Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Physiol Sci ; 71(1): 25, 2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429058

RESUMEN

INTRODUCTION: Adolescence is a critical period in brain development, and it is characterized by persistent maturational alterations in the function of central nervous system. In this respect, many studies show the non-medical use of opioid drugs by adolescents. Although this issue has rather widely been addressed during the last decade, cellular mechanisms through which adolescent opioid exposure may induce long-lasting effects are not duly understood. The present study examined the effect of adolescent morphine exposure on neuronal responses of lateral paragigantocellularis nucleus to naloxone in adult morphine-dependent rats. METHODS: Adolescent male Wistar rats (31 days old) received increasing doses of morphine (from 2.5 to 25 mg/kg, twice daily, s.c.) for 10 days. Control subjects were injected saline with the same protocol. After a drug-free interval (20 days), animals were rendered dependent on morphine during 10 days (10 mg/kg, s.c., twice daily). Then, extracellular single-unit recording was performed to investigate neural response of LPGi to naloxone in adult morphine-dependent rats. RESULTS: Results indicated that adolescent morphine treatment increases the number of excitatory responses to naloxone, enhances the baseline activity and alters the pattern of firing in neurons with excitatory responses in adult morphine-dependent rats. Moreover, the intensity of excitatory responses is reduced following the early life drug intake. CONCLUSION: It seems that prolonged opioid exposure during adolescence induces long-lasting neurobiological changes in LPGi responsiveness to future opioid withdrawal challenges.


Asunto(s)
Dependencia de Morfina , Morfina , Animales , Masculino , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas , Ratas , Ratas Wistar
2.
Behav Brain Res ; 414: 113450, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34265318

RESUMEN

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.


Asunto(s)
Conducta Animal/fisiología , Ritmo Circadiano/fisiología , Condicionamiento Clásico/fisiología , Fenómenos Electrofisiológicos/fisiología , Bulbo Raquídeo/fisiopatología , Dependencia de Morfina/fisiopatología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos/efectos de los fármacos , Masculino , Bulbo Raquídeo/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Wistar
3.
Neurosci Lett ; 724: 134875, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32114118

RESUMEN

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.


Asunto(s)
Bulbo Raquídeo/fisiología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/fisiología , Dependencia de Morfina/prevención & control , Morfina/efectos adversos , Antagonistas de los Receptores de Orexina/administración & dosificación , Animales , Masculino , Bulbo Raquídeo/efectos de los fármacos , Microinyecciones/métodos , Morfina/administración & dosificación , Dependencia de Morfina/fisiopatología , Ratas , Ratas Wistar
4.
Acta Neurol Belg ; 120(3): 653-660, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30132138

RESUMEN

The pain modulatory role of the paragigantocellularis lateralis nucleus (LPGi) and the 17ß-estradiol has thoroughly been probed. This study investigates the contribution of ionotropic glutamate receptors in pain modulatory effect of intra-LPGi injection of 17ß-estradiol. For this purpose, the LPGi nucleus cannulation was performed and drugs were injected into this nucleus, 15 min prior to the formalin test. The duration of formalin-induced flexing and licking behaviors was recorded for 60 min immediately after formalin injection. The results showed that the flexing behavior is significantly decreased by intra-LPGi injection of 0.8 µmol 17ß-estradiol duringboth phases of formalin test (P < 0.001). However, 17ß-estradiol attenuated the licking duration only in the second phase (P < 0.001). Interestingly, NMDA and AMPA/kainate receptor antagonists (AP5 and CNQX, respectively) significantly counteracted the analgesic effect of intra-LPGi injection of 17ß-estradiol in both phases of the formalin test (P < 0.001). Consequently, the revealing results showed that the analgesic effect of intra-LPGi injection of 17ß-estradiol on acute inflammatory pain might be mediated via the activation of ionotropic glutamate receptors.


Asunto(s)
Estradiol/metabolismo , Bulbo Raquídeo/metabolismo , Dolor/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Estradiol/farmacología , Formaldehído/toxicidad , Irritantes/toxicidad , Masculino , Bulbo Raquídeo/efectos de los fármacos , Dolor/inducido químicamente , Dimensión del Dolor , Ratas , Ratas Wistar
5.
Life Sci ; 234: 116784, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31445026

RESUMEN

Tobacco smoking is recognized as a life-threatening risk factor worldwide. Initiation of smoking primarily occurs during adolescence which is a critical developmental phase characterized by specific neurobehavioral alterations. The effect of adolescent nicotine exposure on vulnerability to opioid addiction has not been previously addressed. Furthermore, lateral paragigantocellularis (LPGi) is a key modulator of opiate effects. In this study we investigated the effect of adolescent nicotine treatment on development of morphine tolerance and dependence as well as LPGi neuronal responses to morphine during adulthood. Male Wistar rats received subcutaneous injections of either nicotine or saline during adolescence and then development of morphine tolerance and dependence was assessed during adulthood by tail-flick and withdrawal tests, respectively. In vivo single-unit recording was performed to examine the LPGi neuronal activities. Results indicated that adolescent nicotine exposure significantly facilitates the development of tolerance to analgesic effect of morphine and increases the expression of morphine withdrawal signs in adulthood. Also, it was observed that following adolescent nicotine treatment, the extent of morphine-induced excitation is attenuated in LPGi neurons of adult rats. Moreover, the onset of morphine-induced inhibition was increased in these animals. Neither the baseline, nor the regularity of firing was affected in our observations. It could be concluded that nicotine challenge during adolescence may enhance the future vulnerability to opioid addiction through induction of persistent neuroadaptations in LPGi neurons.


Asunto(s)
Tronco Encefálico/efectos de los fármacos , Dependencia de Morfina/etiología , Neuronas/efectos de los fármacos , Nicotina/efectos adversos , Envejecimiento , Animales , Tronco Encefálico/citología , Tronco Encefálico/fisiopatología , Masculino , Dependencia de Morfina/fisiopatología , Neuronas/patología , Trastornos Relacionados con Opioides/etiología , Trastornos Relacionados con Opioides/fisiopatología , Ratas Wistar , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología
6.
Sleep ; 42(3)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30541142

RESUMEN

Sleep abnormalities are common among children with neurodevelopmental disorders. The human chr16p11.2 microdeletion is associated with a range of neurological and neurobehavioral abnormalities. Previous studies of a mouse model of human chr16p11.2 microdeletion (chr16p11.2df/+) have demonstrated pathophysiological changes at the synapses in the hippocampus and striatum; however, the impact of this genetic abnormality on system level brain functions, such as sleep and neural oscillation, has not been adequately investigated. Here, we show that chr16p11.2df/+ mice have altered sleep architecture, with increased wake time and reduced time in rapid eye movement (REM) and non-REM (NREM) sleep. Importantly, several measurements of REM sleep are significantly changed in deletion mice. The REM bout number and the bout number ratio of REM to NREM are decreased in mutant mice, suggesting a deficit in REM-NREM transition. The average REM bout duration is shorter in mutant mice, indicating a defect in REM maintenance. In addition, whole-cell patch clamp recording of the ventrolateral periaqueductal gray (vlPAG)-projecting gamma-aminobutyric acid (GABA)ergic neurons in the lateral paragigantocellular nucleus of ventral medulla of mutant mice reveal that these neurons, which are important for NREM-REM transition and REM maintenance, have hyperpolarized resting membrane potential and increased membrane resistance. These changes in intrinsic membrane properties suggest that these projection-specific neurons of mutant mice are less excitable, and thereby may play a role in deficient NREM-REM transition and REM maintenance. Furthermore, mutant mice exhibit changes in neural oscillation involving multiple frequency classes in several vigilance states. The most significant alterations occur in the theta frequency during wake and REM sleep.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/fisiopatología , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Neuronas GABAérgicas/fisiología , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Sueño REM/fisiología , Animales , Deleción Cromosómica , Cromosomas Humanos Par 16/genética , Humanos , Masculino , Bulbo Raquídeo/fisiología , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Polisomnografía/métodos
7.
Brain Res Bull ; 137: 178-186, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29246865

RESUMEN

Accumulating evidence support the growing non-medical use of morphine during adolescence. Despite this concern which has recently been addressed in some studies, cellular mechanisms underlying the long-term neurobiological and behavioral effects of opiate exposure during this critical period have still remained largely unexplored. Several reports have proposed that subtle long-lasting neurobiological alterations might be triggered by exposure to opiate derivatives or drugs of abuse particularly when this occurs during a critical phase of brain maturation such as adolescence. The present study was designed to investigate how chronic adolescent morphine exposure could affect the responsiveness of lateral paragigantocellular (LPGi) neurons to acute morphine administration in adult rats. Male Wistar rats received chronic escalating morphine or saline during adolescence (30-39d) for 10 days. During adulthood (65d), the extracellular unit activities of LPGi neurons were recorded in urethane-anesthetized animals. Results indicated that adolescent morphine treatment enhances the baseline activity of LPGi neurons. In addition, morphine-induced inhibition of spontaneous discharge rate was potentiated in adult rats received morphine during adolescence. However, this pretreatment did not affect the extent of morphine excitatory effect, onset or peak of cellular response and regularity of unit discharge in LPGi neurons. Our study supports the hypothesis that adolescent morphine exposure induces long-lasting neurophysiological alterations in brain regions known to play a role in mediating opiate effects. This finding sheds light on the possible effect of opiate pre-exposure on addiction susceptibility in future.


Asunto(s)
Morfina/toxicidad , Narcóticos/toxicidad , Neuronas/efectos de los fármacos , Formación Reticular/efectos de los fármacos , Formación Reticular/crecimiento & desarrollo , Potenciales de Acción/efectos de los fármacos , Animales , Esquema de Medicación , Inyecciones Intraperitoneales , Masculino , Microelectrodos , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Inhibición Neural/efectos de los fármacos , Distribución Aleatoria , Ratas Wistar
8.
Brain Res ; 1578: 14-22, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25010816

RESUMEN

Repetitive administration of opioid agonists is associated with the development of tolerance to the effects of these substances and limits their application. Orexin (also known as hypocretin) is involved in morphine tolerance and dependence. The lateral paragigantocellularis nucleus (LPGi) is a key brain region implicated in the tolerance and dependence to opiates. Orexin type 1 receptor (OXR1) has been detected in LPGi nucleus. In this study the effect of OXR1 blockade was investigated on neural activity of LPGi during the development of morphine tolerance in rats. Male Wistar rats weighing 250-300 g were used in this study. To incite tolerance, morphine sulfate was injected intraperitonealy (10 mg/kg, i.p.) once a day for 6 days. A selective OXR1 antagonist (SB-334867) was microinjected into the right cerebral ventricle (10 µg/10 µl, i.c.v.) immediately before each morphine injection. On day 7, the effect of morphine (10 mg/kg, i.p.) on neural activity of LPGi was investigated using in vivo extracellular single unit recording. In this study morphine injection during 6 days led to the development of morphine tolerance in LPGi neurons which was observed as a significant decrease in responsiveness of LPGi neurons to acute morphine injection. Administration of SB-334867 before each morphine injection could reverse the responses of LPGi neurons to acute morphine injection. This study showed that OXR1 blockade by SB-334867 prevents the development of tolerance to morphine in LPGi neurons. Further studies are required to determine molecular and anatomical mediators which are thought to be involved in this phenomenon.


Asunto(s)
Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiología , Morfina/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Receptores de Orexina/fisiología , Animales , Benzoxazoles/farmacología , Tolerancia a Medicamentos/fisiología , Masculino , Naftiridinas , Antagonistas de los Receptores de Orexina , Ratas , Ratas Wistar , Urea/análogos & derivados , Urea/farmacología
9.
Neurosci Lett ; 558: 62-6, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24211689

RESUMEN

Orexin neuropeptides have been reported to be involved in morphine induced physical dependence and withdrawal. The Lateral Paragigantocellularis (LPGi) is a key brain region implicated in the expression of somatic signs of morphine withdrawal syndrome. Orexin A and orexin type 1 receptor have been found in LPGi neurons but the effect of orexin on the expression of opiate dependence and withdrawal phenomena in this brain structure has not been studied yet. In this study, the effect of intra-LPGi administration of SB 334867 (selective orexin type 1 receptor antagonist) on the behavioral signs of morphine withdrawal syndrome was investigated. Male Wistar rats weighing 250-300 g were rendered dependent by adding morphine sulfate (Temad, Tehran, Iran) to their drinking water in increasing concentrations of 0.1, 0.2, 0.3mg/ml for every 48 h and 0.4 mg/ml during the next 15 days. Behavioral signs of morphine withdrawal were assessed in a transparent cylindrical Plexiglas test chamber (30 cm diameter, 50 cm height) for 25 min. One group of animals received intra-LPGi injection of SB 334867 (0.2 µl, 100 µM) immediately before naloxone. In the control group, SB-334867 vehicle (DMSO 1%, 0.2 µl) was microinjected into LPGi. Our results indicate that intra-LPGi administration of SB 334867 significantly decreases naloxone precipitated morphine withdrawal signs. Thus, it seems that orexin might have a pivotal role in the expression of morphine withdrawal signs through affecting orexin type 1 receptor in LPGi nucleus.


Asunto(s)
Bulbo Raquídeo/efectos de los fármacos , Morfina/efectos adversos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Antagonistas de los Receptores de Orexina , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Masculino , Bulbo Raquídeo/metabolismo , Naloxona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Ratas Wistar , Síndrome de Abstinencia a Sustancias/psicología
10.
Neurosci Lett ; 548: 137-42, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23756176

RESUMEN

Large cholinergic synaptic boutons called "C-terminals" contact motoneurons and regulate their excitability. C-terminals in the spinal somatic motor nuclei originate from cholinergic interneurons in laminae VII and X that express a transcription factor Pitx2. Cranial motor nuclei contain another type of motoneuron: branchiomotor neurons. Although branchiomotor neurons receive abundant C-terminal projections, the neural source of these C-terminals remains unknown. In the present study, we first examined whether cholinergic neurons express Pitx2 in the reticular formation of the adult mouse brainstem, as in the spinal cord. Although Pitx2-positive cholinergic neurons were observed in the magnocellular reticular formation and region around the central canal in the caudal medulla, none was present more rostrally in the brainstem tegmentum. We next explored the origin of C-terminals in the branchiomotor nuclei by using biotinylated dextran amine (BDA). BDA injections into the magnocellular reticular formation of the medulla and pons resulted in the labeling of numerous C-terminals in the branchiomotor nuclei: the ambiguous, facial, and trigeminal motor nuclei. Our results revealed that the origins of C-terminals in the branchiomotor nuclei are cholinergic neurons in the magnocellular reticular formation not only in the caudal medulla, but also at more rostral levels of the brainstem, which lacks Pitx2-positive neurons.


Asunto(s)
Neuronas Colinérgicas/citología , Neuronas Colinérgicas/metabolismo , Proteínas de Homeodominio/metabolismo , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Formación Reticular/citología , Formación Reticular/metabolismo , Factores de Transcripción/metabolismo , Animales , Femenino , Masculino , Ratones Endogámicos ICR , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Proteína del Homeodomínio PITX2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA