kuhn DM. Mcroglial activation is a pharmacologically specific marker for the neurotoxic amphetamines. Neurosci Lett. 2004a;367:349–354. doi: 10.1016/j.neulet.2004.06.065. [DOI] [PubMed] [Google Scholar]
- Thomas DM, Walker PD, Benjamins JA, Geddes TJ, Kuhn DM. Methamphetamine neurotoxicity in dopamine nerve endings of the striatum is associated with microglial activation. J Pharmacol Exp Ther. 2004b;311:1–7. doi: 10.1124/jpet.104.070961. [DOI] [PubMed] [Google Scholar]
- Torres-Platas SG, Hercher C, Davoli MA, Maussion G, Labonte B, Turecki G, et al. Astrocytic hypertrophy in anterior cingulate white matter of depressed suicides. Neuropsychopharmacology. 2011;36:2650–2658. doi: 10.1038/npp.2011.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trantham-Davidson H, LaLumiere RT, Reissner KJ, Kalivas PW, Knackstedt LA. Ceftriaxone normalizes nucleus accumbens synaptic transmission, glutamate transport, and export following cocaine self-administration and extinction training. J Neurosci. 2012;32:12406–12410. doi: 10.1523/JNEUROSCI.1976-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tynan RJ, Weidenhofer J, Hinwood M, Cairns MJ, Day TA, Walker FR. A comparative examination of the anti-inflammatory effects of SSRI and SNRI antidepressants on LPS stimulated microglia. Brain Behav Immun. 2012;26:469–479. doi: 10.1016/j.bbi.2011.12.011. [DOI] [PubMed] [Google Scholar]
- Tyring S, Gottlieb A, Papp K, Gordon K, Leonardi C, Wang A, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet. 2006;367:29–35. doi: 10.1016/S0140-6736(05)67763-X. [DOI] [PubMed] [Google Scholar]
- Udomuksorn W, Mukem S, Kumarnsit E, Vongvatcharanon S, Vongvatcharanon U. Effects of alcohol administration during adulthood on parvalbumin and glial fibrillary acidic protein immunoreactivity in the rat cerebral cortex. Acta Histochem. 2011;113:283–289. doi: 10.1016/j.acthis.2009.11.001. [DOI] [PubMed] [Google Scholar]
- Uranova NA, Vostrikov VM, Orlovskaya DD, Rachmanova VI. Oligodendroglial density in the prefrontal cortex in schizophrenia and mood disorders: a study from the Stanley Neuropathology Consortium. Schizophr Res. 2004;67:269–275. doi: 10.1016/S0920-9964(03)00181-6. [DOI] [PubMed] [Google Scholar]
- Valentine GW, Sanacora G. Targeting glial physiology and glutamate cycling in the treatment of depression. Biochem Pharmacol. 2009;78:431–39. doi: 10.1016/j.bcp.2009.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viana AF, Maciel IS, Dornelles FN, Figueiredo CP, Siqueira JM, Campos MM, et al. Kinin B1 receptors mediate depression-like behavior response in stressed mice treated with systemic E. coli lipopolysaccharide. J Neuroinflammation. 2010;7:98. doi: 10.1186/1742-2094-7-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vongvatcharanon U, Mukem S, Udomuksorn W, Kumarsit E, Vongvatcharanon S. Alcohol administration during adulthood induces alterations of parvalbumin and glial fibrillary acidic protein immunoreactivity in rat hippocampus and cingulate cortex. Acta Histochem. 2010;112:392–401. doi: 10.1016/j.acthis.2009.04.001. [DOI] [PubMed] [Google Scholar]
- Vostrikov VM, Uranova NA, Orlovskaya DD. Deficit of perineuronal oligodendrocytes in the prefrontal cortex in schizophrenia and mood disorders. Schizophr Res. 2007;94:273–280. doi: 10.1016/j.schres.2007.04.014. [DOI] [PubMed] [Google Scholar]
- Watkins LR, Hutchinson MR, Rice KC, Maier SF. The“toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol Sci. 2009;30:581–591. doi: 10.1016/j.tips.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wennstrom M, Hellsten J, Tingstrom A. Electroconvulsive seizures induce proliferation of NG2-expressing glial cells in adult rat amygdala. Biol Psychiatry. 2004;55:464–471. doi: 10.1016/j.biopsych.2003.11.011. [DOI] [PubMed] [Google Scholar]
- Wong WT, Wang M, Li W. Regulation of microglia by ionotropic glutamatergic and GABAergic neurotransmission. Neuron Glia Biol. 2011;7:41–6. doi: 10.1017/S1740925X11000123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, Lee MR, Kim T, Johng S, Rohrback S, Rang N, et al. Regulation of ethanol-sensitive EAAT2 expression through adenosine Al receptor in astrocytes. Biochem Biophys Res Commun. 2011;406:47–52. doi: 10.1016/j.bbrc.2011.01.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye Y, Wang G, Wang H, Wang X. Brain-derived neurotrophic factor (BDNF) infusion restored astrocytic plasticity in the hippocampus of a rat model of depression. Neurosci Lett. 2011;503:15–19. doi: 10.1016/j.neulet.2011.07.055. [DOI] [PubMed] [Google Scholar]
- Zarate CA, Jr., Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63:856–864. doi: 10.1001/archpsyc.63.8.856. [DOI] [PubMed] [Google Scholar]
- Zarate CA, Jr., Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A, et al. Replication of ketamine’s antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol Psychiatry. 2012;71:939–946. doi: 10.1016/j.biopsych.2011.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zarate CA, Jr., Mathews D, Ibrahim L, Chaves JF, Marquardt C, Ukoh I, et al. A randomized trial of a low-trapping N-methyl-d-asparate channel blocker in major depression. Biol Psychiatry. 2013;74:257–264. doi: 10.1016/j.biopsych.2012.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Kitaichi K, Fujimoto Y, Nakayama H, Shimizu E, Iyo M, et al. Protective effects of minocycline on behavioral changes and neurotoxicity in mice after administration of methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:1381–1393. doi: 10.1016/j.pnpbp.2006.05.015. [DOI] [PubMed] [Google Scholar]
- Zhang XQ, Cui Y, Cui Y, Chen Y, Na XD, Chen FY, et al. Activation of p38 signaling in the microglia in the nucleus accumbens contributes to the acquisition and maintenance of morphine-induced conditioned place preference. Brain Behav Immun. 2012;26:318–325. doi: 10.1016/j.bbi.2011.09.017. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Li H, Li Y, Sun X, Zhu M, Hanley G, et al. Essential role of toll-like receptor 2 in morphine-induced microglia activation in mice. Neurosci Lett. 2011;489:43–47. doi: 10.1016/j.neulet.2010.11.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhurov V, Stead JD, Merali Z, Palkovits M, Faludi G, Schild-Poulter C, et al. Molecular pathway reconstruction and analysis of disturbed gene expression in depressed individuals who died by suicide. PLoS One. 2012;7:e47581. doi: 10.1371/journal.pone.0047581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziedonis DM. Integrated treatment of co-occurring mental illness and addiction: clinical intervention, program, and system perspectives. CNS Spectr. 2004;9:892–904. doi: 10.1017/s1092852900009718. 925. [DOI] [PubMed] [Google Scholar]
- Zink M, Vollmayr B, Gebicke-Haerter PJ, Henn FA. Reduced expression of glutamate transporters vGluT1, EAAT2 and EAAT4 in learned helpless rats, an animal model of depression. Neuropharmacology. 2010;58:465–473. doi: 10.1016/j.neuropharm.2009.09.005. [DOI] [PubMed] [Google Scholar]
- Zou JY, Crews FT. TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition. Brain Res. 2005;1034:11–24. doi: 10.1016/j.brainres.2004.11.014. [DOI] [PubMed] [Google Scholar]