Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Biochem Biophys Res Commun ; 720: 150076, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38772224

ABSTRACT

Chronic morphine withdrawal memory formation is a complex process influenced by various molecular mechanisms. In this study, we aimed to investigate the contributions of the basolateral amygdala (BLA) and complement component 1, q subcomponent-like 3 (C1QL3), a secreted and presynaptically targeted protein, to the formation of chronic morphine (repeat dosing of morphine) withdrawal memory using conditioned place aversion (CPA) and chemogenetic methods. We conducted experiments involving the inhibition of the BLA during naloxone-induced withdrawal to assess its impact on CPA scores, providing insights into the significance of the BLA in the chronic morphine memory formation process. We also examined changes in C1ql3/C1QL3 expression within the BLA following conditioning. Immunofluorescence analysis revealed the colocalization of C1QL3 and the G protein-coupled receptor, brain-specific angiogenesis inhibitor 3 (BAI3) in the BLA, supporting their involvement in synaptic development. Moreover, we downregulated C1QL3 expression in the BLA to investigate its role in chronic morphine withdrawal memory formation. Our findings revealed that BLA inhibition during naloxone-induced withdrawal led to a significant reduction in CPA scores, confirming the critical role of the BLA in this memory process. Additionally, the upregulation of C1ql3 expression within the BLA postconditioning suggested its participation in withdrawal memory formation. The colocalization of C1QL3 and BAI3 in the BLA further supported their involvement in synaptic development. Furthermore, downregulation of C1QL3 in the BLA effectively hindered chronic morphine withdrawal memory formation, emphasizing its pivotal role in this process. Notably, we identified postsynaptic density protein 95 (PSD95) as a potential downstream effector of C1QL3 during chronic morphine withdrawal memory formation. Blocking PSD95 led to a significant reduction in the CPA score, and it appeared that C1QL3 modulated the ubiquitination-mediated degradation of PSD95, resulting in decreased PSD95 protein levels. This study underscores the importance of the BLA, C1QL3 and PSD95 in chronic morphine withdrawal memory formation. It provides valuable insights into the underlying molecular mechanisms, emphasizing their significance in this intricate process.


Subject(s)
Basolateral Nuclear Complex , Disks Large Homolog 4 Protein , Memory , Morphine , Substance Withdrawal Syndrome , Animals , Morphine/pharmacology , Substance Withdrawal Syndrome/metabolism , Male , Mice , Memory/drug effects , Disks Large Homolog 4 Protein/metabolism , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/drug effects , Complement C1q/metabolism , Mice, Inbred C57BL , Naloxone/pharmacology
2.
Eur J Neurosci ; 59(12): 3422-3444, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679044

ABSTRACT

Drug dependence is characterized by a switch in motivation wherein a positively reinforcing substance can become negatively reinforcing. Put differently, drug use can transform from a form of pleasure-seeking to a form of relief-seeking. Ventral tegmental area (VTA) GABA neurons form an anatomical point of divergence between two double dissociable pathways that have been shown to be functionally implicated and necessary for these respective motivations to seek drugs. The tegmental pedunculopontine nucleus (TPP) is necessary for opiate conditioned place preferences (CPP) in previously drug-naïve rats and mice, whereas dopaminergic (DA) transmission in the nucleus accumbens (NAc) is necessary for opiate CPP in opiate-dependent and withdrawn (ODW) rats and mice. Here, we show that this switch in functional anatomy is contingent upon the gap junction-forming protein, connexin-36 (Cx36), in VTA GABA neurons. Intra-VTA infusions of the Cx36 blocker, mefloquine, in ODW rats resulted in a reversion to a drug-naïve-like state wherein the TPP was necessary for opiate CPP and where opiate withdrawal aversions were lost. Consistent with these data, conditional knockout mice lacking Cx36 in GABA neurons (GAD65-Cre;Cx36 fl(CFP)/fl(CFP)) exhibited a perpetual drug-naïve-like state wherein opiate CPP was always DA independent, and opiate withdrawal aversions were absent even in mice subjected to an opiate dependence and withdrawal induction protocol. Further, viral-mediated rescue of Cx36 in VTA GABA neurons was sufficient to restore their susceptibility to an ODW state wherein opiate CPP was DA dependent. Our findings reveal a functional role for VTA gap junctions that has eluded prevailing circuit models of addiction.


Subject(s)
Connexins , GABAergic Neurons , Gap Junction delta-2 Protein , Gap Junctions , Opioid-Related Disorders , Ventral Tegmental Area , Animals , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/drug effects , Connexins/metabolism , Connexins/genetics , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Gap Junctions/metabolism , Gap Junctions/drug effects , Male , Rats , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , Mefloquine/pharmacology , Mice , Rats, Sprague-Dawley , Pedunculopontine Tegmental Nucleus/metabolism , Pedunculopontine Tegmental Nucleus/drug effects
3.
Article in English | MEDLINE | ID: mdl-38630316

ABSTRACT

RATIONALE: Corticotropin-releasing factor (CRF), the apical stress-inducing hormone, exacerbates stress and addictive behaviors. TCAP-1 is a peptide that directly inhibits both CRF-mediated stress and addiction-related behaviors; however, the direct action of TCAP-1 on morphine withdrawal-associated behaviors has not previously been examined. OBJECTIVE: To determine whether TCAP-1 administration attenuates behavioral and physiological consequences of morphine withdrawal in mice. METHODS: Mice were administered via subcutaneous route TCAP-1 either before or after initial morphine exposure, after which jumping behavior was quantified to assess the effects of TCAP-1 on naloxone-precipitated morphine withdrawal. As a comparison, mice were treated with nonpeptide CRF1 receptor antagonist CP-154,526. In one experiment, plasma corticosterone (CORT) was also measured as a physiological stress indicator. RESULTS: Pretreatment with TCAP-1 (10-250 nmol/kg) before morphine treatment significantly inhibited the development of naloxone-precipitated withdrawal. TCAP-1 (250-500 nmol/kg) treatment administered after morphine treatment attenuated the behavioral expression of naloxone-precipitated withdrawal. TCAP-1 (250 nmol/kg) treatment during morphine treatment was more effective than the optimal dosing of CP-154,526 (20 mg/kg) at suppressing the behavioral expression of naloxone-precipitated withdrawal, despite similar reduction of withdrawal-induced plasma CORT level increases. CONCLUSIONS: These findings establish TCAP-1 as a potential therapeutic candidate for the prevention and treatment of morphine withdrawal.

4.
Brain Res ; 1835: 148930, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38604556

ABSTRACT

The anxiety caused by morphine protracted abstinence is considered to be an important factor contributes to drug-seeking and relapse. Endoplasmic reticulum (ER) stress plays important roles in many kinds of mental disorders including drug addiction and anxiety, but it is unclear whether ER stress is involved in anxiety-like behaviors induced by morphine withdrawal. In this study, by using behavioral test, western blot, immunofluorescence, electron transmission microscope, we found that: (1) Inhibition of endoplasmic reticulum stress by 4-Phenylbutyric acid (4-PBA) could attenuate anxiety-like behaviors induced by morphine withdrawal. (2) The endoplasmic reticulum stress-related proteins in the lateral habenula (LHb) but not in the nucleus accumbens (NAc), ventral pallidum (VP), basolateral amygdala (BLA) and CA1 of hippocampus was upregulated by morphine withdrawal, upregulation of endoplasmic reticulum stress-related proteins in the lateral habenula induced by morphine withdrawal was inhibited by 4-PBA. (3) Endoplasmic reticulum stress-related protein CHOP and eIF2α were expressed in neurons but not in microglia in the LHb. (4) Morphine withdrawal induced neuronal morphological change in the LHb, which was attenuated by 4-PBA.


