Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1270843, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37795087

RESUMO

Despite the potential of CAR-T therapies for hematological malignancies, their efficacy in patients with relapse and refractory Acute Myeloid Leukemia has been limited. The aim of our study has been to develop and manufacture a CAR-T cell product that addresses some of the current limitations. We initially compared the phenotype of T cells from AML patients and healthy young and elderly controls. This analysis showed that T cells from AML patients displayed a predominantly effector phenotype, with increased expression of activation (CD69 and HLA-DR) and exhaustion markers (PD1 and LAG3), in contrast to the enriched memory phenotype observed in healthy donors. This differentiated and more exhausted phenotype was also observed, and corroborated by transcriptomic analyses, in CAR-T cells from AML patients engineered with an optimized CAR construct targeting CD33, resulting in a decreased in vivo antitumoral efficacy evaluated in xenograft AML models. To overcome some of these limitations we have combined CRISPR-based genome editing technologies with virus-free gene-transfer strategies using Sleeping Beauty transposons, to generate CAR-T cells depleted of HLA-I and TCR complexes (HLA-IKO/TCRKO CAR-T cells) for allogeneic approaches. Our optimized protocol allows one-step generation of edited CAR-T cells that show a similar phenotypic profile to non-edited CAR-T cells, with equivalent in vitro and in vivo antitumoral efficacy. Moreover, genomic analysis of edited CAR-T cells revealed a safe integration profile of the vector, with no preferences for specific genomic regions, with highly specific editing of the HLA-I and TCR, without significant off-target sites. Finally, the production of edited CAR-T cells at a larger scale allowed the generation and selection of enough HLA-IKO/TCRKO CAR-T cells that would be compatible with clinical applications. In summary, our results demonstrate that CAR-T cells from AML patients, although functional, present phenotypic and functional features that could compromise their antitumoral efficacy, compared to CAR-T cells from healthy donors. The combination of CRISPR technologies with transposon-based delivery strategies allows the generation of HLA-IKO/TCRKO CAR-T cells, compatible with allogeneic approaches, that would represent a promising option for AML treatment.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda , Animais , Humanos , Idoso , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/metabolismo , Imunoterapia Adotiva/métodos , Modelos Animais de Doenças
2.
Sci Adv ; 8(39): eabo0514, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36179026

RESUMO

Identification of new markers associated with long-term efficacy in patients treated with CAR T cells is a current medical need, particularly in diseases such as multiple myeloma. In this study, we address the impact of CAR density on the functionality of BCMA CAR T cells. Functional and transcriptional studies demonstrate that CAR T cells with high expression of the CAR construct show an increased tonic signaling with up-regulation of exhaustion markers and increased in vitro cytotoxicity but a decrease in in vivo BM infiltration. Characterization of gene regulatory networks using scRNA-seq identified regulons associated to activation and exhaustion up-regulated in CARHigh T cells, providing mechanistic insights behind differential functionality of these cells. Last, we demonstrate that patients treated with CAR T cell products enriched in CARHigh T cells show a significantly worse clinical response in several hematological malignancies. In summary, our work demonstrates that CAR density plays an important role in CAR T activity with notable impact on clinical response.

3.
Mol Ther Methods Clin Dev ; 25: 137-146, 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35402636

RESUMO

Genome-editing strategies, especially CRISPR-Cas9 systems, have substantially increased the efficiency of innovative therapeutic approaches for monogenic diseases such as primary hyperoxalurias (PHs). We have previously demonstrated that inhibition of glycolate oxidase using CRISPR-Cas9 systems represents a promising therapeutic option for PH type I (PH1). Here, we extended our work evaluating the efficacy of liver-specific inhibition of lactate dehydrogenase (LDH), a key enzyme responsible for converting glyoxylate to oxalate; this strategy would not be limited to PH1, being applicable to other PH subtypes. In this work, we demonstrate a liver-specific inhibition of LDH that resulted in a drastic reduction of LDH levels in the liver of PH1 and PH3 mice after a single-dose delivery of AAV8 vectors expressing the CRISPR-Cas9 system, resulting in reduced urine oxalate levels and kidney damage without signs of toxicity. Deep sequencing analysis revealed that this approach was safe and specific, with no off-targets detected in the liver of treated animals and no on-target/off-tissue events. Altogether, our data provide evidence that in vivo genome editing using CRISPR-Cas9 systems would represent a valuable tool for improved therapeutic approaches for PH.

