Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Hum Mol Genet ; 29(23): 3744-3756, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33084871

RESUMO

Several X-linked neurodevelopmental disorders including Rett syndrome, induced by mutations in the MECP2 gene, and fragile X syndrome (FXS), caused by mutations in the FMR1 gene, share autism-related features. The mRNA coding for methyl CpG binding protein 2 (MeCP2) has previously been identified as a substrate for the mRNA-binding protein, fragile X mental retardation protein (FMRP), which is silenced in FXS. Here, we report a homeostatic relationship between these two key regulators of gene expression in mouse models of FXS (Fmr1 Knockout (KO)) and Rett syndrome (MeCP2 KO). We found that the level of MeCP2 protein in the cerebral cortex was elevated in Fmr1 KO mice, whereas MeCP2 KO mice displayed reduced levels of FMRP, implicating interplay between the activities of MeCP2 and FMRP. Indeed, knockdown of MeCP2 with short hairpin RNAs led to a reduction of FMRP in mouse Neuro2A and in human HEK-293 cells, suggesting a reciprocal coupling in the expression level of these two regulatory proteins. Intra-cerebroventricular injection of an adeno-associated viral vector coding for FMRP led to a concomitant reduction in MeCP2 expression in vivo and partially corrected locomotor hyperactivity. Additionally, the level of MeCP2 in the posterior cortex correlated with the severity of the hyperactive phenotype in Fmr1 KO mice. These results demonstrate that MeCP2 and FMRP operate within a previously undefined homeostatic relationship. Our findings also suggest that MeCP2 overexpression in Fmr1 KO mouse posterior cerebral cortex may contribute to the fragile X locomotor hyperactivity phenotype.


Assuntos
Córtex Cerebral/patologia , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/fisiologia , Síndrome do Cromossomo X Frágil/patologia , Regulação da Expressão Gênica , Proteína 2 de Ligação a Metil-CpG/fisiologia , Fenótipo , Animais , Córtex Cerebral/metabolismo , Feminino , Síndrome do Cromossomo X Frágil/etiologia , Síndrome do Cromossomo X Frágil/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
J Neurol ; 270(4): 2048-2058, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36565348

RESUMO

OBJECTIVE: Neuromyelitis optica spectrum disorders (NMOSD) represent rare autoimmune diseases of the central nervous system largely targeting optic nerve(s) and spinal cord. The present analysis used real-world data to identify clinical and epidemiological correlates of treatment change in patients with NMOSD. METHODS: CIRCLES is a longitudinal, observational study of NMOSD conducted at 15 centers across North America. Patients with ≥ 60 days of follow-up and receiving on-study maintenance treatment were evaluated. The mean annual relapse rate (ARR) was estimated using negative binomial models; the likelihood of treatment change was estimated using Cox proportional hazards models. Relapses were included as time-varying covariates to estimate the relationship to treatment change. RESULTS: Of 542 patients included, 171 (31.5%) experienced ≥ 1 relapse on the study and 133 patients (24.5%) had ≥ 1 change in the treatment regimen. Two categories of variables significantly correlated with the likelihood of treatment change: (1) relapse: any on-study relapse (hazard ratio [HR] = 2.91; p < 0.001), relapse phenotypes (HR range = 2.15-5.49; p < 0.001), and pre-study ARR > 0.75 (HR 2.28; p < 0.001); 2) disease phenotype: brain syndrome only vs transverse myelitis involvement at onset (HR 2.44; p = 0.008), disease duration < 1 vs > 5 years (HR 1.66; p = 0.028), or autoimmune comorbidity (HR 1.55; p = 0.015). A subset of these factors significantly correlated with shorter time to first rituximab discontinuation. CONCLUSIONS: In CIRCLES, relapse patterns and disease phenotype significantly correlated with changes in the maintenance treatment regimen. Such findings may facilitate the identification of patients with NMOSD who are likely to benefit from treatment change to reduce relapse risk or disease burden and enhance the quality of life.


Assuntos
Neuromielite Óptica , Humanos , Neuromielite Óptica/tratamento farmacológico , Neuromielite Óptica/epidemiologia , Qualidade de Vida , Recidiva Local de Neoplasia , Medula Espinal , Estudos Longitudinais , Aquaporina 4 , Estudos Retrospectivos , Autoanticorpos
3.
J Neurosci ; 31(31): 11172-83, 2011 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-21813678

RESUMO

The basolateral amygdala (BLA), ventral tegmental area (VTA), and nucleus accumbens (NAc) play central roles in the processing of opiate-related associative reward learning and memory. The BLA receives innervation from dopaminergic fibers originating in the VTA, and both dopamine (DA) D1 and D2 receptors are expressed in this region. Using a combination of in vivo single-unit extracellular recording in the NAc combined with behavioral pharmacology studies, we have identified a double dissociation in the functional roles of DA D1 versus D2 receptor transmission in the BLA, which depends on opiate exposure state; thus, in previously opiate-naive rats, blockade of intra-BLA D1, but not D2, receptor transmission blocked the acquisition of associative opiate reward memory, measured in an unbiased conditioned place preference procedure. In direct contrast, in rats made opiate dependent and conditioned in a state of withdrawal, intra-BLA D2, but not D1, receptor blockade blocked opiate reward encoding. This functional switch was dependent on cAMP signaling as comodulation of intra-BLA cAMP levels reversed or replicated the functional effects of intra-BLA D1 or D2 transmission during opiate reward processing. Single-unit in vivo extracellular recordings performed in neurons of the NAc confirmed an opiate-state-dependent role for BLA D1/D2 transmission in NAc neuronal response patterns to morphine. Our results characterize and identify a novel opiate addiction switching mechanism directly in the BLA that can control the processing of opiate reward information as a direct function of opiate exposure state via D1 or D2 receptor signaling substrates.


Assuntos
Tonsila do Cerebelo/fisiologia , Memória/fisiologia , Núcleo Accumbens/fisiologia , Receptores Dopaminérgicos/fisiologia , Receptores Opioides/metabolismo , Recompensa , Potenciais de Ação/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Análise de Variância , Animais , Benzazepinas/farmacologia , Condicionamento Operante/efeitos dos fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Modelos Animais de Doenças , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Memória/efeitos dos fármacos , Morfina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/patologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Salicilamidas/farmacologia , Tionucleotídeos/farmacologia
4.
Cereb Cortex ; 21(1): 68-80, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20392811

RESUMO

The medial prefrontal cortex (mPFC) plays a significant role in associative learning and memory formation during the opiate addiction process. Various lines of evidence demonstrate that glutamatergic (GLUT) transmission through the N-methyl D-aspartate (NMDA) receptor can modulate neuronal network activity within the mPFC and influence dopaminergic signaling within the mesocorticolimbic pathway. However, little is known about how modulation of NMDA receptor signaling within the mPFC may regulate associative opiate reward learning and memory formation. Using a conditioned place preference (CPP) procedure, we examined the effects of selective NMDA receptor blockade directly within the prelimbic cortex (PLC) during the acquisition of associative opiate reward learning. NMDA receptor blockade specifically within the PLC caused a strong potentiation in the rewarding effects of either systemic or intra-ventral tegmental area (intra-VTA) morphine administration. This reward potentiation was dose dependently blocked by coadministration of dopamine D1 or D2 receptor antagonists and by blockade of presynaptic GLUT release. In addition, pharmacological inactivation of the basolateral amygdala (BLA) also prevented intra-PLC NMDA receptor blockade-induced potentiation of opiate reward signals, demonstrating a functional interaction between inputs from the VTA and BLA within the PLC, during the encoding and modulation of associative opiate reward information.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Dopamina/fisiologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Recompensa , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiopatologia , Animais , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Modelos Animais de Doenças , Ácido Glutâmico/fisiologia , Masculino , Transtornos Relacionados ao Uso de Opioides/psicologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/fisiologia
5.
J Neurol Sci ; 427: 117553, 2021 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-34224957

RESUMO

BACKGROUND: Neuromyelitis optica spectrum disorder (NMOSD) is a rare autoimmune disease of the central nervous system that often leads to severe disability. Patients with highly active NMOSD have approximately a 10-times higher hospital inpatient admission rate compared with patients without NMOSD. Accurate assessments of the impact of NMOSD treatments on the burdens of illness require quantitative metrics of these burdens, including costs of care. METHODS: This study evaluated claims data from the IBM MarketScan Commercial and Medicare Supplemental Databases between 2014 and 2018. Patients were included based on inpatient or outpatient claims meeting criteria defined for NMOSD. Non-NMOSD controls were matched 5:1 to patients with NMOSD. Total costs of healthcare services in consumer price index-adjusted 2019 US dollars during the 1-year postindex follow-up period were calculated for patients and controls. RESULTS: Patients with NMOSD required more healthcare services and incurred significantly greater costs for inpatient hospitalizations (annual mean [SD] cost: $29,054 [$144,872] vs controls $1521 [$10,759]), outpatient services ($24,881 [$35,463] vs $4761 [$26,447]), and emergency department (ED) visits ($2400 [$7771] vs $408 [$2579]). Almost 12% of patients with NMOSD were further burdened with plasma exchange or intravenous immunoglobulin G treatments, costing an annual median (interquartile range) of $1684 ($566-$3817) and $24,353 ($5425-$42,975), respectively. CONCLUSIONS: Compared with controls, patients with NMOSD had significantly higher costs associated with hospitalizations, ED visits, and prescriptions. These results highlight the considerable economic burden of NMOSD, which may be favorably impacted by disease-modifying therapies that are regulatory-approved to be safe and effective.


Assuntos
Neuromielite Óptica , Idoso , Assistência Ambulatorial , Bases de Dados Factuais , Hospitalização , Humanos , Medicare , Neuromielite Óptica/terapia , Estados Unidos
6.
J Neurol Sci ; 427: 117530, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34111762

RESUMO

BACKGROUND: Neuromyelitis optica spectrum disorder (NMOSD) is associated with various comorbidities, including non-autoimmune and autoimmune conditions. The burden and cost of illness for NMOSD are unclear, particularly in the context of comorbidities. METHODS: Claims data from IBM MarketScan Commercial and Medicare Supplemental Databases between 2014 and 2018 were analyzed. Patients with NMOSD were specified as having inpatient or outpatient claims for NMOSD diagnosis or specific NMOSD symptoms claims and no subsequent claims for multiple sclerosis (MS) or use of MS disease-modifying therapy (DMT). Continuous enrollment ≥ 6 months before and ≥ 1 year after the first claim (index date) was required for study inclusion. Total costs stratified by comorbidities within 12 months post-index date were calculated per patient and compared 1:5 with matched non-NMOSD controls. RESULTS: A total of 162 patients with NMOSD and 810 non-NMOSD controls were evaluated. A significantly higher proportion of NMOSD patients had comorbidities than non-NMOSD controls (66.7% vs 41.5%; P < 0.001). Concomitant autoimmune disease occurred in 19.1% vs 4.9% (P < 0.001) of patients with NMOSD vs non-NMOSD controls. NMOSD patients incurred significantly higher total median (interquartile range) healthcare costs per patient ($68,386.48 [$23,373.54-$160,862.70]) than matched non-NMOSD controls with autoimmune disease ($17,215.13 [$6715.48-$31,441.93]; P < 0.001) or patients with NMOSD without autoimmune comorbidity ($23,905.42 [$8632.82-$67,251.54]; P = 0.022). Similarly, patients with NMOSD and non-autoimmune comorbidities incurred higher median healthcare costs than matched controls. CONCLUSIONS: Patients with NMOSD experience significant disease burden and cost that are amplified by comorbidities. Effective therapies are needed, particularly for patients with concomitant autoimmune disease.


Assuntos
Neuromielite Óptica , Idoso , Aquaporina 4 , Autoanticorpos , Comorbidade , Humanos , Medicare , Neuromielite Óptica/epidemiologia , Neuromielite Óptica/terapia , Estados Unidos/epidemiologia
7.
Epilepsy Res ; 81(1): 44-51, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18502613

RESUMO

Cannabinoid compounds are anticonvulsant since they have inhibitory effects at micromolar doses, which are mediated by activated receptors coupling to G(i/o) proteins. Surprisingly, both the analgesic and anticonvulsant effects of opioids are enhanced by ultra-low doses (nanomolar to picomolar) of the opioid antagonist naltrexone and as opioid and cannabinoid systems interact, it has been shown that ultra-low dose naltrexone also enhances cannabinoid-induced antinociception. Thus, concerning the seizure modulating properties of both classes of receptors this study investigated whether the ultra-low dose opioid antagonist naltrexone influences cannabinoid anticonvulsant effects. The clonic seizure threshold was tested in separate groups of male NMRI mice following injection of vehicle, the cannabinoid selective agonist arachidonyl-2-chloroethylamide (ACEA) and ultra-low doses of the opioid receptor antagonist naltrexone and a combination of ACEA and naltrexone doses in a model of clonic seizure induced by pentylenetetrazole (PTZ). Systemic injection of ultra-low doses of naltrexone (1pg/kg to 1ng/kg, i.p.) significantly potentiated the anticonvulsant effect of ACEA (1mg/kg, i.p.). Moreover, the very low dose of naltrexone (500pg/kg) unmasked a strong anticonvulsant effect for very low doses of ACEA (10 and 100microg/kg). A similar potentiation by naltrexone (500pg/kg) of anticonvulsant effects of non-effective dose of ACEA (1mg/kg) was also observed in the generalized tonic-clonic model of seizure. The present data indicate that the interaction between opioid and cannabinoid systems extends to ultra-low dose levels and ultra-low doses of opioid receptor antagonist in conjunction with very low doses of cannabinoids may provide a potent strategy to modulate seizure susceptibility.


Assuntos
Anticonvulsivantes , Ácidos Araquidônicos/farmacologia , Agonistas de Receptores de Canabinoides , Canabinoides/agonistas , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Animais , Convulsivantes , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Epilepsia Generalizada/induzido quimicamente , Epilepsia Generalizada/prevenção & controle , Epilepsia Tônico-Clônica/induzido quimicamente , Epilepsia Tônico-Clônica/tratamento farmacológico , Injeções Intraperitoneais , Masculino , Camundongos , Pentilenotetrazol , Piperidinas/farmacologia , Pirazóis/farmacologia
8.
Neuropharmacology ; 53(6): 763-70, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17870135

RESUMO

Several lines of evidence suggest that cannabinoid compounds are anticonvulsant since they have inhibitory effects at micromolar doses, which are mediated by activated receptors coupling to Gi/o proteins. Surprisingly, both the analgesic and anticonvulsant effects of opioids are enhanced by ultra-low doses (nanomolar to picomolar) of the opioid antagonist naltrexone and as opioid and cannabinoid systems interact, it has been shown that ultra-low dose naltrexone also enhances cannabinoid-induced antinociception. However, regarding the seizure modulating properties of both classes of receptors this study investigated whether ultra-low dose cannabinoid antagonist AM251 influences cannabinoid anticonvulsant effects. The clonic seizure threshold (CST) was tested in separate groups of male NMRI mice following injection of vehicle, the cannabinoid selective agonist arachidonyl-2-chloroethylamide (ACEA) and ultra-low doses of the cannabinoid CB1 antagonist AM251 and a combination of ACEA and AM251 doses in a model of clonic seizure induced by pentylenetetrazole (PTZ). Systemic administration of ultra-low doses of AM251 (10 fg/kg-100 ng/kg) significantly potentiated the anticonvulsant effect of ACEA at 0.5 and 1 mg/kg. Moreover, inhibition of cannabinoid induced excitatory signaling by AM251 (100 pg/kg) unmasked a strong anticonvulsant effect for very low doses of ACEA (100 ng/kg-100 microg/kg), suggesting that a presumed inhibitory component of cannabinoid receptor signaling can exert strong seizure-protective effects even at very low levels of cannabinoid receptor activation. A similar potentiation by AM251 (100 pg/kg and 1 ng/kg) of anticonvulsant effects of non-effective dose of ACEA (0.5 and 1 mg/kg) was also observed in the generalized tonic-clonic model of seizure. The present data suggest that ultra-low doses of cannabinoid receptor antagonists may provide a potent strategy to modulate seizure susceptibility, especially in conjunction with very low doses of cannabinoids.


Assuntos
Anticonvulsivantes/farmacologia , Antagonistas de Receptores de Canabinoides , Pentilenotetrazol , Piperidinas/farmacologia , Pirazóis/farmacologia , Convulsões/prevenção & controle , Animais , Ácidos Araquidônicos/farmacologia , Agonistas de Receptores de Canabinoides , Convulsivantes , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Masculino , Camundongos , Convulsões/induzido quimicamente
9.
Eur J Pharmacol ; 562(3): 212-20, 2007 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-17379207

RESUMO

We previously had shown that bilateral intrahippocampal infusion of 1 microg nicotine (but not 0.5 microg dose) led to an improvement in spatial memory retention in the Morris water maze task in male rats. We also reported that a similar type of bilateral infusion of H89, a protein kinase AII (PKA II) inhibitor, caused a deficit in spatial memory retention. In the present study, we wished to test the hypothesis that intrahippocampal infusion of dibutyryl cyclic AMP (DB-cAMP also called bucladesine), a membrane permeable selective activator of PKA, into the CA1 region can cause an improvement in spatial memory in this maze task. Indeed, bilateral infusion of 10 and 100 microM bucladesine (but not 1 and 5 microM doses) led to a significant reduction in escape latency and travel distance (showing an improvement in spatial memory) compared to the control. Also, bilateral infusion of 0.5 microg nicotine or 1 microM bucladesine alone did not lead to an improvement in spatial memory. However, such bilateral infusion of bucladesine at 1 and 5 microM concentrations infused within minutes after 0.5 microg nicotine infusion improved spatial memory retention. Taken together, our data suggest that intrahippocampal bucladesine infusions improve spatial memory retention in male rats and that bucladesine can interact synergistically with nicotine to improve spatial memory.


Assuntos
Bucladesina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Retenção Psicológica/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Bucladesina/administração & dosagem , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Hipocampo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Percepção Espacial/efeitos dos fármacos
10.
Behav Brain Res ; 316: 115-124, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27555536

RESUMO

Oxidative stress and mitochondrial dysfunction play indispensable role in memory and learning impairment. Growing evidences have shed light on anti-oxidative role for melatonin in memory deficit. We have previously reported that inhibition of protein kinase A by H-89 can induce memory impairment. Here, we investigated the effect of melatonin on H-89 induced spatial memory deficit and pursued their interactive consequences on oxidative stress and mitochondrial function in Morris Water Maze model. Rats received melatonin (50 and 100µg/kg/side) and H-89(10µM) intra-hippocampally 30min before each day of training. Animals were trained for 4 consecutive days, each containing one block from four trials. Oxidative stress indices, including thiobarbituric acid (TBARS), reactive oxygen species (ROS), thiol groups, and ferric reducing antioxidant power (FRAP) were assessed using spectrophotometer. Mitochondrial function was evaluated through measuring ROS production, mitochondrial membrane potential (MMP), swelling, outer membrane damage, and cytochrome c release. As expected from our previous report, H-89 remarkably impaired memory by increasing the escape latency and traveled distance. Intriguingly, H-89 significantly augmented TBARS and ROS levels, caused mitochondrial ROS production, swelling, outer membrane damage, and cytochrome c release. Moreover, H-89 lowered thiol, FRAP, and MMP values. Intriguingly, melatonin pre-treatment not only effectively hampered H-89-mediated spatial memory deficit at both doses, but also reversed the H-89 effects on mitochondrial and biochemical indices upon higher dose. Collectively, these findings highlight a protective role for melatonin against H-89-induced memory impairment and indicate that melatonin may play a therapeutic role in the treatment of oxidative- related neurodegenerative disorders.


Assuntos
Antioxidantes/uso terapêutico , Isoquinolinas/toxicidade , Melatonina/uso terapêutico , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Inibidores de Proteínas Quinases/toxicidade , Sulfonamidas/toxicidade , Animais , Citocromos c/metabolismo , Modelos Animais de Doenças , Reação de Fuga/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/ultraestrutura , Hipnóticos e Sedativos/uso terapêutico , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Xilazina/uso terapêutico
11.
Hum Gene Ther ; 27(12): 982-996, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27604541

RESUMO

Fragile X mental retardation protein (FMRP) is absent or highly reduced in Fragile X Syndrome, a genetic disorder causing cognitive impairment and autistic behaviors. Previous proof-of-principle studies have demonstrated that restoring FMRP in the brain using viral vectors can improve pathological abnormalities in mouse models of fragile X. However, unlike small molecule drugs where the dose can readily be adjusted during treatment, viral vector-based biological therapeutic drugs present challenges in terms of achieving optimal dosing and expression levels. The objective of this study was to investigate the consequences of expressing varying levels of FMRP selectively in neurons of Fmr1 knockout and wild-type (WT) mice. A wide range of neuronal FMRP transgene levels was achieved in individual mice after intra-cerebroventricular administration of adeno-associated viral vectors coding for FMRP. In all treated knockout mice, prominent FMRP transgene expression was observed in forebrain structures, whereas lower levels were present in more caudal regions of the brain. Reduced levels of the synaptic protein PSD-95, elevated levels of the transcriptional modulator MeCP2, and abnormal motor activity, anxiety, and acoustic startle responses in Fmr1 knockout mice were fully or partially rescued after expression of FMRP at about 35-115% of WT expression, depending on the brain region examined. In the WT mouse, moderate FMRP over-expression of up to about twofold had little or no effect on PSD-95 and MeCP2 levels or on behavioral endophenotypes. In contrast, excessive over-expression in the Fmr1 knockout mouse forebrain (approximately 2.5-6-fold over WT) induced pathological motor hyperactivity and suppressed the startle response relative to WT mice. These results delineate a range of FMRP expression levels in the central nervous system that confer phenotypic improvement in fragile X mice. Collectively, these findings are pertinent to the development of long-term curative gene therapy strategies for treating Fragile X Syndrome and other neurodevelopmental disorders.


Assuntos
Comportamento Animal , Encéfalo/metabolismo , Dependovirus/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/terapia , Terapia Genética , Vetores Genéticos/administração & dosagem , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Brain Res ; 1596: 22-30, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25446451

RESUMO

Fragile X syndrome is the most common inherited form of mental retardation and autism. It is caused by a reduction or elimination of the expression of fragile X mental retardation protein (FMRP). Because fragile X syndrome is a neurodevelopmental disorder, it is important to fully document the cell type expression in the developing CNS to provide a better understanding of the molecular function of FMRP, and the pathogenesis of the syndrome. We investigated FMRP expression in the brain using double-labeling immunocytochemistry and cell type markers for neurons (NeuN), astrocytes (S100ß), microglia (Iba-1), and oligodendrocyte precursor cells (NG2). The hippocampus, striatum, cingulate cortex, retrosplenial cortex, corpus callosum and cerebellum were assessed in wild-type C57/BL6 mice at postnatal days 0, 10, 20, and adult. Our results demonstrate that FMRP is ubiquitously expressed in neurons at all times and brain regions studied, except for corpus callosum where FMRP was predominantly present in astrocytes at all ages. FMRP expression in Iba-1 and NG2-positive cells was detected at postnatal day 0 and 10 and gradually decreased to very low or undetectable levels in postnatal day 20 and adult mice. Our results reveal that in addition to continuous and extensive expression in neurons in the immature and mature brain, FMRP is also present in astrocytes, oligodendrocyte precursor cells, and microglia during the early and mid-postnatal developmental stages of brain maturation. Prominent expression of FMRP in glia during these crucial stages of brain development suggests an important contribution to normal brain function, and in its absence, to the fragile X phenotype.


Assuntos
Encéfalo/citologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Proteínas de Ligação ao Cálcio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Proteoglicanas/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo
13.
Neuropsychopharmacology ; 39(13): 3100-11, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24998620

RESUMO

Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a trinucleotide repeat expansion in the FMR1 gene that codes for fragile X mental retardation protein (FMRP). To determine if FMRP expression in the central nervous system could reverse phenotypic deficits in the Fmr1 knockout (KO) mouse model of FXS, we used a single-stranded adeno-associated viral (AAV) vector with viral capsids from serotype 9 that contained a major isoform of FMRP. FMRP transgene expression was driven by the neuron-selective synapsin-1 promoter. The vector was delivered to the brain via a single bilateral intracerebroventricular injection into neonatal Fmr1 KO mice and transgene expression and behavioral assessments were conducted 22-26 or 50-56 days post injection. Western blotting and immunocytochemical analyses of AAV-FMRP-injected mice revealed FMRP expression in the striatum, hippocampus, retrosplenial cortex, and cingulate cortex. Cellular expression was selective for neurons and reached ∼ 50% of wild-type levels in the hippocampus and cortex at 56 days post injection. The pathologically elevated repetitive behavior and the deficit in social dominance behavior seen in phosphate-buffered saline-injected Fmr1 KO mice were reversed in AAV-FMRP-injected mice. These results provide the first proof of principle that gene therapy can correct specific behavioral abnormalities in the mouse model of FXS.


Assuntos
Encéfalo/metabolismo , Dependovirus/fisiologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/terapia , Animais , Encéfalo/patologia , Dependovirus/genética , Modelos Animais de Doenças , Epilepsia Reflexa/etiologia , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/complicações , Síndrome do Cromossomo X Frágil/patologia , Regulação da Expressão Gênica/genética , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/fisiologia , Fenótipo , Predomínio Social , Estatísticas não Paramétricas , Comportamento Estereotipado/fisiologia , Vocalização Animal
14.
Hum Gene Ther Methods ; 24(4): 205-13, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23808551

RESUMO

Several neurodevelopmental and neurodegenerative disorders affecting the central nervous system are potentially treatable via viral vector-mediated gene transfer. Adeno-associated viral (AAV) vectors have been used in clinical trials because of their desirable properties including a high degree of safety, efficacy, and stability. Major factors affecting tropism, expression level, and cell type specificity of AAV-mediated transgenes include encapsidation of different AAV serotypes, promoter selection, and the timing of vector administration. In this study, we evaluated the ability of single-stranded AAV2 vectors pseudotyped with viral capsids from serotype 9 (AAV2/9) to transduce the brain and target gene expression to specific cell types after intracerebroventricular injection into mice. Titer-matched AAV2/9 vectors encoding the enhanced green fluorescent protein (eGFP) reporter, driven by the cytomegalovirus (CMV) promoter, or the neuron-specific synapsin-1 promoter, were injected bilaterally into the lateral ventricles of C57/BL6 mice on postnatal day 5 (neonatal) or 21 (juvenile). Brain sections were analyzed 25 days after injection, using immunocytochemistry and confocal microscopy. eGFP immunohistochemistry after neonatal and juvenile administration of viral vectors revealed transduction throughout the brain including the striatum, hippocampus, cerebral cortex, and cerebellum, but with different patterns of cell-specific gene expression. eGFP expression was seen in astrocytes after treatment on postnatal day 5 with vectors carrying the CMV promoter, expanding the usefulness of AAVs for modeling and treating diseases involving glial cell pathology. In contrast, injection of AAV2/9-CMV-eGFP on postnatal day 21 resulted in preferential transduction of neurons. Administration of AAV2/9-eGFP with the synapsin-1 promoter on either postnatal day 5 or 21 resulted in widespread neuronal transduction. These results outline efficient methods and tools for gene delivery to the nervous system by direct, early postnatal administration of AAV vectors. Our findings highlight the importance of promoter selection and age of administration on the intensity, distribution, and cell type specificity of AAV transduction in the brain.


Assuntos
Encéfalo/metabolismo , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Transdução Genética/métodos , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/virologia , Dependovirus/metabolismo , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos
15.
PLoS One ; 8(5): e63612, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696837

RESUMO

The consolidation of newly acquired memories involves the temporal transition from a recent, less stable trace to a more permanent consolidated form. Opiates possess potent rewarding effects and produce powerful associative memories. The activation of these memories is associated with opiate abuse relapse phenomena and the persistence of compulsive opiate dependence. However, the neuronal, molecular and temporal mechanisms by which associative opiate reward memories are consolidated are not currently understood. We report that the consolidation of associative opiate reward memories involves a temporal and molecular switch between the basolateral nucleus of the amygdala (BLA) (early consolidation phase) to the medial prefrontal cortex (mPFC) (late consolidation phase). We demonstrate at the molecular, behavioral and neuronal levels that the consolidation of a recently acquired opiate reward memory involves an extracellular signal-related kinase (ERK)-dependent phosphorylation process within the BLA. In contrast, later-stage consolidation of a newly acquired memory is dependent upon a calcium-calmodulin-dependent (CaMKII), ERK-independent, mechanism in the mPFC, over a 12 hr temporal gradient. In addition, using in vivo multi-unit neuronal recordings in the mPFC, we report that protein synthesis within the BLA modulates the consolidation of opiate-reward memory in neuronal mPFC sub-populations, via the same temporal dynamic.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Analgésicos Opioides/farmacologia , Memória/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Masculino , Memória/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Ratos
16.
Eur J Pharmacol ; 670(2-3): 464-70, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21946102

RESUMO

The present study shows interactive effects of pentoxifylline (PTX) as a phosphodiesterase (PDE) inhibitor, H-89 as a protein kinase A (PKA) inhibitor and bucladesine (db-cAMP) as a cAMP agonist on pentylenetetrazol (PTZ)-induced seizure in mice. Different doses of pentoxifylline (25, 50, 100 mg/kg), bucladesine (50, 100, 300 nM/mouse), and H-89 (0.05, 0.1, 0.2 mg/100g) were administered intraperitoneally (i.p.), 30 min before intravenous (i.v.) infusion of PTZ (0.5% w/v). In combination groups, the first and second components were injected 45 and 30 min before PTZ infusion. In all groups, the control animals received an appropriate volume of vehicle. Single administration of PTX had no significant effect on both seizure latency and threshold. Bucladesine significantly decreased seizure latency and threshold only at a high concentration (300 nM/mouse). Intraperitoneal administration of H-89 (0.2 mg/100g) significantly increased seizure latency and threshold in PTZ-treated animals. All applied doses of bucladesine in combination with PTX (50 mg/kg) caused a significant reduction in seizure latency. Pretreatment of animals with PTX (50 and 100 mg/kg) attenuated the anticonvulsant effect of H-89 (0.2 mg/100g) in PTZ-exposed animals. H-89 (0.05, 0.2 mg/100g) prevented the epileptogenic activity of bucladesine (300 nM) with significant increase of seizure latency and seizure threshold. In conclusion, we showed that seizure activities were affected by pentoxifylline, H-89 and bucladesine via interactions with intracellular cAMP and cGMP signaling pathways, cyclic nucleotide-dependent protein kinases, and related neurotransmitters.


Assuntos
Bucladesina/farmacologia , Isoquinolinas/farmacologia , Pentoxifilina/farmacologia , Pentilenotetrazol/efeitos adversos , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Bucladesina/uso terapêutico , Relação Dose-Resposta a Droga , Interações Medicamentosas , Isoquinolinas/uso terapêutico , Masculino , Camundongos , Pentoxifilina/uso terapêutico , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfodiesterase/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Sulfonamidas/uso terapêutico
17.
Psychopharmacology (Berl) ; 204(2): 191-201, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19183965

RESUMO

RATIONALE: Tamoxifen (TMX), a selective estrogen receptor modulator, can affect cognitive functions of the brain. The conditioned place preference (CPP) paradigm involves memory for the association between contextual cues and the rewarding properties produced by a drug. OBJECTIVES: The effects of TMX alone and in combination with estradiol (E2) on reward-related memory of morphine were investigated in adult male mice. MATERIALS AND METHODS: Using an unbiased CPP paradigm, the ability of morphine sulfate (0.5-10 mg/kg, s.c.) to produce CPP was studied. Afterwards, the effects of TMX (1-10 mg/kg, s.c.) on the acquisition, consolidation, and expression of morphine-induced CPP were assessed. We have also evaluated the possible effects of s.c. E2 (10-200 mug/kg) and its co-administration with TMX (10 mg/kg, s.c.) on the consolidation and retrieval of morphine-associated contextual memory. RESULTS: (1) Morphine (0.5-10 mg/kg) significantly induced CPP in a dose-dependent manner. (2) TMX (10 mg/kg) significantly reduced the time spent by mice in the morphine compartment when given immediately after each conditioning session (consolidation) or 30 min before testing for place preference in the absence of morphine (expression), whereas it had no effect when administered 30 min before each training session (acquisition). (3) Post-training or pre-testing administration of E2 increased morphine-induced CPP in a dose-dependent manner. (4) In addition, concomitant administration of E2 with TMX appears to prevent the impairing effect produced by TMX. CONCLUSIONS: TMX appears to disrupt consolidation and retrieval of morphine-associated contextual memory and this impairing effect might be prevented by E2 treatment.


Assuntos
Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Memória/efeitos dos fármacos , Morfina/farmacologia , Entorpecentes/farmacologia , Tamoxifeno/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Interpretação Estatística de Dados , Relação Dose-Resposta a Droga , Estradiol/administração & dosagem , Injeções Subcutâneas , Masculino , Camundongos , Morfina/administração & dosagem , Entorpecentes/administração & dosagem
18.
Planta Med ; 74(5): 485-90, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18404595

RESUMO

In the present study, the time course of the effects of Glycyrrhiza glabra L. (Leguminosae) aqueous extract (GE), administered systemically to rats, on the spatial memory retention in the Morris water maze was investigated. The dose of glycyrrhizin (GL), i. e., 0.5, 2.5 and 5 mg/mL in daily water intake of GE was administered to three groups of rats. The first, second and third groups received GE for 1, 2 and 4 weeks, respectively (each group included 3 subgroups). Three additional control groups of animals received only tap water during the same periods of time. After terminating the treatments, all animals were trained for four days; each day included one block and each block contained four trials. Test trials were conducted 48 h after the completion of the training period. Nicotine (1 microg/side) was infused into the CA1 region of the hippocampus as a positive drug control. GE treatment decreased both escape latency and traveled distance, but not swimming speed, compared with control, suggesting significant spatial memory retention enhancement by GE. Statistical analysis did not show any significant difference between GE-treated animals and the nicotine group in escape latency and traveled distance. At the end of the testing trials plasma samples were collected and the concentrations of glycyrrhetinic acid (GA) as a major metabolite of GL were measured in the different groups of treated rats. The maximum concentration was observed after four weeks of GE administration at 5 mg/mL of GL. These results showed that the enhancement effect of GE on spatial memory retention does not correlate with GA blood levels.


Assuntos
Ácido Glicirretínico/administração & dosagem , Glycyrrhiza , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Comportamento Espacial/efeitos dos fármacos , Animais , Ácido Glicirretínico/sangue , Masculino , Extratos Vegetais/administração & dosagem , Extratos Vegetais/sangue , Ratos , Ratos Wistar , Fatores de Tempo
19.
Pharmacology ; 80(2-3): 158-65, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17534126

RESUMO

We have previously shown that infusion of the PKAII inhibitor H-89 in the CA1 area of the hippocampus impaired spatial memory retention. There is some evidence suggesting the neuroprotective effects of chronic lithium administration including its ability to attenuate a deleterious effect of chronic stress on spatial memory in rats. In the present study, we investigated whether chronic administration of lithium can improve memory as well as influence the inhibitory effect of H-89 on spatial memory retention. Male albino rats were treated systemically with lithium (600 mg/l) for 4 weeks and then trained for 4 days in the Morris water maze. Testing the animals 48 h later showed a significant reduction in escape latency (p < 0.05) and travel distance (p < 0.05) compared to the controls. In separate experiments, the rats were similarly treated with lithium for 4 weeks, followed by similar training for 4 days and then immediately infused bilaterally with vehicle or 5 micromol/l H-89 into the CA1 region of the hippocampus. Animals were then tested 48 h after H-89 infusion in order to assess their spatial memory retention. The lithium treatment caused a significant reduction in escape latency (p < 0.001) and travel distance (p < 0.001) compared to H-89-treated animals. The data suggest that lithium treatment for 4 weeks improved spatial memory retention and that lithium pretreatment prevented or reversed the H-89-induced spatial memory deficits.


Assuntos
Antimaníacos/administração & dosagem , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Isoquinolinas/farmacologia , Cloreto de Lítio/administração & dosagem , Memória/efeitos dos fármacos , Percepção Espacial/efeitos dos fármacos , Sulfonamidas/farmacologia , Análise de Variância , Animais , Antimaníacos/sangue , Comportamento Animal/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Cloreto de Lítio/sangue , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Ratos , Ratos Wistar , Percepção Espacial/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa