Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 967
Filter
1.
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
2.
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
3.
Addict Biol ; 27(1): e13086, 2022 01.
Article in English | MEDLINE | ID: mdl-34382313

ABSTRACT

Repeated morphine exposure has been shown to induce neuronal plasticity in reward-related areas of the brain. miR-132, a CREB-induced and activation-dependent microRNA, has been suggested to be involved in the neuronal plasticity by increasing neuronal dendritic branches and spinogenesis. However, it is still unclear whether miR-132 is related to morphine dependence. Here, we investigate whether miR-132 is involved in morphine dependence and whether it is related to the structural plasticity of the dentate gyrus (DG) neurons. Sprague-Dawley rats are treated with increasing doses of morphine injection for six consecutive days to develop morphine dependence. Our results show that dendritic branching and spinogenesis of the DG neurons of morphine dependent rats are increased. Morphine treatment (24 h) promotes the differentiation of N2a cells stably expressing µ-opioid receptor by up-regulating miR-132 expression. Moreover, inhibiting miR-132 3p (but not 5p) of the DG neurons can reverse the structural plasticity and disrupt the formation of morphine dependence in rats. These findings indicate that miR-132 in the DG neurons is involved in morphine dependence via modifying the neuronal plasticity.


Subject(s)
Dentate Gyrus/drug effects , MicroRNAs/metabolism , Morphine Dependence/physiopathology , Neuronal Plasticity/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects
4.
Pharmacol Res ; 172: 105857, 2021 10.
Article in English | MEDLINE | ID: mdl-34461223

ABSTRACT

Opioid relapse is generally caused by the recurrence of context-induced memory reinstatement of reward. However, the internal mechanisms that facilitate and modify these processes remain unknown. One of the key regions of the reward is the nucleus accumbens (NAc) which receives glutamatergic projections from the dorsal hippocampus CA1 (dCA1). It is not yet known whether the dCA1 projection to the NAc shell regulates the context-induced memory recall of morphine. Here, we used a common model of addiction-related behavior conditioned place preference paradigm, combined with immunofluorescence, chemogenetics, optogenetics, and electrophysiology techniques to characterize the projection of the dCA1 to the NAc shell, in context-induced relapse memory to morphine. We found that glutamatergic neurons of the dCA1 and gamma aminobutyric acidergic (GABA) neurons of the NAc shell are the key brain areas and neurons involved in the context-induced reinstatement of morphine memory. The dCA1-NAc shell glutamatergic input pathway and the excitatory synaptic transmission of the dCA1-NAc shell were enhanced via the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) when mice were re-exposed to environmental cues previously associated with drug intake. Furthermore, chemogenetic and optogenetic inactivation of the dCA1-NAc shell pathway decreased the recurrence of long- and short-term morphine-paired context memory in mice. These results provided evidence that the dCA1-NAc shell glutamatergic projections mediated the context-induced memory recall of morphine.


Subject(s)
Analgesics, Opioid/administration & dosage , CA1 Region, Hippocampal/cytology , Memory , Morphine/administration & dosage , Neurons/drug effects , Nucleus Accumbens/cytology , Reward , Animals , Conditioning, Operant , Glutamic Acid , Male , Mice, Inbred C57BL , Morphine Dependence/physiopathology , Neurons/physiology , Synaptic Transmission/drug effects
5.
Behav Brain Res ; 414: 113450, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34265318

