Next Article in Journal
Maintenance of Ligament Homeostasis of Spheroid-Colonized Embroidered and Functionalized Scaffolds after 3D Stretch
Next Article in Special Issue
Exercise, but Not Metformin Prevents Loss of Muscle Function Due to Doxorubicin in Mice Using an In Situ Method
Previous Article in Journal
Targeting DNA Damage Response and Repair to Enhance Therapeutic Index in Cisplatin-Based Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exercise Pills for Drug Addiction: Forced Moderate Endurance Exercise Inhibits Methamphetamine-Induced Hyperactivity through the Striatal Glutamatergic Signaling Pathway in Male Sprague Dawley Rats

College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(15), 8203; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22158203
Submission received: 4 June 2021 / Revised: 26 July 2021 / Accepted: 28 July 2021 / Published: 30 July 2021
(This article belongs to the Special Issue Molecular and Cellular Exercise Physiology in Metabolism)

Abstract

:
Physical exercise reduces the extent, duration, and frequency of drug use in drug addicts during the drug initiation phase, as well as during prolonged addiction, withdrawal, and recurrence. However, information about exercise-induced neurobiological changes is limited. This study aimed to investigate the effects of forced moderate endurance exercise training on methamphetamine (METH)-induced behavior and the associated neurobiological changes. Male Sprague Dawley rats were subjected to the administration of METH (1 mg/kg/day, i.p.) and/or forced moderate endurance exercise (treadmill running, 21 m/min, 60 min/day) for 2 weeks. Over the two weeks, endurance exercise training significantly reduced METH-induced hyperactivity. METH and/or exercise treatment increased striatal dopamine (DA) levels, decreased p(Thr308)-Akt expression, and increased p(Tyr216)-GSK-3β expression. However, the phosphorylation levels of Ser9-GSK-3β were significantly increased in the exercise group. METH administration significantly increased the expression of NMDAr1, CaMKK2, MAPKs, and PP1 in the striatum, and exercise treatment significantly decreased the expression of these molecules. Therefore, it is apparent that endurance exercise inhibited the METH-induced hyperactivity due to the decrease in GSK-3β activation by the regulation of the striatal glutamate signaling pathway.

1. Introduction

Methamphetamine (METH) is an amphetamine-type stimulant and is one of the most commonly abused drugs in the world [1]. METH causes short-term symptoms related to the sympathetic response of the autonomic nervous system (tachycardia, tachypnea, hypertension, dilatation of the pupils, hyperthermia, reduced fatigue), and increases euphoria, attention, energy, and libido while decreasing appetite [2,3,4,5]. However, repeated METH use can disrupt the central nervous system (CNS), causing neuropsychiatric changes, and in severe cases, death [6,7,8,9]. As drug abuse increases worldwide, deaths from METH misuse are also on the rise [10,11,12]. However, despite the increasing incidence of drug use disorders, there is no effective way to treat addiction [4]. To date, drug therapy, cognitive behavioral therapy, and social culture therapy have been used to treat drug addiction. However, these treatments have limitations. Drug use disorders such as methamphetamine addiction are characterized by consistent neurobiological changes at different stages of addiction [13]. In addition, recovery from addiction takes a long time, and recurrence is frequently observed. Current treatments cannot effectively be used to treat all the stages of drug addiction [14,15]. Therefore, a new approach is needed for drug addiction management.
Over the past few years, a number of studies have been performed on exercise-based interventions for METH abuse management [16]. Physical exercise has been reported to reduce the extent, duration, and frequency of drug use in drug-addicted individuals during the drug initiation phase, as well as during prolonged addiction, withdrawal, and recurrence [17]. Moderate aerobic exercise effectively reduced METH reuse by reducing depression and anxiety and controlled drug cravings in individuals addicted to METH [18,19,20]. In addition, exercise interventions had a positive effect during the METH withdrawal period [21,22,23,24]. Endurance and resistance exercise for eight weeks improved the subjects’ body composition and physical fitness level during the withdrawal period, and these physical changes reduced the drug dependence of the addicted individuals [25,26]. Similar results have been reported in animal studies. METH self-administration was significantly decreased in mice that were subjected to voluntary wheel running (1 h/day) [27], and the amount of METH self-administration also decreased in mice that performed voluntary wheel running prior to the self-administration session [28,29]. It would be important to have information on the neurobiological changes caused by exercise that may have a positive effect on drug-addicted individuals. According to previous studies, the neurobiological changes that take place in METH users through physical exercise may be due to the interaction of various changes, including the regulation of CNS neurochemicals [17,30], improvement of defense mechanisms involving antioxidants [31,32,33,34], neurogenesis [35], glial formation [36], and protection of the blood–brain barrier [37,38]. However, the results presented in these previous studies do not fully explain the effect of exercise on METH-induced neurobiological changes.
METH induces hyperactivity [39,40,41], a phenomenon called context-dependent behavioral sensitization, which plays a key role in the development of drug-seeking behavior [42,43,44]. METH-induced hyperactivity occurs due to the reinforcement of dopaminergic neurotransmission in the mesocorticolimbic system [45,46]. The mesocorticolimbic system consists of dopaminergic cell bodies in the ventral cortical region that project to the nucleus accumbens (NA, ventral striatum), dorsal striatum (caudate putamen), amygdala, hippocampus, and the prefrontal cortex. The neural adaptation process of the mesolimbic dopamine pathway occurs during repetitive drug exposure, affecting synaptic plasticity; this triggers neural adaptation of the neural circuits related to drug-induced learning and behavioral memory, and increases compulsive drug-seeking and recurrence [47,48]. A previous study suggested that glycogen synthase kinase-3 (GSK-3) in the striatum is a critical molecule in the acquisition of stimulant-induced activity and behavioral sensitization [49]. GSK-3 is a serine/threonine phosphorylating enzyme that is expressed throughout the mammalian brain and affects cell structure, motility, cell survival, apoptosis, neuronal differentiation, and DNA transcription [50,51]. It is highly regulated by phosphorylation at Tyr216 or Ser9 [52,53]. The phosphorylation of the Tyr216 residue occurs during the GSK-3β translation process, and results in the synthesis of the fully activated kinase; this activation process occurs in resting neurons to promote substrate access [54]. Ser9 phosphorylation is thought to be the main regulatory modification that occurs during the lifespan of the enzyme [55]. Ser9-phosphorylated GSK-3β remains inhibited, and dephosphorylation of the residue results in the activation of the kinase [56,57,58]. METH-induced GSK-3β activity is regulated by a number of signaling pathways (such as the dopamine, glutamate, and serotonin signaling pathways) [49]. Previous studies have shown that the dopamine receptor D2 (DRD2)–protein kinase B (Akt)–GSK3β signaling pathway is essential for the development of drug-induced behavioral sensitization [59,60,61,62]. Dopamine transporter knockout led to increased dopaminergic neurotransmission and hyperactivity in the knockout mice, which was accompanied by reduced Akt phosphorylation and reduced phosphorylation of the downstream substrates GSK-3α and GSK-3β; this further led to an increase in GSK-3β activity in the striatum [59]. Solis et al. [63] have reported that METH-induced locomotor sensitization is reduced in DRD2 knockout mice, and these mice have reduced locomotor response to a dopamine receptor D1 (DRD1) agonist. In addition, the glutamatergic receptor signaling pathway has also been reported to play an important role in conditioned place preference (CPP) induced by psychostimulants [64,65,66]. The activity of GSK-3β can be regulated by glutamatergic N-methyl-D-aspartate (NMDA) receptors. NMDA application to cultured hippocampal neurons produces a rapid dephosphorylate Ser9-GSK3β, demonstrating that NMDA receptor signaling activates GSK-3β [67].
Neuroadaptation to METH-induced behavioral sensitivity has been studied extensively, but there is little research into the effect of exercise training on METH-induced neuroadaptation. Therefore, this study investigated the effect of forced moderate endurance exercise on METH-induced behavior and the activity of GSK-3β via striatal dopaminergic and glutamatergic signaling pathways. As most individuals with drug addiction live a sedentary life, in this study, rats were subjected to forced exercise rather than voluntary exercise to mimic the conditions of exercise stress found in the clinic [68].

2. Materials and Methods

2.1. Animals

Male Sprague Dawley rats (Hyochang Science, Daegu, Korea) weighing approximately 200 g were housed in pairs. All rats were maintained under standard conditions with a constant room temperature (22 ± 1 °C), relative humidity (50 ± 10%), and a 12 h light/dark cycle (light from 07:00 to 19:00) with food (#2014, Harlan Telkad, Madison, USA) and water ad libitum. All rats were allowed to habituate to the novel environment for 1 week prior to the experiments. The rats were randomly assigned to four groups, as follows: saline control group (CON); methamphetamine injection group (MA); saline + exercise group (Ex); and methamphetamine + exercise group (Mex) (n = 8 in each group). The experiments were carried out in accordance with the guidelines approved by the Institutional Animal Care and Use Committee of Keimyung University (KM2020-008).

2.2. METH Administration and Exercise Training

2.2.1. METH Administration

(+)-S-Methamphetamine hydrochloride (M8750, Sigma-Aldrich, Saint Louis, MO, USA) was dissolved in sterile 0.9% saline to a final concentration of 0.6 mg/mL. METH was freshly prepared before use. Rats in the MA and MEx group were administered 1 mg/kg METH–HCl intraperitoneally [69,70], and in the CON and Ex group, 0.9% saline was administered in the same volume. METH or saline was administrated once a day for 14 days. Every injection was performed between 09:00 and 12:00 p.m. The exercise group was administered METH or saline immediately after exercise training.

2.2.2. Exercise Training

Exercise training was performed between 09:00 and 12:00 p.m. once a day for 2 weeks using an electric treadmill for laboratory animals (FT-200, Techman Soft, Taoyuan City, Taiwan). Exercise intensity was set by partially modifying the research methods of Wang et al. [71] and Kemi et al. [72]. Wang et al. [71] reported that the craving behavior for drugs in individuals addicted to METH was most effectively reduced with exercise of moderate intensity. Accordingly, the exercise training protocol consisted of moderate intensity (60% VO2max, 21 m/min, slope 0%) running on a treadmill for 60 min per day. During the first 7 days, the intensity and duration of the exercise were gradually increased to reach the target amount of exercise. After that, the exercise was performed while maintaining the target exercise amount until the end of the experiment. On the first day, exercise was performed on an electric treadmill at a speed of 10 m/min for 15 min to assist with adaptation to exercise. The duration and intensity of the exercise were then gradually increased by approximately 15% per day for 6 days until the target amount of exercise was reached. Since rats are more active in the dark, the front of the treadmill line was covered with dark paper. No electric shocks were used to reduce the stress effect of running on the treadmill during training sessions.

2.3. Experimental Design

All treatment and behavioral experiments were performed in the light cycle between 09:00 and 14:00. Body weight and food intake were measured daily during the 14-day experimental period. On the 14th day, the locomotor activity test was performed between 9:00 and 15:00. Exercise intervention was not performed on the day of the test, and METH or saline was injected immediately before locomotor activity test. On the 15th day, sampling commenced at 09:00. Before sampling, the Ex and MEx groups exercised lightly for 15 min at low intensity (10 m/min) to prevent loss of the exercise effect (Figure 1). Immediately after exercise, METH (1 mg/kg) or saline was injected and the sample was collected after resting for 2 h. CON and MA groups collected samples after 2 h of rest following saline or METH administration. For sampling, pentobarbital sodium (5 mg/100 g bwt) was administered intraperitoneally. Once rats were fully anesthetized, all blood was completely removed from the abdominal artery using a syringe, and the brain was then isolated by decapitation [73]. The right and left striatum were rapidly dissected out on ice. Tissues were immediately frozen in liquid nitrogen and stored at −80 °C.

2.4. Locomotor Activity

The locomotor activity test was performed from 9:00 on the 14th day of treatment, and the exercise group did not exercise on the day of the test. Rats were habituated to the testing boxes prior to testing by being handled in the colony room for 5 min daily for two days. They were then placed individually in a box for 30 min daily for another two days. On the day of the test, each rat was placed in a black Plexiglas square box (50 × 50 × 50 cm), and after an adaptation period of 15 min, 1 mg/kg of METH or saline was administered intraperitoneally. The locomotor activity was measured and displayed as total distance (cm) traveled during the 90 min period following administration of saline or METH. The apparatus was cleaned with 70% alcohol and dried after each session. The distance was measured by an automated video tracking system (Smart Video Tracking System- SMART 3.0, Panlab, Barcelona, Spain). The video files were analyzed by DigBehv analysis software to calculate motion tracking values, duration by movement speed, and total distance traveled. The speed of movement was divided into resting (<2.5 cm/s), slow (2.5–15 cm/s), and high (>15 cm/s). These classifications were guided by the results of previous studies [74,75].

2.5. Sample Preparation

The left and right striatum were homogenized together in an ice-cold RIPA buffer containing 250 mM sucrose, 10 mM HEPES/1 mM ethylenediaminetetraacetic acid (EDTA, pH 7.4), 1 mM Pefabloc (Roche), 1 mM EDTA, 1 mM NaF, 1 μg/mL aprotinin, 1 μg/mL leupeptin, 1 μg/mL pepstatin, 0.1 mM bpV (phen), 2 mg/mL glycerophosphate, protease inhibitors (#1861280, Fisher Scientific, Rockford, IL, USA), and a phosphatase inhibitor (P2850, Sigma-Aldrich, Saint Louis, MO, USA). Homogenization was performed on ice using a Wheaton tissue grinder (#357535 and 357537, DWK Life Sciences, Millville, NJ, USA). Homogenates were subjected to three freeze/thaw cycles and centrifuged for 15 min at 4 °C, 1000× g. The supernatant was separated and stored at −80 °C until assayed.

2.6. Western Blotting

Protein concentration was determined using the BSA assay (#5000006, Bio-Rad, Hercules, CA, USA). Aliquots were solubilized in Laemmli buffer (#1610747, Bio-Rad) and subjected to SDS/PAGE. A total of 40 ug of protein was separated by SDS-PAGE gel and immunoblotted with antibodies directed against GSK-3β (1:3000, Santa Cruz Bio, Dallas, TX, USA, sc-81462), phospho(Ser9)-GSK3β (1:1000, Santa Cruz Bio, sc-373800), phospho(Tyr279/216)-GSK3α/β (1:1000, Santa Cruz Bio, sc-81496), phospho(Ser473)-AKT (1:1000, Cell signaling, #4060), phospho(Thr308)-AKT (1:1000, Cell signaling, #13038), AKT (1:1000, Cell signaling, #2920), DRD1 (1:1000, Santa Cruz Bio, sc-33660), DRD2 (1:1000, Santa Cruz Bio, sc-5303), NMDA receptor 1 (NMDAr1, 1:1000, Santa Cruz Bio, sc-518053), calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2, 1:1000, Cell signaling, #16810), protein phosphatase 1 (PP1, 1:1000, Santa Cruz Bio, sc-7482), phospho(Thr180/Tyr182)-p38 mitogen-activated protein kinase (MAPK, 1:1000, Cell signaling, #4511), p38 MAPK (1:3000, Cell signaling, #9212), phospho(Thr202/Tyr204)-extracellular signal-regulated kinases 1/2 (Erk1/2, 1:1000, Cell signaling, #9101), Erk1/2 (1:3000, Cell signaling, #9102) and β-actin (1:1000, abcam, ab106814). After incubation with peroxidase-conjugated secondary antibody, blots were subjected to the enhanced chemiluminescent detection method with the ImageQuantTMLas 4000 imaging system (GE Healthcare). The relative intensity of the bands was assessed using SigmaGel (Jandel Scientific Corp., Schimmelbuschstrasse 25, Erkrath, Germany). The loading control was evaluated by the expression level of β-actin. The protein expression levels were normalized against β-actin protein levels and presented in arbitrary units (AU). Proteins activated by phosphorylation were presented as relative ratios of phosphorylation versus total expression level.

2.7. Striatal Dopamine Levels

Striatal dopamine levels were analyzed using a commercially available ELISA for dopamine according to the manufacturer’s instructions (Rat dopamine ELISA-kit, Fine Test, Wuhan Fine Biotech Co., Wuhan, China). The assay was performed in a 96-well plate, and absorbance was recorded using a plate reader (Spectramax). The dopamine levels were quantified and expressed as ng/mg tissue.

2.8. Statistical Analysis

Statistical analyses were conducted using GraphPad Prism 7.0 (GraphPad Software Inc., La Jolla, CA, USA). The results are presented as mean ± SE (standard error). Significant differences between groups were evaluated using one-way analysis of variance (ANOVA), and dietary intake and body weight were evaluated using two-way ANOVA. Post hoc validation was performed using Tukey’s test. A p-value < 0.05 was considered statistically significant. The analysis was performed by a statistician blinded to the experimental groups.

3. Results

3.1. Forced Moderate Endurance Exercise Significantly Reduced METH-Induced Hyperactivity

Dietary intake, body weight, and locomotor activity using an open field were measured to determine the effect of 14 days of exercise and METH administration on body composition and physical excitability. Endurance exercise or METH injection did not affect dietary intake and weight gain during the study period (Figure 2A,B). The administration of METH for 14 days significantly increased locomotor activity compared to the non-administration of METH (p < 0.001), while endurance exercise for 14 days did not affect locomotor activity (Figure 2C). In the MEx group that was administered combined treatment with endurance exercise and METH, the locomotor activity decreased significantly, though it was still significantly higher than that in the CON and Ex groups (p < 0.001, Figure 2C). When the locomotor activity was classified according to speed, the MA group moved exclusively at a fast speed (15 cm/s) for 60 min, whereas the CON and Ex groups moved 60% of the total travel time at a slow speed (2.5–15 cm/s). The animals took a break for the remaining 40% of the time (0–2.5 cm/s). The MEx group moved 5% of the total travel time at a fast speed, 90% at a slow speed, and took a break for the remaining 5% of the time (Figure 2D). In summary, 14 days of endurance exercise significantly reduced METH-induced hyperactivity in the MEx group. However, the activity level was not reduced to that in the CON group.