Subject(s)
Anxiety , Endoplasmic Reticulum Stress , Morphine , Substance Withdrawal Syndrome , Animals , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Male , Morphine/pharmacology , Anxiety/metabolism , Anxiety/drug therapy , Substance Withdrawal Syndrome/metabolism , Mice , Phenylbutyrates/pharmacology , Morphine Dependence/metabolism , Neurons/metabolism , Neurons/drug effects , Mice, Inbred C57BL
5.
Br J Pharmacol ; 181(13): 2070-2084, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38523471

ABSTRACT

BACKGROUND AND PURPOSE: Kratom is a preparation from Mitragyna speciosa, which is used as a natural drug preparation for many purposes around the world. However, an overdose of Kratom may cause addiction-like problems including aversive withdrawal states resulting in cognitive impairments via unknown mechanisms. Its main psychoactive alkaloid is mitragynine, showing opioid-like properties. EXPERIMENTAL APPROACH: Here, we analysed the neuropharmacological effects of mitragynine compared with morphine withdrawal in rats and searched for a pharmacological treatment option that may reverse the occurring cognitive deficits that usually aggravate withdrawal. KEY RESULTS: We found that withdrawal from 14-day mitragynine (1-10 mg·kg-1·day-1) treatment caused dose-dependent behavioural withdrawal signs resembling those of morphine (5 mg·kg-1·day-1) withdrawal. However, mitragynine (5 and 10 mg·kg-1·day-1) withdrawal also induced impairments in a passive avoidance task. Mitragynine withdrawal not only reduced hippocampal field excitatory postsynaptic potential (fEPSP) amplitudes in basal synaptic transmission and long-term potentiation (LTP) but also reduced epigenetic markers, such as histone H3K9 and H4K12 expression. At the same time, it up-regulates HDAC2 expression. Targeting the epigenetic adaptations with the HDAC inhibitor, SAHA, reversed the effects of mitragynine withdrawal on epigenetic dysregulation, hippocampal input/output curves, paired-pulse facilitation, LTP and attenuated the cognitive deficit. However, SAHA amplified the effects of morphine withdrawal. CONCLUSION AND IMPLICATIONS: The data from this work show that changes in histone expression and downstream hippocampal plasticity may explain mitragynine, but not morphine, withdrawal behaviours and cognitive impairments. Thus, it may provide a new treatment approach for aversive Kratom/mitragynine withdrawal and addiction.


Subject(s)
Cognitive Dysfunction , Epigenesis, Genetic , Secologanin Tryptamine Alkaloids , Substance Withdrawal Syndrome , Animals , Secologanin Tryptamine Alkaloids/pharmacology , Substance Withdrawal Syndrome/drug therapy , Epigenesis, Genetic/drug effects , Male , Rats , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/chemically induced , Rats, Sprague-Dawley , Morphine/pharmacology , Mitragyna/chemistry , Behavior, Animal/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Avoidance Learning/drug effects , Dose-Response Relationship, Drug
6.
J Psychiatr Res ; 171: 185-196, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301534

ABSTRACT

Anxiety disorders, common symptoms during morphine withdrawal, are important negative reinforcement factors leading to relapse. Lateral habenula serves as a negative reinforcement center, however its role in morphine withdrawal-induced anxiety remains uncovered. The hyperpolarization activated cyclic nucleotide-gated (HCN) channels have been reported to be important in emotion processing and addiction, but the role of HCN in anxiety from drug protracted abstinence remains elusive. In this study, by using behavioral test, Western blot, immunofluorescence, electrophysiology and virus-mediated regulation of HCN, we found that: (1) Intra-LHb injection of selective HCN blocker ZD7288 alleviated anxiety-like behaviors in morphine protracted abstinent male mice. (2) The LHb neuronal activity was increased by morphine protracted abstinence. (3) LHb neurons were inhibited by ZD7288 and activated by 8-Br-cAMP respectively, which were enhanced by morphine withdrawal. (4) HCN1 in the LHb was upregulated by morphine withdrawal. (5) Virus-mediated overexpression of HCN1 in the LHb was sufficient to produce anxiety-like behaviors in male mice and virus-mediated knockdown of HCN1 in the LHb prevented the anxiety-like behaviors in male mice. The findings reveal that selective blockade of HCN1 channels in the LHb may represent a therapeutic approach to morphine withdrawal-induced anxiety.


