Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 718
Filter
1.
Neurochem Res ; 49(2): 415-426, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37864024

ABSTRACT

Morphine (MPH) is widely used for pain management; however, long-term MPH therapy results in antinociceptive tolerance and physical dependence, limiting its clinical use. Zingerone (ZIN) is a natural phenolic compound with neuroprotective effects. We investigated the effects of single and repeated doses of ZIN on MPH-induced tolerance, dependence, and underlying biochemical mechanisms. After a dose-response experiment, tolerance was developed to MPH (10 mg/kg, i.p.) for seven days. In the single-dose study, ZIN was administered on day seven. In the repeated-dose study, ZIN was administered for seven days. Naloxone (5 mg/kg, i.p., 120 min after MPH) was injected to assess withdrawal signs on day seven. The levels of thiobarbituric acid reactive substances (TBARS), nitric oxide (NO), total thiol (TT), and glutathione peroxidase (GPx) were measured in the prefrontal cortex. The protein levels of interleukin-1 beta (IL-1ß) and NLRP3-ASC-Caspase-1 axis were assessed by ELISA and Western blotting, respectively. Results showed that ZIN (100 mg/kg) had no antinociceptive activity, and subsequent experiments were performed at this dose. Repeated ZIN reversed MPH antinociceptive tolerance, whereas single ZIN did not. Single and repeated ZIN attenuated naloxone-induced jumping. In addition, repeated ZIN significantly inhibited weight loss. Repeated ZIN suppressed the MPH-induced increase in TBARS, NO, IL-1ß, NLRP3, ASC, and Caspase-1. It also inhibited MPH-induced TT and GPx reduction. In contrast, single ZIN had no effect. Findings suggest that ZIN reduces MPH-induced tolerance and dependence by suppressing oxidative stress and NLRP3 inflammasome activation. This study provides a novel therapeutic approach to reduce the side effects of MPH.


Subject(s)
Guaiacol/analogs & derivatives , Morphine Dependence , Morphine , Mice , Animals , Morphine/pharmacology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Thiobarbituric Acid Reactive Substances , Naloxone/pharmacology , Naloxone/therapeutic use , Oxidative Stress , Nitric Oxide/metabolism , Analgesics/therapeutic use , Caspases/metabolism , Morphine Dependence/metabolism
2.
Pharmacol Biochem Behav ; 233: 173660, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37852327

ABSTRACT

Opioid addiction causes some molecular alterations in the brain reward pathway, such as changes in gene expression that may be transferred to the next generation via epigenetic mechanisms such as histone acetylation. This study aimed to evaluate the effect of theophylline as an HDAC (Histone deacetylases) activator on D1 and D2 dopamine receptor expression in the nucleus accumbens (NAc) and anxiety behavior in the offspring of morphine-dependent female rats. Female rats were exposed to escalating doses of morphine for six days and were then treated with theophylline (20 mg/kg) or saline for 10 days before mating with normal male rats. Male and female offspring were tested for anxiety behavior using an elevated plus maze apparatus. Besides, the expression of D1 and D2 dopamine receptors in the NAc was evaluated by real-time PCR (polymerase chain reaction). Results showed that offspring of morphine-dependent female rats had increased expression of both D1 and D2 receptors in the NAc, as well as decreased anxiety behavior, compared to control offspring. However, the mentioned effects were returned to normal levels in the offspring whose morphine-dependent mothers had received theophylline for 10 days before mating. It is concluded that theophylline may be therapeutically effective in minimizing the adverse consequences of maternal morphine dependence on offspring behavior by restoring normal dopamine receptor expression levels and modulating anxiety. To completely comprehend the underlying mechanisms of this phenomenon, more research is required.


Subject(s)
Morphine Dependence , Rats , Male , Female , Animals , Morphine Dependence/metabolism , Theophylline/pharmacology , Morphine/adverse effects , Anxiety/prevention & control , Anxiety/etiology , Anxiety Disorders , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism
3.
Brain Behav Immun ; 113: 328-339, 2023 10.
Article in English | MEDLINE | ID: mdl-37543246

ABSTRACT

Chronic morphine exposure causes the development of addictive behaviors, accompanied by an increase in neuroinflammation in the central nervous system. While previous researches have shown that astrocytes contribute to brain diseases, the role of astrocyte in morphine addiction through induced neuroinflammation remain unexplored. Here we show that morphine-induced inflammation requires the crosstalk among neuron, astrocyte, and microglia. Specifically, astrocytes respond to morphine-induced neuronal activation by increasing glycolytic metabolism. The dysregulation of glycolysis leads to an increased in the generation of mitochondrial reactive oxygen species and causes excessive mitochondrial fragmentation in astrocytes. These fragmented, dysfunctional mitochondria are consequently released into extracellular environment, leading to activation of microglia and release of inflammatory cytokines. We also found that blocking the nicotinamide adenine dinucleotide salvage pathway with FK866 could inhibit astrocytic glycolysis and restore the mitochondrial homeostasis and effectively attenuate neuroinflammatory responses. Importantly, FK866 reversed morphine-induced addictive behaviors in mice. In summary, our findings illustrate an essential role of astrocytic immunometabolism in morphine induced neural and behavioral plasticity, providing a novel insight into the interactions between neurons, astrocytes, and microglia in the brain affected by chronic morphine exposure.


Subject(s)
Morphine Dependence , Mice , Animals , Morphine Dependence/metabolism , Astrocytes/metabolism , Neuroinflammatory Diseases , Morphine/pharmacology , Morphine/metabolism , Microglia/metabolism , Mitochondria
4.
Cells ; 12(15)2023 08 01.
Article in English | MEDLINE | ID: mdl-37566064

ABSTRACT

In addition to the essential pharmacological effects of opioids, situational cues associated with drug addiction memory are key triggers for drug seeking. CircRNAs, an emerging hotspot regulator in crown genetics, play an important role in central nervous system-related diseases. However, the internal mediating mechanism of circRNAs in the field of drug reward and addiction memory remains unknown. Here, we trained mice on a conditional place preference (CPP) model and collected nucleus accumbens (NAc) tissues from day 1 (T0) and day 8 (T1) for high-throughput RNA sequencing. QRT-PCR analysis revealed that circTmeff-1 was highly expressed in the NAc core but not in the NAc shell, suggesting that it plays a role in addiction memory formation. Meanwhile, the down-regulation of circTmeff-1 by adeno-associated viruses in the NAc core or shell could inhibit the morphine CPP scores. Subsequently, the GO and KEGG analyses indicated that circTmeff-1 might regulate the addiction memory via the MAPK and AMPK pathways. These findings suggest that circTmeff-1 in NAc plays a crucial role in morphine-dependent memory formation.


Subject(s)
Morphine Dependence , Mice , Animals , Morphine Dependence/metabolism , RNA, Circular/metabolism , Morphine/pharmacology , Analgesics, Opioid/pharmacology , Nucleus Accumbens/metabolism
5.
Article in English | MEDLINE | ID: mdl-37348641

ABSTRACT

Although the negative effects coming along with opiate withdrawal are in part modulated by L-type calcium channels (LTCCs), the distinctive physiological properties and functions of LTCCs subtypes suggest differential roles of subtypes during withdrawal. The present study aimed to examine the contributions of LTCC subtypes, Cav1.2 and Cav1.3, within the dorsal hippocampus (DH) in naloxone-precipitated morphine withdrawal using the conditioned place aversion (CPA) paradigm. Firstly, we injected the non-specific LTCCs antagonist verapamil into the DH of morphine-dependent rats before conditioning an environment with naloxone-precipitated withdrawal. Our results showed that verapamil blocked the acquisition of CPA. Then, to explore the molecular mechanisms of LTCCs subtypes during withdrawal, we measured the protein expression of Cav1.2 and Cav1.3 in morphine-dependent rats under different conditions. In morphine-dependent rats, conditioning with withdrawal increased Cav1.2 expression in the membrane, while only acute naloxone injection increased the membrane expression of Cav1.3. To further determine the causal roles of LTCCs subtypes in the withdrawal process, we used Cav1.2 siRNA or Cav1.3 shRNA to knock down the expression of subtypes and detected the effects on CPA and somatic withdrawal signs in morphine-dependent rats. Cav1.2 siRNA, but not Cav1.3 shRNA, inhibited the acquirement of CPA and relieved somatic withdrawal symptoms. Together, our findings reveal that Cav1.2, but not Cav1.3 plays an important role in mediating morphine withdrawal, suggesting this subtype may serve as a potential therapeutic target for the treatment of negative effects in opiate dependence.


Subject(s)
Morphine Dependence , Substance Withdrawal Syndrome , Rats , Animals , Morphine/pharmacology , Calcium Channels, L-Type , Naloxone/pharmacology , Morphine Dependence/metabolism , Hippocampus/metabolism , Substance Withdrawal Syndrome/metabolism , Verapamil/pharmacology , Narcotic Antagonists/pharmacology , Avoidance Learning
6.
Behav Brain Res ; 450: 114504, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37209879

ABSTRACT

The misuse of and addiction to opioids are serious public health problems in some countries, such as the USA. Drug addiction is a chronic and relapsing medical condition that involves motivational and memory-related processes due to the strong associations between drugs and consuming-related stimuli. These stimuli usually trigger continuous and compulsive use and are associated with relapses after periods of withdrawal. Several factors contribute to relapse, including withdrawal-induced mood changes. Therefore, drugs attenuating withdrawal-induced affective alterations could be useful alternative treatments for relapse prevention. Cannabidiol (CBD), a non-psychotomimetic component from the Cannabis sativa plant, has anti-anxiety and anti-stress properties and has been investigated as an alternative for the treatment of several mental disorders, including drug addiction. Here, we evaluated if CBD administered 30 min prior to test for a conditioned place aversion (CPA) would attenuate the aversion induced by morphine withdrawal precipitated by the opioid receptor antagonist naloxone in male C57BL/6 mice. We also investigated if this effect involves the activation of 5-HT1A receptors, a mechanism previously associated with CBD anti-aversive effects. As expected, morphine-treated mice spent less time exploring the compartment paired with the naloxone-induced withdrawal, indicating a CPA induced by naloxone-precipitated morphine withdrawal. This effect was not observed in animals treated with CBD, at 30 and 60 mg/kg, prior to the CPA test, indicating that CBD attenuated the expression of CPA induced by naloxone-precipitated morphine withdrawal. Pretreatment with the 5-HT1A receptor antagonist WAY100635 (0.3 mg/kg) blocked CBD effects. Our findings suggest that CBD may reduce the expression of a previously established conditioned aversion induced by morphine withdrawal by a mechanism involving the activation of 5-HT1A receptors. Thus, CBD may be a therapeutic alternative for preventing relapse to opioid addiction by decreasing withdrawal-induced negative affective changes.


Subject(s)
Cannabidiol , Morphine Dependence , Substance Withdrawal Syndrome , Mice , Animals , Naloxone/pharmacology , Morphine/adverse effects , Cannabidiol/pharmacology , Receptor, Serotonin, 5-HT1A , Avoidance Learning , Substance Withdrawal Syndrome/metabolism , Mice, Inbred C57BL , Narcotic Antagonists/pharmacology , Morphine Dependence/drug therapy , Morphine Dependence/metabolism
7.
Behav Brain Res ; 437: 114122, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36174840

ABSTRACT

Gender differences have been observed in the vulnerability to drug abuse and in the different stages of the addictive process. In opiate dependence, differences between sexes have been shown in humans and laboratory animals in various phases of opiate addiction, especially in withdrawal-associated negative affective states. Using a Y-maze conditioned place aversion paradigm, we investigated potential sex differences in the expression and extinction of the aversive memory of precipitated opiate withdrawal state in morphine-dependent rats. No significant difference between sexes was observed in the occurrence of withdrawal signs following naloxone injection during conditioning. Moreover, opiate withdrawal memory expression and extinction following repeated testing was demonstrated in both male and female rats, with no significant differences between sexes. Finally, we report spontaneous recovery following extinction of opiate withdrawal memory. Altogether these data provide further evidence that persistent withdrawal-related memories may be strong drivers of opiate dependence, and demonstrate that both males and females can be used in experimental rodent cohorts to better understand opiate-related effects, reward, aversive state of withdrawal, abstinence and relapse.


Subject(s)
Morphine Dependence , Opiate Alkaloids , Opioid-Related Disorders , Substance Withdrawal Syndrome , Humans , Rats , Animals , Female , Male , Substance Withdrawal Syndrome/metabolism , Avoidance Learning , Naloxone/pharmacology , Analgesics, Opioid/pharmacology , Morphine Dependence/metabolism , Morphine/pharmacology , Narcotic Antagonists/pharmacology
8.
Glia ; 70(7): 1289-1300, 2022 07.
Article in English | MEDLINE | ID: mdl-35275429

ABSTRACT

Mitochondria are abundant in the fine processes of astrocytes, however, potential roles for astrocyte mitochondria remain poorly understood. In the present study, we performed a systematic examination of the effects of abnormal oxidative phosphorylation in astrocytes on several mouse behaviors. Impaired astrocyte oxidative phosphorylation was produced by astrocyte-specific deletion of the nuclear mitochondrial gene, Cox10, that encodes an accessory protein of complex IV, the protoheme:heme-O-farnesyl transferase. As expected, conditional deletion of the Cox10 gene in mice (cKO mice) significantly reduced expression of COX10 and Cytochrome c oxidase subunit I (MTCO1) of Complex IV, resulting in decreased oxidative phosphorylation without significantly affecting glycolysis. No effects of the deletion were observed on locomotor activity, anxiety-like behavior, nociception, or spontaneous alternation. Cox10 cKO female mice exhibited mildly impaired novel object recognition, while Cox10 cKO male mice were moderately deficient in trace fear conditioning. No group-related changes were observed in conditional place preference (CPP) that assessed effects of morphine on reward. In contrast to CPP, Cox10 cKO mice demonstrated significantly increased aversive behaviors produced by naloxone-precipitated withdrawal following chronic exposure to morphine, that is, jumping and avoidance behavior as assessed by conditional place aversion (CPA). Our study suggests that astrocyte oxidative phosphorylation may contribute to behaviors associated with greater cognitive load and/or aversive and stressful conditions.


Subject(s)
Alkyl and Aryl Transferases , Morphine Dependence , Substance Withdrawal Syndrome , Alkyl and Aryl Transferases/metabolism , Animals , Astrocytes/metabolism , Fear , Female , Male , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Morphine/metabolism , Morphine/pharmacology , Morphine Dependence/metabolism , Morphine Dependence/psychology , Naloxone/metabolism , Naloxone/pharmacology , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Respiration , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
9.
Neurochem Int ; 155: 105304, 2022 05.
Article in English | MEDLINE | ID: mdl-35176438

ABSTRACT

Various impacts of exercise on brain performance following the induction of morphine dependence have been documented; however, the underlying neuronal mechanisms are still unclear. The present research was done to investigate the impact of different exercise training modes on apoptosis, neuronal maturation, and synaptic plasticity in the perforant pathway (PP)-dentate gyrus (DG) synapses in the morphine-dependent rats. Five groups, including a control group (Con, ten healthy rats) and forty morphine-dependent rats were considered as follows (n = 10/group): 1) sedentary-dependent (Sed-D); 2) endurance exercise-dependent (En-D); 3) strength exercise-dependent (St-D); and 4) concurrent exercise-dependent (Co-D). The exercise training groups were subjected to endurance, strength, and concurrent training five days a week for ten weeks. After training sessions, the field excitatory postsynaptic potential (fEPSP) slope and population spike (PS) amplitude in the DG were determined in response to high-frequency stimulation (HFS) of the PP. For assessing neurogenesis and apoptosis, NeuroD and Caspase-3 expression levels were evaluated after all experiments. Concurrent training increased PS amplitude and EPSP compared to the control group. NeuroD in the morphine-dependent rats significantly decreased, but concurrent training returned the NeuroD to its levels in healthy rats. Furthermore, Caspase-3 expression levels in morphine-dependent rats remarkably increased and concurrent training significantly reduced Caspase-3 expression levels compared to the Sed-D group. Concurrent training can ameliorate synaptic plasticity impairment in morphine-dependent rats through neurogenesis promotion and apoptosis reduction. According to the results, concurrent training can be an appropriate novel candidate for treating opioid addiction.


Subject(s)
Morphine Dependence , Animals , Dentate Gyrus , Long-Term Potentiation , Morphine/pharmacology , Morphine Dependence/metabolism , Neuronal Plasticity/physiology , Rats , Rats, Wistar
10.
Neurosci Lett ; 774: 136519, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35151827

ABSTRACT

In the present study, to evaluate the role of the basolateral amygdala (BLA) in morphine addiction, the BLA was stimulated electrically, or inactivated temporarily using lidocaine. The electrical stimulation (ES) was delivered to BLA with low or high intensities (LI or HI: 25 or 150 µA, respectively), and five minutes before morphine administration with effective or ineffective doses, lidocaine was microinjected into the BLA. Using a 5-day conditioned place preference (CPP) paradigm, the dependence on morphine was evaluated. The results showed that LI-ES of BLA induced CPP in both the acquisition and expression phases, in the control and the ineffective dose of morphine groups. HI-ES had no effect on CPP acquisition but induced aversion in the expression, with both effective and ineffective doses of morphine. Inactivation of BLA using lidocaine, inhibited morphine-induced CPP in both acquisition and expression phases. The results of the present study indicate the prominent role of BLA in morphine addiction and dependence. Considering the contradictory results of different intensities of ES, it can be inferred that there are different neural circuits in this area of the brain, in relation to the reward responses.


Subject(s)
Basolateral Nuclear Complex , Morphine Dependence , Amygdala/physiology , Animals , Electric Stimulation , Lidocaine/pharmacology , Morphine/metabolism , Morphine/pharmacology , Morphine Dependence/metabolism , Rats , Rats, Wistar , Reward
11.
Neuropharmacology ; 208: 108986, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35134442

ABSTRACT

There is a lack of safe and effective non-opioid medications for the treatment of opioid addiction. Aquaporin-4 (AQP4), a water channel protein expressed in astrocytes, regulates the progression of neurological diseases. Our previous work demonstrated that AQP4 deficiency in mice attenuated morphine-induced physiological dependence. However, the role of AQP4 in the neurobiology of behaviours related to opioid addiction in mice remains unclear. Here, we report that Aqp4-knockout mice exhibited attenuated heroin consumption and heroin-seeking behaviours. Furthermore, Aqp4-knockout mice displayed diminished hyperactivity induced by morphine and heroin and subsequently showed dramatically inhibited morphine-induced behavioural sensitization. This attenuated hyperlocomotion to opioids was accompanied by a decreased dopamine response to the opioid-induced increase in the levels of extracellular dopamine in the NAc. In addition, Aqp4-knockout mice displayed upregulation of dopamine transporters in the striatum, suggesting a probable neurobiological mechanism for uptake of the extracellular dopamine. The present findings suggest that deficiency of AQP4 decreases opiate-induced drug seeking and taking behaviours, and AQP4 may be involved in the treatment of addiction. Therefore, the development of a pharmacological antagonist to AQP4 may be valuable to investigate as opioid addiction therapy.


Subject(s)
Aquaporin 4/metabolism , Behavior, Addictive , Morphine Dependence , Opioid-Related Disorders , Analgesics, Opioid , Animals , Aquaporin 4/genetics , Dopamine/metabolism , Heroin/pharmacology , Mice , Mice, Knockout , Morphine , Morphine Dependence/metabolism , Nucleus Accumbens , Opioid-Related Disorders/metabolism
12.
Behav Brain Res ; 421: 113732, 2022 03 12.
Article in English | MEDLINE | ID: mdl-34990697

ABSTRACT

Chronic morphine impairs cued fear extinction, which may contribute to the high prevalence of anxiety disorders and the replase of opiate addiction. This work investigated the effects of forced exercise with different intensities on cued fear extinction impairment and alternations of hippocampal BDNF and apoptotic proteins induced by chronic morphine. Rats were injected with bi-daily doses of morphine or saline for ten days and then received a cued or contextual fear conditioning training, which was followed by fear extinction training for four consecutive days. Cued, but the not contextual fear response was impaired in morphine-treated rats. Then, different saline or morphine-treated rats underwent forced exercise for 4-weeks with light, moderate or high intensities. Subsequently, rats received a cued fear conditioning followed by four days of extinction training, and the expression of hippocampal BDNF and apoptotic proteins was determined. A relatively long time after the last injection of morphine (35 days), rats again showed cued fear extinction failure and reduced hippocampal BDNF, which recovered by light and moderate, but not high exercise. Light and moderate, but not high-intensity treadmill exercise enhanced the up-regulation of Bcl-2 and down-regulation of the Bax proteins in both saline- and morphine-treated rats, which shifted the balance between pro-apoptotic and anti-apoptotic factors in favor of cell survival. These findings highlight the impact of exercise up to moderate intensity in the recovery of cued extinction failure, more likely via BDNF in addicted individuals.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Extinction, Psychological/physiology , Fear/physiology , Hippocampus/metabolism , Morphine Dependence , Physical Conditioning, Animal/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Behavior, Animal/physiology , Cues , Male , Morphine Dependence/metabolism , Morphine Dependence/physiopathology , Morphine Dependence/rehabilitation , Rats , Rats, Wistar
13.
Int J Neurosci ; 132(3): 283-295, 2022 Mar.
Article in English | MEDLINE | ID: mdl-32783781

ABSTRACT

BACKGROUND: Brain-derived neurotrophic factor (BDNF) mediates opiate dependence phenomenon. In the brain of morphine dependent animals BDNF level is controlled transcriptionally, however, post-transcriptional mechanisms of BDNF regulation in this context remain unknown. Regulation of mRNA by binding of specific proteins to the 3'-untranslated region (3'-UTR) is one of such mechanisms. Among RNA-binding proteins neuronal Hu antigen D (HuD) is the best characterized positive regulator of BDNF, however its involvement in opiate dependence remains obscure. We suggested that HuD binding to the BDNF 3'-UTR may be linked to changes in BDNF expression induced by morphine. The aim of this study was to investigate potential association of HuD with BDNF 3'-UTR in relation to BDNF expression (Exon- and 3'-UTR-specific mRNA variants and protein level) in the frontal cortex and midbrain of male Wistar rats after chronic morphine intoxication and spontaneous withdrawal in dependent animals. RESULTS: After chronic morphine intoxication but not during morphine withdrawal HuD binding to the long BDNF 3'-UTR in the frontal cortex decreased as compared with the corresponding control group, however after intoxication BDNF expression did not change. The level of BDNF Exon I as well as mature BDNF polypeptide increased in the frontal cortex upon morphine withdrawal, while no changes in HuD binding could be detected. CONCLUSION: Thus, contrary to the assumption, HuD-BDNF 3'-UTR interaction and BDNF expression in the frontal cortex differentially change in a manner dependent on the context of morphine action.


Subject(s)
Morphine Dependence , Substance Withdrawal Syndrome , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Frontal Lobe/metabolism , Male , Morphine/pharmacology , Morphine Dependence/genetics , Morphine Dependence/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism
14.
Behav Brain Res ; 419: 113688, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34843742

ABSTRACT

Serotonin neurotransmission has been implicated in behavior deficits that occur during protracted withdrawal from opioids. In addition, studies have highlighted multiple pathways whereby serotonin (5-HT) modulates energy homeostasis, however the underlying metabolic effects of opioid withdrawal have not been investigated. A key metabolic regulator that senses the energy status of the cell and regulates fuel availability is Adenosine Monophosphate-activated Protein Kinase (AMPK). To investigate the interaction between cellular metabolism and serotonin in modulating protracted abstinence from morphine, we depleted AMPK in serotonin neurons. Morphine exposure via drinking water generates dependence in these mice, and both wildtype and serotonergic AMPK knockout mice consume similar amounts of morphine with no changes in body weight. Serotonergic AMPK contributes to baseline differences in open field and social interaction behaviors and blocks abstinence induced reductions in immobility following morphine withdrawal in the tail suspension test. Lastly, morphine locomotor sensitization is blunted in mice lacking AMPK in serotonin neurons. Taken together, our results suggest serotonergic AMPK mediates both baseline and protracted morphine withdrawal-induced behaviors.


Subject(s)
AMP-Activated Protein Kinase Kinases/metabolism , Morphine Dependence/metabolism , Morphine Dependence/physiopathology , Serotonergic Neurons/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
15.
Reprod Toxicol ; 105: 175-183, 2021 10.
Article in English | MEDLINE | ID: mdl-34517100

ABSTRACT

Chronic morphine exerts deleterious effects on testicular function through either suppression of germ cells or somatic including Sertoli cells, probably through the activation of inflammatory, oxidative, and apoptosis biomarkers. Thus, the present study aimed to investigate whether the damaging effects of morphine dependence were reversed by the spontaneous morphine withdrawal or incubation with methadone and/or naloxone in Sertoli (TM4) cells using an in- vitro cell model of morphine dependence. Morphine dependence in TM4 cells was induced by increasing daily doses of morphine for 10 days and then maintained for two weeks in 5 µM. The cAMP levels were measured for an evaluation of morphine dependence. The cell viability and inflammatory, oxidative, apoptosis biomarkers, and glial cell-derived neurotrophic factor (GDNF) were measured after the end of treatment following the incubation of cells with methadone and naloxone and spontaneous withdrawal from morphine. We found that morphine dependence decreased cell viability, GDNF level and increased the levels of pro-oxidant, pro-inflammatory, and apoptotic biomarkers in TM4 cells, while spontaneous withdrawal from morphine and by naloxone decreased the levels of the biomarkers of pro-inflammatory and apoptotic in TM4 cells. Also, despite the low levels of pro-inflammatory factors following morphine withdrawal by methadone, it increased the cleaved/pro-caspase3 ratio in TM4 cells. This study showed that morphine dependence increased apoptosis probably via oxidative stress and inflammation pathways in TM4 cells. Also, it seems likely that spontaneous and naloxone withdrawal have beneficial consequences in the treatment of morphine dependence than methadone therapy, although they may require longer incubation periods.


Subject(s)
Morphine Dependence/metabolism , Sertoli Cells/metabolism , Substance Withdrawal Syndrome/metabolism , Analgesics, Opioid/pharmacology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Line , Cyclic AMP/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Inflammation , Male , Methadone/pharmacology , Mice , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oxidative Stress/drug effects , Oxidoreductases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Sertoli Cells/drug effects
16.
Neuropharmacology ; 198: 108764, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34450116

ABSTRACT

Self-control problems are a typical character of drug addiction and excessive food consumption and it has been shown that natural rewards and drugs of abuse share parts of the same neural substrate and reward processing in the brain. Different brain areas are involved in natural and drug reward processing including the mesolimbic pathway, amygdala, nucleus accumbens (NAc), and prefrontal cortex. Considering the important role of orexins in the addictive behavior and the presence of orexin-1 subtype receptors (Orx1R) in the medial prefrontal cortex (mPFC), this study investigated the role of mPFC in natural- and drug-reward seeking behaviors to deepen our understanding of possible similarities or differences. To induce food- or morphine-conditioned place preference (CPP), adult male Wistar rats underwent CPP testing and received intra-mPFC doses of SB334867 (3, 10, or 30 nM/0.5 µl DMSO 12%), as an Orx1R antagonist, during the acquisition or expression phases of the CPP test. Results indicated that microinjection of Orx1R antagonist into the mPFC had similar effects on both morphine- and food-induced CPP and attenuated CPP scores in the acquisition and expression phases of the CPP test. The data demonstrated that Orx1Rs in the mPFC regulate the reward-related effects of morphine- and food-induced reward.


Subject(s)
Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Food , Morphine Dependence/metabolism , Morphine/pharmacology , Orexin Receptors/metabolism , Animals , Benzoxazoles/pharmacology , Drug-Seeking Behavior , Male , Naphthyridines/pharmacology , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Reward , Urea/analogs & derivatives , Urea/pharmacology
17.
Neurochem Int ; 150: 105157, 2021 11.
Article in English | MEDLINE | ID: mdl-34390773

ABSTRACT

Besides their clinical application, chronic misuse of opioids has often been associated to drug addiction due to their addictive properties, underlying neuroadaptations of AMPA glutamate-receptor-dependent synaptic plasticity. Topiramate (TPM), an AMPAR antagonist, has been used to treat psychostimulants addiction, despite its harmful effects on memory. This study aimed to evaluate the effects of a novel topiramate nanosystem on molecular changes related to morphine reinstatement. Rats were previously exposed to morphine in conditioned place preference (CPP) paradigm and treated with topiramate-chitosan nanoparticles (TPM-CS-NP) or non-encapsulated topiramate in solution (S-TPM) during CPP extinction; following memory performance evaluation, they were re-exposed to morphine reinstatement. While morphine-CPP extinction was comparable among all experimental groups, TPM-CS-NP treatment prevented morphine reinstatement, preserving memory performance, which was impaired by both morphine-conditioning and S-TPM treatment. In the NAc, morphine increased D1R, D2R, D3R, DAT, GluA1 and MOR immunoreactivity. It also increased D1R, DAT, GluA1 and MOR in the dorsal hippocampus. TPM-CS-NP treatment decreased D1R, D3R and GluA1 and increased DAT in the NAc, decreasing GluA1 and increasing D2 and DAT in the dorsal hippocampus. Taken together, we may infer that TPM-CS-NP treatment was able to prevent the morphine reinstatement without memory impairment. Therefore, TPM-CS-NP may be considered an innovative therapeutic tool due to its property to prevent opioid reinstatement because it acts modifying both dopaminergic and glutamatergic neurotransmission, which are commonly related to morphine addiction.


Subject(s)
Chitosan/administration & dosage , Dopamine/metabolism , Glutamic Acid/metabolism , Morphine Dependence/metabolism , Nanoparticles/administration & dosage , Topiramate/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Therapy, Combination , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Memory/drug effects , Memory/physiology , Morphine/pharmacology , Morphine Dependence/prevention & control , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, Dopamine/metabolism
18.
Mediators Inflamm ; 2021: 9828995, 2021.
Article in English | MEDLINE | ID: mdl-34220336

ABSTRACT

Prenatal opioid exposure might disturb epigenetic programming in the brain of neonatal offspring with various consequences for gene expressions and behaviors. This study determined whether altered trimethylation of histone 3 at lysine 4 (H3K4me3) in the promoter of the tumor necrosis factor-α (tnf-α) gene with neural cell apoptosis was involved in the ventral-medial striatum, an important brain region for withdrawal symptoms, of neonatal rat offspring from morphine-addicted mothers. Female adult rats were injected with morphine before gestation and until 14 days after giving birth. On postnatal day 14 (P14), rat offspring from morphine-addicted mothers were subjected to an opioid-withdrawal protocol and were analyzed 2 or 8 h after administration of that protocol. Expressions of the TNF-α protein, H3K4me3 in the tnf-α promoter gene, and neural cell apoptosis within the ventral-medial striatum of neonatal rat offspring were evaluated. In the absence of significant opioid withdrawal (2 h after initiation of the opioid-withdrawal protocol on P14), prenatal morphine exposure led to increased levels of H3K4me3 in the tnf-α promoter gene, of the TNF-α protein, and of neural cell apoptosis within the ventral-medial striatum of neonatal rat offspring. Following opioid withdrawal (8 h after initiation of the opioid-withdrawal protocol on P14), differential expression of H3K4me3 in the tnf-α promoter gene locus and upregulation of the level of TNF-α protein expression were further enhanced in these offspring. In addition, increased levels of caspase-3 and neural cell apoptosis were also observed. Taken together, this study revealed that prenatal opioid exposure can activate an epigenetic histone mechanism which regulates proinflammatory factor generation, which hence, led to cell apoptotic damage within the ventral-medial striatum of neonatal rat offspring from morphine-addicted mothers. More importantly, the opioid-withdrawal episode may provide augmented effects for the abovementioned alterations and could lead to deleterious effects in the neonatal brain of such offspring.


Subject(s)
Apoptosis , Histones/metabolism , Morphine Dependence/metabolism , Morphine , Pregnancy, Animal , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/genetics , Analgesics, Opioid , Animals , Animals, Newborn , Brain/pathology , Caspase 3/metabolism , Corpus Striatum , Epigenesis, Genetic , Female , Maternal Exposure , Methylation , Pregnancy , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/pathology
19.
Behav Brain Res ; 414: 113478, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34302875

ABSTRACT

Morphine-induced analgesic tolerance and dependence are significant limits of pain control; however, the exact molecular mechanisms underlying morphine tolerance and dependence have remained unclear. The role of long non-coding RNAs (lncRNAs) in morphine tolerance and dependence is yet to be determined. We aimed to explore the association of specific lncRNAs expression in key brain reward regions after repeated injection of morphine. Male Wistar rats received subcutaneous injections of twice-daily morphine (10 mg/kg) or saline (1 mL/kg) for eight days. On day 8 of the repeated injections, induction of morphine analgesic tolerance and dependence was confirmed through a hotplate test and a naloxone-precipitated withdrawal analysis, respectively. Expression of H19, BC1, MIAT1, and MALAT1 lncRNAs was determined from the midbrain, striatum, hypothalamus, prefrontal cortex (PFC), and hippocampus by real-time PCR on day 8 of the repeated injections. The H19 expression was significantly different between morphine-treated and control saline-treated rats in all investigated areas except for the hippocampus. The BC1 expression significantly altered in the midbrain, hypothalamus, and hippocampus, but not in the striatum and PFC after repeated morphine treatment. The MIAT1 and MALAT1 expression site-specifically altered in the midbrain, hypothalamus, and striatum; however, no significant changes were detected in their expression in the PFC and hippocampus after repeated morphine treatment. We conclude that alterations in the expression of these lncRNAs in the brain reward regions especially in the midbrain, striatum and hypothalamus may have critical roles in the development of morphine dependence and tolerance, which need to be considered in future researches.


Subject(s)
Brain/metabolism , Drug Tolerance , Morphine Dependence/metabolism , RNA, Long Noncoding/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Disease Models, Animal , Male , Morphine/administration & dosage , Narcotics/administration & dosage , Rats , Rats, Wistar , Reward
20.
Exp Neurol ; 343: 113787, 2021 09.
Article in English | MEDLINE | ID: mdl-34153321

ABSTRACT

Opioid addiction can produce severe side effects including physical dependence and withdrawal. Perturbations of the gut microbiome have recently been shown to alter opioid-induced side-effects such as addiction, tolerance and dependence. In the present study, we investigated the influence of the gut microbiome on opioid withdrawal by evaluating the effects of fecal microbiota transplantation (FMT), antibiotic and probiotic treatments, and pharmacological inhibition of gut permeability in a mouse model of opioid dependence. Repeated intraperitoneal (i.p.) morphine treatment produced physical dependence that was quantified by measuring somatic signs of withdrawal (i.e. number of jumps) precipitated using the opioid antagonist naloxone. Morphine-dependent mice that received FMT from morphine-treated donor mice exhibited fewer naloxone-precipitated jumps compared to morphine-dependent counterparts receiving FMT from saline-treated donor mice. Microbial contents in the mouse cecum were altered by morphine treatment but were not differentially impacted by FMT. A broad-spectrum antibiotic cocktail (ABX) regimen reduced the bacterial load and attenuated naloxone-precipitated morphine withdrawal in morphine-dependent mice, whereas commercially available probiotic strains did not reliably alter somatic signs of opioid withdrawal. ML-7, a pharmacological inhibitor of gut permeability, reduced the morphine-induced increase in gut permeability in vivo but did not reliably alter somatic signs of naloxone-precipitated opioid withdrawal. Our results suggest that the gut microbiome impacts the development of physical dependence induced by chronic morphine administration, and that therapeutic manipulations of the gut microbiome may reduce opioid withdrawal.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Fecal Microbiota Transplantation/methods , Morphine Dependence/therapy , Naloxone/toxicity , Narcotic Antagonists/toxicity , Substance Withdrawal Syndrome/therapy , Analgesics, Opioid/administration & dosage , Animals , Combined Modality Therapy/methods , Male , Mice , Mice, Inbred C57BL , Morphine Dependence/drug therapy , Morphine Dependence/metabolism , Probiotics/administration & dosage , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...