4.
Stem Cell Res ; 41: 101626, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31715429

RESUMO

Primary Hyperoxaluria Type I (PH1) is a rare autosomal recessive metabolic disorder characterized by defects in enzymes involved in glyoxylate metabolism. PH1 is a life-threatening disease caused by the absence, deficiency or mistargeting of the hepatic alanine-glyoxylate aminotransferase (AGT) enzyme. A human induced pluripotent stem cell (iPSC) line was generated from dermal fibroblasts of a PH1 patient being compound heterozygous for the most common mutation c.508G>A (G170R), a mistargeting mutation, and c.364C>T (R122*), a previously reported nonsense mutation in AGTX. This iPSC line offers a useful resource to study the disease pathophysiology and a cell-based model for drug development.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem Celular/patologia , Hiperoxalúria Primária/genética , Hiperoxalúria Primária/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Mutação/genética , Transaminases/genética , Adulto , Sequência de Bases , Humanos , Masculino , Reprodutibilidade dos Testes
5.
Elife ; 82019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31090538

RESUMO

Synaptophysins 1 and 2 and synaptogyrins 1 and 3 constitute a major family of synaptic vesicle membrane proteins. Unlike other widely expressed synaptic vesicle proteins such as vSNAREs and synaptotagmins, the primary function has not been resolved. Here, we report robust elevation in the probability of release of readily releasable vesicles with both high and low release probabilities at a variety of synapse types from knockout mice missing all four family members. Neither the number of readily releasable vesicles, nor the timing of recruitment to the readily releasable pool was affected. The results suggest that family members serve as negative regulators of neurotransmission, acting directly at the level of exocytosis to dampen connection strength selectively when presynaptic action potentials fire at low frequency. The widespread expression suggests that chemical synapses may play a frequency filtering role in biological computation that is more elemental than presently envisioned. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Assuntos
Neurônios/metabolismo , Vesículas Sinápticas/metabolismo , Sinaptogirinas/deficiência , Sinaptofisina/deficiência , Animais , Camundongos Knockout , Transmissão Sináptica
6.
Nat Commun ; 9(1): 5454, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575740

RESUMO

CRISPR/Cas9 technology offers novel approaches for the development of new therapies for many unmet clinical needs, including a significant number of inherited monogenic diseases. However, in vivo correction of disease-causing genes is still inefficient, especially for those diseases without selective advantage for corrected cells. We reasoned that substrate reduction therapies (SRT) targeting non-essential enzymes could provide an attractive alternative. Here we evaluate the therapeutic efficacy of an in vivo CRISPR/Cas9-mediated SRT to treat primary hyperoxaluria type I (PH1), a rare inborn dysfunction in glyoxylate metabolism that results in excessive hepatic oxalate production causing end-stage renal disease. A single systemic administration of an AAV8-CRISPR/Cas9 vector targeting glycolate oxidase, prevents oxalate overproduction and kidney damage, with no signs of toxicity in Agxt1-/- mice. Our results reveal that CRISPR/Cas9-mediated SRT represents a promising therapeutic option for PH1 that can be potentially applied to other metabolic diseases caused by the accumulation of toxic metabolites.


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Sistemas CRISPR-Cas , Terapia Genética/métodos , Hiperoxalúria Primária/terapia , Oxalatos/urina , Oxirredutases do Álcool/genética , Animais , Modelos Animais de Doenças , Edição de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Nefrocalcinose/prevenção & controle
7.
Neurobiol Dis ; 93: 47-56, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27072890

RESUMO

Age-inappropriate expression of juvenile NMDA receptors (NMDARs) containing GluN3A subunits has been linked to synapse loss and death of spiny projection neurons of the striatum (SPNs) in Huntington's disease (HD). Here we show that suppressing GluN3A expression prevents a multivariate synaptic transmission phenotype that precedes morphological signs at early prodromal stages. We start by confirming that afferent fiber stimulation elicits larger synaptic responses mediated by both AMPA receptors and NMDARs in SPNs in the YAC128 mouse model of HD. We then show that the enhancement mediated by both is fully prevented by suppressing GluN3A expression. Strong fiber-stimulation unexpectedly elicited robust NMDAR-mediated electrogenic events (termed "upstates" or "NMDA spikes"), and the effective threshold for induction was more than 2-fold lower in YAC128 SPNs because of the enhanced synaptic transmission. The threshold could be restored to control levels by suppressing GluN3A expression or by applying the weak NMDAR blocker memantine. However, the threshold was not affected by preventing glutamate spillover from synaptic clefts. Instead, long-lasting NMDAR responses interpreted previously as activation of extrasynaptic receptors by spilled-over glutamate were caused by NMDA spikes occurring in voltage clamp mode as escape potentials. Together, the results implicate GluN3A reactivation in a broad spectrum of early-stage synaptic transmission deficits in YAC128 mice; question the current concept that NMDAR mislocalization is the pathological trigger in HD; and introduce NMDA spikes as a new candidate mechanism for coupling NMDARs to neurodegeneration.


Assuntos
Doença de Huntington/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica , Animais , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Doença de Huntington/genética , Memantina/farmacologia , Camundongos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos
8.
Nat Med ; 19(8): 1030-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23852340

RESUMO

Huntington's disease is caused by an expanded polyglutamine repeat in the huntingtin protein (HTT), but the pathophysiological sequence of events that trigger synaptic failure and neuronal loss are not fully understood. Alterations in N-methyl-D-aspartate (NMDA)-type glutamate receptors (NMDARs) have been implicated. Yet, it remains unclear how the HTT mutation affects NMDAR function, and direct evidence for a causative role is missing. Here we show that mutant HTT redirects an intracellular store of juvenile NMDARs containing GluN3A subunits to the surface of striatal neurons by sequestering and disrupting the subcellular localization of the endocytic adaptor PACSIN1, which is specific for GluN3A. Overexpressing GluN3A in wild-type mouse striatum mimicked the synapse loss observed in Huntington's disease mouse models, whereas genetic deletion of GluN3A prevented synapse degeneration, ameliorated motor and cognitive decline and reduced striatal atrophy and neuronal loss in the YAC128 Huntington's disease mouse model. Furthermore, GluN3A deletion corrected the abnormally enhanced NMDAR currents, which have been linked to cell death in Huntington's disease and other neurodegenerative conditions. Our findings reveal an early pathogenic role of GluN3A dysregulation in Huntington's disease and suggest that therapies targeting GluN3A or pathogenic HTT-PACSIN1 interactions might prevent or delay disease progression.


Assuntos
Comportamento Animal , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Morte Celular/efeitos dos fármacos , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Deleção de Genes , Células HEK293 , Humanos , Doença de Huntington/fisiopatologia , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Atividade Motora/efeitos dos fármacos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/toxicidade , Neostriado/metabolismo , Neostriado/patologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeos/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Teste de Desempenho do Rota-Rod , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura
9.
Neurobiol Dis ; 48(3): 290-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22801082

RESUMO

Excitotoxicity due to excessive activation of glutamate receptors is a primary mediator of cell death in acute and chronic neurological disorders, and NMDA-type glutamate receptors (NMDARs) are thought to be involved. NMDARs assemble from heteromeric combinations of GluN1, GluN2 and GluN3 subunits, yielding a variety of receptor subtypes that differ in biophysical properties, signaling, and synaptic targeting. Inclusion of inhibitory GluN3 subunits reduces Ca2+ influx via NMDAR channels and alters their synaptic targeting, thus modifying the two hallmarks of NMDARs that are critical for their roles on neuronal death and survival. Here we evaluated the neuroprotective potential of GluN3A subunits by analyzing the susceptibility to striatal excitotoxic damage of transgenic mice overexpressing GluN3A. We found that mild GluN3A overexpression protected susceptible striatal neurons from lesions induced by the neurotoxin 3-nitropropionic acid (3-NP), an inhibitor of mitochondrial complex II/succinate dehydrogenase. GluN3A-mediated neuroprotection was dose-dependent, and correlated with the levels of transgenic GluN3A expressed by two different mice strains. Neuroprotection was associated with a potent reduction of the activation of calpain, a Ca2+-dependent protease, which was measured as a decrease in 3-NP-induced fodrin and STEP cleavage in GluN3A transgenic mice relative to controls. We further show that transgenic GluN3A subunits incorporate into extrasynaptic compartments in mouse striatum, suggesting that reductions of toxic calpain activation might be linked to inhibition by GluN3A of pathological extrasynaptic NMDAR activity.


Assuntos
Calpaína/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Western Blotting , Convulsivantes/toxicidade , Corpo Estriado/patologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Nitrocompostos/toxicidade , Propionatos/toxicidade , Subunidades Proteicas/metabolismo
10.
J Neurosci ; 31(32): 11563-77, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21832187

RESUMO

At least two rate-limiting mechanisms in vesicle trafficking operate at mouse Schaffer collateral synapses, but their molecular/physical identities are unknown. The first mechanism determines the baseline rate at which reserve vesicles are supplied to a readily releasable pool. The second causes the supply rate to depress threefold when synaptic transmission is driven hard for extended periods. Previous models invoked depletion of a reserve vesicle pool to explain the reductions in the supply rate, but the mass-action assumption at their core is not compatible with kinetic measurements of neurotransmission under maximal-use conditions. Here we develop a new theoretical model of rate-limiting steps in vesicle trafficking that is compatible with previous and new measurements. A physical interpretation is proposed where local reserve pools consisting of four vesicles are tethered to individual release sites and are replenished stochastically in an all-or-none fashion. We then show that the supply rate depresses more rapidly in synapsin knock-outs and that the phenotype can be fully explained by changing the value of the single parameter in the model that would specify the size of the local reserve pools. Vesicle-trafficking rates between pools were not affected. Finally, optical imaging experiments argue against alternative interpretations of the theoretical model where vesicle trafficking is inhibited without reserve pool depletion. This new conceptual framework will be useful for distinguishing which of the multiple molecular and cell biological mechanisms involved in vesicle trafficking are rate limiting at different levels of synaptic throughput and are thus candidates for physiological and pharmacological modulation.


Assuntos
Modelos Neurológicos , Sinapsinas/deficiência , Sinapsinas/metabolismo , Vesículas Sinápticas/fisiologia , Potenciais de Ação/genética , Animais , Células Cultivadas , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Transporte Proteico/genética , Vesículas Sinápticas/genética
11.
Neuron ; 63(3): 342-56, 2009 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-19679074

RESUMO

NR3A is the only NMDA receptor (NMDAR) subunit that downregulates sharply prior to the onset of sensitive periods for plasticity, yet the functional importance of this transient expression remains unknown. To investigate whether removal/replacement of juvenile NR3A-containing NMDARs is involved in experience-driven synapse maturation, we used a reversible transgenic system that prolonged NR3A expression in the forebrain. We found that removal of NR3A is required to develop strong NMDAR currents, full expression of long-term synaptic plasticity, a mature synaptic organization characterized by more synapses and larger postsynaptic densities, and the ability to form long-term memories. Deficits associated with prolonged NR3A were reversible, as late-onset suppression of transgene expression rescued both synaptic and memory impairments. Our results suggest that NR3A behaves as a molecular brake to prevent the premature strengthening and stabilization of excitatory synapses and that NR3A removal might thereby initiate critical stages of synapse maturation during early postnatal neural development.


Assuntos
Regulação para Baixo/fisiologia , Memória/fisiologia , Neurônios/citologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/fisiologia , Animais , Animais Recém-Nascidos , Biofísica , Proteína 4 Homóloga a Disks-Large , Estimulação Elétrica/métodos , Preferências Alimentares/fisiologia , Proteínas de Fluorescência Verde/genética , Guanilato Quinases , Hipocampo/citologia , Imunoprecipitação/métodos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão/métodos , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Neurônios/ultraestrutura , Técnicas de Patch-Clamp/métodos , Receptores de N-Metil-D-Aspartato/genética , Reconhecimento Psicológico/fisiologia , Coloração pela Prata/métodos , Comportamento Social , Potenciais Sinápticos/genética , Potenciais Sinápticos/fisiologia
12.
Biochem Biophys Res Commun ; 353(3): 750-5, 2007 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-17194442

RESUMO

Recent data suggest that the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor subtype plays a pivotal role in the pathogenesis of effective disorders and in the action of antidepressant drugs. After chronic treatment with the antidepressants desipramine or paroxetine, we measured by immunoprecipitation and Western blotting, the changes in the interaction of AMPA receptor subunits with proteins involved in trafficking and/or stabilization of the subunits into synaptic membranes of the hippocampus. Both antidepressants increased the interaction of GluR1 subunit with stargazin and of GluR2/3 with NSF. Paroxetine increased the interaction of GluR1 with Rab4A, and desipramine markedly increased the interaction of GluR1 with SAP97. Paroxetine, but not desipramine, also increased membrane levels of CaMKII, autophosphorylated CaMKII and GluR1 phosphorylated at the CaMKII site. Interactions of GluR1 and GluR2/3 with proteins implicated in AMPA receptor trafficking and with scaffolding proteins appear to account for the enhanced membrane expression of AMPA receptors in the hippocampus after antidepressant treatment.


Assuntos
Antidepressivos/farmacologia , Hipocampo/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de AMPA/efeitos dos fármacos , Sinapses/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Desipramina/farmacologia , Masculino , Paroxetina/farmacologia , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Transporte Proteico , Ratos , Ratos Wistar , Receptores de AMPA/fisiologia
13.
Neurosci Lett ; 402(1-2): 126-30, 2006 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-16644117

RESUMO

The serotonergic neurotoxin 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy") produces rapid serotonin (5-HT) depletion in different areas of the forebrain after acute administration to rats and other animal species. We previously found that 5-HT depletion induced by acute MDMA treatment was transient in the frontal cortex, but not in the hippocampus, and recovery of cortical 5-HT levels correlated with an induction of CRE-binding activity and increased expression of tryptophan-hydroxylase (TPH), the rate-limiting enzyme in 5-HT biosynthesis. As the brain-derived neurotrophic factor (BDNF) stimulates the growth and sprouting of serotonergic neurons, we sought the possible involvement of this neurotrophin in the region-specific increase in TPH mRNA expression induced by MDMA. We here report that, 24-48 h after acute MDMA treatment, the expression of BDNF in the frontal cortex is increased by approximately 33-70%, and the levels of the transcription factor phospho-CREB are also increased. In the hippocampus, however, a time-dependent decrease in BDNF mRNA expression (maximal decrease of approximately 73%) is found in all subfields examined 2-7 days after treatment in spite of increased phospho-CREB levels, perhaps as a consequence of corticosterone release by the serotonergic neurotoxin. The differential regulation of BDNF mRNA expression in the two brain regions examined appears to account for the enhanced TPH expression and the recovery of 5-HT levels in the frontal cortex, but not in the hippocampus, after neurotoxin treatment.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Lobo Frontal/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Alucinógenos/farmacologia , Hipocampo/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Análise de Variância , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Serotonina/metabolismo , Fatores de Tempo , Triptofano Hidroxilase/metabolismo
14.
Neuropharmacology ; 49(8): 1178-88, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16143352

RESUMO

Increased expression of brain-derived neurotrophic factor (BDNF) appears to be involved in the mechanism of action of antidepressant drugs. It has also been proposed that potentiation of the AMPA receptor (AMPAR) function may be useful in the treatment of depression. Here we looked for the time course of the effect of different doses of two antidepressants, desipramine (DMI) and paroxetine (PAR), which differentially affect monoamine reuptake, on BDNF mRNA expression in hippocampal subfields (CA1, CA3 and dentate gyrus) and levels of AMPAR subunits in total and membrane-enriched extracts from rat hippocampus. Acute antidepressant treatment changed neither BDNF mRNA expression nor AMPAR subunit levels. In chronic treatments, rats were treated daily with the antidepressants for 7-21 days. PAR produced a time- and dose-dependent increase of BDNF expression in the three hippocampal subfields examined. On the contrary, the effect of DMI on BDNF mRNA was neither dose- nor time-dependent. In rats receiving the same chronic antidepressant treatments, PAR produced a dose-dependent increase of GluR1 and GluR2/3 levels in the membrane fraction after a 3-week treatment, and not at earlier times. DMI increased the membrane levels of AMPAR subunits after a 3-week treatment with the lower dose tested. However, a higher dose, 15 mg/kg, did not produce any change in AMPAR subunits and reduced membrane levels of alpha-tubulin and PSD-95, possibly indicating a disorganization of membrane scaffolding proteins. The results suggest that paroxetine, but not desipramine, enhances synaptic plasticity in the hippocampus by increasing BDNF mRNA expression, which determines a later AMPAR subunit trafficking to synaptic membranes.


Assuntos
Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Hipocampo/metabolismo , RNA Mensageiro/biossíntese , Receptores de AMPA/efeitos dos fármacos , Sinapses/metabolismo , Animais , Antidepressivos de Segunda Geração/farmacologia , Antidepressivos Tricíclicos/farmacologia , Autorradiografia , Western Blotting , Desipramina/farmacologia , Proteína 4 Homóloga a Disks-Large , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Paroxetina/farmacologia , Ratos , Ratos Wistar , Receptores de AMPA/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sinapses/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
15.
Neuropharmacology ; 43(8): 1230-7, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12527472

RESUMO

It has been proposed that potentiation of AMPA receptor (AMPAR) function may be useful in the treatment of depression. Here we studied the acute and chronic effect of the antidepressants desipramine and paroxetine, which differentially affect monoamine reuptake, on the expression of the AMPAR subunits GluR1 and GluR2/3, analyzed by Western blot, both in total and in membrane-enriched extracts from rat hippocampus. Acute antidepressant treatment did not produce any change in the expression of AMPAR subunits. In chronic treatments, rats were daily treated with the antidepressants (10 mg/kg/day) for 7, 14, or 21 days. In rats receiving either of the two antidepressant treatments for 21 consecutive days and killed 24 h after the last injection, an increase in GluR1 and GluR2/3 levels was found in the membrane fraction, with no significant change in the total extract, suggesting a trafficking of the AMPAR subunits from intracellular pools to synaptic sites in the hippocampus. This appeared to be a region-specific effect since no change in AMPAR subunit expression was found in the frontal cortex. Previously reported modifications in phosphorylating enzymes by chronic antidepressants could perhaps play a role in hippocampal membrane insertion of AMPAR subunits. When the survival time after the 21-day-treatment was longer - 72 instead of 24 h - the hippocampal membrane expression of GluR1, but not of GluR2/3 subunits, was still increased, as could be expected from the distinct mechanisms operating in synaptic delivery of GluR1 and GluR2/3 subunits. The antidepressant-induced increase in the number of GluR1- and GluR2/3-containing AMPARs at the synapses may indicate an enhanced AMPAR-mediated synaptic transmission which could help to counteract the alterations in neuronal connectivity which appear to underlie the pathophysiology of mood disorders.


Assuntos
Antidepressivos/administração & dosagem , Membrana Celular/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Receptores de AMPA/biossíntese , Animais , Membrana Celular/metabolismo , Desipramina/farmacologia , Hipocampo/metabolismo , Masculino , Paroxetina/farmacologia , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA