Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a brain-enriched immediate early gene that regulates important mechanisms implicated in learning and memory. Arc levels are controlled through a balance of induction and degradation in an activity-dependent manner. Arc further undergoes multiple post-translational modifications that regulate its stability, localization and function. Recent studies demonstrate that these features of Arc can be pharmacologically manipulated. In this review, we discuss some of these compounds, with an emphasis on drugs of abuse and psychotropic drugs. We also discuss inflammatory states that regulate Arc.
Keywords: Arc/Arg3.1, Addiction, Antidepressants, Antipsychotics, BDNF, Drugs of abuse, Inflammation
Graphical abstract
Highlights
-
•
Arc is an immediate early gene whose expression is disrupted in neurological conditions.
-
•
Arc expression in the brain can be modulated by drugs of abuse and psychotropic drugs.
-
•
Arc might be a viable therapeutic target for neurological conditions.
1. Introduction
Activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene (IEG) product whose levels rapidly increase and then decline as a response to changes in neural activity (Guzowski et al., 1999). Arc is elevated during memory acquisition, consolidation, and retrieval and is involved in the regulation of mood and reward-reinforcement (Li et al., 2015; Zhang and Bramham, 2020; Bramham et al., 2008; Ramirez-Amaya et al., 2005; Plath et al., 2006). Arc temporal dynamics are also critical for cognitive flexibility (Wall et al., 2018). Not surprisingly, Arc modulates Hebbian and non-Hebbian forms of synaptic plasticity including long-term potentiation (LTP) (Wang et al., 2016,Guzowski, 2000, Plath et al., 2006), long-term depression (LTD) (Park et al., 2008; Waung et al., 2008), inverse synaptic tagging (Okuno et al., 2012), and homeostatic plasticity (Shepherd et al., 2006; Mabb and Ehlers, 2018). Changes in Arc expression in the prelimbic, infralimbic, anterior cingulate, prefrontal cortex, and amygdala have also been shown to play a role in mood regulation (Li et al., 2015). Arc regulation in the mesocorticolimbic system, including the nucleus accumbens (NAc), medial prefrontal cortex (mPFC), and dorsomedial striatum modulates the reward and reinforcement system and affects drug-seeking behaviors (Li et al., 2013). Modulation of Arc has further been implicated in several neurological disorders including Alzheimer's disease (AD), schizophrenia, depression, autism, Angelman syndrome, and Fragile X syndrome (Gallo et al., 2018; Wilkerson et al., 2018).
Given Arc's involvement in neurological and neuropsychiatric conditions, along with recent findings for its role in drug seeking behavior, it is viable to propose that Arc modulation at the transcriptional, translational and posttranslational levels may be a relevant therapeutic target. Here, we discuss how Arc can be pharmacologically manipulated, with a specific focus on neuropsychotropic and inflammatory pathways that modulate Arc in key brain regions that are involved in addiction, reinforcement and reward.
2. Arc structure
To establish Arc as a potential clinical target, it is important to discuss recent findings regarding its genetic organization and protein structure. The mouse Arc promotor has a major synaptic activity-responsive element (SARE) located >5 kb upstream of its transcription initiation site, which is necessary and sufficient to replicate endogenous Arc transcription. The promoter consists of a cluster of neuronal activity-dependent cis-regulatory elements of closely localized binding sites for cAMP-response element binding protein (CREB), myocyte enhancer factor 2 (MEF2) and serum response factor (SRF). This SARE falls within a region that is highly conserved in humans (Kawashima et al., 2009). Based on this discovery, an enhanced synthetic activity-regulated promotor was generated (E-SARE) with a more than 20-fold higher expression and 30-fold higher induction ratio than the c-fos promoter. This E-SARE was used in mice to map active neuronal ensembles in multiple brain regions (Kawashima et al., 2013). SRF, a transcription factor that is regulated by synaptic activity (Knoll and Nordheim, 2009), binds to the Arc SARE at two locations that are approximately 1.1 and 6.9 kb upstream from the translation start site. SRF binding at location 6.9 is required for the late phase of LTD in mouse cultured cerebellar Purkinje cells (Smith-Hicks et al., 2010). The rapid induction of Arc is due to the phenomenon of “promotor proximal Poll II stalling” that is mediated by negative elongation factor (NELF), where RNA polymerase II (Pol II) is recruited to the transcription start site (TSS) and stalls in the proximity of the promotor until heightened neuronal activity releases the stalled Pol II (Saha et al., 2011).
Arc mRNA is unique among other IEGs in that newly transcribed Arc is rapidly localized to dendrites, selectively in domains contacted by active synapses (Lyford et al., 1995; Link et al., 1995). Constructs that contain the 3′ untranslated terminal region (UTR) of Arc localize with high precision to a small domain at the base of the dendritic spine even when the transcript is tethered to a different protein product suggesting that the 3’ UTR of Arc is sufficient for its dendritic localization. Additionally, translation inhibitors do not interfere with Arc mRNA localization, indicating that targeting of Arc is a function of its mRNA rather than its protein (Dynes and Steward, 2007, 2012; Steward et al., 1998). Similarly, Arc mRNA degradation is activity-dependent, where both localization and degradation appear to rely on N-methyl D-aspartate receptor (NMDAR) activation (Farris et al., 2014). Arc also accumulates at inactive synapses through an interaction with calmodulin-dependent kinase II (CaMKII) beta (Okuno et al., 2012). Both paired pulse and metabotropic glutamate receptor (mGluR)-dependent LTD rapidly induce Arc translation (Park et al., 2008).
Structurally, Arc is composed of two lobes; the N-lobe and the C-lobe, which together form the GAG domain, sharing similarities to capsid-forming Gag proteins (Zhang et al., 2015; Pastuzyn et al., 2018). The N-lobe contains a matrix domain and forms a hydrophobic pocket, allowing it to selectively bind to synaptic scaffolding proteins such as CaMKII and the transmembrane AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor regulatory protein, TARPγ2 (also known as Stargazin) (Zhang et al., 2015). The acidic C-lobe of Arc (amino acids 280–396 in rat) contains a capsid-like domain with an adjacent disordered C-terminal tail (Boldridge et al., 2020; Nielsen et al., 2019). Given this domain organization, Arc has been shown to assemble into viral-like capsids that are proposed to form via electrostatic interactions of the C-lobes (Pastuzyn et al., 2018; Boldridge et al., 2020; Nielsen et al., 2019). A recent study demonstrated that a 28-amino acid helical coil motif in the N-terminal region is necessary and sufficient for Arc to self-assemble into viral-like capsids (Eriksen et al., 2020). Arc can exist as a monomer but can also undergo self-oligomerization, which can be reversible depending on external conditions. In high ionic environments, Arc oligomerizes, leading to stabilization of its C-lobe (amino acids 209–365 in human) (Myrum et al., 2015). These structural features allow Arc to directly interact with endogenous binding partners that control synaptic function like TARPγ2, NMDA receptor subunits, and CaMKII (Zhang et al., 2015; Nielsen et al., 2019; Jackson and Nicoll, 2011).
Arc also undergoes various post-translational modifications (PTMs) including ubiquitination, SUMOylation, phosphorylation, acetylation, and palmitoylation (Mabb and Ehlers, 2018; Carmichael and Henley, 2018). These PTMs play important roles in Arc functions that include oligomerization, proteasome-dependent degradation, AMPA receptor trafficking, association with the cytoskeleton, consolidation of LTP, mGluR-LTD, and subcellular localization (Greer et al., 2010; Craig et al., 2012; Nair et al., 2017; Nikolaienko et al., 2017; Mabb et al., 2014; Zhang et al., 2019; Gozdz et al., 2017; Barylko et al., 2018; Lalonde et al., 2017). For example, phosphorylation within the GAG domain of Arc by CaMKIIα regulates its oligomerization (Zhang et al., 2019). Deacetylation of Arc by a protein lysine deacetylase, or phosphorylation of Arc by GSK3α and GSK3β, promotes Arc degradation (Gozdz et al., 2017; Lalonde et al., 2017).
3. Drugs of abuse and Arc
Several studies have demonstrated an increase in Arc mRNA in the striatum and various other brain areas in response to amphetamine and cocaine (Tan et al., 2000; Fosnaugh et al., 1995; Fumagalli et al., 2006; Kodama et al., 1998). Arc is increased in the rat striatum within 15 min of cocaine administration and returns to basal levels by 2 h. This effect is suppressed by pretreatment with reserpine, which depletes catecholamines and dopamine (DA) stores at 24 and 3 h before cocaine treatment. Suppression is also observed following intrastriatal injections of the selective DA neurotoxin 6-hydroxydopamine (6-OHDA) or the DA receptor D1 antagonist SCH- 23390. Interestingly, DA D2 antagonists such as haloperidol, spiperone and eticlopride also induce Arc in the striatum. Collectively, these findings demonstrate a role for DA D1 antagonists in inducing Arc in response to cocaine (Fosnaugh et al., 1995; Fumagalli et al., 2006). Similarly, within 1 h of a single methamphetamine (METH) intraperitoneal injection, Arc significantly increases in the parietal, orbital and medial prefrontal cortex (mPFC) and to a lesser degree in medium-sized striatal neurons and the hippocampus, returning to basal levels by 6 h. Pretreatment with SCH- 23390 or the NMDA receptor antagonist MK-801 attenuates these effects suggesting a synergistic role for D1 and NMDA receptors in Arc modulation. However, a repeated METH regimen does not affect basal Arc (Kodama et al., 1998; Fujiyama et al., 2003) suggesting a potential role for Arc in synaptic tuning associated with an acute response to METH. The METH-induced increase in Arc in the cerebral cortex suggests that it may also play a role in the induction and maintenance of behavioral sensitization due to METH (Kodama et al., 1998). Interestingly, pretreatment with an intrastriatal infusion of the mu opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-Pen-Thr-NH2 (CTAP) attenuates the METH-induced increase in Arc (Horner et al., 2010) suggesting that opioid signaling can modulate dopaminergic responses to METH in the striatum.
Recreational drugs such as 3,4-methylenedioxymethamphetamine (MDMA/ecstasy) and delta-9-tetrahydrocannabinol (THC/Cannabis) are commonly abused by adolescents (Malone et al., 2010). MDMA increases serotonin release and inhibits its reuptake, while THC activates CB1 cannabinoid receptors (Cunningham et al., 2009; Rodriguez de Fonseca et al., 1993). Rats treated with MDMA and THC in adolescence (postnatal day 28–45) show long-term decreases in Arc that persist into adulthood in the cortex and the hippocampus in a sex-dependent manner. THC reduces Arc in the hippocampus of both males and females, while reductions in Arc in the PFC are observed only in females. MDMA also selectively reduces Arc in the female hippocampus (Llorente-Berzal et al., 2013).
Arc is upregulated in the mPFC at 1 day of abstinence in heroin self-administrating rats compared to rats yoked to heroin, but these changes do not persist following 14 days, which suggests that Arc might be involved in establishing the memory of the self-administration behavior (Kuntz et al., 2008). Runway training with heroin, in which rats are trained to run to a goal area to self-administer the drug, significantly increases Arc in the mPFC, the nucleus accumbens (NAc) and the striatum, with differences in the temporal expression of Arc between the dorsomedial and ventrolateral striatum, suggesting the involvement of Arc in goal-directed behavior in the ventrolateral striatum. SCH2 3390 and MK-801 abolish this increase and suppress drug seeking behavior (Li et al., 2013).
Withdrawal from psychostimulants such as amphetamines produce manic and depressive symptoms (Mamelak, 1978). Acute D-amphetamine challenge significantly increases Arc in the frontal cortex, visual cortex, CA1 and amygdala (Pathak et al., 2015). Moreover, Arc mRNA significantly increases in the insular frontal cortex, visual cortex, entorhinal cortex, CA3, CA1, subiculum and striatum on day 17 of amphetamine withdrawal (Pathak et al., 2015). This withdrawl-induced increase is associated with a decrease in functional connectivity between frontal cortex and striatum and an increase in functional connectivity between the amygdala and the hippocampus. The observed changes in connectivity are coupled to behavioral signs of amphetamine sensitization which include hyperactive locomotor responses to restraint stress and amphetamine challenge, reduction in sucrose consumption, impaired memory consolidation and impaired nest building (Pathak et al., 2015). In prepubertal rats, Arc is increased in the striatum and cingulate cortex in response to daily treatment of methylphenidate (MPH) for 14 days and is also induced with a challenge MPH dose following a 4-week drug-free period (Chase et al., 2007). An increase in Arc in the PFC is also observed following cocaine self-administration training (Fumagalli et al., 2009a) suggesting a role for Arc in drug-related learning. Increases in Arc are also found in c-fos positive PFC neurons following cue-induced heroin seeking (Fanous et al., 2013). Infusion of Arc antisense oligodeoxynucleotides into the striatum attenuates extinction of cocaine-seeking (Hearing et al., 2011), suggesting that Arc induction in the striatum is required for drug-seeking behaviors. Recently, Penrod et al. described a role for Arc in volitional cocaine-taking intravenous self-administration (IVSA). They found that despite Arc Knock-out (KO) mice showing deficits in fear conditioning, they have intact contextual and reward learning along with normal appetitive classical and operant conditioning. However, Arc KO mice have a flat dose-response curve, that unlike WT mice, continue to self-administer cocaine at all doses, suggesting a decrease in the hedonic set-point and in the reinforcing effects of the drug (Penrod et al., 2020). The consequences of selective Arc disruption in the PFC on cocaine self-administration have yet to be established.
Arc is implicated in the reinstatement of drug seeking behavior. An increase in Arc is observed in subregions of the PFC upon re-exposure to cocaine-related cues (Zavala et al., 2008). In rats, cocaine-related context-induced relapse to drug seeking increases Arc in the mPFC, orbitofrontal cortex (OFC), striatum, NAc, basolateral amygdala (BLA) and dorsal hippocampus (Hearing et al., 2008a, 2008b, 2010). Visual cue-elicited reinstatement of cocaine seeking is also associated with increases in Arc in the mPFC and BLA, while priming-induced reinstatement causes a more widespread Arc induction in cortical regions (especially the anterior cingulate and motor cortex) (Ziolkowska et al., 2011). These findings suggest a unique brain-specific pattern of Arc induction in different forms of drug relapse that include drug-paired stimulus-induced relapse (cue) and drug slip/lapse (priming) (Bossert et al., 2013; Beardsley and Shelton, 2012).
Following chronic morphine treatment, Arc mRNA and protein are increased in the rat striatum (Marie-Claire et al., 2004). Upon naloxone-precipitated withdrawal from morphine, Arc remains elevated in the frontal cortex, suggesting that persistent changes during opiate dependence may be due to a permanence in synaptic remodeling (Ammon et al., 2003). Additionally, Arc is induced upon morphine withdrawal in the central and basolateral amygdala along with the basolateral amygdala during reactivation of opiate withdrawal memories by re-exposure to the withdrawal-paired environment (Lucas et al., 2008). In the NAc, Arc protein significantly increases in the core after 2 h of morphine treatment, while it increases in the shell but not the core after context-induced drug seeking. Arc in the NAc is also essential for morphine-related addiction memory. Knockdown of Arc in the core blocks the acquisition and expression of morphine conditioned-place preference (CPP) and reinstatement, while knockdown in the shell only impairs the expression of morphine CPP (Lv et al., 2011). In a recent study, activation of neural activity patterns in the striatum was measured using cellular compartment analysis of temporal activity by fluorescence in-situ hybridization (catFISH) for Arc and Homer1a. Although separate injections of cocaine and heroin induce Arc and Homer1a with a similar time course, injection of cocaine followed by heroin 25 min later engages distinct patterns of neurons in the striatum, providing supporting evidence that these two drugs operate through different mechanisms in the brain and encode distinct representations in the striatum (Vassilev et al., 2020).
Arc is also modulated in models of alcoholism, where acute ethanol exposure through intraperitoneal injections in rats increases Arc protein and dendritic spine density in the central and medial amygdala, while withdrawal after long term exposure is associated with a decrease in Arc and dendritic spine density in these same areas. Changes in brain derived neurotrophic factor (BDNF) and its receptor tyrosine Kinase B (trkB) mirror the changes in Arc. Additionally, infusion of BDNF into the central amygdala normalizes Arc levels and attenuates the onset of the withdrawal-related anxiety, suggesting that BDNF-Arc signaling in the central and medial amygdala is involved in alcohol dependence and anxiety related to drinking behavior (Pandey et al., 2008).
Intraperitoneal injections of caffeine, an adenosine (A) receptor antagonist, or the A1 antagonist 1,3-dipropyl-8 cyclopentylxanthine (DPCPX) induce Arc mRNA and protein in striatonigral and striatopallidal neurons. This effect appears to be under the control of the dopaminergic system, where pretreatment with the D2/D3 agonist quinpirole almost completely blocks this caffeine-induced increase in Arc, while the D1 antagonist SCH2 3390 attenuates it in a dose-dependent manner (Dassesse et al., 1999). In juvenile rats, acute nicotine exposure by intraperitoneal injections at postnatal day (P) 5,7 and 10 lead to an increase in Arc in cingulate cortex, hippocampus, caudate and central amygdala, while Arc increases in the bed nucleus of stria terminalis after acute exposure to nicotine at P10 only, indicating that age is an important factor in nicotine-induced regulation of Arc (Schmitt et al., 2008).
4. Psychotropic drugs
Arc is affected by serotonin (5-HT) in various brain areas. While the 5-HT precursor L-tryptophan by itself fails to induce an increase in Arc, its combined intraperitoneal injection with the monoamine oxidase inhibitor (MAOI), tranylcypromine increases Arc in the orbital, frontal and parietal cortices the striatum and decreases in the CA1 region of the hippocampus. The 5-HT releasing agent p-chloroamphetamine and the 5-HT2 agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) also increase Arc in cortical and striatal areas. 5-HT depletion using the tryptophan hydroxylase inhibitor p-chlorophenylalanine and pretreatment with the 5-HT2 antagonist ketanserin attenuate increases in the cortices and striatum, with no effect on the reduction observed in CA1. This suggests that Arc is regulated by endogenous 5-HT release in the cortex and striatum, where 5-HT2 appears to play an important role in the cortex (Pei et al., 2000). Several drugs that manipulate 5-HT are used to treat mood disorders and need several weeks to achieve their therapeutic effect (Machado-Vieira et al., 2010). Consistent with this, twice-daily injections for 14 days with the MAOIs paroxetine, desipramine and venlafaxine increase Arc in the parietal, frontal, orbital, cingulate cortex and CA1 of the hippocampus, with no changes detected in the caudate putamen or the dentate gyrus, while tranylcypromine increases Arc in the dentate gyrus. However, acute administration of these drugs has no effect on Arc, possibly because acute administration of antidepressant drugs causes a compensatory decrease in 5-HT due to feedback regulatory mechanisms involving 5-HT1A receptors (Blier and Ward, 2003), although tranylcypromine does cause an increase in Arc in the parietal cortex (Pei et al., 2003). S35966A, a dual α2-adrenoceptor antagonist and 5-HT-noradrenaline reuptake inhibitor more potently increases Arc compared to the selective serotonin and norepinephrine reuptake inhibitor (SSNRI) venlafaxine in the cortical and hippocampal areas following acute treatment (Serres et al., 2012). Duloxetine, also an SSNRI, induces Arc primarily in the frontal cortex after chronic treatment suggesting a role for Arc in long-term adaptive changes with chronic antidepressant treatment (Molteni et al., 2008).
Selective 5-HT1A receptor antagonists such as WAY 100635 and NDA-299 combined with the selective serotonin reuptake inhibitor (SSRI) paroxetine also increase Arc in frontal, parietal and piriform cortices and the caudate putamen, while none of these drugs have an effect on Arc when administered without paroxetine, suggesting the involvement of other 5-HT receptor subtypes (Tordera et al., 2003; Castro et al., 2003). Agomelatine, an MT1/MT2 melatonergic agonist and the 5-HT2c receptor antagonist, also upregulates Arc in the hippocampus (Calabrese et al., 2011).
Interestingly, electroconvulsive stimulation (ECS), which remains one of the most effective treatments against severe depression (Berton and Nestler, 2006), strongly induces Arc at 1 and 4 h post exposure (Lyford et al., 1995), and causes a significant increase in DNA methylation of the Arc gene promoter at 24 h suggesting repression of Arc expression for a long period post-ECS (Dyrvig et al., 2012). A genetic rat model of depression called Flinders sensitive line, displays depression-like behaviors accompanied by reduction of Arc in the prefrontal cortex and hippocampus as well as reduction in mitogen-activated protein kinase (MEK) activity. Chronic treatment with the SSRI escitalopram as well as increasing 5-HT postsynaptic function via stimulation of the cAMP/mitogen-activated protein kinase (MAPK) signaling cascades restores some cognitive functions such as memory performance and Arc, suggesting a role for the 5-HT/MEK cascade in regulating Arc in depression (Eriksson et al., 2012).
Unlike other antidepressants, the non-selective NMDA receptor antagonist ketamine produces antidepressant effects within hours of its administration that last up to 7 days (Berman et al., 2000). In rat models of depression, a low dose of 10 mg/kg ketamine activates the mTOR signaling pathway in the PFC, leading to an increase in synaptic proteins that peak at 2–6 h and remain elevated up to 72 h, with Arc protein showing a more transient increase. The selective NR2B antagonist Ro 25-6981, which also activates the mTOR pathway, causes a more rapid and transient induction of Arc at 1 h that returns to normal by 6 h, and produces rapid antidepressant effects in the forced swim and novelty-suppressed feeding tests (Li et al., 2010).
Both typical and atypical antipsychotic drugs block D2 receptors (Rampino et al., 2018) and have been shown to modulate Arc (Bruins Slot et al., 2009; de Bartolomeis et al., 2013). The typical (first generation) antipsychotic haloperidol increases Arc in the NAc and striatum at 1 h post treatment, while the atypical (second generation) drug clozapine significantly reduces Arc at 6 h in the thalamus and hypothalamus (Robbins et al., 2008). Another study compared haloperidol to a different atypical drug, olanzapine. A single injection of haloperidol induces Arc in the striatum at 30, 60, and 120 min, while olanzapine induces Arc only at the 30 min time point. In contrast, both drugs cause a sharp decline in Arc in the frontal cortex at 60 min. The atypical D2 antagonist raclopride produces similar effects to haloperidol and olanzapine, inducing Arc in the striatum and reducing it in the frontal cortex, while the D2 agonist quinpirole (which has lower affinity to D2 than olanzapine) produces no change in Arc (Fumagalli et al., 2009b). One study compared the effect of acute and chronic treatment (once daily for 21 days and 24 h following withdrawal of haloperidol and clozapine) in rats on Arc protein. In the caudate nucleus, Arc increases following acute and chronic treatment with haloperidol, while clozapine has no effect. In the NAc, haloperidol increases Arc following acute treatment but decreases it following chronic treatment, while clozapine increases Arc in the NAc shell but not the core following acute treatment. In the mPFC and cingulate cortex, only clozapine has an effect where it decreases Arc following acute and chronic administration. Amisulpride, a benzamide which is known for fewer motor side effects compared to haloperidol despite being a high-affinity D2/D3 receptor blocker (Grunder et al., 2009), has a region-selective effect on Arc in the striatum and induces significantly lower levels of Arc than those produced by haloperidol that is restricted to the medial striatum (de Bartolomeis et al., 2013). Interestingly, the first-generation anti-psychotics thioridazine and trifluoperazine have been shown to inhibit the Arc N-lobe binding to TARPγ2 through mechanisms independent of their action on D2 and their antipsychotic function (Zhang et al., 2015).
Several studies point out that schizophrenia patients consume increased amounts of nicotine and caffeine (Williams and Gandhi, 2008). Coffee drinking and tobacco smoking can modulate the haloperidol-induced changes in Arc mRNA and protein expression. One study showed that cotreatment of rats with caffeine and haloperidol reduces the increase in Arc in the striatum compared to treatment with haloperidol alone, while cotreatment with nicotine and haloperidol produces significantly higher Arc in the cortex compared to haloperidol alone (de Bartolomeis et al., 2018). Phencyclidine (PCP), an NMDA antagonist with psychomimetic effects that resemble positive and negative symptoms of schizophrenia, produces opposite effects on Arc mRNA in juvenile and adult rats in the mPFC, orbitofrontal cortex and NAc shell, where it decreases expression in juveniles and increases it in adults when administered for 5 days (Thomsen et al., 2010). Pretreatment with atypical antipsychotics 1 h before administration of PCP in adult rats significantly reduces the PCP-induced increase in Arc in the mPFC and NAc suggesting that Arc modulation could be a key factor in the therapeutic properties of these drugs (Nakahara et al., 2000).
5. Brain derived neurotrophic factor (BDNF) and Arc
BDNF is a neurotrophic factor that regulates the differentiation and survival of neurons through activation of tyrosine receptor kinase B (TrkB) (Haapasalo et al., 2002). Infusion of BDNF into the dentate gyrus of rats increases Arc mRNA in the granular and molecular layers at 2 h, with a three-fold increase in Arc protein at 3 h. This effect is abolished with the MEK inhibitor U0126 (Ying et al., 2002). The BDNF-induced increase in Arc in synaptoneurosomes is also partially inhibited by pretreatment of the tyrosine kinase non-specific inhibitor K252-alpha, and to a greater extent with the NMDA antagonist MK801 suggesting a role for NMDA receptors (Yin et al., 2002). BDNF also induces Arc in rat neuronal cultures and organotypic slices, an effect that can be potentiated by inhibiting AMPARs, which negatively regulate Arc transcription (Rao et al., 2006). One study demonstrated the importance of the serum-response element 1 (SRE1) of the Arc gene regulatory region in modulating the BDNF-induced Arc response, where SRE1 binds SRF in an activity-dependent manner to regulate Arc transcription (Pintchovski et al., 2009). Arc is necessary for BDNF-induced LTP induction and consolidation, where infusion of Arc antisense into the medial perforant path before BDNF infusion blocks the induction of BDNF-LTP and causes rapid reversal of ongoing BDNF-LTP when infused 2 h after BDNF (Messaoudi et al., 2007). BDNF also plays an important role in adult neurogenesis in the hippocampus (Scharfman et al., 2005) and infusion of Arc antisense into the rat dentate gyrus blocks the pro-neurogenic effects of BDNF (Kuipers et al., 2016).
6. Interfering with Arc removal
As discussed above, Arc PTMs play an important role in regulating the dynamics of Arc turnover. The proteasome inhibitor MG-132 interferes with removal of Arc by the ubiquitin proteasome system (UPS) (Mabb et al., 2014; Rao et al., 2006). Mutations of Arc ubiquitination sites (ArcKR) interfere with its proteasome-dependent removal, leading to abnormal elevations in Arc and deficits in cognitive flexibility (Wall et al., 2018). The glycogen synthase kinases α and β (GSK3α/β) are serine/threonine kinases that catalyze Arc phosphorylation and degradation. The GSK3α/β inhibitor CHIR 98014 (CH98) augments the effect of NMDA on Arc and slows down Arc degradation (Gozdz et al., 2017). Protein lysine deacetylase (KDAC) inhibitors, such as AK-7 and oxamflatin can increase Arc by limiting its degradation (Lalonde et al., 2017). A reduction in autophagy and accumulation of ubiquitinated Arc is observed in Fragile X syndrome model mice. Inhibiting autophagosome formation by addition of 3-methyladenine (3-MA), and/or the lysosomal inhibitor leupeptin in combination with ammonium chloride inhibits the autophagy pathway and leads to Arc accumulation in hippocampal neurons. Enhancing autophagy by removing the inhibitory autophagy signaling protein Raptor using shRNA corrects excessive Arc and rescues synaptic and cognitive deficits in Fragile X syndrome model mice (Yan et al., 2018).
7. Stress, the immune system and Arc
Arc has recently been shown to play a role in the inflammatory response inside and outside the central nervous system (Rosi, 2011). Neuroinflammation alters patterns of Arc in the brain. In rats, chronic infusion with lipopolysaccharide (LPS) into the fourth ventricle for 28 days results in elevated exploration-induced Arc and an increase in the percentage of neurons expressing Arc in the dentate gyrus and CA3 regions, which is accompanied by activation of microglia as revealed by OX-6 immunoreactivity. However, LPS-induced inflammation does not alter basal Arc nor affect Arc translation and removal (Rosi et al., 2005). Interestingly, memantine, the NMDA receptor antagonist widely used to manage AD, prevents these alterations when infused with LPS at concentrations that produce similar plasma levels to those produced by therapeutic doses in humans, and results in significant improvement in acquisition and retention in the water maze, suggesting these alterations are mediated through NMDA receptors (Rosi et al., 2006). Arc catFISH analysis demonstrates unstable CA3 pyramidal neuron activity between two exposures to the same contextual environments during LPS-induced chronic neuroinflammation, an effect that is partially normalized by memantine (Rosi et al., 2009). Conversely, mice receiving whole brain irradiation, similar to therapeutic irradiation commonly used to treat brain tumors, show a reduction in the percentage of neurons expressing Arc mRNA and protein, which is associated with a significant increase in activated microglia at 2 months post-irradiation (Rosi et al., 2008).
While Arc has been conventionally studied in the context of its expression and function in neurons, it has recently come into the limelight as a mediator outside of the nervous system. Arc plays an important role in regulating peripheral cellular stress. Exposure of cells to toxic chemicals or oxidative stress launches the cellular defense mechanism known as the heat shock response (HSR), which is characterized by the induction of heat shock proteins (Hsps) (Neef et al., 2011). Diamide, the disulfide crosslinker and sodium arsenite, which is used as a pesticide and causes protein misfolding, induces Arc transiently in thermotolerant HeLa cells where it is rapidly degraded by the ubiquitin proteasome system (UPS) with a half-life of approximately 30 min. Arc then inhibits the activation of heat shock factor 1 (HSF1) leading to a decrease in the expression of Hsp27 and Hsp70, suggesting that Arc is part of a feedback loop to regulate the HSR (Park et al., 2019).
Arc is also a mediator of inflammatory migratory dendritic cell (migDC) migration from the skin to draining lymph nodes for inflammation-mediated T cell activation. Arc is enriched in four different subsets of migDC as well as Langerhans cells in the skin and draining lymph nodes (Tintelnot et al., 2019). The functional sphingosine 1-phosphate receptor antagonist fingolimod (FTY720), which impairs migDC influx into lymph nodes and is used in the treatment of multiple sclerosis, reduces Arc mRNA in migDC and in draining lymph nodes (Ufer et al., 2016). A recent preprint showed a new role for Arc in mediating neuroinflammatory responses in the skin, where Arc increases in dorsal root ganglion neurons in response to the inflammatory mediators neurotrophic growth factor (NGF) and interleukin-6 (IL-6), accumulating in the skin. Arc null mice exhibit an exaggerated inflammatory response that is reversed by addition of extracellular vesicles (exosomes) containing Arc (Barragan-Iglesias et al., 2020).
8. Conclusion
Functions of Arc are tightly related to glutamatergic transmission via NMDARs (Balu and Coyle, 2014), AMPARs (Chowdhury et al., 2006) and metabotropic glutamate receptor 5 (mGluR5s) (Kumar et al., 2012). The serotonergic system also induces Arc through mechanisms that involve both 5-HT1A and 5-HT2A receptors (Pei et al., 2003; Tordera et al., 2003). Additionally, Arc is regulated by the dopaminergic system, particularly in the striatum and the NAc, brain regions that play an important role in reward and reinforcement. Several drugs of abuse alter Arc in these regions suggesting a role for Arc in drug-induced learning, drug-seeking behavior and drug withdrawal (Pathak et al., 2015; Zavala et al., 2008) (Table 1).
Table 1.
List of drugs of abuse, clinical names, general mechanism of action, and effect on Arc.
| Drugs of Abuse | ||||
|---|---|---|---|---|
| Drug | Clinical/Trade Name | General Mechanism | Effect on Arc | Reference |
| alcohol | binds to ACh, 5-HT, GABA, and NMDA receptors; mainly increases effects of GABA | acute exposure increases Arc in central and medial amygdala; withdrawal after long-term exposure decreases Arc in central and medial amygdala | Pandey et al. (2008) | |
| amphetamine | Adderall, Dexedrine | increases release of dopamine, NE, 5-HT from presynaptic terminal | acute administration increases Arc in frontal cortex, visual cortex, CA1 hippocampus, and amygdala; withdrawal increases Arc in insular frontal cortex, visual cortex, entorhinal cortex, CA3 hippocampus, CA1 hippocampus, subiculum, and striatum | (Tan et al., 2000; Pathak et al., 2015) |
| caffeine | adenosine receptor antagonist | increases Arc and Arc in striatonigral and striatopallidal neurons | Dassesse et al. (1999) | |
| cocaine | blocks reuptake of dopamine, NE, 5-HT | increases Arc in striatum, PFC; relapse or cue-induced reinstatement increases Arc in medial PFC, OFC, striatum, NAc, BLA, dHP, anterior cingulate, and motor cortex | (Fosnaugh et al., 1995; Fumagalli et al., 2006, 2009a; Zavala et al., 2008; Hearing et al., 2008a, 2008b, 2010; Ziolkowska et al., 2011) | |
| heroin | opioid receptor agonist | increases Arc in mPFC, NAc, and striatum | (Kuntz et al., 2008; Li et al., 2013; Fanous et al., 2013) | |
| 3,4-methyl enedioxy methamphetamine (MDMA) | increases serotonin release and inhibits serotonin reuptake | sex-dependent changes in Arc protein in cortex and hippocampus | Llorente-Berzal et al. (2013) | |
| methamphetamine (METH) | Desoxyn, Methedrine | induces release of dopamine, NE, 5-HT | increases Arc in parietal, orbital and medial PFC, striatum, and hippocampus | (Kodama et al., 1998; Fujiyama et al., 2003) |
| methylphenidate (MPH) | Ritalin, Ritalin SR, Ritalin LA, Aptensio XR, Concerta, Daytrana, Metadate, Metadate CD, Metadate ER, Methylin, Quillivant, QuilliChew ER | blocks reuptake of NE and dopamine | increases Arc in striatum and cingulate cortex | Chase et al. (2007) |
| morphine | AVINza, Kadian, Kadian ER, Morphabond, MS Contin, Oramorph SR, Roxanol, Roxanol-T | opioid analgesic | treatment increases Arc and Arc in striatum, and increases Arc in the NAc core; withdrawl increases Arc in central and basolateral amygdala | (Marie-Claire et al., 2004; Lucas et al., 2008; Lv et al., 2011) |
| nicotine | blocks nicotonic acetylcholine receptors | increases Arc in cingulate cortex, hippocampus, caudate, central amygdala, and bed nucleus of stria terminalis | Schmitt et al. (2008) | |
| tetrahydrocannabinol (THC) | activates cannabinoid-1 receptors | sex-dependent changes in Arc protein in cortex and hippocampus | Llorente-Berzal et al. (2013) | |
| Drugs of Abuse in Combination with Other Compounds | ||||
| BDNF and alcohol | neurotrophic factor, activates TrkB receptors | infusion of BDNF in central amygdala normalizes Arc levels after alcohol exposure | Pandey et al. (2008) | |
| CTAP and METH | mu-opioid receptor antagonist | pretreatment of CTAP attenuates METH-induced increase in Arc | Horner et al. (2010) | |
| 6-hydroxydopamine (Oxidopamine) and cocaine | dopamine and noradrenergic neurotoxin | pretreatment of oxidopamine before cocaine administration suppresses increase in Arc in the striatum | (Fosnaugh et al., 1995; Fumagalli et al., 2006) | |
| MK-801 and METH/heroin | Dizocilpine (MK-801) | NMDA receptor antagonist | pretreatment of MK-801 before METH or heroin administration abolishes increases in Arc or Arc in certain brain regions | (Kodama et al., 1998; Fujiyama et al., 2003; Li et al., 2013) |
| naloxone and morphine | Narcan, Evzio (naloxone) | opioid antagonist | naloxone-precipitated withdrawal from morphine maintains elevated levels of Arc in the frontal cortex | Ammon et al. (2003) |
| quinpirole and caffeine | dopamine 2 and 3 receptor agonist | pretreatment of quinpirole blocks caffeine-induced increase in Arc and Arc in striatonigral and striatopallidal neurons; direct administration of quinpirole has no change in Arc | (Dassesse et al., 1999; Fumagalli et al., 2009b) | |
| reserpine and cocaine | Serpasil (Reserpine) | blocks reuptake of NE, dopamine, 5-HT | pretreatment of reserpine before cocaine administration suppresses increase in Arc in the striatum | (Fosnaugh et al., 1995; Fumagalli et al., 2006) |
| SCH- 23390 (halobenzazepine) and cocaine/METH/heroin/caffeine | dopamine receptor D1 antagonist | pretreatment of SCH- 23390 before cocaine, METH, heroin, or caffeine administration abolishes increase in Arc or Arc in certain brain regions | (Fosnaugh et al., 1995; Fumagalli et al., 2006; Kodama et al., 1998; Fujiyama et al., 2003; Li et al., 2013; Dassesse et al., 1999) | |
Abbreviations: 5-hydroxytryptamine (5-HT), acetylcholine (ACh), basolateral amygdala (BLA), brain-derived neurotrophic factor (BDNF), dorsal hippocampus (dHP), gamma-aminobutyric acid (GABA), H-D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP), methamphetamine (METH), N-methyl-D-aspartate (NMDA), norepinephrine (NE), nucleus accumbens (NAc), orbitofrontal cortex (OFC), prefrontal cortex (PFC), tropomyosin receptor kinase B (TrkB).
Several drugs used in the management of Major Depressive Disorder (MDD) such as SSRIs and ketamine also affect Arc (Table 2). Repeated exposure to stress is a well-known risk factor for MDD (Wurtman, 2005). Acute and chronic stress are associated with upregulation of Arc and disruptions in glutamatergic transmission, and Arc levels are dysregulated in various models of MDD with depression-like behavior (Li et al., 2015; Penrod et al., 2019). Antidepressants alter Arc mainly in the cortex and striatum. Interestingly, their effect on Arc appears to be somewhat similar to their therapeutic course in humans, where little change is observed with acute treatments and more pronounced effects with a chronic course of treatment (Pei et al., 2003; Ferres-Coy et al., 2013; De Foubert et al., 2004; Alme et al., 2007), unlike the non-selective NMDAR antagonist ketamine, which is characterized by its rapid antidepressant effects, and induces Arc within hours (Li et al., 2010). The Arc gene has also been implicated in schizophrenia (Fromer et al., 2014; Purcell et al., 2014), with altered Arc mRNA detected in the PFC of schizophrenia patients (Guillozet-Bongaarts et al., 2014). Arc disruptions produce schizophrenia-like symptoms in mice through modulation of the dopaminergic system in the PFC and striatum (Manago et al., 2016). Both typical and atypical antipsychotic drugs alter Arc in the striatum, PFC and NAc through their actions on D2 receptors, suggesting that modulation of Arc may underlie some of their therapeutic effects (Robbins et al., 2008; Fumagalli et al., 2009b).
Table 2.
List of psychotropic drugs, clinical names, general mechanism of action, and effect on Arc.
| Psychotropic Drugs | ||||
|---|---|---|---|---|
| Drug | Clinical/Trade Name | General Mechanism | Effect on Arc | Reference |
| agomelatine | Valdoxan, Thymanax, Agoprex, Melitor, Vestin, Alodil, etc. | melatonergic MT1/MT2 receptor agonist and 5-HT2C receptor antagonist | upregulates Arc in hippocampus | Calabrese et al. (2011) |
| amisulpride | Solian | high-affinity dopamine 2/3 receptor blocker | induces lower levels of Arc in medial striatum than haloperidol | de Bartolomeis et al. (2013) |
| clozapine | Clozaril, FazaClo ODT, Versacloz | binds serotonin, dopamine, and GABAB receptors | decreases Arc in thalamus and hypothalamus; decreases Arc in medial PFC and cingulate cortex; increases Arc in NAc shell | (Robbins et al., 2008; Fumagalli et al., 2009b) |
| desipramine | Norpramin | monoamine oxidase inhibitor | chronic administration increases Arc in parietal, frontal, orbital, cingulate cortex, and CA1 hippocampus | ( Pei et al., 2003) |
| 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) | 5-HT2 agonist | increases Arc in cortex and striatum | Pei et al. (2000) | |
| duloxetine | Cymbalta | serotonin-norepinephrine reuptake inhibitor | chronic treatment induces Arc in frontal cortex | Molteni et al. (2008) |
| escitalopram | Cipralex, Lexapro | selective serotonin reuptake inhibitor | chronic treatment of escitalopram along with stimulation of MAPK signaling cascades restores Arc levels in the PFC and hippocampus in Flinders sensitive line rats | Eriksson et al. (2012) |
| haloperidol | Haldol, Haldol Decanoate, Haloperidol LA, Peridol | dopamine receptor D2 antagonist | increases Arc in striatum, Arc and Arc (acute treatment) in NAc, and Arc in caudate nucleus; decreases Arc in frontal cortex | (Fosnaugh et al., 1995; Fumagalli et al., 2006, 2009b; Robbins et al., 2008) |
| ketamine | Ketalar, LidoProfen | non-selective NMDA receptor antagonist, activates mTOR signaling pathway | increases Arc in PFC | Li et al. (2010) |
| L-tryptophan | Tryptan | increase 5-HT synthesis | no change in Arc | Pei et al. (2000) |
| olanzapine | Zyprexa, Zyprexa Relprevv, Zyprexa Zydis | blocks serotonin, dopamine, and muscarinic receptors | increases Arc in striatum, decreases Arc in frontal cortex | Fumagalli et al. (2009b) |
| paroxetine | Paxil, Brisdelle, Paxil CR, Pexeva | serotonin reuptake inhibitor; monoamine oxidase inhibitor | chronic administration increases Arc in parietal, frontal, orbital, cingulate cortex, and CA1 hippocampus | ( Pei et al., 2003) |
| phencyclidine (PCP) | Sernyl, Sernylan | NMDA antagonist | alters Arc in medial PFC, orbitofrontal cortex, and NAc shell | (Thomsen et al., 2010; Nakahara et al., 2000) |
| spiperone | Spiropitan | dopamine receptor D2 antagonist | increases Arc in striatum | (Fosnaugh et al., 1995; Fumagalli et al., 2006) |
| thioridazine | Mellaril | inhibit Arc N-lobe binding, blocks dopamine 1/2 receptors, serotonin receptors, H1 receptors, and alpha-adrenergic receptors | inhibits TARPγ2 binding to Arc N-lobe | Zhang et al. (2015) |
| tranylcypromine | Parnate | monoamine oxidase inhibitor (MAOI) | chronic administration increases Arc in dentate gyrus, acute administration increases Arc in parietal cortex | (Pei et al., 2003) |
| trifluoperazine | Stelazine | inhibit Arc N-lobe binding, blocks dopamine 1/2 receptors | inhibits TARPγ2 binding to Arc N-lobe | Zhang et al. (2015) |
| venlafaxine | Effexor, Effexor XR | monoamine oxidase inhibitor | chronic administration increases Arc in parietal, frontal, orbital, cingulate cortex, and CA1 hippocampus | ( Pei et al., 2003) |
| Psychotropic Drugs in Combination with Other Compounds | ||||
| caffeine and haloperidol | cotreatment with caffeine and haloperidol reduces increase in Arc in striatum caused by haloperidol treatment alone | de Bartolomeis et al. (2018) | ||
| ketanserin and tranylcypromine with L-tryptophan | Sufrexal (ketanserin) | 5-HT2 receptor antagonist | administration of ketanserin attenuates increase in Arc in cortex and striatum from tranylcypromine and L-tryptophan administration | Pei et al. (2000) |
| NDA-299 and paroxetine | 5-HT1A receptor antagonist | administration of NDA-99 with paroxetine increases Arc in caudate putamen, frontal, parietal, and piriform cortices | (Tordera et al., 2003; Castro et al., 2003) | |
| nicotine and haloperidol | cotreatment with nicotine and haloperidol increases Arc in cortex than haloperidol treatment alone | de Bartolomeis et al. (2018) | ||
| p-chlorophenylalanine and tranylcypromine with L-tryptophan | Fenclonine (p-chlorophenylalanine) | tryptophan hydroxylase inhibitor | administration of p-chlorophenylalanine attenuates increase in Arc in cortex and striatum from tranylcypromine and L-tryptophan administration | Pei et al. (2000) |
| tranylcypromine and L-tryptophan | Parnate (tranylcypromine) | monoamine oxidase inhibitor (MAOI) | combined administration of tranylcypromine with L-tryptophan increases Arc in striatum, orbital cortex, frontal cortex, and parietal cortex; decreases Arc in CA1 hippocampus | Pei et al. (2000) |
| WAY 100635 and paroxetine | Nefazodone (WAY 100635) | 5-HT1A receptor antagonist | administration of WAY 100635 with paroxetine increases Arc in caudate putamen, frontal, parietal, and piriform cortices | (Tordera et al., 2003; Castro et al., 2003) |
Abbreviations: 5-hydroxytryptamine (5-HT), gamma-aminobutyric acid (GABA), histamine 1 (H1), mammalian target of rapamycin (mTOR), N-methyl-D-aspartate (NMDA), nucleus accumbens (NAc), prefrontal cortex (PFC), transmembrane AMPA receptor regulatory protein subunit γ 2 (TARPγ2).
Arc is modulated by BDNF which regulates neuronal survival and differentiation (Yin et al., 2002). BDNF has been extensively investigated as a potential therapeutic tool for spinal cord injuries (Awad et al., 2015), neurodegenerative diseases (Sampaio et al., 2017) and stem cell therapy (Pramanik et al., 2017) with unclear results in human studies (Zoladz and Pilc, 2010; Palasz et al., 2020). Furthermore, Arc is implicated in several neurodegenerative diseases including AD, where it is found to be dysregulated by Aβ oligomers (Lacor et al., 2004). Studies show abnormal levels of Arc in response to neuronal activation in various AD mouse models (Kerrigan and Randall, 2013; Morin et al., 2016) and in human postmortem AD brains (Wu et al., 2011). Arc binds to presenilin 1 (PSI), the catalytic subunit of the gamma-secretase complex, and disruption of the Arc-PS1 interaction prevents activity-dependent increases in soluble hAβ40 (Wu et al., 2011). AD has been characterized as a chronic inflammatory state (Kinney et al., 2018). Intriguingly, memantine, an NMDA antagonist widely used to treat symptoms of mild to moderate AD (Folch et al., 2018), reverses the disruptions of behaviorally-induced Arc in chronic inflammation, which appear to be mediated via NMDAR-related mechanisms, perhaps adding another layer to Arc's proposed neuronal functions in AD (Rosi et al., 2005, 2006). Additionally, Arc levels increase in response to inflammatory mediators such as IL-6 and NGF in dorsal root ganglia (Barragan-Iglesias et al., 2020) (Table 3).
Table 3.
List of other compounds, clinical names, general mechanism of action, and effect on Arc.
| Other Compounds | ||||
|---|---|---|---|---|
| Compound | Clinical/Trade Name | General Mechanism | Effect on Arc | Reference |
| AK-7 | protein lysine deacetylase inhibitor | increases Arc by limiting its degradation in dentate gyrus and BDNF-treated primary cortical neuron cultures | Lalonde et al. (2017) | |
| Brain-derived neurotrophic factor (BDNF) | neurotrophic factor, activates TrkB receptors | increases Arc and Arc in dentate gyrus; induces Arc in primary cortical neuronal cultures and hippocampal organotypic slices | (Ying et al., 2002; Rao et al., 2006) | |
| CHIR 98014 (CH98) | GSK3α/β inhibitor | decreases Arc degradation | Gozdz et al. (2017) | |
| 1,3-dipropyl-8 cyclopentylxanthine (DPCPX) | adenosine 1 antagonist | increases Arc and Arc in striatonigral and striatopallidal neurons | Dassesse et al. (1999) | |
| diamide | disulfide crosslinker | increases Arc (observed in HeLa cells) | Park et al. (2019) | |
| eticlopride | dopamine receptor D2 antagonist | increases Arc in striatum | (Fosnaugh et al., 1995; Fumagalli et al., 2006) | |
| FTY720 | Fingolimod | functional sphingosine 1-phosphate receptor antagonist | decreases Arc in migratory dendritic cells | Ufer et al. (2016) |
| interleukin 6 (IL-6) | neuroinflammatory agent | increases Arc in dorsal root ganglion neurons | Barragan-Iglesias et al. (2020) | |
| K252-alpha | tyrosine kinase inhibitor | pretreatment of K252-alpha before BDNF infusion inhibits BDNF-induced increase in Arc in synaptoneurosomes | Yin et al. (2002) | |
| leupeptin | lysosomal inhibitor | leupeptin along with ammonium chloride inhibits autophagy pathway leading to Arc accumulation in hippocampal neurons | Yan et al. (2018) | |
| lipopolysaccharide (LPS) | endotoxin that binds to TLR4 receptor, results in secretion of cytokines | induces Arc in dentate gyrus and CA3 hippocampus; no change in basal Arc or Arc translation/removal | (Rosi et al., 2005, 2006, 2009) | |
| 3-methyladenine (3-MA) | inhibits autophagosome formation | inhibits autophagy pathway leading to Arc accumulation in hippocampal neurons | Yan et al. (2018) | |
| memantine | Namenda, Namenda XR | NMDA receptor antagonist | abolishes increase in Arc from LPS administration | (Rosi et al., 2006, 2009) |
| MG-132 | proteasome inhibitor | decreases Arc degradation | (Mabb et al., 2014; Rao et al., 2006) | |
| MK-801 | Dizocilpine | NMDA receptor antagonist | pretreatment of MK-801 before BDNF infusion inhibits BDNF-induced increase in Arc in synaptoneurosomes | Yin et al. (2002) |
| nerve growth factor (NGF) | regulates cell growth and survival, released by mast cells in periphery | increases Arc in dorsal root ganglion neurons | Barragan-Iglesias et al. (2020) | |
| oxamflatin | protein lysine deacetylase inhibitor | increases Arc by limiting its degradation in BDNF-treated primary cortical neuron cultures | Lalonde et al. (2017) | |
| p-chloroamphetamine | 5-HT releasing agent | increases Arc in cortex and striatum | Pei et al. (2000) | |
| raclopride | dopamine 2 receptor antagonist | increases Arc in striatum, decreases Arc in frontal cortex | Fumagalli et al. (2009b) | |
| Ro25-6981 | NR2B antagonist, activates mTOR pathway | increases Arc in PFC | Li et al. (2010) | |
| S35966A | dual α2-adrenoceptor antagonist and 5-HT-noradrenaline reuptake inhibitor | increases Arc in cortex and hippocampus | Serres et al. (2012) | |
| sodium arsenite | binds to telomeric sequences to induce apoptosis | increases Arc (observed in HeLa cells) | Park et al. (2019) | |
| U0126 | mitogen-activated protein kinase inhibitor | abolishes increase in Arc observed from BDNF infusion | Ying et al. (2002) | |
Abbreviations: 5-hydroxytryptamine (5-HT), brain-derived neurotrophic factor (BDNF), glycogen synthase kinase 3α/β (GSK3α/β), lipopolysaccharide (LPS), mammalian target of rapamycin (mTOR), N-methyl-D-aspartate (NMDA), N-methyl-D-aspartate receptor subtype 2B (NR2B), prefrontal cortex (PFC), toll-like receptor 4 (TLR4), tropomyosin receptor kinase B (TrkB).
Taken together, these studies suggest that reversing abnormalities in Arc expression and function might be a key therapeutic strategy for various disorders that disrupt cognitive functions. Arc is pivotal to memory, learning and cognitive processing where Arc knock-out mouse models display behavioral phenotypes related to schizophrenia (Manago et al., 2016). To this end, Lalonde et al. performed a chemogenomic screen in search for small molecules capable of modulating Arc levels and function through BDNF signaling revealing that the majority of the drugs considered as “Arc suppressers” were drugs with antipsychotic or antidepressant activity, while the majority of “Arc potentiators” had neuroprotective and/or nootropic properties (Lalonde et al., 2017). It remains unclear whether modulating Arc in itself is sufficient to achieve the full therapeutic effects of these drugs. To the extent of our knowledge, only the first-generation anti-psychotics thioridazine and trifluoperazine can directly bind Arc and hold promise for pharmacological use to interfere with Arc binding partners (Zhang et al., 2015). Additionally, while injecting drugs that can modulate Arc into target brain areas is feasible in animal models, this remains a challenge in humans to avoid side-effects that might result from undesired widespread modulation of Arc in the brain. Addressing these challenges is key to the potential of Arc as a therapeutic target.
Funding
This work was supported by a NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation Research Partners Program (P&S Fund Investigator) and a Whitehall Foundation (Grant 2017-05-35) to A.M.M. D.W.Y. was supported by a Georgia State University Neurogenomics 2CI Fellowship. N.S. was supported by the Brains & Behavior summer scholars program and a Goldwater Scholarship.
CRediT authorship contribution statement
Dina W. Yakout: Conceptualization, Writing - original draft, Writing - review & editing. Nitheyaa Shree: Writing - original draft, Writing - review & editing. Angela M. Mabb: Conceptualization, Supervision, Writing - original draft, Writing - review & editing.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
We would like to thank Dr. Marise Parent for providing critical feedback on this manuscript.
References
- Alme M.N., et al. Chronic fluoxetine treatment induces brain region-specific upregulation of genes associated with BDNF-induced long-term potentiation. Neural Plast. 2007;2007:26496. doi: 10.1155/2007/26496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ammon S., et al. Microarray analysis of genes expressed in the frontal cortex of rats chronically treated with morphine and after naloxone precipitated withdrawal. Brain Res Mol Brain Res. 2003;112(1–2):113–125. doi: 10.1016/s0169-328x(03)00057-3. [DOI] [PubMed] [Google Scholar]
- Awad B.I., Carmody M.A., Steinmetz M.P. Potential role of growth factors in the management of spinal cord injury. World Neurosurg. 2015;83(1):120–131. doi: 10.1016/j.wneu.2013.01.042. [DOI] [PubMed] [Google Scholar]
- Balu D.T., Coyle J.T. Chronic D-serine reverses arc expression and partially rescues dendritic abnormalities in a mouse model of NMDA receptor hypofunction. Neurochem. Int. 2014;75:76–78. doi: 10.1016/j.neuint.2014.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barragan-Iglesias P., et al. bioRxiv; 2020. Intercellular Arc Signaling Regulates Vasodilation. 2020.08.13.250209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barylko B., et al. Palmitoylation and membrane binding of Arc/Arg3.1: a potential role in synaptic depression. Biochemistry. 2018;57(5):520–524. doi: 10.1021/acs.biochem.7b00959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beardsley P.M., Shelton K.L. Prime-, stress-, and cue-induced reinstatement of extinguished drug-reinforced responding in rats: cocaine as the prototypical drug of abuse. Curr Protoc Neurosci. 2012:39. doi: 10.1002/0471142301.ns0939s61. (Chapter 9): p. Unit 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berman R.M., et al. Antidepressant effects of ketamine in depressed patients. Biol. Psychiatr. 2000;47(4):351–354. doi: 10.1016/s0006-3223(99)00230-9. [DOI] [PubMed] [Google Scholar]
- Berton O., Nestler E.J. New approaches to antidepressant drug discovery: beyond monoamines. Nat. Rev. Neurosci. 2006;7(2):137–151. doi: 10.1038/nrn1846. [DOI] [PubMed] [Google Scholar]
- Blier P., Ward N.M. Is there a role for 5-HT1A agonists in the treatment of depression? Biol. Psychiatr. 2003;53(3):193–203. doi: 10.1016/s0006-3223(02)01643-8. [DOI] [PubMed] [Google Scholar]
- Boldridge M., et al. Characterization of the C-terminal tail of the Arc protein. PloS One. 2020;15(9) doi: 10.1371/journal.pone.0239870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bossert J.M., et al. The reinstatement model of drug relapse: recent neurobiological findings, emerging research topics, and translational research. Psychopharmacology (Berlin) 2013;229(3):453–476. doi: 10.1007/s00213-013-3120-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bramham C.R., et al. The immediate early gene arc/arg3.1: regulation, mechanisms, and function. J. Neurosci. 2008;28(46):11760–11767. doi: 10.1523/JNEUROSCI.3864-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruins Slot L.A., et al. F15063, a potential antipsychotic with dopamine D(2)/D(3) receptor antagonist and 5-HT(1A) receptor agonist properties: influence on immediate-early gene expression in rat prefrontal cortex and striatum. Eur. J. Pharmacol. 2009;620(1–3):27–35. doi: 10.1016/j.ejphar.2009.08.019. [DOI] [PubMed] [Google Scholar]
- Calabrese F., et al. Modulation of neuroplastic molecules in selected brain regions after chronic administration of the novel antidepressant agomelatine. Psychopharmacology (Berlin) 2011;215(2):267–275. doi: 10.1007/s00213-010-2129-8. [DOI] [PubMed] [Google Scholar]
- Carmichael R.E., Henley J.M. Transcriptional and post-translational regulation of Arc in synaptic plasticity. Semin. Cell Dev. Biol. 2018;77:3–9. doi: 10.1016/j.semcdb.2017.09.007. [DOI] [PubMed] [Google Scholar]
- Castro E., et al. Use of Arc expression as a molecular marker of increased postsynaptic 5-HT function after SSRI/5-HT1A receptor antagonist co-administration. J. Neurochem. 2003;85(6):1480–1487. doi: 10.1046/j.1471-4159.2003.01782.x. [DOI] [PubMed] [Google Scholar]
- Chase T., et al. Methylphenidate regulates activity regulated cytoskeletal associated but not brain-derived neurotrophic factor gene expression in the developing rat striatum. Neuroscience. 2007;144(3):969–984. doi: 10.1016/j.neuroscience.2006.10.035. [DOI] [PubMed] [Google Scholar]
- Chowdhury S., et al. Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron. 2006;52(3):445–459. doi: 10.1016/j.neuron.2006.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Craig T.J., et al. Homeostatic synaptic scaling is regulated by protein SUMOylation. J. Biol. Chem. 2012;287(27):22781–22788. doi: 10.1074/jbc.M112.356337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunningham J.I., et al. MDMA pretreatment leads to mild chronic unpredictable stress-induced impairments in spatial learning. Behav. Neurosci. 2009;123(5):1076–1084. doi: 10.1037/a0016716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dassesse D., et al. Caffeine-mediated induction of c-fos, zif-268 and arc expression through A1 receptors in the striatum: different interactions with the dopaminergic system. Eur. J. Neurosci. 1999;11(9):3101–3114. doi: 10.1046/j.1460-9568.1999.00725.x. [DOI] [PubMed] [Google Scholar]
- de Bartolomeis A., et al. Imaging brain gene expression profiles by antipsychotics: region-specific action of amisulpride on postsynaptic density transcripts compared to haloperidol. Eur. Neuropsychopharmacol. 2013;23(11):1516–1529. doi: 10.1016/j.euroneuro.2012.11.014. [DOI] [PubMed] [Google Scholar]
- de Bartolomeis A., et al. Nicotine and caffeine modulate haloperidol-induced changes in postsynaptic density transcripts expression: translational insights in psychosis therapy and treatment resistance. Eur. Neuropsychopharmacol. 2018;28(4):538–559. doi: 10.1016/j.euroneuro.2018.01.006. [DOI] [PubMed] [Google Scholar]
- De Foubert G., et al. Fluoxetine-induced change in rat brain expression of brain-derived neurotrophic factor varies depending on length of treatment. Neuroscience. 2004;128(3):597–604. doi: 10.1016/j.neuroscience.2004.06.054. [DOI] [PubMed] [Google Scholar]
- Dynes J.L., Steward O. Dynamics of bidirectional transport of Arc mRNA in neuronal dendrites. J. Comp. Neurol. 2007;500(3):433–447. doi: 10.1002/cne.21189. [DOI] [PubMed] [Google Scholar]
- Dynes J.L., Steward O. Arc mRNA docks precisely at the base of individual dendritic spines indicating the existence of a specialized microdomain for synapse-specific mRNA translation. J. Comp. Neurol. 2012;520(14):3105–3119. doi: 10.1002/cne.23073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dyrvig M., et al. Epigenetic regulation of Arc and c-Fos in the hippocampus after acute electroconvulsive stimulation in the rat. Brain Res. Bull. 2012;88(5):507–513. doi: 10.1016/j.brainresbull.2012.05.004. [DOI] [PubMed] [Google Scholar]
- Eriksen M.S., et al. Arc self-association and formation of virus-like capsids are mediated by an N-terminal helical coil motif. FEBS J. 2020 doi: 10.1111/febs.15618. [DOI] [PubMed] [Google Scholar]
- Eriksson T.M., et al. Emotional memory impairments in a genetic rat model of depression: involvement of 5-HT/MEK/Arc signaling in restoration. Mol. Psychiatr. 2012;17(2):173–184. doi: 10.1038/mp.2010.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fanous S., et al. Unique gene alterations are induced in FACS-purified Fos-positive neurons activated during cue-induced relapse to heroin seeking. J. Neurochem. 2013;124(1):100–108. doi: 10.1111/jnc.12074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farris S., et al. Selective localization of arc mRNA in dendrites involves activity- and translation-dependent mRNA degradation. J. Neurosci. 2014;34(13):4481–4493. doi: 10.1523/JNEUROSCI.4944-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferres-Coy A., et al. RNAi-mediated serotonin transporter suppression rapidly increases serotonergic neurotransmission and hippocampal neurogenesis. Transl. Psychiatry. 2013;3:e211. doi: 10.1038/tp.2012.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Folch J., et al. Memantine for the treatment of dementia: a review on its current and future applications. J Alzheimers Dis. 2018;62(3):1223–1240. doi: 10.3233/JAD-170672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fosnaugh J.S., et al. Activation of arc, a putative "effector" immediate early gene, by cocaine in rat brain. J. Neurochem. 1995;64(5):2377–2380. doi: 10.1046/j.1471-4159.1995.64052377.x. [DOI] [PubMed] [Google Scholar]
- Fromer M., et al. De novo mutations in schizophrenia implicate synaptic networks. Nature. 2014;506(7487):179–184. doi: 10.1038/nature12929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fujiyama K., et al. Differential regulation by stimulants of neocortical expression of mrt1, arc, and homer1a mRNA in the rats treated with repeated methamphetamine. Synapse. 2003;49(3):143–149. doi: 10.1002/syn.10220. [DOI] [PubMed] [Google Scholar]
- Fumagalli F., et al. Corticostriatal up-regulation of activity-regulated cytoskeletal-associated protein expression after repeated exposure to cocaine. Mol. Pharmacol. 2006;70(5):1726–1734. doi: 10.1124/mol.106.026302. [DOI] [PubMed] [Google Scholar]
- Fumagalli F., et al. Single session of cocaine intravenous self-administration shapes goal-oriented behaviours and up-regulates Arc mRNA levels in rat medial prefrontal cortex. Int. J. Neuropsychopharmacol. 2009;12(3):423–429. doi: 10.1017/S1461145708009681. [DOI] [PubMed] [Google Scholar]
- Fumagalli F., et al. Antipsychotic drugs modulate Arc expression in the rat brain. Eur. Neuropsychopharmacol. 2009;19(2):109–115. doi: 10.1016/j.euroneuro.2008.09.001. [DOI] [PubMed] [Google Scholar]
- Gallo F.T., et al. Immediate early genes, memory and psychiatric disorders: focus on c-fos, Egr1 and Arc. Front. Behav. Neurosci. 2018;12:79. doi: 10.3389/fnbeh.2018.00079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gozdz A., et al. GSK3alpha and GSK3beta phosphorylate Arc and regulate its degradation. Front. Mol. Neurosci. 2017;10:192. doi: 10.3389/fnmol.2017.00192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greer P.L., et al. The Angelman Syndrome protein Ube3A regulates synapse development by ubiquitinating arc. Cell. 2010;140(5):704–716. doi: 10.1016/j.cell.2010.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grunder G., Hippius H., Carlsson A. The 'atypicality' of antipsychotics: a concept re-examined and re-defined. Nat. Rev. Drug Discov. 2009;8(3):197–202. doi: 10.1038/nrd2806. [DOI] [PubMed] [Google Scholar]
- Guillozet-Bongaarts A.L., et al. Altered gene expression in the dorsolateral prefrontal cortex of individuals with schizophrenia. Mol. Psychiatr. 2014;19(4):478–485. doi: 10.1038/mp.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guzowski J.F., et al. Environment-specific expression of the immediate-early gene Arc in hippocampal neuronal ensembles. Nat. Neurosci. 1999;2(12):1120–1124. doi: 10.1038/16046. [DOI] [PubMed] [Google Scholar]
- Guzowski J, et al. Inhibition of activity-dependent arc protein expression in the rat hippocampus impairs the maintenance of long-term potentiation and the consolidation of long-term memory. Journal of Neuroscience. 2000:3993–4001. doi: 10.1523/JNEUROSCI.20-11-03993.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haapasalo A., et al. Regulation of TRKB surface expression by brain-derived neurotrophic factor and truncated TRKB isoforms. J. Biol. Chem. 2002;277(45):43160–43167. doi: 10.1074/jbc.M205202200. [DOI] [PubMed] [Google Scholar]
- Hearing M.C., et al. Relapse to cocaine seeking increases activity-regulated gene expression differentially in the prefrontal cortex of abstinent rats. Psychopharmacology (Berlin) 2008;198(1):77–91. doi: 10.1007/s00213-008-1090-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hearing M.C., See R.E., McGinty J.F. Relapse to cocaine-seeking increases activity-regulated gene expression differentially in the striatum and cerebral cortex of rats following short or long periods of abstinence. Brain Struct. Funct. 2008;213(1–2):215–227. doi: 10.1007/s00429-008-0182-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hearing M.C., et al. Context-driven cocaine-seeking in abstinent rats increases activity-regulated gene expression in the basolateral amygdala and dorsal hippocampus differentially following short and long periods of abstinence. Neuroscience. 2010;170(2):570–579. doi: 10.1016/j.neuroscience.2010.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hearing M.C., Schwendt M., McGinty J.F. Suppression of activity-regulated cytoskeleton-associated gene expression in the dorsal striatum attenuates extinction of cocaine-seeking. Int. J. Neuropsychopharmacol. 2011;14(6):784–795. doi: 10.1017/S1461145710001173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horner K.A., Noble E.S., Gilbert Y.E. Methamphetamine-induced stereotypy correlates negatively with patch-enhanced prodynorphin and arc mRNA expression in the rat caudate putamen: the role of mu opioid receptor activation. Pharmacol. Biochem. Behav. 2010;95(4):410–421. doi: 10.1016/j.pbb.2010.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson A.C., Nicoll R.A. Stargazing from a new vantage--TARP modulation of AMPA receptor pharmacology. J. Physiol. 2011;589(Pt 24):5909–5910. doi: 10.1113/jphysiol.2011.223495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawashima T., et al. Synaptic activity-responsive element in the Arc/Arg3.1 promoter essential for synapse-to-nucleus signaling in activated neurons. Proc. Natl. Acad. Sci. U. S. A. 2009;106(1):316–321. doi: 10.1073/pnas.0806518106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawashima T., et al. Functional labeling of neurons and their projections using the synthetic activity-dependent promoter E-SARE. Nat. Methods. 2013;10(9):889–895. doi: 10.1038/nmeth.2559. [DOI] [PubMed] [Google Scholar]
- Kerrigan T.L., Randall A.D. A new player in the "synaptopathy" of Alzheimer's disease - arc/arg 3.1. Front. Neurol. 2013;4:9. doi: 10.3389/fneur.2013.00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kinney J.W., et al. Inflammation as a central mechanism in Alzheimer's disease. Alzheimers Dement (N Y) 2018;4:575–590. doi: 10.1016/j.trci.2018.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knoll B., Nordheim A. Functional versatility of transcription factors in the nervous system: the SRF paradigm. Trends Neurosci. 2009;32(8):432–442. doi: 10.1016/j.tins.2009.05.004. [DOI] [PubMed] [Google Scholar]
- Kodama M., et al. A robust increase in expression of arc gene, an effector immediate early gene, in the rat brain after acute and chronic methamphetamine administration. Brain Res. 1998;796(1–2):273–283. doi: 10.1016/s0006-8993(98)00349-7. [DOI] [PubMed] [Google Scholar]
- Kuipers S.D., et al. BDNF-induced LTP is associated with rapid Arc/Arg3.1-dependent enhancement in adult hippocampal neurogenesis. Sci. Rep. 2016;6:21222. doi: 10.1038/srep21222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar V., et al. Activation of intracellular metabotropic glutamate receptor 5 in striatal neurons leads to up-regulation of genes associated with sustained synaptic transmission including Arc/Arg3.1 protein. J. Biol. Chem. 2012;287(8):5412–5425. doi: 10.1074/jbc.M111.301366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuntz K.L., et al. Heroin self-administration: II. CNS gene expression following withdrawal and cue-induced drug-seeking behavior. Pharmacol. Biochem. Behav. 2008;90(3):349–356. doi: 10.1016/j.pbb.2008.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lacor P.N., et al. Synaptic targeting by Alzheimer's-related amyloid beta oligomers. J. Neurosci. 2004;24(45):10191–10200. doi: 10.1523/JNEUROSCI.3432-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lalonde J., et al. Chemogenomic analysis reveals key role for lysine acetylation in regulating Arc stability. Nat. Commun. 2017;8(1):1659. doi: 10.1038/s41467-017-01750-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li N., et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959–964. doi: 10.1126/science.1190287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M., et al. Differential expression of Arc in the mesocorticolimbic system is involved in drug and natural rewarding behavior in rats. Acta Pharmacol. Sin. 2013;34(8):1013–1024. doi: 10.1038/aps.2013.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y., et al. A critical evaluation of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1)'s putative role in regulating dendritic plasticity, cognitive processes, and mood in animal models of depression. Front. Neurosci. 2015;9:279. doi: 10.3389/fnins.2015.00279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Link W., et al. Somatodendritic expression of an immediate early gene is regulated by synaptic activity. Proc. Natl. Acad. Sci. U. S. A. 1995;92(12):5734–5738. doi: 10.1073/pnas.92.12.5734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Llorente-Berzal A., et al. Sex-dependent psychoneuroendocrine effects of THC and MDMA in an animal model of adolescent drug consumption. PloS One. 2013;8(11) doi: 10.1371/journal.pone.0078386. e78386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lucas M., et al. Reactivity and plasticity in the amygdala nuclei during opiate withdrawal conditioning: differential expression of c-fos and arc immediate early genes. Neuroscience. 2008;154(3):1021–1033. doi: 10.1016/j.neuroscience.2008.04.006. [DOI] [PubMed] [Google Scholar]
- Lv X.F., et al. Expression of activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) in the nucleus accumbens is critical for the acquisition, expression and reinstatement of morphine-induced conditioned place preference. Behav. Brain Res. 2011;223(1):182–191. doi: 10.1016/j.bbr.2011.04.029. [DOI] [PubMed] [Google Scholar]
- Lyford G.L., et al. Arc, a growth factor and activity-regulated gene, encodes a novel cytoskeleton-associated protein that is enriched in neuronal dendrites. Neuron. 1995;14(2):433–445. doi: 10.1016/0896-6273(95)90299-6. [DOI] [PubMed] [Google Scholar]
- Mabb A.M., Ehlers M.D. Arc ubiquitination in synaptic plasticity. Semin. Cell Dev. Biol. 2018;77:10–16. doi: 10.1016/j.semcdb.2017.09.009. [DOI] [PubMed] [Google Scholar]
- Mabb A.M., et al. Triad3A regulates synaptic strength by ubiquitination of Arc. Neuron. 2014;82(6):1299–1316. doi: 10.1016/j.neuron.2014.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Machado-Vieira R., et al. The timing of antidepressant effects: a comparison of diverse pharmacological and somatic treatments. Pharmaceuticals. 2010;3(1):19–41. doi: 10.3390/ph3010019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malone D.T., Hill M.N., Rubino T. Adolescent cannabis use and psychosis: epidemiology and neurodevelopmental models. Br. J. Pharmacol. 2010;160(3):511–522. doi: 10.1111/j.1476-5381.2010.00721.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mamelak M. An amphetamine model of manic depressive illness. Int. Pharmacopsychiatr. 1978;13(4):193–208. doi: 10.1159/000468341. [DOI] [PubMed] [Google Scholar]
- Manago F., et al. Genetic disruption of Arc/Arg3.1 in mice causes alterations in dopamine and neurobehavioral phenotypes related to schizophrenia. Cell Rep. 2016;16(8):2116–2128. doi: 10.1016/j.celrep.2016.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marie-Claire C., et al. Cytoskeletal genes regulation by chronic morphine treatment in rat striatum. Neuropsychopharmacology. 2004;29(12):2208–2215. doi: 10.1038/sj.npp.1300513. [DOI] [PubMed] [Google Scholar]
- Messaoudi E., et al. Sustained Arc/Arg3.1 synthesis controls long-term potentiation consolidation through regulation of local actin polymerization in the dentate gyrus in vivo. J. Neurosci. 2007;27(39):10445–10455. doi: 10.1523/JNEUROSCI.2883-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molteni R., et al. Basal and stress-induced modulation of activity-regulated cytoskeletal associated protein (Arc) in the rat brain following duloxetine treatment. Psychopharmacology (Berlin) 2008;201(2):285–292. doi: 10.1007/s00213-008-1276-7. [DOI] [PubMed] [Google Scholar]
- Morin J.P., et al. Decreased levels of NMDA but not AMPA receptors in the lipid-raft fraction of 3xTg-AD model of Alzheimer's disease: relation to Arc/Arg3.1 protein expression. Neurochem. Int. 2016;100:159–163. doi: 10.1016/j.neuint.2016.09.013. [DOI] [PubMed] [Google Scholar]
- Myrum C., et al. Arc is a flexible modular protein capable of reversible self-oligomerization. Biochem. J. 2015;468(1):145–158. doi: 10.1042/BJ20141446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair R.R., et al. Dynamic arc SUMOylation and selective interaction with F-Actin-Binding protein drebrin A in LTP consolidation in vivo. Front. Synaptic Neurosci. 2017;9:8. doi: 10.3389/fnsyn.2017.00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakahara T., et al. Effect of atypical antipsychotics on phencyclidine-induced expression of arc in rat brain. Neuroreport. 2000;11(3):551–555. doi: 10.1097/00001756-200002280-00025. [DOI] [PubMed] [Google Scholar]
- Neef D.W., Jaeger A.M., Thiele D.J. Heat shock transcription factor 1 as a therapeutic target in neurodegenerative diseases. Nat. Rev. Drug Discov. 2011;10(12):930–944. doi: 10.1038/nrd3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nielsen L.D., et al. The capsid domain of Arc changes its oligomerization propensity through direct interaction with the NMDA receptor. Structure. 2019;27(7):1071–1081. doi: 10.1016/j.str.2019.04.001. e5. [DOI] [PubMed] [Google Scholar]
- Nikolaienko O., et al. Stimulus-evoked ERK-dependent phosphorylation of activity-regulated cytoskeleton-associated protein (Arc) regulates its neuronal subcellular localization. Neuroscience. 2017;360:68–80. doi: 10.1016/j.neuroscience.2017.07.026. [DOI] [PubMed] [Google Scholar]
- Okuno H., et al. Inverse synaptic tagging of inactive synapses via dynamic interaction of Arc/Arg3.1 with CaMKIIbeta. Cell. 2012;149(4):886–898. doi: 10.1016/j.cell.2012.02.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palasz E., et al. BDNF as a promising therapeutic agent in Parkinson's disease. Int. J. Mol. Sci. 2020;21(3) doi: 10.3390/ijms21031170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pandey S.C., et al. Effector immediate-early gene arc in the amygdala plays a critical role in alcoholism. J. Neurosci. 2008;28(10):2589–2600. doi: 10.1523/JNEUROSCI.4752-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park S., et al. Elongation factor 2 and fragile X mental retardation protein control the dynamic translation of Arc/Arg3.1 essential for mGluR-LTD. Neuron. 2008;59(1):70–83. doi: 10.1016/j.neuron.2008.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park A.Y., et al. Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is transiently expressed after heat shock stress and suppresses heat shock factor 1. Sci. Rep. 2019;9(1):2592. doi: 10.1038/s41598-019-39292-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pastuzyn E.D., et al. The neuronal gene arc encodes a repurposed retrotransposon Gag protein that mediates intercellular RNA transfer. Cell. 2018;173(1):275. doi: 10.1016/j.cell.2018.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pathak G., et al. Amphetamine sensitization in mice is sufficient to produce both manic- and depressive-related behaviors as well as changes in the functional connectivity of corticolimbic structures. Neuropharmacology. 2015;95:434–447. doi: 10.1016/j.neuropharm.2015.04.026. [DOI] [PubMed] [Google Scholar]
- Pei Q., et al. Serotonergic regulation of mRNA expression of Arc, an immediate early gene selectively localized at neuronal dendrites. Neuropharmacology. 2000;39(3):463–470. doi: 10.1016/s0028-3908(99)00148-3. [DOI] [PubMed] [Google Scholar]
- Pei Q., et al. Antidepressant drug treatment induces Arc gene expression in the rat brain. Neuroscience. 2003;121(4):975–982. doi: 10.1016/s0306-4522(03)00504-9. [DOI] [PubMed] [Google Scholar]
- Penrod R.D., et al. Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) regulates anxiety- and novelty-related behaviors. Gene Brain Behav. 2019;18(7) doi: 10.1111/gbb.12561. e12561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Penrod R.D., et al. The activity-regulated cytoskeleton-associated protein, Arc/Arg3.1, influences mouse cocaine self-administration. Pharmacol. Biochem. Behav. 2020;188:172818. doi: 10.1016/j.pbb.2019.172818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pintchovski S.A., et al. The serum response factor and a putative novel transcription factor regulate expression of the immediate-early gene Arc/Arg3.1 in neurons. J. Neurosci. 2009;29(5):1525–1537. doi: 10.1523/JNEUROSCI.5575-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plath N., et al. Arc/Arg3.1 is essential for the consolidation of synaptic plasticity and memories. Neuron. 2006;52(3):437–444. doi: 10.1016/j.neuron.2006.08.024. [DOI] [PubMed] [Google Scholar]
- Pramanik S., Sulistio Y.A., Heese K. Neurotrophin signaling and stem cells-implications for neurodegenerative diseases and stem cell therapy. Mol. Neurobiol. 2017;54(9):7401–7459. doi: 10.1007/s12035-016-0214-7. [DOI] [PubMed] [Google Scholar]
- Purcell S.M., et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506(7487):185–190. doi: 10.1038/nature12975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramirez-Amaya V., et al. Spatial exploration-induced Arc mRNA and protein expression: evidence for selective, network-specific reactivation. J. Neurosci. 2005;25(7):1761–1768. doi: 10.1523/JNEUROSCI.4342-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rampino A., et al. Antipsychotic drug responsiveness and dopamine receptor signaling; old players and new prospects. Front. Psychiatr. 2018;9:702. doi: 10.3389/fpsyt.2018.00702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rao V.R., et al. AMPA receptors regulate transcription of the plasticity-related immediate-early gene Arc. Nat. Neurosci. 2006;9(7):887–895. doi: 10.1038/nn1708. [DOI] [PubMed] [Google Scholar]
- Robbins M.J., et al. Differential expression of IEG mRNA in rat brain following acute treatment with clozapine or haloperidol: a semi-quantitative RT-PCR study. J. Psychopharmacol. 2008;22(5):536–542. doi: 10.1177/0269881107081521. [DOI] [PubMed] [Google Scholar]
- Rodriguez de Fonseca F., et al. Presence of cannabinoid binding sites in the brain from early postnatal ages. Neuroreport. 1993;4(2):135–138. doi: 10.1097/00001756-199302000-00005. [DOI] [PubMed] [Google Scholar]
- Rosi S. Neuroinflammation and the plasticity-related immediate-early gene Arc. Brain Behav. Immun. 2011;25(Suppl. 1):S39–S49. doi: 10.1016/j.bbi.2011.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosi S., et al. Neuroinflammation alters the hippocampal pattern of behaviorally induced Arc expression. J. Neurosci. 2005;25(3):723–731. doi: 10.1523/JNEUROSCI.4469-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosi S., et al. Memantine protects against LPS-induced neuroinflammation, restores behaviorally-induced gene expression and spatial learning in the rat. Neuroscience. 2006;142(4):1303–1315. doi: 10.1016/j.neuroscience.2006.08.017. [DOI] [PubMed] [Google Scholar]
- Rosi S., et al. Cranial irradiation alters the behaviorally induced immediate-early gene arc (activity-regulated cytoskeleton-associated protein) Canc. Res. 2008;68(23):9763–9770. doi: 10.1158/0008-5472.CAN-08-1861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosi S., et al. Accuracy of hippocampal network activity is disrupted by neuroinflammation: rescue by memantine. Brain. 2009;132(Pt 9):2464–2477. doi: 10.1093/brain/awp148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saha R.N., et al. Rapid activity-induced transcription of Arc and other IEGs relies on poised RNA polymerase II. Nat. Neurosci. 2011;14(7):848–856. doi: 10.1038/nn.2839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampaio T.B., et al. Neurotrophic factors in Alzheimer's and Parkinson's diseases: implications for pathogenesis and therapy. Neural Regen Res. 2017;12(4):549–557. doi: 10.4103/1673-5374.205084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scharfman H., et al. Increased neurogenesis and the ectopic granule cells after intrahippocampal BDNF infusion in adult rats. Exp. Neurol. 2005;192(2):348–356. doi: 10.1016/j.expneurol.2004.11.016. [DOI] [PubMed] [Google Scholar]
- Schmitt H.F., et al. Acute nicotine activates c-fos and activity-regulated cytoskeletal associated protein mRNA expression in limbic brain areas involved in the central stress-response in rat pups during a period of hypo-responsiveness to stress. Neuroscience. 2008;157(2):349–359. doi: 10.1016/j.neuroscience.2008.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serres F., Millan M.J., Sharp T. Molecular adaptation to chronic antidepressant treatment: evidence for a more rapid response to the novel alpha(2)-adrenoceptor antagonist/5-HT-noradrenaline reuptake inhibitor (SNRI), S35966, compared to the SNRI, venlafaxine. Int. J. Neuropsychopharmacol. 2012;15(5):617–629. doi: 10.1017/S1461145711000733. [DOI] [PubMed] [Google Scholar]
- Shepherd J.D., et al. Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron. 2006;52(3):475–484. doi: 10.1016/j.neuron.2006.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith-Hicks C., et al. SRF binding to SRE 6.9 in the Arc promoter is essential for LTD in cultured Purkinje cells. Nat. Neurosci. 2010;13(9):1082–1089. doi: 10.1038/nn.2611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steward O., et al. Synaptic activation causes the mRNA for the IEG Arc to localize selectively near activated postsynaptic sites on dendrites. Neuron. 1998;21(4):741–751. doi: 10.1016/s0896-6273(00)80591-7. [DOI] [PubMed] [Google Scholar]
- Tan A., et al. The activity-regulated cytoskeletal-associated protein arc is expressed in different striosome-matrix patterns following exposure to amphetamine and cocaine. J. Neurochem. 2000;74(5):2074–2078. doi: 10.1046/j.1471-4159.2000.0742074.x. [DOI] [PubMed] [Google Scholar]
- Thomsen M.S., Hansen H.H., Mikkelsen J.D. Opposite effect of phencyclidine on activity-regulated cytoskeleton-associated protein (Arc) in juvenile and adult limbic rat brain regions. Neurochem. Int. 2010;56(2):270–275. doi: 10.1016/j.neuint.2009.10.011. [DOI] [PubMed] [Google Scholar]
- Tintelnot J., et al. Arc/Arg3.1 defines dendritic cells and Langerhans cells with superior migratory ability independent of phenotype and ontogeny in mice. Eur. J. Immunol. 2019;49(5):724–736. doi: 10.1002/eji.201847797. [DOI] [PubMed] [Google Scholar]
- Tordera R., et al. Effect of different 5-HT1A receptor antagonists in combination with paroxetine on expression of the immediate-early gene Arc in rat brain. Neuropharmacology. 2003;44(7):893–902. doi: 10.1016/s0028-3908(03)00096-0. [DOI] [PubMed] [Google Scholar]
- Ufer F., et al. Arc/Arg3.1 governs inflammatory dendritic cell migration from the skin and thereby controls T cell activation. Sci Immunol. 2016;1(3):eaaf8665. doi: 10.1126/sciimmunol.aaf8665. [DOI] [PubMed] [Google Scholar]
- Vassilev P., et al. Distinct populations of neurons activated by heroin and cocaine in the striatum as assessed by catFISH. eNeuro. 2020;7(1) doi: 10.1523/ENEURO.0394-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wall M.J., et al. The temporal dynamics of Arc expression regulate cognitive flexibility. Neuron. 2018;98(6):1124–1132. doi: 10.1016/j.neuron.2018.05.012. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H., et al. Metabotropic glutamate receptors induce a form of LTP controlled by translation and Arc signaling in the Hippocampus. J. Neurosci. 2016;36(5):1723–1729. doi: 10.1523/JNEUROSCI.0878-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waung M.W., et al. Rapid translation of Arc/Arg3.1 selectively mediates mGluR-dependent LTD through persistent increases in AMPAR endocytosis rate. Neuron. 2008;59(1):84–97. doi: 10.1016/j.neuron.2008.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilkerson J.R., Albanesi J.P., Huber K.M. Roles for Arc in metabotropic glutamate receptor-dependent LTD and synapse elimination: implications in health and disease. Semin. Cell Dev. Biol. 2018;77:51–62. doi: 10.1016/j.semcdb.2017.09.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams J.M., Gandhi K.K. Use of caffeine and nicotine in people with schizophrenia. Curr. Drug Abuse Rev. 2008;1(2):155–161. doi: 10.2174/1874473710801020155. [DOI] [PubMed] [Google Scholar]
- Wu J., et al. Arc/Arg3.1 regulates an endosomal pathway essential for activity-dependent beta-amyloid generation. Cell. 2011;147(3):615–628. doi: 10.1016/j.cell.2011.09.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wurtman R.J. Genes, stress, and depression. Metabolism. 2005;54(5 Suppl. 1):16–19. doi: 10.1016/j.metabol.2005.01.007. [DOI] [PubMed] [Google Scholar]
- Yan J., et al. Activation of autophagy rescues synaptic and cognitive deficits in fragile X mice. Proc. Natl. Acad. Sci. U. S. A. 2018;115(41):E9707–E9716. doi: 10.1073/pnas.1808247115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin Y., Edelman G.M., Vanderklish P.W. The brain-derived neurotrophic factor enhances synthesis of Arc in synaptoneurosomes. Proc. Natl. Acad. Sci. U. S. A. 2002;99(4):2368–2373. doi: 10.1073/pnas.042693699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ying S.W., et al. Brain-derived neurotrophic factor induces long-term potentiation in intact adult hippocampus: requirement for ERK activation coupled to CREB and upregulation of Arc synthesis. J. Neurosci. 2002;22(5):1532–1540. doi: 10.1523/JNEUROSCI.22-05-01532.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zavala A.R., et al. Upregulation of Arc mRNA expression in the prefrontal cortex following cue-induced reinstatement of extinguished cocaine-seeking behavior. Synapse. 2008;62(6):421–431. doi: 10.1002/syn.20502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H., Bramham C.R. Arc/Arg3.1 function in long-term synaptic plasticity: emerging mechanisms and unresolved issues. Eur. J. Neurosci. 2020 doi: 10.1111/ejn.14958. [DOI] [PubMed] [Google Scholar]
- Zhang W., et al. Structural basis of arc binding to synaptic proteins: implications for cognitive disease. Neuron. 2015;86(2):490–500. doi: 10.1016/j.neuron.2015.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W., et al. Arc oligomerization is regulated by CaMKII phosphorylation of the GAG domain: an essential mechanism for plasticity and memory formation. Mol. Cell. 2019;75(1):13–25. doi: 10.1016/j.molcel.2019.05.004. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziolkowska B., et al. Regulation of the immediate-early genes arc and zif268 in a mouse operant model of cocaine seeking reinstatement. J. Neural. Transm. 2011;118(6):877–887. doi: 10.1007/s00702-011-0583-z. [DOI] [PubMed] [Google Scholar]
- Zoladz J.A., Pilc A. The effect of physical activity on the brain derived neurotrophic factor: from animal to human studies. J. Physiol. Pharmacol. 2010;61(5):533–541. [PubMed] [Google Scholar]