ABSTRACT

Investigations have shown that the circadian rhythm can affect the mechanisms associated with drug dependence. In this regard, we sought to assess the negative consequence of morphine withdrawal syndrome on conditioned place aversion (CPA) and lateral paragigantocellularis (LPGi) neuronal activity in morphine-dependent rats during light (8:00-12:00) and dark (20:00-24:00) cycles. Male Wistar rats (250-300 g) were received 10 mg/kg morphine or its vehicle (Saline, 2 mL/kg/12 h, s.c.) in 13 consecutive days for behavioral assessment tests. Then, naloxone-induced conditioned place aversion and physical signs of withdrawal syndrome were evaluated during light and dark cycles. In contrast to the behavioral part, we performed in vivo extracellular single-unit recording for investigating the neural response of LPGi to naloxone in morphine-dependent rats on day 10 of morphine/saline exposure. Results showed that naloxone induced conditioned place aversion in both light and dark cycles, but the CPA score during the light cycle was larger. Moreover, the intensity of physical signs of morphine withdrawal syndrome was more severe during the light cycle (rest phase) compare to the dark one. In electrophysiological experiments, results indicated that naloxone evoked both excitatory and inhibitory responses in LPGi neurons and the incremental effect of naloxone on LPGi activity was stronger in the light cycle. Also, the neurons with the excitatory response exhibited higher baseline activity in the dark cycle, but the neurons with the inhibitory response showed higher baseline activity in the light cycle. Interestingly, the baseline firing rate of neurons recorded in the light cycle was significantly different in response (excitatory/inhibitory) -dependent manner. We concluded that naloxone-induced changes in LPGi cellular activity and behaviors of morphine-dependent rats can be affected by circadian rhythm and the internal clock.


Subject(s)
Behavior, Animal/physiology , Circadian Rhythm/physiology , Conditioning, Classical/physiology , Electrophysiological Phenomena/physiology , Medulla Oblongata/physiopathology , Morphine Dependence/physiopathology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Substance Withdrawal Syndrome/physiopathology , Animals , Behavior, Animal/drug effects , Conditioning, Classical/drug effects , Disease Models, Animal , Electrophysiological Phenomena/drug effects , Male , Medulla Oblongata/drug effects , Neurons/physiology , Rats , Rats, Wistar
6.
Pharmacol Res ; 170: 105722, 2021 08.
Article in English | MEDLINE | ID: mdl-34116208

ABSTRACT

A progressive increase in drug craving following drug exposure is an important trigger of relapse. CircularRNAs (CircRNAs), key regulators of gene expression, play an important role in neurological diseases. However, the role of circRNAs in drug craving is unclear. In the present study, we trained mice to morphine conditioned place preference (CPP) and collected the nucleus accumbens (NAc) sections on abstinence day 1 (AD1) and day 14 (AD14) for RNA-sequencing. CircTmeff-1, which was highly expressed in the NAc core, was associated with incubation of context-induced morphine craving. The gain- and loss- of function showed that circTmeff-1 was a positive regulator of incubation. Simultaneously, the expression of miR-541-5p and miR-6934-3p were down-regulated in the NAc core during the incubation period. The dual luciferase reporter, RNA pulldown, and fluorescence insitu hybridization assays confirmed that miR-541-5p and miR-6934-3p bind to circTmeff-1 selectively. Furthermore, bioinformatics and western blot analysis suggested that vesicle-associated membrane protein 1 (VAMP1) and neurofascin (NFASC), both overlapping targets of miR-541-5p and miR-6934-3p, were highly expressed during incubation. Lastly, AAV-induced down-regulation of circTmeff-1 decreased VAMP1 and NFASC expression and incubation of morphine craving. These findings suggested that circTmeff-1, a novel circRNA, promotes incubation of context-induced morphine craving by sponging miR-541/miR-6934 in the NAc core. Thus, circTmeff-1 represents a potential therapeutic target for context-induced opioid craving, following prolonged abstinence.


Subject(s)
Behavior, Animal , Craving , Drug-Seeking Behavior , Morphine Dependence/metabolism , Nucleus Accumbens/metabolism , RNA, Circular/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cues , Disease Models, Animal , Gene Expression Regulation , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Morphine Dependence/genetics , Morphine Dependence/physiopathology , Morphine Dependence/psychology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nucleus Accumbens/physiopathology , RNA, Circular/genetics , Vesicle-Associated Membrane Protein 1/genetics , Vesicle-Associated Membrane Protein 1/metabolism
7.
Mol Neurobiol ; 58(9): 4628-4638, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34148215

ABSTRACT

Drug addiction is a global health problem and continues to place an enormous financial burden on society. This addiction is characterized by drug dependence sensitization and craving. Morphine has been widely used for pain relief, but chronic administration of morphine causes analgesic tolerance, hyperalgesia, and addiction, all of which limit its clinical usage. Alterations of multiple molecular pathways have been reported to be involved in the development of drug addiction, including mitochondrial dysfunction, excessive oxidative stress and nitric oxide stress, and increased levels of apoptosis, autophagy, and neuroinflammation. Preclinical and clinical studies have shown that the co-administration of melatonin with morphine leads to a reversal of these affected pathways. In addition, murine models have shown that melatonin improves morphine-induced analgesic tolerance and addictive behaviors, such as behavioral sensitization, reward effect, and physical dependence. In this review, we attempt to summarize the recent findings about the beneficial effect and molecular mechanism of melatonin on mitochondrial dysfunction, uncontrolled autophagy, and neuroinflammation in morphine addiction and morphine analgesic tolerance. We propose that melatonin might be a useful supplement in the treatment opiate abuse.


Subject(s)
Drug Tolerance , Melatonin/pharmacology , Morphine Dependence/physiopathology , Morphine/pharmacology , Neuroprotective Agents/pharmacology , Autophagy/drug effects , Humans , Mitochondria/drug effects , Pain Measurement
8.
Neurosci Lett ; 760: 136076, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34153368

ABSTRACT

A growing body of evidence has shown that abused drugs could simultaneously induce the paradoxical effect-reward and aversion. Moreover, the medial prefrontal cortex (mPFC), amygdala, and hippocampus were involved in this paradoxical effect by abused drugs. However, no research examined whether neuroinflammatory changes in the mPFC [including cingulate cortex area 1 (Cg1); prelimbic cortex (PrL); infralimbic cortex (IL)], basolateral amygdala, and hippocampus [e.g., CA1, CA2, CA3, and dentate gyrus (DG)] after morphine-induced reward in conditioned place preference (CPP) and aversion in conditioned taste aversion (CTA). The results showed that after morphine administration, the consumption of a 0.1% saccharin solution decreased; the mean time spent in the morphine-paired side compartment of the CPP box increased, indicating that morphine simultaneously induced the paradoxical effects of reward and aversion. The PrL and IL of the mPFC, the BLA of the amygdala, the CA1, CA2, CA3, and DG of the hippocampus but not the Cg1 presented hyperactive IL-1ß expression in response to morphine's aversion and reward. The mPFC, amygdala, and hippocampus may appear neuroinflammation activity following morphine-induced paradoxical effect-reward in CPP and aversion in CTA. The present data may provide a better understanding of the relationship between neuroinflammation and morphine addiction.


Subject(s)
Interleukin-1beta/metabolism , Morphine Dependence/immunology , Morphine/adverse effects , Neuroinflammatory Diseases/immunology , Reward , Amygdala/metabolism , Amygdala/pathology , Amygdala/physiopathology , Animals , Conditioning, Operant , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Male , Morphine/administration & dosage , Morphine Dependence/pathology , Morphine Dependence/physiopathology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/physiopathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Rats , Saccharin/administration & dosage , Signal Transduction/immunology
9.
Neurosci Lett ; 759: 135996, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34062194

ABSTRACT

The lateral habenula (LHb) plays a principal role in response to aversive stimuli and negative emotional states. In this study, we have evaluated the effects of unilateral electrical stimulation (e-stim) of the LHb on morphine-conditioned place preference (CPP), before or after bilateral injections of Gamma-aminobutyric acid-B receptor (GABABR) antagonist, phaclofen, in male rats. Morphine (5 mg/kg; s.c.) induced a significant CPP, using a 5-day CPP paradigm. Intra-LHb microinjection of phaclofen or the LHb e-stim decreased only the acquisition of CPP. The 150 µA stimulation plus phaclofen significantly suppressed the expression phase but induced aversion in the acquisition of CPP, and an e-stim of 25 µA in combination with the antagonist, significantly prevented only the acquisition phase. The findings of this study confirm the possible role of GABABRs in the LHb on the acquisition and the expression of CPP. These results show that e-stim of LHb alone or plus phaclofen may change the GABA transmission, involving into CPP. Therefore, the GABAergic system, especially through GABABRs, may play a prominent role in the behavioral responses to morphine-induced CPP by LHb stimulation.


Subject(s)
Conditioning, Classical/physiology , Electric Stimulation/methods , GABA-B Receptor Antagonists/pharmacology , Habenula/physiology , Morphine Dependence/physiopathology , Animals , Conditioning, Classical/drug effects , Male , Morphine/pharmacology , Narcotics/pharmacology , Rats , Rats, Wistar
10.
Neurosci Lett ; 755: 135909, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33892002

ABSTRACT

Orexin neuropeptides are implicated in the expression of morphine dependence. Locus coeruleus (LC) nucleus is an important brain area involving in the development of withdrawal signs of morphine and contains high expression of orexin type 1 receptors (OX1Rs). Despite extensive considerations, effects of immediate inhibition of OX1Rs by a single dose administration of SB-334867 prior to the naloxone-induced activation of LC neurons remains unknown. Therefore, we examined the direct effects of OX1Rs acute blockade on the neuronal activity of the morphine-dependent rats which underwent naloxone administration. Adult male rats underwent subcutaneous administration of 10 mg/kg morphine (two times/day) for a ten-day period. On the last day of experiment, intra-cerebroventricular administration of 10 µg/µl antagonist of OX1Rs, SB-334867, was performed just before intra-peritoneal injection of 2 mg/kg naloxone. Thereafter, in vivo extracellular single unit recording was employed to evaluate the electrical activity of LC neuronal cells. The outcomes demonstrated that morphine tolerance developed following ten-day of injection. Then, naloxone administration causes hyperactivity of LC neuronal cells, whereas a single dose administration of SB-334867 prior to naloxone prevented the enhanced activity of neurons upon morphine withdrawal. Our findings indicate that increased response of LC neuronal cells to applied naloxone could be prevented by the acute inhibition of the OX1Rs just before the naloxone treatment.


Subject(s)
Locus Coeruleus/physiology , Morphine Dependence/physiopathology , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Orexin Receptor Antagonists/administration & dosage , Orexin Receptors/physiology , Action Potentials/drug effects , Action Potentials/physiology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Benzoxazoles/administration & dosage , Injections, Intraperitoneal , Injections, Intraventricular , Locus Coeruleus/drug effects , Male , Morphine/administration & dosage , Morphine/adverse effects , Morphine Dependence/drug therapy , Naphthyridines/administration & dosage , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar , Urea/administration & dosage , Urea/analogs & derivatives
11.
Mar Drugs ; 19(1)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478061

ABSTRACT

N-methyl-D-aspartate receptor (NMDAR) antagonists have been found to be effective to inhibit morphine dependence. However, the discovery of the selective antagonist for NMDAR GluN2B with low side-effects still remains challenging. In the present study, we report a selective NMDAR GluN2B antagonist con-T[M8Q](a conantokin-T variant) that potently inhibits the naloxone-induced jumping and conditioned place preference of morphine-dependent mice at nmol/kg level, 100-fold higher than ifenprodil, a classical NMDAR NR2B antagonist. Con-T[M8Q] displays no significant impacts on coordinated locomotion function, spontaneous locomotor activity, and spatial memory mice motor function at the dose used. Further molecular mechanism experiments demonstrate that con-T[M8Q] effectively inhibited the transcription and expression levels of signaling molecules related to NMDAR NR2B subunit in hippocampus, including NR2B, p-NR2B, CaMKII-α, CaMKII-ß, CaMKIV, pERK, and c-fos. The high efficacy and low side effects of con-T[M8Q] make it a good lead compound for the treatment of opiate dependence and for the reduction of morphine usage.


Subject(s)
Conotoxins/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Morphine Dependence/drug therapy , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Conotoxins/administration & dosage , Conotoxins/toxicity , Disease Models, Animal , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Locomotion/drug effects , Male , Mice , Morphine Dependence/physiopathology , Naloxone/pharmacology , Piperidines/pharmacology , Spatial Memory/drug effects
12.
Brain Res Bull ; 169: 128-135, 2021 04.
Article in English | MEDLINE | ID: mdl-33482287

ABSTRACT

Early life stress can serve as one of the principle sources leading to individual differences in confronting challenges throughout the lifetime. Maternal deprivation (MD), a model of early life stress, can cause persistent alterations in brain function, and it may constitute a risk factor for later incidence of drug addiction. It is becoming more apparent that early life MD predisposes opiate abuse in adulthood. Although several behavioral and molecular studies have addressed this issue, changes in electrophysiological features of the neurons are yet to be understood. The lateral paragigantocellularis (LPGi) nucleus, which participates in the mediation of opiate dependence and withdrawal, may be susceptible to modifications following MD. This study sought to find whether early life MD can alter the discharge activity of LPGi neurons and their response to acute morphine administration in adult rats. Male Wistar rats experienced MD on postnatal days (PNDs) 1-14 for three h per day. Afterward, they were left undisturbed until PND 70, during which the extracellular activities of LPGi neurons were recorded in anesthetized animals at baseline and in response to acute morphine. In both MD and control groups, acute morphine administration induced heterogeneous (excitatory, inhibitory, and no effect) responses in LPGi neurons. At baseline recording, the interspike interval variability of the LPGi neurons was attenuated in both inhibitory and excitatory responses in animals with the history of MD. The extent of morphine-induced discharge inhibition was also lower in deprived animals compared to the control group. These findings suggest that early life MD induces long-term alterations in LPGi neuronal activity in response to acute administration of morphine. Therefore, the MD may alter the vulnerability to develop opiate abuse in adulthood.


Subject(s)
Maternal Deprivation , Medulla Oblongata/drug effects , Morphine/pharmacology , Narcotics/pharmacology , Neural Inhibition/drug effects , Neurons/drug effects , Animals , Male , Medulla Oblongata/physiopathology , Morphine Dependence/physiopathology , Rats , Rats, Wistar
13.
Exp Neurol ; 338: 113600, 2021 04.
Article in English | MEDLINE | ID: mdl-33453215

ABSTRACT

Drug relapse can be mainly ascribed to the retrieval of drug withdrawal memory induced by conditioned context. Previous studies have shown that the central nucleus of the amygdala lateral division (CeL) could be activated by conditioned context. However, what source of input that activates the CeL during conditioned context-induced retrieval of morphine-withdrawal memory remains unknown. In this study, using retrograde labeling, immunohistochemistry, local microinjection and chemogenetic technologies, we found that (1) Conditioned context induced an activation of the CeL and the inhibition of the CeL inhibited the context-induced retrieval of morphine-withdrawal memory; (2) the inhibition of the paraventricular nucleus of thalamus (PVT) or PVT-CeL projection neurons caused an attenuation of the activation of the CeL by conditioned context and conditioned place aversion (CPA); (3) the inhibition of the locus coeruleus (LC) or LC-CeL projection neurons decreased the activation of the CeL by conditioned context and CPA. These results suggest that the CeL is necessary for conditioned context-induced retrieval of morphine-withdrawal memory and inputs from PVT and LC contribute to the activation of the CeL during context-induced retrieval of morphine withdrawal memory.


Subject(s)
Central Amygdaloid Nucleus/physiopathology , Locus Coeruleus/physiopathology , Memory/physiology , Midline Thalamic Nuclei/physiopathology , Morphine Dependence/physiopathology , Animals , Conditioning, Psychological/physiology , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiopathology , Substance Withdrawal Syndrome/physiopathology
14.
Bull Exp Biol Med ; 170(1): 30-34, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33222084

ABSTRACT

Activity of compound GSB-106, a low-molecular mimetic of loop 4 of the brain neurotrophic factor (BDNF), was studied in experimental morphine withdrawal syndrome simulated in outbred rats. Single and subchronic (5 intraperitoneal injections) administration of GSB-106 in a dose of 0.1 mg/kg significantly reduced the total index of morphine withdrawal syndrome by 55.2 and 45.6%, respectively. GSB-106 reduced the severity of some behavioral signs (piloerection, gnashing of teeth, wet-dog shaking, and runaway attempts), but had no effect on mechanical allodynia formed in the rats with dependence. Subchronic treatment with GSB-106 prevented the increase in the content of ΔFosB (product of early response gene) in the striatum induced by morphine withdrawal. The results confirmed the concept on the involvement of neurotrophins, specifically BDNF and its analogs, in the mechanisms associated with the formation of opiate dependence.


Subject(s)
Dipeptides/pharmacology , Morphine Dependence/drug therapy , Morphine/antagonists & inhibitors , Narcotic Antagonists/pharmacology , Peptidomimetics/pharmacology , Substance Withdrawal Syndrome/drug therapy , Animals , Animals, Outbred Strains , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Gene Expression , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Injections, Intraperitoneal , Male , Morphine/adverse effects , Morphine Dependence/genetics , Morphine Dependence/metabolism , Morphine Dependence/physiopathology , Narcotics/adverse effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology
15.
Drug Alcohol Depend ; 216: 108310, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33017752

ABSTRACT

BACKGROUND: Made as a tea, the Thai traditional drug "kratom" reportedly possesses pharmacological actions that include both a coca-like stimulant effect and opium-like depressant effect. Kratom has been used as a substitute for opium in physically-dependent subjects. The objective of this study was to evaluate the antinociception, somatic and physical dependence produced by kratom tea, and then assess if the tea ameliorated withdrawal in opioid physically-dependent subjects. METHODS: Lyophilized kratom tea (LKT) was evaluated in C57BL/6J and opioid receptor knockout mice after oral administration. Antinociceptive activity was measured in the 55 °C warm-water tail-withdrawal assay. Potential locomotor impairment, respiratory depression and locomotor hyperlocomotion, and place preference induced by oral LKT were assessed in the rotarod, Comprehensive Lab Animal Monitoring System, and conditioned place preference assays, respectively. Naloxone-precipitated withdrawal was used to determine potential physical dependence in mice repeatedly treated with saline or escalating doses of morphine or LKT, and LKT amelioration of morphine withdrawal. Data were analyzed using one- and two-way ANOVA. RESULTS: Oral administration of LKT resulted in dose-dependent antinociception (≥1 g/kg, p.o.) absent in mice lacking the mu-opioid receptor (MOR) and reduced in mice lacking the kappa-opioid receptor. These doses of LKT did not alter coordinated locomotion or induce conditioned place preference, and only briefly reduced respiration. Repeated administration of LKT did not produce physical dependence, but significantly decreased naloxone-precipitated withdrawal in morphine dependent mice. CONCLUSIONS: The present study confirms the MOR agonist activity and therapeutic effect of LKT for the treatment of pain and opioid physical dependence.


Subject(s)
Mitragyna , Morphine Dependence/drug therapy , Plant Extracts/administration & dosage , Receptors, Opioid, mu/agonists , Tea , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Freeze Drying/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/administration & dosage , Morphine Dependence/physiopathology , Morphine Dependence/psychology , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Pain Measurement/methods , Plant Extracts/isolation & purification , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, mu/deficiency
16.
Proc Natl Acad Sci U S A ; 117(32): 19556-19565, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32694207

ABSTRACT

Opioid addiction is a chronic, relapsing disorder associated with persistent changes in brain plasticity. Reconfiguration of neuronal connectivity may explain heightened abuse liability in individuals with a history of chronic drug exposure. To characterize network-level changes in neuronal activity induced by chronic opiate exposure, we compared FOS expression in mice that are morphine-naïve, morphine-dependent, or have undergone 4 wk of withdrawal from chronic morphine exposure, relative to saline-exposed controls. Pairwise interregional correlations in FOS expression data were used to construct network models that reveal a persistent reduction in connectivity strength following opiate dependence. Further, we demonstrate that basal gene expression patterns are predictive of changes in FOS correlation networks in the morphine-dependent state. Finally, we determine that regions of the hippocampus, striatum, and midbrain are most influential in driving transitions between opiate-naïve and opiate-dependent brain states using a control theoretic approach. This study provides a framework for predicting the influence of specific therapeutic interventions on the state of the opiate-dependent brain.


Subject(s)
Brain/physiopathology , Morphine Dependence/physiopathology , Nerve Net/physiopathology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Brain/drug effects , Brain/metabolism , Connectome , Gene Expression Profiling , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Models, Neurological , Morphine/administration & dosage , Morphine/adverse effects , Morphine Dependence/metabolism , Nerve Net/drug effects , Nerve Net/metabolism , Neuronal Plasticity/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology
17.
Neurosci Lett ; 724: 134875, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32114118

ABSTRACT

Orexin neuropeptides are involved in opiate-induced physical dependence and expression of withdrawal signs following drug abstinence. Orexin type-1 receptors (OXR1) are expressed throughout the rostroventrolateral medulla (RVLM), particularly in the lateral paragigantocellularis (LPGi) nucleus. The present study examined whether blocking OXR1 in LPGi region could affect the development of morphine dependence and so behavioral manifestations induced by morphine withdrawal in rats. Male Wistar rats weighing 250-300 g were used. In order to induce drug dependence, morphine was injected subcutaneously (s.c.) (6, 16, 26, 36, 46, 56, and 66 mg/kg, 2 ml/kg) at an interval of 24 h for 7 days. Animals were divided into experimental groups in which the orexin type-1 receptor antagonist, SB-334867 (0.2 µl, 3 mM), or its vehicle were injected into the LPGi nucleus for 7 days before each morphine injection. On day 8, naloxone (2.5 mg/kg, i.p.) was administered and morphine withdrawal behaviors were monitored for 25 min. Our results indicated that the inhibition of OXR1 in LPGi nucleus significantly reduces the development of morphine dependence and behavioral signs elicited by the administration of naloxone in morphine-dependent rats.


Subject(s)
Medulla Oblongata/physiology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/physiology , Morphine Dependence/prevention & control , Morphine/adverse effects , Orexin Receptor Antagonists/administration & dosage , Animals , Male , Medulla Oblongata/drug effects , Microinjections/methods , Morphine/administration & dosage , Morphine Dependence/physiopathology , Rats , Rats, Wistar
18.
Psychopharmacology (Berl) ; 237(2): 375-384, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31712968

ABSTRACT

RATIONALE: Acute naloxone-precipitated morphine withdrawal (MWD) produces a conditioned place aversion (CPA) in rats even after one or two exposures to high-dose (20 mg/kg, sc) morphine followed 24-h later by naloxone (1 mg/kg, sc). However, the somatic withdrawal reactions produced by acute naloxone-precipitated MWD in rats have not been investigated. A recently discovered fatty acid amide, N-oleoylglycine (OlGly), which has been suggested to act as a fatty acid amide hydrolase (FAAH) inhibitor and as a peroxisome proliferator-activated receptor alpha (PPARα) agonist, was previously shown to interfere with a naloxone-precipitated MWD-induced CPA in rats. OBJECTIVES: The aims of these studies were to examine the somatic withdrawal responses produced by acute naloxone-precipitated MWD and determine whether OlGly can also interfere with these responses. RESULTS: Here, we report that following two exposures to morphine (20 mg/kg, sc) each followed by naloxone (1 mg/kg, sc) 24 h later, rats display nausea-like somatic reactions of lying flattened on belly, abdominal contractions and diarrhea, and display increased mouthing movements and loss of body weight. OlGly (5 mg/kg, ip) interfered with naloxone-precipitated MWD-induced abdominal contractions, lying on belly, diarrhea and mouthing movements in male Sprague-Dawley rats, by both a cannabinoid 1 (CB1) and a PPARα mechanism of action. Since these withdrawal reactions are symptomatic of nausea, we evaluated the potential of OlGly to interfere with lithium chloride (LiCl)-induced and MWD-induced conditioned gaping in rats, a selective measure of nausea; the suppression of MWD-induced gaping reactions by OlGly was both CB1 and PPARα mediated. CONCLUSION: These results suggest that the aversive effects of acute naloxone-precipitated MWD reflect nausea, which is suppressed by OlGly.


Subject(s)
Glycine/analogs & derivatives , Morphine/adverse effects , Naloxone/toxicity , Narcotic Antagonists/toxicity , Nausea/drug therapy , Oleic Acids/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Animals , Female , Glycine/pharmacology , Glycine/therapeutic use , Male , Medically Unexplained Symptoms , Morphine Dependence/drug therapy , Morphine Dependence/physiopathology , Nausea/chemically induced , Nausea/physiopathology , Oleic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Shrews , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/physiopathology
19.
eNeuro ; 6(6)2019.
Article in English | MEDLINE | ID: mdl-31801741

ABSTRACT

The paraventricular thalamic nucleus (PVT) is a brain region involved in regulating arousal, goal-oriented behaviors, and drug seeking, all key factors playing a role in substance use disorder. Given this, we investigated the temporal effects of administering morphine, an opioid with strongly addictive properties, on PVT neuronal function in mice using acute brain slices. Here, we show that morphine administration and electrophysiological recordings that occur during periods of animal inactivity (light cycle) elicit increases in PVT neuronal function during a 24-h abstinence time point. Furthermore, we show that morphine-induced increases in PVT neuronal activity at 24-h abstinence are occluded when morphine administration and recordings are performed during an animals' active state (dark cycle). Based on our electrophysiological results combined with previous findings demonstrating that PVT neuronal activity regulates drug-seeking behaviors, we investigated whether timing morphine administration with periods of vigilance (dark cycle) would decrease drug-seeking behaviors in an animal model of substance use disorder. We found that context-induced morphine-seeking behaviors were intact regardless of the time morphine was administered (e.g., light cycle or dark cycle). Our electrophysiological results suggest that timing morphine with various states of arousal may impact the firing of PVT neurons during abstinence. Although, this may not impact context-induced drug-seeking behaviors.


Subject(s)
Midline Thalamic Nuclei/drug effects , Morphine Dependence/physiopathology , Morphine/administration & dosage , Narcotics/administration & dosage , Neurons/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Administration Schedule , Drug-Seeking Behavior/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice, Inbred C57BL , Midline Thalamic Nuclei/physiopathology , Neurons/physiology , Photoperiod , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors , Tissue Culture Techniques
20.
Pharmacol Biochem Behav ; 187: 172793, 2019 12.
Article in English | MEDLINE | ID: mdl-31639387

ABSTRACT

Gender- and age-dependent effects on the severity of morphine dependence are still controversial. The aim of this study was to investigate the effects of age and sex on the severity of physical and psychological dependence in morphine-dependent rats. The adult/aged male and female Wistar rats were chronically treated with bi-daily doses (10 mg/kg, at 12 h intervals) of morphine for 14 days. Then, rats were tested for the severity of physical dependence on morphine (spontaneous withdrawal signs), anxiety-like (the elevated plus maze), depressive-like (sucrose preference test) and grooming behaviors after spontaneous morphine withdrawal. We found that the morphine withdrawal signs decreased after 3 and 7 days of withdrawal in female and male rats respectively, while there was no significant difference in overall dependence severity between the two sexes or ages. Also, we found that the withdrawal of morphine led to increased anxiety, depression and obsessive-compulsive behavior in the D (dependent)/Adult male and female rats. Also, the D/aged female and male rats exhibited a reduction in depressive-like behavior than the D/Adult rats. Moreover, the D/female rats exhibited a decreased obsessive-compulsive behavior in both age groups than male rats. We conclude that age has no effect on the duration of withdrawal from morphine and overall dependence severity. While, the duration of withdrawal from morphine was lower in female than male rats. Our results showed a sex difference on the duration of morphine withdrawal and an age difference in the expression of psychological dependence on morphine. Thus, therapeutic strategies may be different for opiate-dependent individuals in physical and psychological dimensions.


Subject(s)
Morphine Dependence/physiopathology , Morphine Dependence/psychology , Severity of Illness Index , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology , Age Factors , Animals , Anxiety/psychology , Behavior, Animal , Depression/psychology , Disease Models, Animal , Female , Male , Maze Learning , Rats , Rats, Wistar , Sex Factors , Sucrose/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...