Subject(s)
Habenula , Morphine , Mice , Male , Animals , Morphine/pharmacology , Habenula/physiology , Neurons , Anxiety/chemically induced , Anxiety/drug therapy , Anxiety Disorders
7.
Neurobiol Dis ; 186: 106279, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37661023

ABSTRACT

Opioid withdrawal generates extremely unpleasant physical symptoms and negative affective states. A rapid relief of opioid withdrawal-induced anxiety has obvious clinical relevance but has been rarely reported. We have shown that injection of ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) leads to a rapid alleviation of anxiety-like behaviors in male mice undergoing chronic morphine withdrawal. Here we investigated the contribution of nucleus accumbens shell (sNAc) parvalbumin (PV)-neurons to this process. Chronic morphine withdrawal was associated with higher intrinsic excitability of sNAc PV-neurons via reduced voltage-dependent potassium currents. Chemogenetic inhibition of sNAc PV-neurons reversed the enhanced excitability of PV-neurons and anxiety-like behaviors in these morphine withdrawal male mice, while activation of sNAc PV-neurons induced anxiety-like behaviors in naive male mice. (2R,6R)-HNK reversed the altered potassium currents and intrinsic excitability of sNAc PV-neurons. Our findings demonstrate an important contribution of sNAc PV-neurons to modulating morphine withdrawal-induced anxiety-like behaviors and rapid relief of anxiety-like behaviors by (2R,6R)-HNK, this newly identified target may have therapeutic potentials in treating opioid addiction and anxiety disorders.


Subject(s)
Ketamine , Male , Animals , Mice , Parvalbumins , Morphine , Analgesics, Opioid , Nucleus Accumbens , Anxiety/chemically induced , Anxiety/drug therapy , Anxiety Disorders , Neurons , Potassium
8.
Cells ; 12(10)2023 05 17.
Article in English | MEDLINE | ID: mdl-37408246

ABSTRACT

Astrocytes express mu/µ opioid receptors, but the function of these receptors remains poorly understood. We evaluated the effects of astrocyte-restricted knockout of µ opioid receptors on reward- and aversion-associated behaviors in mice chronically exposed to morphine. Specifically, one of the floxed alleles of the Oprm1 gene encoding µ opioid receptor 1 was selectively deleted from brain astrocytes in Oprm1 inducible conditional knockout (icKO) mice. These mice did not exhibit changes in locomotor activity, anxiety, or novel object recognition, or in their responses to the acute analgesic effects of morphine. Oprm1 icKO mice displayed increased locomotor activity in response to acute morphine administration but unaltered locomotor sensitization. Oprm1 icKO mice showed normal morphine-induced conditioned place preference but exhibited stronger conditioned place aversion associated with naloxone-precipitated morphine withdrawal. Notably, elevated conditioned place aversion lasted up to 6 weeks in Oprm1 icKO mice. Astrocytes isolated from the brains of Oprm1 icKO mice had unchanged levels of glycolysis but had elevated oxidative phosphorylation. The basal augmentation of oxidative phosphorylation in Oprm1 icKO mice was further exacerbated by naloxone-precipitated withdrawal from morphine and, similar to that for conditioned place aversion, was still present 6 weeks later. Our findings suggest that µ opioid receptors in astrocytes are linked to oxidative phosphorylation and they contribute to long-term changes associated with opioid withdrawal.


Subject(s)
Astrocytes , Morphine , Mice , Animals , Morphine/adverse effects , Receptors, Opioid , Narcotic Antagonists/pharmacology , Naloxone/pharmacology , Mice, Knockout , Receptors, Opioid, mu/genetics
9.
Brain Res ; 1813: 148428, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37263551

ABSTRACT

Protracted opioid withdrawal is considered to be a traumatic event with many adverse effects. However, little attention is paid to its consequences on the protein expression in the rat brain. A better understanding of the changes at the molecular level is essential for designing future innovative drug therapies. Our previous proteomic data indicated that long-term morphine withdrawal is associated with altered proteins functionally involved in energy metabolism, cytoskeletal changes, oxidative stress, apoptosis, or signal transduction. In this study, we selected peroxiredoxin II (PRX II) as a marker of oxidative stress, 14-3-3 proteins as adaptors, and creatine kinase-B (CK-B) as a marker of energy metabolism to detect their amounts in the brain cortex and hippocampus isolated from rats after 3-month (3 MW) and 6-month morphine withdrawal (6 MW). Methodically, our work was based on immunoblotting accompanied by 2D resolution of PRX II and 14-3-3 proteins. Our results demonstrate significant upregulation of PRX II in the rat brain cortex (3-fold) and hippocampus (1.3-fold) after 3-month morphine abstinence, which returned to the baseline six months since the drug was withdrawn. Interestingly, the level of 14-3-3 proteins was downregulated in both brain areas in 3 MW samples and remained decreased only in the brain cortex of 6 MW. Our findings suggest that the rat brain cortex and hippocampus exhibit the oxidative stress-induced vulnerability represented by compensatory upregulation of PRX II after three months of morphine withdrawal.


Subject(s)
Morphine Dependence , Substance Withdrawal Syndrome , Rats , Animals , Morphine/metabolism , 14-3-3 Proteins/metabolism , Up-Regulation , Proteomics , Peroxiredoxins/metabolism , Peroxiredoxins/pharmacology , Hippocampus/metabolism , Brain/metabolism , Substance Withdrawal Syndrome/metabolism
10.
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
11.
Psychoneuroendocrinology ; 151: 106080, 2023 05.
Article in English | MEDLINE | ID: mdl-36931057

ABSTRACT

One negative emotional state from morphine protracted abstinence is anxiety which can drive craving and relapse risk in opioid addicts. Although the orexinergic system has been reported to be important in mediating emotion processing and addiction, the role of orexinergic system in anxiety from drug protracted abstinence remains elusive. In this study, by using behavioral test, western blot, electrophysiology and virus-mediated regulation of orexin receptor 1 (OX1R), we found that: (1) Intraperitoneal and intra-VTA administration of a selective OX1R antagonist SB334867 alleviated anxiety-like behaviors in open field test (OFT) but not in elevated plus maze test (EPM) in morphine protracted abstinent male mice. (2) OX1R expression in the VTA was upregulated by morphine withdrawal. (3) Virus-mediated knockdown of OX1R in the VTA prevented morphine abstinence-induced anxiety-like behaviors and virus-mediated overexpression of OX1R in the VTA was sufficient to produce anxiety-like behaviors in male mice. (4) The VTA neuronal activity was increased significantly induced by morphine protracted abstinence, which was mediated by OX1R. (5) OX1R was widely distributed in the neuronal soma and processes of dopaminergic and non-dopaminergic neurons in the VTA. The findings revealed that the OX1R mediates morphine abstinence-induced anxiety-like behaviors and the VTA plays a critical role in this effect.


Subject(s)
Anxiety , Morphine , Mice , Male , Animals , Morphine/pharmacology , Morphine/metabolism , Orexin Receptors/metabolism , Anxiety/metabolism , Anxiety Disorders/metabolism , Neurons/metabolism
12.
Acta Pharmacol Sin ; 44(3): 538-545, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36127507

ABSTRACT

Aversive emotion of opioid withdrawal generates motivational state leading to compulsive drug seeking and taking. Kappa opioid receptor (KOR) and its endogenous ligand dynorphin have been shown to participate in the regulation of aversive emotion. In the present study, we investigated the role of dynorphin/KOR system in the aversive emotion following opioid withdrawal in acute morphine-dependent mice. We found that blockade of KORs before pairing by intracerebroventricular injection of KOR antagonist norBNI (20, 40 µg) attenuated the development of morphine withdrawal-induced conditioned place aversion (CPA) behavior. We further found that morphine withdrawal increased dynorphin A expression in the dorsal hippocampus, but not in the amygdala, prefrontal cortex, nucleus accumbens, and thalamus. Microinjection of norBNI (20 µg) into the dorsal hippocampus significantly decreased morphine withdrawal-induced CPA behavior. We further found that p38 MAPK was significantly activated in the dorsal hippocampus after morphine withdrawal, and the activation of p38 MAPK was blocked by pretreatment with norBNI. Accordingly, microinjection of p38 MAPK inhibitor SB203580 (5 µg) into the dorsal hippocampus significantly decreased morphine withdrawal-produced CPA behavior. This study demonstrates that upregulation of dynorphin/KOR system in the dorsal hippocampus plays a critical role in the formation of aversive emotion associated with morphine withdrawal, suggesting that KOR antagonists may have therapeutic value for the treatment of opioid withdrawal-induced mood-related disorders.


Subject(s)
Dynorphins , Substance Withdrawal Syndrome , Mice , Animals , Dynorphins/metabolism , Receptors, Opioid, kappa , Morphine , Analgesics, Opioid/pharmacology , Up-Regulation , Narcotic Antagonists/pharmacology , Hippocampus/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Int J Neuropsychopharmacol ; 25(9): 774-785, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35797010

ABSTRACT

BACKGROUND: Anxiety is a negative emotion that contributes to craving and relapse during drug withdrawal. Sirtuins 1 (SIRT1) has been reported to be critical in both negative emotions and drug addiction. However, it remains incompletely elucidated whether SIRT1 is involved in morphine withdrawal-associated anxiety. METHODS: We established a mouse model of anxiety-like behaviors induced by morphine withdrawal and then detected neuronal activity with immunofluorescence and mitochondrial morphology with electron microscopy, mitochondrial DNA contents with quantitative real-time PCR, and mitochondrial function with the ATP content detection kit and the Mitochondrial Complex IV Activity Kit in the basolateral amygdala (BLA). The mitochondrial molecules were detected by western blot. Then we used virus-mediated downregulation and overexpression of SIRT1 in BLA to investigate the effect of SIRT1 on anxiety and mitochondrial function. Finally, we examined the effects of pharmacological inhibition of SIRT1 on anxiety and mitochondrial function. RESULTS: We found that BLA neuronal activity, mitochondrial function, and mtDNA content were significantly higher in morphine withdrawal mice. Furthermore, the expression levels of mitochondrial molecules increased in BLA cells. Virus-mediated downregulation of SIRT1 in BLA prevented anxiety-like behaviors in morphine withdrawal mice, whereas overexpression of SIRT1 in BLA facilitated anxiety-like behaviors in untreated mice through the SIRT1/ peroxisome proliferator activated receptor gamma coactivator 1-alpha pathway. Intra-BLA infusion of selective SIRT1 antagonist EX527 effectively ameliorated anxiety-like behaviors and mitochondrial dysfunction in mice with morphine withdrawal. CONCLUSION: Our results implicate a causal role for SIRT1 in the regulation of anxiety through actions on mitochondrial biogenesis. Inhibitors targeting SIRT1 may have therapeutic potential for the treatment of opioid withdrawal-associated anxiety.


Subject(s)
Basolateral Nuclear Complex , Sirtuin 1 , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Analgesics, Opioid/pharmacology , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Basolateral Nuclear Complex/metabolism , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/pharmacology , Mice , Mitochondria/metabolism , Morphine/pharmacology , Organelle Biogenesis , PPAR gamma/metabolism , PPAR gamma/pharmacology , Sirtuin 1/metabolism
14.
Physiol Behav ; 254: 113868, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35724926

ABSTRACT

Morphine withdrawal can trigger disruptions in neuronal pathways involved in the modulation and expression of anxiety and fear-related behaviors, particularly those involved in associative learning. When it comes to contextual fear, specific subdivisions of the medial prefrontal cortex (mPFC) regulate the expression of defensive behaviors through projections to specific amygdala (AM) nuclei, such as the prelimbic cortex (PrL). The basolateral nucleus (BLA) of the AM has been shown to be involved in the modulation and expression of associative memories of fear, including those associated with opiate withdrawal-related aversive events. The purpose of this study is to determine the role of GABA mechanisms in the PrL and BLA in startle potentiation and freezing behavior caused by morphine-precipitated withdrawal. Our findings show that morphine withdrawal promotes the emergence of contextual conditioned fear in animals when they are exposed to the same environment where the withdrawal sessions were performed. This suggests that the neural circuits underlying the organism's response to conditioned stressors and the circuits modulating the negative affective states induced by drug withdrawal may overlap. The pharmacological manipulation of GABAergic neurotransmission in the PrL and BLA can reverse contextual fear in morphine-withdrawn rats, an effect that appears to be mediated, at least in part, by GABAA receptors.


Subject(s)
Basolateral Nuclear Complex , Amygdala , Animals , Fear/physiology , Morphine/adverse effects , Prefrontal Cortex/physiology , Rats , Receptors, GABA-A , gamma-Aminobutyric Acid
15.
Molecules ; 27(8)2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35458676

ABSTRACT

(1) Background: Recent data indicate that receptors for GLP-1 peptide are involved in the activity of the mesolimbic system. Thus, the purpose of the present study was to examine the effect of the selective dipeptidyl peptidase-4 (DPP-4) inhibitor, linagliptin, on morphine dependence in mice. (2) Methods: Morphine dependence in mice was obtained by administration of increasing doses of morphine for eight consecutive days, twice a day. On the 9th day of the experiment, the naloxone-induced (2 mg/kg, ip) morphine withdrawal signs (jumping) were assessed. Moreover, behavioral effects of short-term (60 h after morphine discontinuation) and long-term (14 days after morphine discontinuation) morphine withdrawal were observed. In terms of behavioral effects, the depressive effect in the forced swim test and anxiety in the elevated plus maze test were investigated. Locomotor activity of mice was also studied. (3) Results: The administration of linagliptin (10 and 20 mg/kg, ip) for 8 consecutive days before morphine injections significantly diminished the number of naloxone-induced morphine withdrawal signs (jumping) in mice. In addition, the cessation of morphine administration induced depressive behavior in mice which were observed during short- and long-term morphine withdrawal. Linagliptin administered during morphine withdrawal significantly reduced the depressive behavior in studied mice. Furthermore, the short-term morphine withdrawal evoked anxiety which also was reduced by linagliptin in mice. (4) Conclusions: The present study reveals that GLP-1 receptors are involved in morphine dependence. What is more, linagliptin might be a valuable drug in attenuating the physical symptoms of morphine dependence. It might be also a useful drug in reducing emotional disturbances which may develop during the morphine withdrawal period.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Morphine Dependence , Substance Withdrawal Syndrome , Animals , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Glucagon-Like Peptide 1 , Hypoglycemic Agents/pharmacology , Linagliptin/pharmacology , Mice , Morphine/adverse effects , Morphine Dependence/drug therapy , Naloxone/pharmacology , Substance Withdrawal Syndrome/drug therapy
16.
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
17.
Biomedicines ; 10(3)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35327388

ABSTRACT

Despite their indisputable efficacy for pain management, opiate prescriptions remain highly controversial partially due to their elevated addictive potential. Relapse in drug use is one of the principal problems for addiction treatment, with drug-associated memories being among its main triggers. Consequently, the extinction of these memories has been proposed as a useful therapeutic tool. Hence, by using the conditioned place aversion (CPA) paradigm in rats, we investigated some of the molecular mechanisms that occurr during the retrieval and extinction of morphine withdrawal memories in the basolateral amygdala (BLA) and the hippocampal dentate gyrus (DG), which control emotional and episodic memories, respectively. The retrieval of aversive memories associated with the abstinence syndrome paralleled with decreased mTOR activity and increased Arc and GluN1 expressions in the DG. Additionally, Arc mRNA levels in this nucleus very strongly correlated with the CPA score exhibited by the opiate-treated rats. On the other hand, despite the unaltered mTOR phosphorylation, Arc levels augmented in the BLA. After the extinction test, Arc and GluN1 expressions were raised in both the DG and BLA of the control and morphine-treated animals. Remarkably, Homer1 expression in both areas correlated almost perfectly with the extinction showed by morphine-dependent animals. Moreover, Arc expression in the DG correlated strongly with the extinction of the CPA manifested by the group treated with the opiate. Finally, our results support the coordinated activity of some of these neuroplastic proteins for the extinction of morphine withdrawal memories in a regional-dependent manner. Present data provide evidence of differential expression and activity of synaptic molecules during the retrieval and extinction of aversive memories of opiate withdrawal in the amygdalar and hippocampal regions that will likely permit the development of therapeutic strategies able to minimize relapses induced by morphine withdrawal-associated aversive memories.

