Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Neuropharmacology ; 240: 109713, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37689261

RESUMEN

2-(4-Bromo-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)etanoamine (25B-NBOMe) is a highly selective 5-HT2A receptor agonist, exhibiting a potent hallucinogenic activity. In the present study, we investigated the effect of a 7-day treatment with 25B-NBOMe in a dose of 0.3 mg/kg on the following: the neurotransmitter release in vivo using microdialysis in freely moving animals, hallucinogenic activity measured in the Wet Dog Shake (WDS) test, anxiety level as measured in the light/dark box (LDB) and locomotor activity in the open field (OF) test, DNA damage with the comet assay, and on a number of neuronal and glial cells with immunohistochemistry. Repeated administration of 25B-NBOMe decreased the response to a challenge dose (0.3 mg/kg) on DA, 5-HT and glutamatergic neurons in the rats' frontal cortex, striatum, and nucleus accumbens. The WDS response dropped drastically after the second day of treatment, suggesting a rapid development of tolerance. LDB and OF tests showed that the effect of 25B-NBOMe on anxiety depends on the treatment and environmental settings. Results obtained with the comet assay indicate a genotoxic properties in the frontal cortex and hippocampus. An increase in immunopositive glial but not neuronal cells was observed in the cortical regions but not in the hippocampus. In conclusion, our study showed that a chronic administration of 25B-NBOMe produces the development of tolerance observed in the neurotransmitters release and hallucinogenic activity. The oxidative damage of cortical and hippocampal DNA implies the generation of free radicals by the drug, resulting in genotoxicity but rather not in neurotoxic tissue damage. Behavioral tests show that 25B-NBOMe exerts anxiogenic effect after single and repeated treatment.

2.
Brain Sci ; 13(3)2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36979236

RESUMEN

Schizophrenia is regarded as a neurodevelopmental disorder with its course progressing throughout life. However, the aetiology and development of schizophrenia are still under investigation. Several data suggest that the dysfunction of epigenetic mechanisms is known to be involved in the pathomechanism of this mental disorder. The present article revised the epigenetic background of schizophrenia based on the data available in online databases (PubMed, Scopus). This paper focused on the role of epigenetic regulation, such as DNA methylation, histone modifications, and interference of non-coding RNAs, in schizophrenia development. The article also reviewed the available data related to epigenetic regulation that may modify the severity of the disease as a possible target for schizophrenia pharmacotherapy. Moreover, the effects of antipsychotics on epigenetic malfunction in schizophrenia are discussed based on preclinical and clinical results. The obtainable data suggest alterations of epigenetic regulation in schizophrenia. Moreover, they also showed the important role of epigenetic modifications in antipsychotic action. There is a need for more data to establish the role of epigenetic mechanisms in schizophrenia therapy. It would be of special interest to find and develop new targets for schizophrenia therapy because patients with schizophrenia could show little or no response to current pharmacotherapy and have treatment-resistant schizophrenia.

3.
J Neurochem ; 164(5): 598-612, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36161462

RESUMEN

Noradrenergic neurotransmission is a critical mediator of stress responses. In turn, exposure to stress induces noradrenergic system adaptations, some of which are implicated in the etiology of stress-related disorders. Adrenergic receptors (ARs) in the ventral tegmental area (VTA) have been demonstrated to regulate phasic dopamine (DA) release in the forebrain, necessary for behavioral responses to conditional cues. However, the impact of stress on noradrenergic modulation of the VTA has not been previously explored. We demonstrate that ARs in the VTA regulate dopaminergic activity in the VTA-BLA (basolateral amygdala) circuit, a key system for processing stress-related stimuli; and that such control is altered by acute stress. We utilized fast-scan cyclic voltammetry to assess the effects of intra-VTA microinfusion of α1 -AR and α2 -AR antagonists (terazosin and RX-821002, respectively), on electrically evoked phasic DA release in the BLA in stress-naïve and stressed (unavoidable electric shocks - UES) anesthetized male Sprague-Dawley rats. In addition, we used western blotting to explore UES-induced alterations in AR protein level in the VTA. Intra-VTA terazosin or RX-821002 dose-dependently attenuated DA release in the BLA. Interestingly, UES decreased the effects of intra-VTA α2 -AR blockade on DA release (24 h but not 7 days after stress), while the effects of terazosin were unchanged. Despite changes in α2 -AR physiological function in the VTA, UES did not alter α2 -AR protein levels in either intracellular or membrane fractions. These findings demonstrate that NA-ergic modulation of the VTA-BLA circuit undergoes significant alterations in response to acute stress, with α2 -AR signaling indicated as a key target.


Asunto(s)
Transducción de Señal , Área Tegmental Ventral , Ratas , Animales , Masculino , Área Tegmental Ventral/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Transmisión Sináptica , Dopamina/metabolismo , Norepinefrina/metabolismo
4.
Int J Mol Sci ; 25(1)2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38203271

RESUMEN

The pathophysiology of depression is related to the reduced volume of the hippocampus and amygdala and hypertrophy of the nucleus accumbens. The mechanism of these changes is not well understood; however, clinical studies have shown that the administration of the fast-acting antidepressant ketamine reversed the decrease in hippocampus and amygdala volume in depressed patients, and the magnitude of this effect correlated with the reduction in depressive symptoms. In the present study, we attempted to find out whether the psychedelic substance psilocybin affects neurotransmission in the limbic system in comparison to ketamine. Psilocybin and ketamine increased the release of dopamine (DA) and serotonin (5-HT) in the nucleus accumbens of naive rats as demonstrated using microdialysis. Both drugs influenced glutamate and GABA release in the nucleus accumbens, hippocampus and amygdala and increased ACh levels in the hippocampus. The changes in D2, 5-HT1A and 5-HT2A receptor density in the nucleus accumbens and hippocampus were observed as a long-lasting effect. A marked anxiolytic effect of psilocybin in the acute phase and 24 h post-treatment was shown in the open field test. These data provide the neurobiological background for psilocybin's effect on stress, anxiety and structural changes in the limbic system and translate into the antidepressant effect of psilocybin in depressed patients.


Asunto(s)
Ketamina , Psilocibina , Humanos , Animales , Ratas , Psilocibina/farmacología , Ketamina/farmacología , Sistema Límbico , Ácido Glutámico , Antidepresivos/farmacología
5.
Int J Mol Sci ; 23(12)2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35743159

RESUMEN

Clinical studies provide evidence that ketamine and psilocybin could be used as fast-acting antidepressants, though their mechanisms and toxicity are still not fully understood. To address this issue, we have examined the effect of a single administration of ketamine and psilocybin on the extracellular levels of neurotransmitters in the rat frontal cortex and reticular nucleus of the thalamus using microdialysis. The genotoxic effect and density of glutamate receptor proteins was measured with comet assay and Western blot, respectively. An open field test, light-dark box test and forced swim test were conducted to examine rat behavior 24 h after drug administration. Ketamine (10 mg/kg) and psilocybin (2 and 10 mg/kg) increased dopamine, serotonin, glutamate and GABA extracellular levels in the frontal cortex, while psilocybin also increased GABA in the reticular nucleus of the thalamus. Oxidative DNA damage due to psilocybin was observed in the frontal cortex and from both drugs in the hippocampus. NR2A subunit levels were increased after psilocybin (10 mg/kg). Behavioral tests showed no antidepressant or anxiolytic effects, and only ketamine suppressed rat locomotor activity. The observed changes in neurotransmission might lead to genotoxicity and increased NR2A levels, while not markedly affecting animal behavior.


Asunto(s)
Ketamina , Animales , Antidepresivos/farmacología , Conducta Animal , Encéfalo/metabolismo , ADN/farmacología , Ketamina/farmacología , Neurotransmisores/farmacología , Psilocibina/farmacología , Ratas , Receptores de Glutamato/metabolismo , Ácido gamma-Aminobutírico/metabolismo
6.
Sci Rep ; 12(1): 2939, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-35190675

RESUMEN

4-Iodo-2,5-dimethoxy-N-(2-methoxybenzyl)phenethylamine (25I-NBOMe) is a new psychoactive substance with strong hallucinogenic properties. Our previous data reported increased release of dopamine, serotonin, and glutamate after acute injections and a tolerance development in the neurotransmitters release and rats' behavior after chronic treatment with 25I-NBOMe. The recreational use of 25I-NBOMe is associated with severe intoxication and deaths in humans. There is no data about 25I-NBOMe in vivo toxicity towards the brain tissue. In this article 25I-NBOMe-crossing through the blood-brain barrier (BBB), the impact on DNA damage, apoptosis induction, and changes in the number of cortical and hippocampal cells were studied. The presence of 25I-NBOMe in several brain regions shortly after the drug administration and its accumulation after multiple injections was found. The DNA damage was detected 72 h after the chronic treatment. On the contrary, at the same time point apoptotic signal was not identified. A decrease in the number of glial but not in neural cells in the frontal (FC) and medial prefrontal cortex (mPFC) was observed. The obtained data indicate that 25I-NBOMe passes easily across the BBB and accumulates in the brain tissue. Observed oxidative DNA damage may lead to the glial cells' death.


Asunto(s)
Encéfalo/efectos de los fármacos , Dimetoxifeniletilamina/análogos & derivados , Alucinógenos/toxicidad , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Daño del ADN/efectos de los fármacos , Dimetoxifeniletilamina/administración & dosificación , Dimetoxifeniletilamina/metabolismo , Dimetoxifeniletilamina/toxicidad , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Humanos , Inyecciones , Neuroglía/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Serotonina/metabolismo
7.
Int J Mol Sci ; 22(16)2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34445411

RESUMEN

BACKGROUND: The present study investigated the role of proteins from the bromodomain and extra-terminal (BET) family in schizophrenia-like abnormalities in a neurodevelopmental model of schizophrenia induced by prenatal methylazoxymethanol (MAM) administration (MAM-E17). METHODS: An inhibitor of BET proteins, JQ1, was administered during adolescence on postnatal days (P) 23-P29, and behavioural responses (sensorimotor gating, recognition memory) and prefrontal cortical (mPFC) function (long-term potentiation (LTP), molecular and proteomic analyses) studies were performed in adult males and females. RESULTS: Deficits in sensorimotor gating and recognition memory were observed only in MAM-treated males. However, adolescent JQ1 treatment affected animals of both sexes in the control but not MAM-treated groups and reduced behavioural responses in both sexes. An electrophysiological study showed LTP impairments only in male MAM-treated animals, and JQ1 did not affect LTP in the mPFC. In contrast, MAM did not affect activity-dependent gene expression, but JQ1 altered gene expression in both sexes. A proteomic study revealed alterations in MAM-treated groups mainly in males, while JQ1 affected both sexes. CONCLUSIONS: MAM-induced schizophrenia-like abnormalities were observed only in males, while adolescent JQ1 treatment affected memory recognition and altered the molecular and proteomic landscape in the mPFC of both sexes. Thus, transient adolescent inhibition of the BET family might prompt permanent alterations in the mPFC.


Asunto(s)
Azepinas/administración & dosificación , Acetato de Metilazoximetanol/análogos & derivados , Corteza Prefrontal/crecimiento & desarrollo , Esquizofrenia/fisiopatología , Triazoles/administración & dosificación , Adolescente , Desarrollo del Adolescente/efectos de los fármacos , Animales , Azepinas/farmacología , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Acetato de Metilazoximetanol/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteómica , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/inducido químicamente , Esquizofrenia/metabolismo , Caracteres Sexuales , Triazoles/farmacología
8.
Elife ; 92020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32687056

RESUMEN

Cannabinoid CB2 receptor (CB2) agonists are potential analgesics void of psychotropic effects. Peripheral immune cells, neurons and glia express CB2; however, the involvement of CB2 from these cells in neuropathic pain remains unresolved. We explored spontaneous neuropathic pain through on-demand self-administration of the selective CB2 agonist JWH133 in wild-type and knockout mice lacking CB2 in neurons, monocytes or constitutively. Operant self-administration reflected drug-taking to alleviate spontaneous pain, nociceptive and affective manifestations. While constitutive deletion of CB2 disrupted JWH133-taking behavior, this behavior was not modified in monocyte-specific CB2 knockouts and was increased in mice defective in neuronal CB2 knockouts suggestive of increased spontaneous pain. Interestingly, CB2-positive lymphocytes infiltrated the injured nerve and possible CB2transfer from immune cells to neurons was found. Lymphocyte CB2depletion also exacerbated JWH133 self-administration and inhibited antinociception. This work identifies a simultaneous activity of neuronal and lymphoid CB2that protects against spontaneous and evoked neuropathic pain.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Cannabinoides/farmacología , Neuralgia/tratamiento farmacológico , Sustancias Protectoras/farmacología , Receptores de Cannabinoides/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Distribución Aleatoria , Autoadministración
9.
Neurotox Res ; 37(4): 800-814, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32026358

RESUMEN

Disturbances in the function of the mesostriatal dopamine system may contribute to the development and maintenance of chronic pain, including its sensory and emotional/cognitive aspects. In the present study, we assessed the influence of chronic constriction injury (CCI) of the sciatic nerve on the expression of genes coding for dopamine and opioid receptors as well as opioid propeptides in the mouse mesostriatal system, particularly in the nucleus accumbens. We demonstrated bilateral increases in mRNA levels of the dopamine D1 and D2 receptors (the latter accompanied by elevated protein level), opioid propeptides proenkephalin and prodynorphin, as well as delta and kappa (but not mu) opioid receptors in the nucleus accumbens at 7 to 14 days after CCI. These results show that CCI-induced neuropathic pain is accompanied by a major transcriptional dysregulation of molecules involved in dopaminergic and opioidergic signaling in the striatum/nucleus accumbens. Possible functional consequences of these changes include opposite effects of upregulated enkephalin/delta opioid receptor signaling vs. dynorphin/kappa opioid receptor signaling, with the former most likely having an analgesic effect and the latter exacerbating pain and contributing to pain-related negative emotional states.


Asunto(s)
Neuralgia/metabolismo , Dimensión del Dolor/métodos , Prosencéfalo/metabolismo , Receptores Dopaminérgicos/biosíntesis , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Animales , Cuerpo Estriado/metabolismo , Encefalinas/biosíntesis , Encefalinas/genética , Expresión Génica , Masculino , Ratones , Neuralgia/genética , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Receptores Dopaminérgicos/genética , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/biosíntesis , Receptores Opioides mu/genética
10.
Br J Pharmacol ; 177(5): 1187-1205, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31655493

RESUMEN

BACKGROUND AND PURPOSE: Mu and delta opioid receptors(MOP, DOP) contribution to the manifestations of pathological pain is not understood. We used genetic approaches to investigate the opioid mechanisms modulating neuropathic pain and its comorbid manifestations. EXPERIMENTAL APPROACH: We generated conditional knockout mice with MOP or DOP deletion in sensoryNav1.8-positive neurons (Nav1.8), in GABAergic forebrain neurons (DLX5/6) orconstitutively (CMV). Mutant mice and wild-type littermates were subjected topartial sciatic nerve ligation (PSNL) or sham surgery and their nociception wascompared. Anxiety-, depressivelike behaviour and cognitive performance were also measured. Opioid receptor mRNA expression, microgliosis and astrocytosis were assessed in the dorsalroot ganglia (DRG) and/or the spinal cord (SC). KEY RESULTS: Constitutive CMV-MOP knockouts after PSNL displayed reduced mechanical allodynia and enhanced heat hyperalgesia. This phenotype was accompanied by increased DOP expression in DRG and SC, and reduced microgliosis and astrocytosis in deep dorsal horn laminae. Conditional MOP knockouts and control mice developed similar hypersensitivity after PSNL, except for anenhanced heat hyperalgesia by DLX5/6-MOP male mice. Neuropathic pain-induced anxiety was aggravated in CMV-MOP and DLX5/6-MOP knockouts. Nerve-injured CMV-DOP mice showed increased mechanical allodynia, whereas Nav1.8-DOP and DLX5/8-DOP mice had partial nociceptive enhancement. CMV-DOP and DLX5/6-DOP mutants showed increased depressive-like behaviour after PSNL. CONCLUSIONS AND IMPLICATIONS: MOP activity after nerve injury increased anxiety-like responses involving forebrain GABAergic neurons and enhanced mechanical pain sensitivity along with repression of DOP expression and spinal cord gliosis. In contrast, DOP shows a protective function limiting nociceptive and affective manifestations of neuropathic pain.


Asunto(s)
Nocicepción , Receptores Opioides delta , Animales , Hiperalgesia , Masculino , Ratones , Ratones Mutantes Neurológicos , Receptores Opioides , Receptores Opioides delta/genética , Receptores Opioides mu/genética
11.
Transl Psychiatry ; 8(1): 255, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30487639

RESUMEN

Stress elicits the release of glucocorticoids (GCs) that regulate energy metabolism and play a role in emotional memory. Astrocytes express glucocorticoid receptors (GR), but their contribution to cognitive effects of GC's action in the brain is unknown. To address this question, we studied how astrocyte-specific elimination of GR affects animal behavior known to be regulated by stress. Mice with astrocyte-specific ablation of GR presented impaired aversive memory expression in two different paradigms of Pavlovian learning: contextual fear conditioning and conditioned place aversion. These mice also displayed compromised regulation of genes encoding key elements of the glucose metabolism pathway upon GR stimulation. In particular, we identified that the glial, but not the neuronal isoform of a crucial stress-response molecule, Sgk1, undergoes GR-dependent regulation in vivo and demonstrated the involvement of SGK1 in regulation of glucose uptake in astrocytes. Together, our results reveal astrocytes as a central element in GC-dependent formation of aversive memory and suggest their relevance for stress-induced alteration of brain glucose metabolism. Consequently, astrocytes should be considered as a cellular target of therapies of stress-induced brain diseases.


Asunto(s)
Astrocitos/metabolismo , Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Memoria/fisiología , Nocicepción/fisiología , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/fisiología , Estrés Psicológico/metabolismo , Animales , Proteínas Inmediatas-Precoces/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/metabolismo
12.
Neuroscience ; 390: 293-302, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30176322

RESUMEN

Clinical studies have reported lower effectivity of opioid drugs in therapy of neuropathic pain. Therefore, to determine the changes in endogenous opioid systems in this pain more precisely, we have studied the changes in the pain-related behavior on days 1, 14, and 28 following a chronic constriction injury (CCI) to the sciatic nerve in mice. In parallel, we have studied the changes of µ-(MOP), δ-(DOP) and κ-(KOP) receptors, proenkephalin (PENK) and prodynorphin (PDYN) mRNA levels, as well as GTPγS binding of opioid receptors on the ipsi- and contralateral parts of the spinal cord and thalamus on the 14th day following CCI, as on this day the greatest manifestation of pain-related behavior was observed. On ipsilateral spinal cord, the decrease in MOP/DOP/KOP receptor and increase in PDYN/PENK mRNA expression was observed. In thalamus, MOP/DOP/KOP receptor expression decreased contralaterally. On ipsilateral side, there were no changes in PDYN/PENK or DOP/KOP receptor expression, but MOP mRNA decreased. The spinal GTPγS binding of MOP/DOP/KOP receptor ligands decreased on the ipsilateral side, yet the effect was less pronounced for DOP receptor ligands. In thalamus, a decrease was observed on the contralateral side for all opioid receptor ligands, especially for DOP ligand. A less pronounced decrease in GTPγS binding of spinal DOP ligands may indicate a weaker stimulation of ascending nociceptive pathways, which could explain the absence of decreased activity of DOP receptor ligands in neuropathy. These findings may suggest that drugs with a higher affinity for the DOP receptor will perform better in neuropathic pain.


Asunto(s)
Encefalinas/metabolismo , Neuralgia/metabolismo , Precursores de Proteínas/metabolismo , Receptores Opioides/metabolismo , Médula Espinal/metabolismo , Tálamo/metabolismo , Animales , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Ratones , Umbral del Dolor , ARN Mensajero/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/metabolismo
13.
Neurotox Res ; 33(3): 532-548, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29134560

RESUMEN

MDMA (3,4-methylenedioxymethamphetamine) is a psychostimulant popular as a recreational drug because of its effect on mood and social interactions. MDMA acts at dopamine (DA) transporter (DAT) and serotonin (5-HT) transporter (SERT) and is known to induce damage of dopamine and serotonin neurons. MDMA is often ingested with caffeine. Caffeine as a non-selective adenosine A1/A2A receptor antagonist affects dopaminergic and serotonergic transmissions. The aim of the present study was to determine the changes in DA and 5-HT release in the mouse striatum induced by MDMA and caffeine after their chronic administration. To find out whether caffeine aggravates MDMA neurotoxicity, the content of DA and 5-HT, density of brain DAT and SERT, and oxidative damage of nuclear DNA were determined. Furthermore, the effect of caffeine on MDMA-induced changes in striatal dynorphin and enkephalin and on behavior was assessed. The DA and 5-HT release was determined with in vivo microdialysis, and the monoamine contents were measured by HPLC with electrochemical detection. DNA damage was assayed with the alkaline comet assay. DAT and SERT densities were determined by immunohistochemistry, while prodynorphin (PDYN) and proenkephalin were determined by quantitative PCR reactions. The behavioral changes were measured by the open-field (OF) test and novel object recognition (NOR) test. Caffeine potentiated MDMA-induced DA release while inhibiting 5-HT release in the mouse striatum. Caffeine also exacerbated the oxidative damage of nuclear DNA induced by MDMA but diminished DAT decrease in the striatum and worsened a decrease in SERT density produced by MDMA in the frontal cortex. Neither the striatal PDYN expression, increased by MDMA, nor exploratory and locomotor activities of mice, decreased by MDMA, were affected by caffeine. The exploration of novel object in the NOR test was diminished by MDMA and caffeine. Our data provide evidence that long-term caffeine administration has a powerful influence on functions of dopaminergic and serotonergic neurons in the mouse brain and on neurotoxic effects evoked by MDMA.


Asunto(s)
Cafeína/administración & dosificación , Estimulantes del Sistema Nervioso Central/administración & dosificación , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Alucinógenos/administración & dosificación , N-Metil-3,4-metilenodioxianfetamina/administración & dosificación , Animales , Ensayo Cometa/métodos , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Conducta Exploratoria/efectos de los fármacos , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Serotonina/metabolismo
14.
Addict Biol ; 22(6): 1802-1816, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27578564

RESUMEN

Chronic exposure to opioids induces adaptations in brain function that lead to the formation of the behavioral and physiological symptoms of drug dependence and addiction. Animal models commonly used to test these symptoms typically last less than two weeks, which is presumably too short to observe the alterations in the brain that accompany drug addiction. Here, we analyzed the phenotypic and molecular effects of nearly lifelong morphine or saccharin intake in C57BL/6J mice. We used multiple paradigms to evaluate the symptoms of compulsive drug intake: a progressive ratio schedule, intermittent access and a schedule involving a risk of punishment were programmed into an automated IntelliCage system. Gene expression profiles were evaluated in the striatum using whole-genome microarrays and further validated using quantitative polymerase chain reaction in the striatum and the prefrontal cortex. Mice voluntary self-administering morphine showed addiction-related behavioral pattern that included: higher motivation to work for a drug reward, increased reward seeking and increased craving. The analysis of molecular changes revealed a tolerance effect in the transcriptional response to morphine injection (20 mg/kg, ip), as well as some long-lasting alterations in gene expression profiles between the analyzed groups of animals. Interestingly, among the morphine-drinking animals, certain transcriptional profiles were found to be associated with alterations in behavior. In conclusion, our model represents a novel approach for investigating the behavioral and molecular mechanisms underlying opioid addiction. Prolonged morphine intake caused adaptive processes in the brain that manifested as altered behavior and transcriptional sensitivity to opioids.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Trastornos Relacionados con Opioides/fisiopatología , Analgésicos Opioides/administración & dosificación , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Autoadministración
15.
Behav Brain Res ; 292: 95-101, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26051817

RESUMEN

The molecular mechanisms underlying the susceptibility or resilience to trauma-related disorders remain incompletely understood. Opioids modulate emotional learning, but the roles of specific receptors are unclear. Here, we aimed to analyze the contribution of the opioid system to fear responses in two inbred mouse strains exhibiting distinct behavioral phenotypes. SWR/J and C57BL/6J mice were subjected to five consecutive electric footshocks (1mA each), and the contextual freezing time was measured. Stress-induced alterations in gene expression were analyzed in the amygdala and the hippocampus. In both strains, the fear response was modulated using pharmacological tools. SWR/J mice did not develop conditioned fear but exhibited increased transcriptional expression of Pdyn and Penk in the amygdala region. Blocking opioid receptors prior to the footshocks using naltrexone (2 mg/kg) or naltrindole (5 mg/kg) increased the freezing responses in these animals. The C57BL/6J strain displayed high conditioned fear, although no alteration in the mRNA abundance of genes encoding opioid precursors was observed. Double-injection of morphine (20 mg/kg) following stress and upon context re-exposure prevented the enhancement of freezing. Moreover, selective delta and kappa agonists caused a reduction in conditioned fear responses. To summarize, the increased expression of the Pdyn and Penk genes corresponded to reduced intensity of fear responses. Blockade of the endogenous opioid system restored freezing behavior in stress-resistant animals. The pharmacological stimulation of the kappa and delta opioid receptors in stress-susceptible individuals may alleviate fear. Thus, subtype-selective opioid receptor agonists may protect against the development of trauma-related disorders.


Asunto(s)
Analgésicos Opioides/farmacología , Miedo/efectos de los fármacos , Miedo/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Condicionamiento Clásico , Encefalinas/biosíntesis , Encefalinas/genética , Expresión Génica/efectos de los fármacos , Estudios de Asociación Genética , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Distribución Aleatoria , Receptores Opioides/agonistas , Estrés Psicológico/genética , Estrés Psicológico/psicología
16.
Eur J Neurosci ; 30(6): 1101-10, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19735294

RESUMEN

Persistent changes that take place during the development of opioid addiction are thought to be due to reorganization of synaptic connections in relevant brain circuits. This neuronal plasticity requires trafficking of signaling molecules that are controlled by kinesins. In neurons, kinesin light chain 1 (KLC1) acts as the primary regulator of kinesin action. We observed that KLC1 was enriched in sub-cortical regions of the brain in C57Bl/6J mice. KLC1 expression was especially enriched in the striatum, hippocampus and amygdala, which are known to be involved in opioid addiction. Our study revealed that conditioning of C57Bl/6J mice with morphine elevated KLC1 levels in the amygdala, frontal cortex and hippocampus, but not in the striatum. Further study revealed that alterations in KLC1 protein levels in the studied brain regions correlated with the expression of morphine-induced conditioned place preference. In the cortex, hippocampus and amygdala, KLC1 co-localized with calcium/calmodulin-dependent protein kinase II (CaMKII), suggesting that KLC1 was present in the cell bodies and dendrites of pyramidal neurons. Our findings indicate that KLC1, a molecule involved in dendritic and axonal transport in the brain, is affected during chronic morphine treatment and may be involved in the development of opioid addiction.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Cuerpo Estriado/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Morfina/farmacología , Recompensa , Análisis de Varianza , Animales , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Cinesinas , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/metabolismo , Actividad Motora/efectos de los fármacos , Neuronas/metabolismo
17.
Eur J Pharmacol ; 623(1-3): 65-72, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19766105

RESUMEN

The pharmacological attenuation of glial activation represents a novel approach for controlling neuropathic pain, but the role of microglial and astroglial cells is not well established. To better understand the potential role of two types of glial cells, microglia and astrocytes, in the pathogenesis of neuropathic pain, we examined markers associated with them by quantitative RT-PCR, western blot and immunohistochemical analyses in the dorsal horn of the lumbar spinal cord 7days after chronic constriction injury (CCI) to the sciatic nerve in mice. The mRNA and protein of microglial cells were labeled with C1q and OX42(CD11b/c), respectively. The mRNA and protein of astrocytes were labeled with GFAP. The RT-PCR results indicated an increase in C1q mRNA that was more pronounced than the increased expression of GFAP mRNA ipsilateral to the injury in the dorsal spinal cord. Similarly, western blot and immunohistochemical analyses demonstrated an ipsilateral upregulation of OX42-positive cells (72 and 20%, respectively) and no or little (8% upregulation) change in GFAP-positive cells in the ipsilateral dorsal lumbar spinal cord. We also found that chronic intraperitoneal injection of the minocycline (microglial inhibitor) and pentoxifylline (cytokine inhibitor) attenuated CCI-induced activation of microglia, and both, but not fluorocitrate (astroglial inhibitor), diminished neuropathic pain symptoms and tactile and cold sensitivity. Our findings indicate that spinal microglia are more activated than astrocytes in peripheral injury-induced neuropathic pain. These findings implicate a glial regulation of the pain response and suggest that pharmacologically targeting microglia could effectively prevent clinical pain syndromes in programmed and/or anticipated injury.


Asunto(s)
Astrocitos/fisiología , Modelos Animales de Enfermedad , Microglía/fisiología , Neuralgia/fisiopatología , Nociceptores/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Médula Espinal/citología , Analgésicos/uso terapéutico , Animales , Biomarcadores/metabolismo , Citratos/uso terapéutico , Complemento C1q/genética , Complemento C1q/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones , Minociclina/uso terapéutico , Neuralgia/tratamiento farmacológico , Especificidad de Órganos , Dimensión del Dolor , Pentoxifilina/uso terapéutico , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico
18.
Brain Behav Immun ; 23(1): 75-84, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18684397

RESUMEN

We have previously demonstrated that glial inhibitors reduce the development of allodynia and hyperalgesia, potentiating the effect of a single morphine dose in a neuropathic pain model. This study explores the effects of two glial activation inhibitors, minocycline and pentoxifylline, on the development of tolerance to morphine in naive and chronic constriction injury (CCI)-exposed mice. Administration of morphine to naive (20 mg/kg; i.p.) and CCI-exposed mice (40 mg/kg; i.p.) twice daily resulted in tolerance to its anti-nociceptive effect after 6 days. Injections of morphine were combined with minocycline (30 mg/kg, i.p.) or pentoxifylline (20 mg/kg, i.p.) administered as two preemptive doses before first morphine administration in naive or pre-injury in CCI-exposed mice, and repeated twice daily 30 min before each morphine administration. With treatment, development of morphine tolerance was delayed by 5 days (from 6 to 11 days), as measured by the tail-flick test in naive and by tail-flick, von Frey, and cold plate tests in CCI-exposed mice. Western blot analysis of CD11b/c and GFAP protein demonstrated that minocycline and pentoxifylline, at doses delaying development of tolerance to morphine analgesia, significantly diminished the morphine-induced increase in CD11b/c protein level. We found that repeated systemic administration of glial inhibitors significantly delays development of morphine tolerance by attenuating the level of this microglial marker under normal and neuropathic pain conditions. Our results support the idea that targeting microglial activation during morphine therapy/treatment is a novel and clinically promising method for enhancing morphine's analgesic effects, especially in neuropathic pain.


Asunto(s)
Minociclina/farmacología , Morfina/farmacología , Pentoxifilina/farmacología , Neuropatía Ciática/fisiopatología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Western Blotting , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Proteína Ácida Fibrilar de la Glía/metabolismo , Inyecciones Intraperitoneales , Masculino , Ratones , Minociclina/administración & dosificación , Morfina/administración & dosificación , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Dolor/etiología , Dolor/fisiopatología , Dolor/prevención & control , Dimensión del Dolor/métodos , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Pentoxifilina/administración & dosificación , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Nervio Ciático/lesiones , Neuropatía Ciática/etiología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
19.
FEBS J ; 275(15): 3836-49, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18616461

RESUMEN

Extracellular signal-regulated kinases (ERKs) have been shown to be activated by opioids and functionally linked to addiction. Morphine-associated changes in ERK activity seem to be the characteristic features of opioid action. In this study, we observed a rapid and severe increase in ERK1/2 activity after a 5 min morphine treatment of HEK-MOR cells (transfected with the rat mu-opioid receptor MOR1) expressing mu-opioid receptor. Cellular adaptations to chronic (72 h) morphine treatment were manifested by a slight and sustained increase in ERK1/2 activity. Withdrawal caused by an opioid receptor antagonist - naloxone - attenuated phosphorylation of ERK1/2. Little information is available on the precise mechanism of ERK activity regulation. Using RNA interference technology, we generated stably transfected cells with silenced expression of cAMP-responsive element binding factor (CREB) and Ets-like protein-1 (Elk-1) transcription factors, which are known targets for activated ERK1/2. In these cells, ERK1/2 activity regulation was altered. Silencing of CREB or Elk-1 significantly increased ERK activation observed after 5 min of morphine stimulation. The initial level of activated ERKs in these cells was also augmented. Moreover, the cellular response to withdrawal signals and chronic opioid treatment was diminished. These differences suggest that both CREB-dependent and Elk-1-dependent transcription contribute to the expression of proteins regulating morphine-induced ERK activity (particular phosphatases, upstream kinases or their activatory proteins).


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfina/farmacología , ARN Interferente Pequeño/fisiología , Transcripción Genética/fisiología , Proteína Elk-1 con Dominio ets/fisiología , Western Blotting , Línea Celular , Silenciador del Gen , Humanos , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Neuropharmacology ; 54(8): 1239-46, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18456291

RESUMEN

alpha-Synuclein is a presynaptic protein proposed to serve as a negative regulator of dopaminergic neurotransmission. Recent research has implicated alpha-synuclein in chronic neuroadaptations produced by psychostimulant and opiate use, as well as in genetically determined susceptibility to alcoholism in humans. The aim of our study was to characterize the changes in alpha-synuclein expression after short-term abstinence from chronic alcohol drinking in mice. Male C57BL/6J mice were allowed to drink increasing concentrations of alcohol in the two-bottle choice procedure. Then the mice were given constant access to an 8% alcohol solution and water for 32 days, and were sacrificed 2 h, 24 h or 48 h after alcohol withdrawal. RT-PCR, in situ hybridization and Western blotting techniques were used to measure alpha-synuclein mRNA and protein levels in the brain and blood. alpha-Synuclein protein levels were elevated by up to 80% in the amygdala of mice withdrawn from alcohol for 24 h or 48 h. No changes in alpha-synuclein levels were found in the mesencephalon or striatum/accumbens. The levels of alpha-synuclein mRNA remained unchanged in all brain regions examined (the striatum, nucleus accumbens, amygdala, substantia nigra, ventral tegmental area). alpha-Synuclein mRNA was up-regulated in the whole blood 48 h after alcohol withdrawal. The accumulation of alpha-synuclein in the amygdala, observed in this study, seems to be a common feature of alcohol and opiate abstinence. This finding suggests a role of alpha-synuclein in common neuroadaptations produced by long-term alcohol and drug use. Although alpha-synuclein expression in the blood seems unrelated to that in the brain, it may serve as a peripheral biomarker of chronic alcohol consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Química Encefálica , Síndrome de Abstinencia a Sustancias/psicología , alfa-Sinucleína/biosíntesis , Animales , Western Blotting , Encéfalo/patología , Procesamiento de Imagen Asistido por Computador , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Abstinencia a Sustancias/genética , alfa-Sinucleína/sangre , alfa-Sinucleína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...