3.2. Forced Moderate Endurance Exercise Did Not Affect the METH-Induced Dopaminergic Signaling Pathway

Most of the drug-induced sensitization is considered to be regulated in the striatum that receive efferent processes from the dopaminergic mesocorticolimbic system [14,76]. The release of the neurotransmitters dopamine and glutamate in these core brain regions is essential for addiction-induced behavioral changes [77,78]. As per a previous study, we analyzed changes in striatal dopaminergic signaling molecules to understand the mechanism underlying the inhibition of METH-induced hyperactivity by endurance exercise training. METH administration for 14 days and/or endurance exercise significantly increased dopamine levels in the striatum (p < 0.01, Figure 3A). The expression levels of DRD1 and DRD2 were not significantly different between the four groups (Figure 3B,C). The expression level of p(Ser473)-Akt was not significantly different between the four groups (Figure 3D). However, the expression of p(Thr308)-Akt was significantly decreased in the MA, Ex, and MEx groups compared to CON (p < 0.05, Figure 3E). A significant increase in the expression of p(Tyr279)-GSK3α and p(Tyr216)-GSK-3β was observed in MA and MEx groups compared to CON (p < 0.01, 0.05, Figure 3F,G). The expression of p(Ser9)-GSK-3β, which is an inhibited from of GSK-3β, was not significantly altered in the MA group compared with the CON group, while it was significantly higher in the Ex and MEx groups than in the MA group (p < 0.05). The expression of p(Ser9)-GSK-3β in the Ex group was significantly higher than that in the CON group (p < 0.05, Figure 3H). To sum up the above results, 14-day METH and/or exercise treatment increased striatal dopamine levels, decreased the expression of p(Thr308)-Akt and increased the expression of p(Tyr206)-GSK-3β. However, the expression of p(Ser9)-GSK-3β, which inhibits the activity of GSK-3β, was significantly increased only in the exercise groups.

3.3. Forced Moderate Endurance Exercise Affected the METH-Induced Glutamatergic Signaling Pathway

To analyze the effect of endurance exercise on METH-induced hyperactivity via the glutamate signaling system, changes in the glutamate receptor and subsignal factors in the striatum were measured. According to previous studies, repeated administration of psychostimulants causes behavior sensitization, and the adaptation of dopaminergic and glutamatergic neurotransmission plays an important role in the expression of behavior sensitization [49,79,80]. In this study, two weeks of METH administration significantly increased the expression of NMDAr1 in the striatum compared to the other three groups (p < 0.01, 0.05, Figure 4A), and the expression levels of CaMKK2 and PP1, downstream of NMDAr1, were also significantly increased in the MA group (p < 0.05, Figure 4B,C). However, exercise intervention for two weeks significantly decreased the expression level of NMDAr1/CaMKK2/PP1 (p < 0.001, 0.01, 0.05, Figure 4A–C).
Previous studies have reported that Ca2+ enters cells through NMDA receptors by METH stimulation and activates MAPKs in the striatum [81]. Accordingly, we also analyzed the changes in the expression of MAPKs (p38, p42, p44). The level of p-p38MAPK expression was significantly increased in the MA group compared to the CON group, and decreased significantly in the Ex and MEx groups compared to the MA group (p < 0.05, 0.01, Figure 4D). The expression of p-ERK1 (p44MEPK) was not significantly different between the CON, MA, and MEx groups, but decreased significantly in the Ex group compared to the CON and MA groups (p < 0.05, Figure 4E). There was no significant difference in the expression of p-ERK2 (p42MEPK) between the MA and CON groups. However, p-ERK2 (p42MEPK) expression was significantly decreased in the Ex group compared to the CON and MA groups, and significantly decreased in the MEx group compared to the MA group (p < 0.01, 0.05, Figure 4F). In summary, two weeks of METH treatment increased the expression of NMDAr1/CaMKK2/PP1 and MAPKs, such as p-p38MAPK and p-ERK2, but endurance exercise training suppressed the expression of these molecules to the levels of the CON group.

4. Discussion

We investigated the effect of moderate endurance exercise (as used commonly in clinical studies) on METH-induced hyperactivity, and attempted to elucidate the underlying neurobiological mechanisms. METH administration for 14 days induced hyperactivity in SD rats, increased striatal dopamine levels, and increased GSK-3β activity by decreasing p(Thr308)-Akt expression and increasing p(Tyr216)-GSK3β expression levels. Interestingly, our results showed that the changes in the dopaminergic signaling pathway were similar between METH administration and METH administration combined with exercise. The combination of METH and endurance exercise increased the DA level in the striatum, decreased p(Thr308)-Akt, and increased the activity of GSK-3β by increasing p(Tyr216)-GSK3β. Previous studies have reported that physical exercise, similar to METH, can increase extracellular dopamine levels in the striatum and activate the neural circuits that underlie the reward and reinforcing effects [82,83]. Therefore, similar to the effect of chronic drug addiction, continuous physical exercise causes hedonic symptoms by activating the reward system [82,84]. In animal studies, the negative effects and symptoms of drug withdrawal were also observed in physical exercise withdrawal [85]. In mice exposed to wheel running, when access to the wheel was prohibited, systolic and diastolic blood pressure and body temperature decreased [86]; additionally, depression and behavioral symptoms such as anxiety were also observed [87,88,89]. However, the concept of “exercise addiction” is not yet clear and peer-reviewed evidence is lacking, so it has not been classified as a mental illness [90]. This study observed the effect of endurance exercise on METH-induced behavioral sensitization. Although the effect of exercise on METH-induced reward and reinforcement can be estimated, it is difficult to clarify the causal relationship.
In our study, the exercise group showed a significant increase in the expression of pSer9-GSK3β, unlike the METH treatment group. Thus, we may conclude that the inhibition of METH-induced hyperactivity caused by exercise is regulated by signaling pathways other than dopaminergic signaling pathways. Although the glutamate receptor is not a direct molecular target of METH, METH activates DRD1 to induce glutamate release [91,92], and the increase in extracellular glutamate activates the NMDA receptor; this in turn increases the intracellular influx of calcium [93,94]. The increase in intracellular calcium activates CaMK2, triggers calcineurin (PP2B), and subsequently dephosphorylates inhibitor-1 to induce PP1 activation [67]. PP1 directly increases GSK-3β activity by dephosphorylating Akt and Ser9-GSK3β [95,96,97,98]. In this study, METH administration for 14 days significantly increased the expression of NMDAr1 and PP1 in the striatum, and endurance exercise reduced these molecules to control (CON group) levels. Although DRD1 activity was not measured, we considered that METH-induced NMDAr1-PP1-GSK3β activity was inhibited by endurance exercise, and thus it may have acted as one of the regulatory factors of METH-induced hyperactivity. GSK-3β activation is regulated by a variety of factors, including the MAPK/ERK pathway [99]. METH administration for 14 days increased p38MAPK and ERK-2 phosphorylation in the striatum in this study, and endurance exercise inhibited METH-induced activation of the MAPKs. MAPK is a key signaling molecule in the regulation of pro-inflammatory cytokines and cellular responses to external stress. Rawas et al. [100] have reported that p38MAPK in the NA is involved in abnormal behavioral responses induced by drug abuse. Acute or chronic administration of psychostimulants such as cocaine or METH activates ERK throughout the striatum [101,102,103,104]. Gerdjikov et al. [105] suggest that ERK and p38 may be necessary for the establishment of NA amphetamine-produced CPP and may also mediate other forms of reward-related learning dependent on NA. It has been suggested that the activated MAPK cascade is involved in a major burst of gene expression that underlies long-term behavioral changes to drug exposure [106]. In this study, 14 days of METH administration increased the expression of NMDAr1, CaMKK2, and MAPK in the striatum, and endurance exercise suppressed the expression of these molecules. Therefore, these molecular changes may be involved in METH-induced hyperactivity. In summary, endurance exercise and METH had similar effects on the molecular expression of striatal dopaminergic markers, but opposite effects on glutamatergic markers. Although these molecular changes cannot fully explain the mechanism of the regulation of METH hyperactivity by exercise, it is possible to conclude that they can be implicated in this process.
The exercise intervention in this study involved forced moderate-intensity endurance exercise. In animal studies, voluntary exercise, in which the animal has access to freely running wheels, or forced exercise, in which the animal is made to run on a treadmill or swim in a closed pool, is employed. These methods are similar to human exercise patterns in a specific way, where voluntary exercise represents exercising according to an individual’s own will, and forced exercise represents standardized exercise formulated for the prevention or treatment of certain disorders [107]. According to a previous study, regular swimming exercise (45 min/day, 5 days/week, 14 days) decreased METH consumption in mice, and anxiety and depressive behavior decreased when METH administration was stopped [108]. In addition, CPP for amphetamine was inhibited after forced treadmill running [109], and both voluntary and forced wheel running were effective in reducing uncontrollable stress-induced learning deficits in mice [110]. There is very little research on the intensity of exercise therapy for the treatment of drug addiction. However, as most drug-addicted individuals have a sedentary lifestyle, low to moderate-intensity exercise is more likely to be preferred initially, rather than vigorous intensity exercise; moderate-intensity exercise has also been reported to be associated with higher compliance [111,112,113]. Based on these previous studies, we used moderate-intensity forced endurance exercise, and this exercise regimen effectively reduced METH-induced hyperactivity. However, it is difficult to formulate conclusions on the intensity and type of exercise that would be useful for drug addiction treatment based on the results of this study alone. Therefore, further research on the intensity, frequency, duration, and type of exercise is needed to formulate a therapeutic exercise regimen for individuals with METH addiction.
This study has a limitation in that it only targeted male rats. Numerous studies in humans [114,115,116,117,118,119] and rodents [120,121,122,123,124] have demonstrated gender differences in the effects of exercise on drug addiction. For example, female rats had reduced cocaine conditioned place preference (CPP) after chronic forced treadmill exercise, whereas male rats had complete inhibition of drug CPP after exercise [125]. In contrast, Ehringer et al. [120] reported a decrease in alcohol consumption after exercise in female rats, but not in male rats. In addition, other studies have shown a decrease in cocaine self-administration under extended-access conditions after exercise in both men and women [126,127]. The reasons for these conflicting results are not yet known, but may be related to the type of addictive drug, the type of motor activity [114,117], and gender-specific neurobiological mechanisms [123,128,129,130]. Overall, gender differences in the preventive and therapeutic effects of exercise on drug addiction are unclear and further investigations across both genders are needed.

5. Conclusions

Drug addiction is caused by a combination of psychological, physiological, and social factors, and an intervention combining various treatments will be effective. Therefore, we investigated the neurobiological effects of physical exercise on METH addiction to understand its therapeutic potential (“exercise pills”) for the treatment of drug addiction and prevention of recurrence. The results of this study show that forced moderate endurance exercise significantly reduces METH-induced hyperactivity in rats, which may be due to the regulation of the striatal glutamatergic signaling molecules leading to a decrease in GSK-3β activation (Figure 5). However, the results of this study alone cannot fully explain the mechanism of the effect of exercise on METH addiction. Future studies on the combined effect of various GSK-3β inhibitors (such as lithium, arylindolemaleimide, and thiadiazolidindiones) and exercise can help in the formulation of a combination therapy that can be used as an effective adjuvant treatment for drug addiction. In addition, using exercise as a therapeutic intervention requires additional research that can provide accurate guidelines (e.g., intensity, frequency, duration, type of exercise) for various types of drug-use disorders.

Author Contributions

Conceptualization, S.J. and S.L.; methodology, S.J., Y.K., M.K., M.S., S.K. (Suji Kim) and S.K. (Seungju Kim); formal analysis, S.J.; investigation, S.J.; resources, S.L.; writing—original draft preparation, S.J.; writing—review and editing, S.J. and S.L.; supervision, S.L.; project administration, S.J.; funding acquisition, S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Basic Science Research Program (NRF-2016R1A6A1A03011325 and NRF-2018R1D1A1B07048159) through the National Research Foundation funded by the Ministry of Education in Korea.

Institutional Review Board Statement

The study was carried out in accordance with the guidelines approved by the Institutional Animal Care and Use Committee of Keimyung University (KM2020-008).

Informed Consent Statement

Not applicable.

Data Availability Statement

Available data are presented in the manuscript.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. United Nations Office on Drugs and Crime. World Drug Report-2016; United Nations: New York, NY, USA, 2016. [Google Scholar]
  2. Courtney, K.E.; Ray, L.A. Methamphetamine: An update on epidemiology, pharmacology, clinical phenomenology, and treatment literature. Drug Alcohol Depend. 2014, 143, 11–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Rawson, R.A. Current research on the epidemiology, medical and psychiatric effects, and treatment of methamphetamine use. J. Food Drug Anal. 2013, 21, S77–S81. [Google Scholar] [CrossRef] [Green Version]
  4. Morais, A.P.D.; Pita, I.R.; Fontes-Ribeiro, C.A.; Pereira, F.C. The neurobiological mechanisms of physical exercise in methamphetamine addiction. CNS Neurosci. Ther. 2018, 24, 85–97. [Google Scholar] [CrossRef] [PubMed]
  5. Hwang, J.S.; Cha, E.H.; Park, B.; Ha, E.; Seo, J.H. PBN inhibits a detrimental effect of methamphetamine on brain endothelial cells by alleviating the generation of reactive oxygen species. Arch. Pharm. Res. 2020, 43, 1347–1355. [Google Scholar] [CrossRef]
  6. Kirkpatrick, M.G.; Haney, M.; Vosburg, S.K.; Comer, S.D.; Foltin, R.W.; Hart, C.L. Methamphetamine self-administration by humans subjected to abrupt shift and sleep schedule changes. Psychopharmacology 2009, 203, 771–780. [Google Scholar] [CrossRef]
  7. Mendelson, J.; Rawson, R.; Newton, T.; Galloway, G.; de Wit, H.; Dewey, S.L.; Hart, C.L.; Epstein, D.H. Treatment of methamphetamine dependence. Mayo Clin. Proc. 2008, 83, 369–370. [Google Scholar] [CrossRef] [Green Version]
  8. Perez, A.Y.; Kirkpatrick, M.G.; Gunderson, E.W.; Marrone, G.; Silver, R.; Foltin, R.W.; Hart, C.L. Residual effects of intranasal methamphetamine on sleep, mood, and performance. Drug Alcohol Depend. 2008, 94, 258–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Prakash, M.D.; Tangalakis, K.; Antonipillai, J.; Stojanovska, L.; Nurgali, K.; Apostolopoulos, V. Methamphetamine: Effects on the brain, gut and immune system. Pharmacol. Res. 2017, 120, 60–67. [Google Scholar] [CrossRef] [Green Version]
  10. Halpin, L.E.; Collins, S.A.; Yamamoto, B.K. Neurotoxicity of methamphetamine and 3,4-methylenedioxymethamphetamine. Life Sci. 2014, 97, 37–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Dean, A.C.; Groman, S.M.; Morales, A.M.; London, E.D. An evaluation of the evidence that methamphetamine abuse causes cognitive decline in humans. Neuropsychopharmacology 2013, 38, 259–274. [Google Scholar] [CrossRef] [Green Version]
  12. Seo, M.J.; Song, S.H.; Kim, S.; Jang, W.J.; Jeong, C.H.; Lee, S. Characteristics of Korean patients with methamphetamine use disorder based on the quantitative analysis of methamphetamine and amphetamine in hair. Arch. Pharm. Res. 2020, 43, 798–807. [Google Scholar] [CrossRef] [PubMed]
  13. Somkuwar, S.S.; Staples, M.C.; Fannon, M.J.; Ghofranian, A.; Mandyam, C.D. Evaluating Exercise as a Therapeutic Intervention for Methamphetamine Addiction-Like Behavior. Brain Plast. 2015, 1, 63–81. [Google Scholar] [CrossRef] [Green Version]
  14. Koob, G.F.; Volkow, N.D. Neurocircuitry of addiction. Neuropsychopharmacology 2010, 35, 217–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kalivas, P.W.; Volkow, N.D. The neural basis of addiction: A pathology of motivation and choice. Am. J. Psychiatry 2005, 162, 1403–1413. [Google Scholar] [CrossRef]
  16. Brown, R.A.; Abrantes, A.M.; Read, J.P.; Marcus, B.H.; Jakicic, J.; Strong, D.R.; Oakley, J.R.; Ramsey, S.E.; Kahler, C.W.; Stuart, G.G.; et al. A Pilot Study of Aerobic Exercise as an Adjunctive Treatment for Drug Dependence. Ment. Health Phys. Act. 2010, 3, 27–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Lynch, W.J.; Peterson, A.B.; Sanchez, V.; Abel, J.; Smith, M.A. Exercise as a novel treatment for drug addiction: A neurobiological and stage-dependent hypothesis. Neurosci. Biobehav. Rev. 2013, 37, 1622–1644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Rawson, R.A.; Chudzynski, J.; Mooney, L.; Gonzales, R.; Ang, A.; Dickerson, D.; Penate, J.; Salem, B.A.; Dolezal, B.; Cooper, C.B. Impact of an exercise intervention on methamphetamine use outcomes post-residential treatment care. Drug Alcohol Depend. 2015, 156, 21–28. [Google Scholar] [CrossRef] [Green Version]
  19. Rawson, R.A.; Chudzynski, J.; Gonzales, R.; Mooney, L.; Dickerson, D.; Ang, A.; Dolezal, B.; Cooper, C.B. The Impact of Exercise On Depression and Anxiety Symptoms Among Abstinent Methamphetamine-Dependent Individuals in A Residential Treatment Setting. J. Subst. Abus. Treat. 2015, 57, 36–40. [Google Scholar] [CrossRef] [Green Version]
  20. Haglund, M.; Ang, A.; Mooney, L.; Gonzales, R.; Chudzynski, J.; Cooper, C.B.; Dolezal, B.A.; Gitlin, M.; Rawson, R.A. Predictors of depression outcomes among abstinent methamphetamine-dependent individuals exposed to an exercise intervention. Am. J. Addict. 2015, 24, 246–251. [Google Scholar] [CrossRef]
  21. Muller, A.E.; Clausen, T. Group exercise to improve quality of life among substance use disorder patients. Scand. J. Public Health 2015, 43, 146–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Koob, G.F. Addiction is a Reward Deficit and Stress Surfeit Disorder. Front. Psychiatry 2013, 4, 72. [Google Scholar] [CrossRef] [Green Version]
  23. Jones, E.M.; Knutson, D.; Haines, D. Common problems in patients recovering from chemical dependency. Am. Fam. Phys. 2003, 68, 1971–1978. [Google Scholar]
  24. Polter, A.M.; Kauer, J.A. Stress and VTA synapses: Implications for addiction and depression. Eur. J. Neurosci. 2014, 39, 1179–1188. [Google Scholar] [CrossRef] [PubMed]
  25. Dolezal, B.A.; Chudzynski, J.; Dickerson, D.; Mooney, L.; Rawson, R.A.; Garfinkel, A.; Cooper, C.B. Exercise training improves heart rate variability after methamphetamine dependency. Med. Sci. Sports Exerc. 2014, 46, 1057–1066. [Google Scholar] [CrossRef] [Green Version]
  26. Dolezal, B.A.; Chudzynski, J.; Storer, T.W.; Abrazado, M.; Penate, J.; Mooney, L.; Dickerson, D.; Rawson, R.A.; Cooper, C.B. Eight weeks of exercise training improves fitness measures in methamphetamine-dependent individuals in residential treatment. J. Addict. Med. 2013, 7, 122–128. [Google Scholar] [CrossRef]
  27. Miller, M.L.; Vaillancourt, B.D.; Wright, M.J., Jr.; Aarde, S.M.; Vandewater, S.A.; Creehan, K.M.; Taffe, M.A. Reciprocal inhibitory effects of intravenous d-methamphetamine self-administration and wheel activity in rats. Drug Alcohol Depend. 2012, 121, 90–96. [Google Scholar] [CrossRef] [Green Version]
  28. Aarde, S.M.; Miller, M.L.; Creehan, K.M.; Vandewater, S.A.; Taffe, M.A. One day access to a running wheel reduces self-administration of D-methamphetamine, MDMA and methylone. Drug Alcohol Depend. 2015, 151, 151–158. [Google Scholar] [CrossRef] [Green Version]
  29. Engelmann, A.J.; Aparicio, M.B.; Kim, A.; Sobieraj, J.C.; Yuan, C.J.; Grant, Y.; Mandyam, C.D. Chronic wheel running reduces maladaptive patterns of methamphetamine intake: Regulation by attenuation of methamphetamine-induced neuronal nitric oxide synthase. Brain Struct. Funct. 2014, 219, 657–672. [Google Scholar] [CrossRef] [Green Version]
  30. O’Dell, S.J.; Galvez, B.A.; Ball, A.J.; Marshall, J.F. Running wheel exercise ameliorates methamphetamine-induced damage to dopamine and serotonin terminals. Synapse 2012, 66, 71–80. [Google Scholar] [CrossRef]
  31. Radak, Z.; Chung, H.Y.; Goto, S. Systemic adaptation to oxidative challenge induced by regular exercise. Free Radic Biol. Med. 2008, 44, 153–159. [Google Scholar] [CrossRef] [PubMed]
  32. Aguiar, A.S., Jr.; Tuon, T.; Soares, F.S.; da Rocha, L.G.; Silveira, P.C.; Pinho, R.A. The effect of n-acetylcysteine and deferoxamine on exercise-induced oxidative damage in striatum and hippocampus of mice. Neurochem. Res. 2008, 33, 729–736. [Google Scholar] [CrossRef] [PubMed]
  33. Camiletti-Moirón, D.; Aparicio, V.A.; Aranda, P.; Radak, Z. Does exercise reduce brain oxidative stress? A systematic review. Scand. J. Med. Sci. Sports 2013, 23, e202–e212. [Google Scholar] [CrossRef] [Green Version]
  34. Speck, A.E.; Tromm, C.B.; Pozzi, B.G.; Paganini, C.S.; Tuon, T.; Silveira, P.C.; Aguiar, A.S., Jr.; Pinho, R.A. The dose-dependent antioxidant effects of physical exercise in the hippocampus of mice. Neurochem. Res. 2014, 39, 1496–1501. [Google Scholar] [CrossRef]
  35. Sobieraj, J.C.; Kim, A.; Fannon, M.J.; Mandyam, C.D. Chronic wheel running-induced reduction of extinction and reinstatement of methamphetamine seeking in methamphetamine dependent rats is associated with reduced number of periaqueductal gray dopamine neurons. Brain Struct. Funct. 2016, 221, 261–276. [Google Scholar] [CrossRef] [Green Version]
  36. Mandyam, C.D.; Wee, S.; Eisch, A.J.; Richardson, H.N.; Koob, G.F. Methamphetamine self-administration and voluntary exercise have opposing effects on medial prefrontal cortex gliogenesis. J. Neurosci. 2007, 27, 11442–11450. [Google Scholar] [CrossRef] [PubMed]
  37. Park, M.; Levine, H.; Toborek, M. Exercise protects against methamphetamine-induced aberrant neurogenesis. Sci. Rep. 2016, 6, 34111. [Google Scholar] [CrossRef] [Green Version]
  38. Wang, D.; Wang, Y.; Wang, Y.; Li, R.; Zhou, C. Impact of physical exercise on substance use disorders: A meta-analysis. PLoS ONE 2014, 9, e110728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Filip, M.; Frankowska, M.; Gołda, A.; Zaniewska, M.; Vetulani, J.; Przegaliński, E. Various GABA-mimetic drugs differently affect cocaine-evoked hyperlocomotion and sensitization. Eur. J. Pharmacol. 2006, 541, 163–170. [Google Scholar] [CrossRef]
  40. Fukushima, S.; Shen, H.; Hata, H.; Ohara, A.; Ohmi, K.; Ikeda, K.; Numachi, Y.; Kobayashi, H.; Hall, F.S.; Uhl, G.R.; et al. Methamphetamine-induced locomotor activity and sensitization in dopamine transporter and vesicular monoamine transporter 2 double mutant mice. Psychopharmacology 2007, 193, 55–62. [Google Scholar] [CrossRef]
  41. Shimosato, K.; Ohkuma, S. Simultaneous monitoring of conditioned place preference and locomotor sensitization following repeated administration of cocaine and methamphetamine. Pharmacol. Biochem. Behav. 2000, 66, 285–292. [Google Scholar] [CrossRef]
  42. Hooks, M.S.; Jones, G.H.; Hemby, S.E.; Justice, J.B., Jr. Environmental and pharmacological sensitization: Effects of repeated administration of systemic or intra-nucleus accumbens cocaine. Psychopharmacology 1993, 111, 109–116. [Google Scholar] [CrossRef]
  43. Mattingly, B.A.; Rice, L.L.; Langfels, M.; Fields, S.E. Repeated treatments with 7-OH-DPAT: Context-independent behavioral sensitization and conditioned hyperactivity. Pharmacol. Biochem. Behav. 2000, 65, 241–246. [Google Scholar] [CrossRef]
  44. Shen, F.; Meredith, G.E.; Napier, T.C. Amphetamine-induced place preference and conditioned motor sensitization requires activation of tyrosine kinase receptors in the hippocampus. J. Neurosci. 2006, 26, 11041–11051. [Google Scholar] [CrossRef]
  45. Bello, E.P.; Mateo, Y.; Gelman, D.M.; Noaín, D.; Shin, J.H.; Low, M.J.; Alvarez, V.A.; Lovinger, D.M.; Rubinstein, M. Cocaine supersensitivity and enhanced motivation for reward in mice lacking dopamine D2 autoreceptors. Nat. Neurosci. 2011, 14, 1033–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Pak, A.C.; Ashby, C.R., Jr.; Heidbreder, C.A.; Pilla, M.; Gilbert, J.; Xi, Z.X.; Gardner, E.L. The selective dopamine D3 receptor antagonist SB-277011A reduces nicotine-enhanced brain reward and nicotine-paired environmental cue functions. Int. J. Neuropsychopharmacol. 2006, 9, 585–602. [Google Scholar] [CrossRef]
  47. Bisagno, V.; González, B.; Urbano, F.J. Cognitive enhancers versus addictive psychostimulants: The good and bad side of dopamine on prefrontal cortical circuits. Pharmacol. Res. 2016, 109, 108–118. [Google Scholar] [CrossRef] [PubMed]
  48. Nestler, H.P. International Proteolysis Society—Drug discovery and application. iDrugs 2001, 4, 1365–1367. [Google Scholar] [PubMed]
  49. Barr, J.L.; Unterwald, E.M. Glycogen synthase kinase-3 signaling in cellular and behavioral responses to psychostimulant drugs. Biochim. Biophys. Acta Mol. Cell Res. 2020, 1867, 118746. [Google Scholar] [CrossRef]
  50. Jope, R.S.; Johnson, G.V. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem. Sci. 2004, 29, 95–102. [Google Scholar] [CrossRef]
  51. Woodgett, J.R. Judging a protein by more than its name: GSK-3. Sci. STKE 2001, 2001, re12. [Google Scholar] [CrossRef]
  52. Wang, Q.M.; Fiol, C.J.; DePaoli-Roach, A.A.; Roach, P.J. Glycogen synthase kinase-3 beta is a dual specificity kinase differentially regulated by tyrosine and serine/threonine phosphorylation. J. Biol. Chem. 1994, 269, 14566–14574. [Google Scholar] [CrossRef]
  53. Hughes, K.; Nikolakaki, E.; Plyte, S.E.; Totty, N.F.; Woodgett, J.R. Modulation of the glycogen synthase kinase-3 family by tyrosine phosphorylation. EMBO J. 1993, 12, 803–808. [Google Scholar] [CrossRef]
  54. Hur, E.M.; Zhou, F.Q. GSK3 signalling in neural development. Nat. Rev. Neurosci. 2010, 11, 539–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Beurel, E.; Grieco, S.F.; Jope, R.S. Glycogen synthase kinase-3 (GSK3): Regulation, actions, and diseases. Pharmacol. Ther. 2015, 148, 114–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Xu, C.M.; Wang, J.; Wu, P.; Xue, Y.X.; Zhu, W.L.; Li, Q.Q.; Zhai, H.F.; Shi, J.; Lu, L. Glycogen synthase kinase 3β in the nucleus accumbens core is critical for methamphetamine-induced behavioral sensitization. J. Neurochem. 2011, 118, 126–139. [Google Scholar] [CrossRef]
  57. Duda, P.; Hajka, D.; Wójcicka, O.; Rakus, D.; Gizak, A. GSK3β: A Master Player in Depressive Disorder Pathogenesis and Treatment Responsiveness. Cells 2020, 9, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Hong, M.; Lee, V.M. Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J. Biol. Chem. 1997, 272, 19547–19553. [Google Scholar] [CrossRef] [Green Version]
  59. Beaulieu, J.M.; Sotnikova, T.D.; Yao, W.D.; Kockeritz, L.; Woodgett, J.R.; Gainetdinov, R.R.; Caron, M.G. Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade. Proc. Natl. Acad. Sci. USA 2004, 101, 5099–5104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Gould, T.D.; Manji, H.K. Glycogen synthase kinase-3: A putative molecular target for lithium mimetic drugs. Neuropsychopharmacology 2005, 30, 1223–1237. [Google Scholar] [CrossRef] [Green Version]
  61. Koros, E.; Dorner-Ciossek, C. The role of glycogen synthase kinase-3beta in schizophrenia. Drug News Perspect. 2007, 20, 437–445. [Google Scholar] [CrossRef]
  62. Lei, G.; Xia, Y.; Johnson, K.M. The role of Akt-GSK-3beta signaling and synaptic strength in phencyclidine-induced neurodegeneration. Neuropsychopharmacology 2008, 33, 1343–1353. [Google Scholar] [CrossRef] [Green Version]
  63. Solís, O.; García-Sanz, P.; Martín, A.B.; Granado, N.; Sanz-Magro, A.; Podlesniy, P.; Trullas, R.; Murer, M.G.; Maldonado, R.; Moratalla, R. Behavioral sensitization and cellular responses to psychostimulants are reduced in D2R knockout mice. Addict. Biol. 2021, 26, e12840. [Google Scholar] [CrossRef] [PubMed]
  64. Anderson, S.M.; Pierce, R.C. Cocaine-induced alterations in dopamine receptor signaling: Implications for reinforcement and reinstatement. Pharmacol. Ther. 2005, 106, 389–403. [Google Scholar] [CrossRef] [PubMed]
  65. Cervo, L.; Samanin, R. Effects of dopaminergic and glutamatergic receptor antagonists on the acquisition and expression of cocaine conditioning place preference. Brain Res. 1995, 673, 242–250. [Google Scholar] [CrossRef]
  66. Kim, H.S.; Park, W.K.; Jang, C.G.; Oh, S. Inhibition by MK-801 of cocaine-induced sensitization, conditioned place preference, and dopamine-receptor supersensitivity in mice. Brain Res. Bull. 1996, 40, 201–207. [Google Scholar] [CrossRef]
  67. Luo, H.R.; Hattori, H.; Hossain, M.A.; Hester, L.; Huang, Y.; Lee-Kwon, W.; Donowitz, M.; Nagata, E.; Snyder, S.H. Akt as a mediator of cell death. Proc. Natl. Acad. Sci. USA 2003, 100, 11712–11717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Linke, S.E.; Ussher, M. Exercise-based treatments for substance use disorders: Evidence, theory, and practicality. Am. J. Drug Alcohol Abus. 2015, 41, 7–15. [Google Scholar] [CrossRef]
  69. Kim, S.; Jang, E.Y.; Song, S.H.; Kim, J.S.; Ryu, I.S.; Jeong, C.H.; Lee, S. Brain Microdialysis Coupled to LC-MS/MS Revealed That CVT-10216, a Selective Inhibitor of Aldehyde Dehydrogenase 2, Alters the Neurochemical and Behavioral Effects of Methamphetamine. ACS Chem. Neurosci. 2021, 12, 1552–1562. [Google Scholar] [CrossRef]
  70. Gu, S.M.; Kim, S.Y.; Lamichhane, S.; Hong, J.T.; Yun, J. Limonene Inhibits Methamphetamine-Induced Sensitizations via the Regulation of Dopamine Receptor Supersensitivity. Biomol. Ther. 2019, 27, 357–362. [Google Scholar] [CrossRef]
  71. Wang, D.; Zhou, C.; Zhao, M.; Wu, X.; Chang, Y.K. Dose-response relationships between exercise intensity, cravings, and inhibitory control in methamphetamine dependence: An ERPs study. Drug Alcohol Depend. 2016, 161, 331–339. [Google Scholar] [CrossRef]
  72. Kemi, O.J.; Loennechen, J.P.; Wisløff, U.; Ellingsen, Ø. Intensity-controlled treadmill running in mice: Cardiac and skeletal muscle hypertrophy. J. Appl. Physiol. 2002, 93, 1301–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ko, M.J.; Mulia, G.E.; van Rijn, R.M. Commonly Used Anesthesia/Euthanasia Methods for Brain Collection Differentially Impact MAPK Activity in Male and Female C57BL/6 Mice. Front. Cell Neurosci. 2019, 13, 96. [Google Scholar] [CrossRef] [PubMed]
  74. Jiang, P.E.; Lang, Q.H.; Yu, Q.Y.; Tang, X.Y.; Liu, Q.Q.; Li, X.Y.; Feng, X.Z. Behavioral Assessments of Spontaneous Locomotion in a Murine MPTP-induced Parkinson’s Disease Model. J. Vis. Exp. 2019, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Brudzynski, S.M.; Krol, S. Analysis of locomotor activity in the rat: Parallelism index, a new measure of locomotor exploratory pattern. Physiol. Behav. 1997, 62, 635–642. [Google Scholar] [CrossRef]
  76. Crombag, H.S.; Bossert, J.M.; Koya, E.; Shaham, Y. Context-induced relapse to drug seeking: A review. Philos. Trans. R. Soc. B Biol. Sci. 2008, 363, 3233–3243. [Google Scholar] [CrossRef]
  77. Koob, G.F. Stress, corticotropin-releasing factor, and drug addiction. Ann. N. Y. Acad. Sci. 1999, 897, 27–45. [Google Scholar] [CrossRef]
  78. Knackstedt, L.A.; Kalivas, P.W. Glutamate and reinstatement. Curr. Opin. Pharmacol. 2009, 9, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Karler, R.; Calder, L.D.; Thai, L.H.; Bedingfield, J.B. A dopaminergic-glutamatergic basis for the action of amphetamine and cocaine. Brain Res. 1994, 658, 8–14. [Google Scholar] [CrossRef]
  80. McCreary, A.C.; Marsden, C.A. Cocaine-induced behaviour: Dopamine D1 receptor antagonism by SCH 23390 prevents expression of conditioned sensitisation following repeated administration of cocaine. Neuropharmacology 1993, 32, 387–391. [Google Scholar] [CrossRef]
  81. Choe, E.S.; Chung, K.T.; Mao, L.; Wang, J.Q. Amphetamine increases phosphorylation of extracellular signal-regulated kinase and transcription factors in the rat striatum via group I metabotropic glutamate receptors. Neuropsychopharmacology 2002, 27, 565–575. [Google Scholar] [CrossRef] [Green Version]
  82. Lett, B.T.; Grant, V.L.; Koh, M.T.; Flynn, G. Prior experience with wheel running produces cross-tolerance to the rewarding effect of morphine. Pharmacol. Biochem. Behav. 2002, 72, 101–105. [Google Scholar] [CrossRef]
  83. Smith, M.A.; Gergans, S.R.; Iordanou, J.C.; Lyle, M.A. Chronic exercise increases sensitivity to the conditioned rewarding effects of cocaine. Pharmacol. Rep. 2008, 60, 561–565. [Google Scholar]
  84. Epling, W.F.; Pierce, W.D. Solving the Anorexia Puzzle: A Scientific Approach; Hogrefe & Huber Pub.: Toronto, ON, Canada, 1992. [Google Scholar]
  85. Egorov, A.Y.; Szabo, A. The exercise paradox: An interactional model for a clearer conceptualization of exercise addiction. J. Behav. Addict. 2013, 2, 199–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kolb, E.M.; Kelly, S.A.; Garland, T., Jr. Mice from lines selectively bred for high voluntary wheel running exhibit lower blood pressure during withdrawal from wheel access. Physiol. Behav. 2013, 112–113, 49–55. [Google Scholar] [CrossRef] [PubMed]
  87. Perrine, S.A.; Sheikh, I.S.; Nwaneshiudu, C.A.; Schroeder, J.A.; Unterwald, E.M. Withdrawal from chronic administration of cocaine decreases delta opioid receptor signaling and increases anxiety- and depression-like behaviors in the rat. Neuropharmacology 2008, 54, 355–364. [Google Scholar] [CrossRef] [Green Version]
  88. Anraku, T.; Ikegaya, Y.; Matsuki, N.; Nishiyama, N. Withdrawal from chronic morphine administration causes prolonged enhancement of immobility in rat forced swimming test. Psychopharmacology 2001, 157, 217–220. [Google Scholar] [CrossRef]
  89. Malisch, J.L.; Breuner, C.W.; Kolb, E.M.; Wada, H.; Hannon, R.M.; Chappell, M.A.; Middleton, K.M.; Garland, T., Jr. Behavioral despair and home-cage activity in mice with chronically elevated baseline corticosterone concentrations. Behav. Genet. 2009, 39, 192–201. [Google Scholar] [CrossRef] [PubMed]
  90. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders: DSM-5, 5th ed.; American Psychiatric Association: Washingotn, DC, USA, 2013; p. 947. [Google Scholar]
  91. Mark, K.A.; Soghomonian, J.J.; Yamamoto, B.K. High-dose methamphetamine acutely activates the striatonigral pathway to increase striatal glutamate and mediate long-term dopamine toxicity. J. Neurosci. 2004, 24, 11449–11456. [Google Scholar] [CrossRef] [Green Version]
  92. Nash, J.F.; Yamamoto, B.K. Methamphetamine neurotoxicity and striatal glutamate release: Comparison to 3,4-methylenedioxymethamphetamine. Brain Res. 1992, 581, 237–243. [Google Scholar] [CrossRef]
  93. Krasnova, I.N.; Cadet, J.L. Methamphetamine toxicity and messengers of death. Brain Res. Rev. 2009, 60, 379–407. [Google Scholar] [CrossRef] [Green Version]
  94. Randall, R.D.; Thayer, S.A. Glutamate-induced calcium transient triggers delayed calcium overload and neurotoxicity in rat hippocampal neurons. J. Neurosci. 1992, 12, 1882–1895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Szatmari, E.; Habas, A.; Yang, P.; Zheng, J.J.; Hagg, T.; Hetman, M. A positive feedback loop between glycogen synthase kinase 3beta and protein phosphatase 1 after stimulation of NR2B NMDA receptors in forebrain neurons. J. Biol. Chem. 2005, 280, 37526–37535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Mulkey, R.M.; Endo, S.; Shenolikar, S.; Malenka, R.C. Involvement of a calcineurin/inhibitor-1 phosphatase cascade in hippocampal long-term depression. Nature 1994, 369, 486–488. [Google Scholar] [CrossRef]
  97. Peineau, S.; Taghibiglou, C.; Bradley, C.; Wong, T.P.; Liu, L.; Lu, J.; Lo, E.; Wu, D.; Saule, E.; Bouschet, T.; et al. LTP inhibits LTD in the hippocampus via regulation of GSK3beta. Neuron 2007, 53, 703–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Hernández, F.; Langa, E.; Cuadros, R.; Avila, J.; Villanueva, N. Regulation of GSK3 isoforms by phosphatases PP1 and PP2A. Mol. Cell Biochem. 2010, 344, 211–215. [Google Scholar] [CrossRef]
  99. Nagini, S.; Sophia, J.; Mishra, R. Glycogen synthase kinases: Moonlighting proteins with theranostic potential in cancer. Semin. Cancer Biol. 2019, 56, 25–36. [Google Scholar] [CrossRef]
  100. El Rawas, R.; Amaral, I.M.; Hofer, A. Is p38 MAPK Associated to Drugs of Abuse-Induced Abnormal Behaviors? Int. J. Mol. Sci. 2020, 21, 4833. [Google Scholar] [CrossRef]
  101. Valjent, E.; Corvol, J.C.; Pages, C.; Besson, M.J.; Maldonado, R.; Caboche, J. Involvement of the extracellular signal-regulated kinase cascade for cocaine-rewarding properties. J. Neurosci. 2000, 20, 8701–8709. [Google Scholar] [CrossRef]
  102. Choe, E.S.; Wang, J.Q. CaMKII regulates amphetamine-induced ERK1/2 phosphorylation in striatal neurons. Neuroreport 2002, 13, 1013–1016. [Google Scholar] [CrossRef]
  103. Berhow, M.T.; Hiroi, N.; Nestler, E.J. Regulation of ERK (extracellular signal regulated kinase), part of the neurotrophin signal transduction cascade, in the rat mesolimbic dopamine system by chronic exposure to morphine or cocaine. J. Neurosci. 1996, 16, 4707–4715. [Google Scholar] [CrossRef] [Green Version]
  104. Freeman, W.M.; Nader, M.A.; Nader, S.H.; Robertson, D.J.; Gioia, L.; Mitchell, S.M.; Daunais, J.B.; Porrino, L.J.; Friedman, D.P.; Vrana, K.E. Chronic cocaine-mediated changes in non-human primate nucleus accumbens gene expression. J. Neurochem. 2001, 77, 542–549. [Google Scholar] [CrossRef] [PubMed]
  105. Gerdjikov, T.V.; Ross, G.M.; Beninger, R.J. Place preference induced by nucleus accumbens amphetamine is impaired by antagonists of ERK or p38 MAP kinases in rats. Behav. Neurosci. 2004, 118, 740–750. [Google Scholar] [CrossRef] [Green Version]
  106. Pierce, R.C.; Pierce-Bancroft, A.F.; Prasad, B.M. Neurotrophin-3 contributes to the initiation of behavioral sensitization to cocaine by activating the Ras/Mitogen-activated protein kinase signal transduction cascade. J. Neurosci. 1999, 19, 8685–8695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Alkadhi, K.A. Exercise as a Positive Modulator of Brain Function. Mol. Neurobiol. 2018, 55, 3112–3130. [Google Scholar] [CrossRef] [PubMed]
  108. Damghani, F.; Bigdeli, I.; Miladi-Gorji, H.; Fadaei, A. Swimming exercise attenuates psychological dependence and voluntary methamphetamine consumption in methamphetamine withdrawn rats. Iran. J. Basic Med. Sci. 2016, 19, 594–600. [Google Scholar]
  109. Fontes-Ribeiro, C.A.; Marques, E.; Pereira, F.C.; Silva, A.P.; Macedo, T.R. May exercise prevent addiction? Curr. Neuropharmacol. 2011, 9, 45–48. [Google Scholar] [CrossRef] [Green Version]
  110. Greenwood, B.N.; Spence, K.G.; Crevling, D.M.; Clark, P.J.; Craig, W.C.; Fleshner, M. Exercise-induced stress resistance is independent of exercise controllability and the medial prefrontal cortex. Eur. J. Neurosci. 2013, 37, 469–478. [Google Scholar] [CrossRef] [Green Version]
  111. Williams, D.M.; Dunsiger, S.; Whiteley, J.A.; Ussher, M.H.; Ciccolo, J.T.; Jennings, E.G. Acute effects of moderate intensity aerobic exercise on affective withdrawal symptoms and cravings among women smokers. Addict. Behav. 2011, 36, 894–897. [Google Scholar] [CrossRef] [Green Version]
  112. Abrantes, A.M.; Battle, C.L.; Strong, D.R.; Ing, E.; Dubreuil, M.E.; Gordon, A.; Brown, R.A. Exercise preferences of patients in substance abuse treatment. Ment. Health Phys. Act. 2011, 4, 79–87. [Google Scholar] [CrossRef] [Green Version]
  113. Williams, D.M.; Whiteley, J.A.; Dunsiger, S.; Jennings, E.G.; Albrecht, A.E.; Ussher, M.H.; Ciccolo, J.T.; Parisi, A.F.; Marcus, B.H. Moderate intensity exercise as an adjunct to standard smoking cessation treatment for women: A pilot study. Psychol. Addict. Behav. 2010, 24, 349–354. [Google Scholar] [CrossRef] [Green Version]
  114. Aaron, D.J.; Dearwater, S.R.; Anderson, R.; Olsen, T.; Kriska, A.M.; Laporte, R.E. Physical activity and the initiation of high-risk health behaviors in adolescents. Med. Sci. Sports Exerc. 1995, 27, 1639–1645. [Google Scholar] [CrossRef]
  115. Pate, R.R.; Trost, S.G.; Levin, S.; Dowda, M. Sports participation and health-related behaviors among US youth. Arch. Pediatr. Adolesc. Med. 2000, 154, 904–911. [Google Scholar] [CrossRef] [PubMed]
  116. Kulig, K.; Brener, N.D.; McManus, T. Sexual activity and substance use among adolescents by category of physical activity plus team sports participation. Arch. Pediatr. Adolesc. Med. 2003, 157, 905–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Moore, M.J.; Werch, C.E. Sport and physical activity participation and substance use among adolescents. J. Adolesc. Health 2005, 36, 486–493. [Google Scholar] [CrossRef]
  118. Ströhle, A.; Höfler, M.; Pfister, H.; Müller, A.G.; Hoyer, J.; Wittchen, H.U.; Lieb, R. Physical activity and prevalence and incidence of mental disorders in adolescents and young adults. Psychol. Med. 2007, 37, 1657–1666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Korhonen, T.; Kujala, U.M.; Rose, R.J.; Kaprio, J. Physical activity in adolescence as a predictor of alcohol and illicit drug use in early adulthood: A longitudinal population-based twin study. Twin Res. Hum. Genet. 2009, 12, 261–268. [Google Scholar] [CrossRef]
  120. Ehringer, M.A.; Hoft, N.R.; Zunhammer, M. Reduced alcohol consumption in mice with access to a running wheel. Alcohol 2009, 43, 443–452. [Google Scholar] [CrossRef]
  121. Cosgrove, K.P.; Hunter, R.G.; Carroll, M.E. Wheel-running attenuates intravenous cocaine self-administration in rats: Sex differences. Pharmacol. Biochem. Behav. 2002, 73, 663–671. [Google Scholar] [CrossRef]
  122. Brocardo, P.S.; Boehme, F.; Patten, A.; Cox, A.; Gil-Mohapel, J.; Christie, B.R. Anxiety- and depression-like behaviors are accompanied by an increase in oxidative stress in a rat model of fetal alcohol spectrum disorders: Protective effects of voluntary physical exercise. Neuropharmacology 2012, 62, 1607–1618. [Google Scholar] [CrossRef]
  123. Peterson, A.B.; Hivick, D.P.; Lynch, W.J. Dose-dependent effectiveness of wheel running to attenuate cocaine-seeking: Impact of sex and estrous cycle in rats. Psychopharmacology 2014, 231, 2661–2670. [Google Scholar] [CrossRef] [PubMed]
  124. Zlebnik, N.E.; Saykao, A.T.; Carroll, M.E. Effects of combined exercise and progesterone treatments on cocaine seeking in male and female rats. Psychopharmacology 2014, 231, 3787–3798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Thanos, P.K.; Tucci, A.; Stamos, J.; Robison, L.; Wang, G.J.; Anderson, B.J.; Volkow, N.D. Chronic forced exercise during adolescence decreases cocaine conditioned place preference in Lewis rats. Behav. Brain. Res. 2010, 215, 77–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Smith, M.A.; Pitts, E.G. Access to a running wheel inhibits the acquisition of cocaine self-administration. Pharmacol. Biochem. Behav. 2011, 100, 237–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Smith, M.A.; Pitts, E.G. Wheel running decreases the positive reinforcing effects of heroin. Pharmacol. Rep. 2012, 64, 960–964. [Google Scholar] [CrossRef] [Green Version]
  128. Berchtold, N.C.; Kesslak, J.P.; Pike, C.J.; Adlard, P.A.; Cotman, C.W. Estrogen and exercise interact to regulate brain-derived neurotrophic factor mRNA and protein expression in the hippocampus. Eur. J. Neurosci. 2001, 14, 1992–2002. [Google Scholar] [CrossRef] [PubMed]
  129. Lynch, W.J.; Roth, M.E.; Carroll, M.E. Biological basis of sex differences in drug abuse: Preclinical and clinical studies. Psychopharmacology 2002, 164, 121–137. [Google Scholar] [CrossRef] [PubMed]
  130. Becker, J.B.; Hu, M. Sex differences in drug abuse. Front. Neuroendocr. 2008, 29, 36–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic view of the experimental design. METH, methamphetamine; D, day; w/o, without; w/, with.
Figure 1. Schematic view of the experimental design. METH, methamphetamine; D, day; w/o, without; w/, with.
Ijms 22 08203 g001
Figure 2. Forced moderate endurance exercise significantly reduced methamphetamine (METH)-induced hyperactivity. (A) Changes in food consumption (n = 8 per group, F(39, 261) = 1.275). (B) Changes in body weight (n = 8 per group, F(39, 248) = 1.744). (C) Total distance covered by the animal during the locomotor test conducted for 90 min (n = 7 per group, F(3, 24) = 67.96). (D) The classification ratio according to the movement speed within the total travel time (n = 7 per group). Values are means ± SE, *** p < 0.001, ** p < 0.01. CON, saline control; METH, methamphetamine; MA, METH administration; Ex, saline + exercise; MEx, METH + exercise.
Figure 2. Forced moderate endurance exercise significantly reduced methamphetamine (METH)-induced hyperactivity. (A) Changes in food consumption (n = 8 per group, F(39, 261) = 1.275). (B) Changes in body weight (n = 8 per group, F(39, 248) = 1.744). (C) Total distance covered by the animal during the locomotor test conducted for 90 min (n = 7 per group, F(3, 24) = 67.96). (D) The classification ratio according to the movement speed within the total travel time (n = 7 per group). Values are means ± SE, *** p < 0.001, ** p < 0.01. CON, saline control; METH, methamphetamine; MA, METH administration; Ex, saline + exercise; MEx, METH + exercise.
Ijms 22 08203 g002
Figure 3. Forced moderate endurance exercise did not affect the methamphetamine (METH)-induced dopaminergic signaling pathway. Protein content was determined by Western blotting in the striatum of the brains of experimental rats. (A) Striatal dopamine level (n = 8 per group, F(3, 28) = 10.25). (B) Dopamine receptor D1 (DRD1) expression level (n = 8 per group, F(3, 28) = 0.2384). (C) Dopamine receptor D2 (DRD2) expression level (n = 8 per group, F(3, 28) = 1.347). (D) p(Ser743)/t-Akt ratio (n = 8 per group, F(3, 28) = 0.4629). (E) p(Thr308)/t-Akt ratio (n = 8 per group, F(3, 28) = 6.311). (F) p(Tyr279)/t-GSK-3α ratio (n = 8 per group, F(3, 28) = 3.433). (G) p(Tyr216)/t-GSK-3β ratio (n = 8 per group, (F(3, 22) = 4.072), (H) p(Ser9)/t-GSK-3β ratio (n = 8 per group, F(3, 28) = 3.241). Values are means ± SE, * p < 0.05, ** p < 0.01. AU, arbitrary unit; CON, saline control; METH, methamphetamine; MA, METH administration; Ex, saline + exercise; GSK-3, glycogen synthase kinase-3; MEx, METH + exercise.
Figure 3. Forced moderate endurance exercise did not affect the methamphetamine (METH)-induced dopaminergic signaling pathway. Protein content was determined by Western blotting in the striatum of the brains of experimental rats. (A) Striatal dopamine level (n = 8 per group, F(3, 28) = 10.25). (B) Dopamine receptor D1 (DRD1) expression level (n = 8 per group, F(3, 28) = 0.2384). (C) Dopamine receptor D2 (DRD2) expression level (n = 8 per group, F(3, 28) = 1.347). (D) p(Ser743)/t-Akt ratio (n = 8 per group, F(3, 28) = 0.4629). (E) p(Thr308)/t-Akt ratio (n = 8 per group, F(3, 28) = 6.311). (F) p(Tyr279)/t-GSK-3α ratio (n = 8 per group, F(3, 28) = 3.433). (G) p(Tyr216)/t-GSK-3β ratio (n = 8 per group, (F(3, 22) = 4.072), (H) p(Ser9)/t-GSK-3β ratio (n = 8 per group, F(3, 28) = 3.241). Values are means ± SE, * p < 0.05, ** p < 0.01. AU, arbitrary unit; CON, saline control; METH, methamphetamine; MA, METH administration; Ex, saline + exercise; GSK-3, glycogen synthase kinase-3; MEx, METH + exercise.
Ijms 22 08203 g003
Figure 4. Forced endurance exercise training significantly reduced the methamphetamine (METH)-induced striatal glutamatergic signaling pathway. Protein content was determined by Western blotting in the striatum of brains of experimental rats. (A) NMDA receptor 1 (NMDAr1) expression level (n = 8 per group, F(3, 27) = 6.718). (B) CaMKK2 expression level (n = 8 per group, F(3, 26) = 2.489). (C) PP1 expression level (n = 8 per group, F(3, 27) = 19.68). (D) p/t-p38MAPK ratio (n = 7 per group, F(3, 24) = 7.042). (E) p/t ERK1 ratio (n = 7 per group, F(3, 21) = 2.118). (F) p/t ERK2 ratio (n = 7 per group, F(3, 23) = 5.471). Values are means ± SE, * p < 0.05, ** p < 0.01, *** p < 0.001. AU, arbitrary unit; CON, saline control; METH, methamphetamine; MA, METH administration; Ex, saline + exercise training; MEx, METH + exercise; NMDA, n-methyl-D-aspartate; CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2; PPI, protein phosphatase 1; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase.
Figure 4. Forced endurance exercise training significantly reduced the methamphetamine (METH)-induced striatal glutamatergic signaling pathway. Protein content was determined by Western blotting in the striatum of brains of experimental rats. (A) NMDA receptor 1 (NMDAr1) expression level (n = 8 per group, F(3, 27) = 6.718). (B) CaMKK2 expression level (n = 8 per group, F(3, 26) = 2.489). (C) PP1 expression level (n = 8 per group, F(3, 27) = 19.68). (D) p/t-p38MAPK ratio (n = 7 per group, F(3, 24) = 7.042). (E) p/t ERK1 ratio (n = 7 per group, F(3, 21) = 2.118). (F) p/t ERK2 ratio (n = 7 per group, F(3, 23) = 5.471). Values are means ± SE, * p < 0.05, ** p < 0.01, *** p < 0.001. AU, arbitrary unit; CON, saline control; METH, methamphetamine; MA, METH administration; Ex, saline + exercise training; MEx, METH + exercise; NMDA, n-methyl-D-aspartate; CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2; PPI, protein phosphatase 1; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase.
Ijms 22 08203 g004
Figure 5. Summary of research results. METH, methamphetamine; NMDAr1, N-methyl-D-aspartate receptor 1; GSK, glycogen synthase kinase-3; MAPK, mitogen-activated protein kinase; CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2; PPI, protein phosphatase 1.
Figure 5. Summary of research results. METH, methamphetamine; NMDAr1, N-methyl-D-aspartate receptor 1; GSK, glycogen synthase kinase-3; MAPK, mitogen-activated protein kinase; CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2; PPI, protein phosphatase 1.
Ijms 22 08203 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jung, S.; Kim, Y.; Kim, M.; Seo, M.; Kim, S.; Kim, S.; Lee, S. Exercise Pills for Drug Addiction: Forced Moderate Endurance Exercise Inhibits Methamphetamine-Induced Hyperactivity through the Striatal Glutamatergic Signaling Pathway in Male Sprague Dawley Rats. Int. J. Mol. Sci. 2021, 22, 8203. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22158203

AMA Style

Jung S, Kim Y, Kim M, Seo M, Kim S, Kim S, Lee S. Exercise Pills for Drug Addiction: Forced Moderate Endurance Exercise Inhibits Methamphetamine-Induced Hyperactivity through the Striatal Glutamatergic Signaling Pathway in Male Sprague Dawley Rats. International Journal of Molecular Sciences. 2021; 22(15):8203. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22158203

Chicago/Turabian Style

Jung, Suryun, Youjeong Kim, Mingyu Kim, Minjae Seo, Suji Kim, Seungju Kim, and Sooyeun Lee. 2021. "Exercise Pills for Drug Addiction: Forced Moderate Endurance Exercise Inhibits Methamphetamine-Induced Hyperactivity through the Striatal Glutamatergic Signaling Pathway in Male Sprague Dawley Rats" International Journal of Molecular Sciences 22, no. 15: 8203. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22158203

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

Article Metrics

Back to TopTop