18.
Behav Brain Res ; 427: 113859, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35337941

ABSTRACT

Opiates are among the widely abused substances worldwide. Also, the clinical use of opioids can cause unwanted and potentially severe consequences such as developing tolerance and dependence. This study simultaneously measured the changes induced after morphine dependence and naloxone-induced withdrawal syndrome on the resting-state functional connectivity (rsFC) and Local Field Potential (LFP) power in the prefrontal cortex of the rat. The obtained results revealed that acute morphine administration significantly increased the LFP power in all frequency bands, as well as the rsFC strength of the prefrontal cortex, and naloxone injection reversed this effect. In contrast, chronic morphine administration reduced neural activity and general correlation values in intrinsic signals, as well as the LFP power in all frequency bands. In morphine-dependent rats, after each morphine administration, the LFP power in all frequency bands and the rsFC strength of the prefrontal cortex were increased, and these effects were further enhanced after naloxone precipitated withdrawal syndrome. The present study concludes that general correlation merely reflects the field activity of the local cortices imaged.


Subject(s)
Morphine Dependence , Substance Withdrawal Syndrome , Analgesics, Opioid/pharmacology , Animals , Morphine/adverse effects , Naloxone/pharmacology , Naloxone/therapeutic use , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Prefrontal Cortex , Rats
19.
J Neurosci Res ; 100(1): 309-321, 2022 01.
Article in English | MEDLINE | ID: mdl-32725663

ABSTRACT

Poor sociability and aggressive behavior are key clinical features of opioid use disorders. The corticotropin-releasing factor (CRF) system may mediate behavioral effects of substances of abuse but its implication in substance-induced social behavior deficits and outward-directed hostility remains largely unknown. CRF signaling is mediated by two receptor types, termed CRF1 and CRF2 . The present study aimed at understanding the role for the CRF1 receptor in social and aggressive behavior induced by withdrawal from repeated opiate administration. Thus, wild-type (CRF1 +/+), CRF1 receptor heterozygous (CRF1 +/-), and null mutant (CRF1 -/-) female and male mice were treated with saline or escalating doses of morphine (20-100 mg/kg, i.p.) during six consecutive days and tested in the three-chamber task for sociability (i.e., preference for an unfamiliar same-sex conspecific vs. an object) 7 days after the last administration. Moreover, aggressive biting behavior toward the unfamiliar conspecific was assessed during the three-chamber test. Opiate withdrawal disrupted sociability in CRF1 +/+ and CRF1 +/-, but not in CRF1 -/-, female mice, without affecting aggressive biting behavior in any genotype. In contrast, opiate withdrawal did not affect sociability but increased aggressive biting behavior in male mice, independently of CRF1 receptor-deficiency. Nevertheless, in opiate-withdrawn CRF1 +/+, but not CRF1 +/- and CRF1 -/-, male mice, sociability directly correlated with aggressive biting behavior, suggesting a role for the CRF1 receptor in hostility-linked social approach. These findings demonstrate the implication of the CRF1 receptor in social behavior deficits associated with repeated opiate administration and withdrawal, revealing a new potential target for the treatment of opioid use disorders.


Subject(s)
Opiate Alkaloids , Receptors, Corticotropin-Releasing Hormone , Social Behavior , Substance Withdrawal Syndrome , Animals , Corticotropin-Releasing Hormone , Female , Male , Mice , Mice, Knockout , Receptors, Corticotropin-Releasing Hormone/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...