Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 723
Filtrar
1.
PLoS Biol ; 22(7): e3002716, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39008526

RESUMEN

Heterologous sensitization of adenylyl cyclase (AC) results in elevated cAMP signaling transduction that contributes to drug dependence. Inhibiting cullin3-RING ligases by blocking the neddylation of cullin3 abolishes heterologous sensitization, however, the modulating mechanism remains uncharted. Here, we report an essential role of the potassium channel tetramerization domain (KCTD) protein 2, 5, and 17, especially the dominant isoform KCTD5 in regulating heterologous sensitization of AC1 and morphine dependence via working with cullin3 and the cullin-associated and neddylation-dissociated 1 (CAND1) protein. In cellular models, we observed enhanced association of KCTD5 with Gß and cullin3, along with elevated dissociation of Gß from AC1 as well as of CAND1 from cullin3 in heterologous sensitization of AC1. Given binding of CAND1 inhibits the neddylation of cullin3, we further elucidated that the enhanced interaction of KCTD5 with both Gß and cullin3 promoted the dissociation of CAND1 from cullin3, attenuated the inhibitory effect of CAND1 on cullin3 neddylation, ultimately resulted in heterologous sensitization of AC1. The paraventricular thalamic nucleus (PVT) plays an important role in mediating morphine dependence. Through pharmacological and biochemical approaches, we then demonstrated that KCTD5/cullin3 regulates morphine dependence via modulating heterologous sensitization of AC, likely AC1 in PVT in mice. In summary, the present study revealed the underlying mechanism of heterologous sensitization of AC1 mediated by cullin3 and discovered the role of KCTD proteins in regulating morphine dependence in mice.


Asunto(s)
Adenilil Ciclasas , Proteínas Cullin , Dependencia de Morfina , Animales , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/genética , Proteínas Cullin/metabolismo , Ratones , Dependencia de Morfina/metabolismo , Células HEK293 , Humanos , Canales de Potasio/metabolismo , Canales de Potasio/genética , Ratones Endogámicos C57BL , Masculino , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Morfina/farmacología , Ratones Noqueados , Transducción de Señal , AMP Cíclico/metabolismo
2.
Psychopharmacology (Berl) ; 241(8): 1565-1575, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38630316

RESUMEN

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.


Asunto(s)
Corticosterona , Morfina , Naloxona , Antagonistas de Narcóticos , Síndrome de Abstinencia a Sustancias , Animales , Masculino , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Naloxona/farmacología , Naloxona/administración & dosificación , Ratones , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/administración & dosificación , Morfina/administración & dosificación , Morfina/farmacología , Corticosterona/sangre , Corticosterona/administración & dosificación , Relación Dosis-Respuesta a Droga , Pirroles/farmacología , Pirroles/administración & dosificación , Proteínas del Tejido Nervioso/metabolismo , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Conducta Animal/efectos de los fármacos , Dependencia de Morfina/metabolismo , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/prevención & control , Pirrolidinas/farmacología , Pirrolidinas/administración & dosificación , Inyecciones Subcutáneas , Hormona Liberadora de Corticotropina/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Pirimidinas
3.
Brain Res ; 1835: 148930, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38604556

RESUMEN

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.


Asunto(s)
Ansiedad , Estrés del Retículo Endoplásmico , Morfina , Síndrome de Abstinencia a Sustancias , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Masculino , Morfina/farmacología , Ansiedad/metabolismo , Ansiedad/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Ratones , Fenilbutiratos/farmacología , Dependencia de Morfina/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratones Endogámicos C57BL
4.
Psychopharmacology (Berl) ; 241(7): 1435-1446, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38503843

RESUMEN

RATIONALE: Transgenerational effects of preconception morphine exposure in female rats have been reported which suggest that epigenetic modifications triggered by female opioid exposure, even when that exposure ends several weeks prior to pregnancy, has significant ramifications for their future offspring. OBJECTIVE: The current study compares two mouse strains with well-established genetic variation in their response to mu opioid receptor agonists, C57BL/6J (BL6) and 129S1/svlmJ (129) to determine whether genetic background modifies the impact of preconception opioid exposure. METHODS: Adolescent females from both strains were injected daily with morphine for a total of 10 days using an increasing dosing regimen with controls receiving saline. Several weeks after their final injection, aged-matched BL6 and 129 morphine (Mor-F0) or saline (Sal-F0) females were mated with drug naïve males to generate Mor-F1 and Sal-F1 offspring, respectively. As adults, F1 mice were made morphine dependent using thrice daily morphine injections for 4 days. On day 5, mice were administered either saline or morphine followed 3 h later by naloxone. Behavioral and physiological signs of withdrawal were then measured. RESULTS: Regardless of strain or sex, morphine-dependent Mor-F1 mice had significantly lower levels of withdrawal-induced corticosterone but significantly higher glucose levels when compared to Sal-F1 controls. In contrast, both strain- and preconception opioid exposure effects on physical signs of morphine dependence were observed.


Asunto(s)
Analgésicos Opioides , Ratones Endogámicos C57BL , Dependencia de Morfina , Morfina , Receptores Opioides mu , Síndrome de Abstinencia a Sustancias , Animales , Femenino , Morfina/farmacología , Morfina/administración & dosificación , Síndrome de Abstinencia a Sustancias/metabolismo , Ratones , Masculino , Dependencia de Morfina/metabolismo , Embarazo , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Receptores Opioides mu/metabolismo , Receptores Opioides mu/genética , Ratones de la Cepa 129 , Naloxona/farmacología , Naloxona/administración & dosificación , Especificidad de la Especie , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/administración & dosificación , Corticosterona/sangre , Efectos Tardíos de la Exposición Prenatal/metabolismo
5.
Neurochem Res ; 49(2): 415-426, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37864024

RESUMEN

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.


Asunto(s)
Guayacol/análogos & derivados , Dependencia de Morfina , Morfina , Ratones , Animales , Morfina/farmacología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico , Naloxona/farmacología , Naloxona/uso terapéutico , Estrés Oxidativo , Óxido Nítrico/metabolismo , Analgésicos/uso terapéutico , Caspasas/metabolismo , Dependencia de Morfina/metabolismo
6.
Mol Psychiatry ; 29(3): 793-808, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38145987

RESUMEN

Context-induced retrieval of drug withdrawal memory is one of the important reasons for drug relapses. Previous studies have shown that different projection neurons in different brain regions or in the same brain region such as the basolateral amygdala (BLA) participate in context-induced retrieval of drug withdrawal memory. However, whether these different projection neurons participate in the retrieval of drug withdrawal memory with same or different molecular pathways remains a topic for research. The present results showed that (1) BLA neurons projecting to the prelimbic cortex (BLA-PrL) and BLA neurons projecting to the nucleus accumbens (BLA-NAc) participated in context-induced retrieval of morphine withdrawal memory; (2) there was an increase in the expression of Arc and pERK in BLA-NAc neurons, but not in BLA-PrL neurons during context-induced retrieval of morphine withdrawal memory; (3) pERK was the upstream molecule of Arc, whereas D1 receptor was the upstream molecule of pERK in BLA-NAc neurons during context-induced retrieval of morphine withdrawal memory; (4) D1 receptors also strengthened AMPA receptors, but not NMDA receptors, -mediated glutamatergic input to BLA-NAc neurons via pERK during context-induced retrieval of morphine withdrawal memory. These results suggest that different projection neurons of the BLA participate in the retrieval of morphine withdrawal memory with diverse molecular pathways.


Asunto(s)
Complejo Nuclear Basolateral , Morfina , Neuronas , Núcleo Accumbens , Síndrome de Abstinencia a Sustancias , Animales , Complejo Nuclear Basolateral/metabolismo , Masculino , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Morfina/farmacología , Neuronas/metabolismo , Núcleo Accumbens/metabolismo , Memoria/fisiología , Receptores AMPA/metabolismo , Ratas , Dependencia de Morfina/metabolismo , Amígdala del Cerebelo/metabolismo , Ratas Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Vías Nerviosas/metabolismo , Corteza Prefrontal/metabolismo
7.
Pharmacol Biochem Behav ; 233: 173660, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37852327

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Ratas , Masculino , Femenino , Animales , Dependencia de Morfina/metabolismo , Teofilina/farmacología , Morfina/efectos adversos , Ansiedad/prevención & control , Ansiedad/etiología , Trastornos de Ansiedad , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo
8.
Cells ; 12(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37566064

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Ratones , Animales , Dependencia de Morfina/metabolismo , ARN Circular/metabolismo , Morfina/farmacología , Analgésicos Opioides/farmacología , Núcleo Accumbens/metabolismo
9.
Brain Behav Immun ; 113: 328-339, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37543246

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Ratones , Animales , Dependencia de Morfina/metabolismo , Astrocitos/metabolismo , Enfermedades Neuroinflamatorias , Morfina/farmacología , Morfina/metabolismo , Microglía/metabolismo , Mitocondrias
10.
Artículo en Inglés | MEDLINE | ID: mdl-37348641

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Ratas , Animales , Morfina/farmacología , Canales de Calcio Tipo L , Naloxona/farmacología , Dependencia de Morfina/metabolismo , Hipocampo/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Verapamilo/farmacología , Antagonistas de Narcóticos/farmacología , Reacción de Prevención
11.
Behav Brain Res ; 450: 114504, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37209879

RESUMEN

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.


Asunto(s)
Cannabidiol , Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Ratones , Animales , Naloxona/farmacología , Morfina/efectos adversos , Cannabidiol/farmacología , Receptor de Serotonina 5-HT1A , Reacción de Prevención , Síndrome de Abstinencia a Sustancias/metabolismo , Ratones Endogámicos C57BL , Antagonistas de Narcóticos/farmacología , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/metabolismo
12.
Behav Brain Res ; 437: 114122, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36174840

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Alcaloides Opiáceos , Trastornos Relacionados con Opioides , Síndrome de Abstinencia a Sustancias , Humanos , Ratas , Animales , Femenino , Masculino , Síndrome de Abstinencia a Sustancias/metabolismo , Reacción de Prevención , Naloxona/farmacología , Analgésicos Opioides/farmacología , Dependencia de Morfina/metabolismo , Morfina/farmacología , Antagonistas de Narcóticos/farmacología
13.
Glia ; 70(7): 1289-1300, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35275429

RESUMEN

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.


Asunto(s)
Transferasas Alquil y Aril , Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Transferasas Alquil y Aril/metabolismo , Animales , Astrocitos/metabolismo , Miedo , Femenino , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Morfina/metabolismo , Morfina/farmacología , Dependencia de Morfina/metabolismo , Dependencia de Morfina/psicología , Naloxona/metabolismo , Naloxona/farmacología , Antagonistas de Narcóticos/metabolismo , Antagonistas de Narcóticos/farmacología , Respiración , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/psicología
14.
Neurosci Lett ; 774: 136519, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35151827

RESUMEN

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.


Asunto(s)
Complejo Nuclear Basolateral , Dependencia de Morfina , Amígdala del Cerebelo/fisiología , Animales , Estimulación Eléctrica , Lidocaína/farmacología , Morfina/metabolismo , Morfina/farmacología , Dependencia de Morfina/metabolismo , Ratas , Ratas Wistar , Recompensa
15.
Neuropharmacology ; 208: 108986, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35134442

RESUMEN

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.


Asunto(s)
Acuaporina 4/metabolismo , Conducta Adictiva , Dependencia de Morfina , Trastornos Relacionados con Opioides , Analgésicos Opioides , Animales , Acuaporina 4/genética , Dopamina/metabolismo , Heroína/farmacología , Ratones , Ratones Noqueados , Morfina , Dependencia de Morfina/metabolismo , Núcleo Accumbens , Trastornos Relacionados con Opioides/metabolismo
16.
Neurochem Int ; 155: 105304, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35176438

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Animales , Giro Dentado , Potenciación a Largo Plazo , Morfina/farmacología , Dependencia de Morfina/metabolismo , Plasticidad Neuronal/fisiología , Ratas , Ratas Wistar
17.
Behav Brain Res ; 421: 113732, 2022 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-34990697

RESUMEN

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.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Extinción Psicológica/fisiología , Miedo/fisiología , Hipocampo/metabolismo , Dependencia de Morfina , Condicionamiento Físico Animal/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Conducta Animal/fisiología , Señales (Psicología) , Masculino , Dependencia de Morfina/metabolismo , Dependencia de Morfina/fisiopatología , Dependencia de Morfina/rehabilitación , Ratas , Ratas Wistar
18.
Behav Brain Res ; 419: 113688, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34843742

RESUMEN

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.


Asunto(s)
Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Dependencia de Morfina/metabolismo , Dependencia de Morfina/fisiopatología , Neuronas Serotoninérgicas/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Int J Neurosci ; 132(3): 283-295, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32783781

RESUMEN

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.


Asunto(s)
Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Lóbulo Frontal/metabolismo , Masculino , Morfina/farmacología , Dependencia de Morfina/genética , Dependencia de Morfina/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/metabolismo
20.
Reprod Toxicol ; 105: 175-183, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34517100

RESUMEN

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.


Asunto(s)
Dependencia de Morfina/metabolismo , Células de Sertoli/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Analgésicos Opioides/farmacología , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Inflamación , Masculino , Metadona/farmacología , Ratones , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células de Sertoli/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA