Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Mol Psychiatry ; 8(2): 135-44, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12610644

RESUMO

The clinical efficacy of lithium in the prophylaxis of recurrent affective episodes in bipolar disorder is characterized by a lag in onset and remains for weeks to months after discontinuation. Thus, the long-term therapeutic effect of lithium likely requires reprogramming of gene expression. Protein kinase C and glycogen synthase kinase-3 signal transduction pathways are perturbed by chronic lithium at therapeutically relevant concentrations and have been implicated in modulating synaptic function in nerve terminals. These signaling pathways offer an opportunity to model critical signals for altering gene expression programs that underlie adaptive responses of neurons to long-term lithium exposure. While the precise physiological events critical for the clinical efficacy of lithium remain unknown, we propose that linking lithium-responsive genes as a regulatory network will provide a strategy to identify signature gene expression patterns that distinguish between therapeutic and nontherapeutic actions of lithium.


Assuntos
Antimaníacos/uso terapêutico , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular , Lítio/uso terapêutico , Proteínas de Membrana , Transdução de Sinais/efeitos dos fármacos , Transtorno Bipolar/genética , Proteínas de Ligação ao Cálcio , Expressão Gênica/efeitos dos fármacos , Glucosidases , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Substrato Quinase C Rico em Alanina Miristoilada , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo
2.
J Neurochem ; 79(4): 816-25, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11723174

RESUMO

The gene (Macs) for the mouse myristoylated alanine-rich C kinase substrate (MARCKS) encodes a prominent substrate for protein kinase C that has been implicated in processes requiring signal dependent changes in actin-membrane plasticity and cytoskeletal restructuring. We have previously demonstrated that MARCKS protein is significantly down-regulated in rat hippocampus and in an immortalized hippocampal cell line (HN33.dw) following long-term exposure to lithium at clinically relevant concentrations (1 mM). Our current studies have examined transcriptional and post-transcriptional events that may underlie the lithium-induced down-regulation of MARCKS protein in the cultured hippocampal cell model system. MARCKS mRNA and protein expression were found to be concomitantly down-regulated following exposure of the HN33.dw cells to chronic lithium. Whereas the stability of MARCKS mRNA remained unchanged in the presence of lithium, nuclear run-off assay indicated that the transcription of nascent MARCKS mRNA was significantly reduced (approximately 50%) in the cells that had been treated with lithium for 7 days. Transient transfection of HN33.dw cells with a mouse cloned Macs promoter (993-bp) showed that the Macs promoter activity was attenuated to the same extent after chronic (7-10 days), but not subacute (24 h), lithium exposure. The inhibition of the Macs promoter was found to be dependent upon the presence of a 280-bp promoter region between -993-bp and -713-bp relative to the translation start site, suggesting that this region is a potential lithium-responsive region of Macs promoter (LRR). Mutant promoter lacking the LRR not only did not respond to chronic lithium exposure but also had significantly reduced promoter activity, suggesting that chronic lithium exposure represses the transcriptional activity of activator(s) bound to the promoter. Taken together, our data indicate that transcriptional inhibition of the Macs gene underlies the lithium-induced down-regulation of MARCKS expression in the immortalized hippocampal cells.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Lítio/farmacologia , Proteínas de Membrana , Proteínas/metabolismo , Animais , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Genes Reguladores/efeitos dos fármacos , Hipocampo/citologia , Camundongos , Mutagênese Sítio-Dirigida , Substrato Quinase C Rico em Alanina Miristoilada , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas/genética , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
3.
J Biol Chem ; 276(25): 22351-8, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11294839

RESUMO

We recently identified conformational changes that occur upon phosphorylation of myristoylated alanine-rich protein kinase C substrate (MARCKS) that preclude efficient cross-linking of actin filaments (Bubb, M. R., Lenox, R. H., and Edison, A. S. (1999) J. Biol. Chem. 274, 36472-36478). These results implied that the phosphorylation site domain of MARCKS has two actin-binding sites. We now present evidence for the existence of two actin-binding sites that not only mutually compete but also specifically compete with the actin-binding proteins thymosin beta(4) and actobindin to bind to actin. The effects of substitution of alanine for phenylalanine within a repeated hexapeptide segment suggest that the noncharged region of the domain contributes to binding affinity, but the binding affinity of peptides corresponding to each binding site has a steep dependence on salt concentration, consistent with presumed electrostatic interactions between these polycationic peptides and the polyanionic N terminus of actin. Phosphorylation decreases the site-specific affinity by no more than 0.7 kcal/mol, which is less than the effect of alanine substitution. However, phosphorylation has a much greater effect than alanine substitution on the loss of actin filament cross-linking activity. These results are consistent with the hypothesis that the compact structure resulting from conformational changes due to phosphorylation, in addition to modest decreases in site-specific affinity, explains the loss of cross-linking activity in phosphorylated MARCKS.


Assuntos
Actinas/metabolismo , Proteínas Contráteis , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Proteínas de Transporte/metabolismo , Proteínas dos Microfilamentos/metabolismo , Dados de Sequência Molecular , Substrato Quinase C Rico em Alanina Miristoilada , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação , Profilinas , Proteínas/química , Proteínas de Protozoários , Coelhos , Timosina/metabolismo
4.
J Neurosci Res ; 62(3): 416-26, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11054811

RESUMO

In the mature hippocampus, kainic acid seizures lead to excitotoxic cell death and synaptic reorganization in which granule cell axons (mossy fibers) form ectopic synapses on granule cell dendrites. In the present study, we examined the expression of four major, developmentally regulated protein kinase C (PKC) substrates (MARCKS, MLP, GAP-43, RC3), which have different subcellular and regional localizations in the hippocampus at several time points (6 hr, 12 hr, 18 hr, 24 hr, 48 hr, 5 days, or 15 days) following kainic acid seizures using in situ hybridization. Consistent with previous reports, following kainate seizures, GAP-43 mRNA expression exhibited a delayed and protracted elevation in the granule cell layer, which peaked at 24 hr, whereas expression in fields CA1 and CA3 remained relatively unchanged. Conversely, RC3 mRNA expression exhibited a delayed reduction in the granule cell layer that was maximal at 18 hr, as well as a reduction CA1 at 48 hr, whereas CA3 levels did not change. MARCKS mRNA expression in the granule cell layer and CA1 remained stable following kainate, although an elevation was observed in subfield CA3c at 12 hr. Similarly, MLP mRNA expression did not change in the granule cell layer or CA1 following kainate but exhibited a protracted elevation in subfields CA3b,c beginning at 6 hr post-kainate. Collectively these data demonstrate that different PKC substrate mRNAs exhibit unique expression profiles and regulation in the different cell fields of the mature hippocampus following kainic acid seizures and during subsequent synaptic reorganization. The expression profiles following kainate seizures bear resemblance to those observed during postnatal hippocampal development, which may indicate the recruitment of common regulatory mechanisms.


Assuntos
Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Convulsões/metabolismo , Sinapses/metabolismo , Animais , Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Hipocampo/patologia , Ácido Caínico , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microglia/metabolismo , Fibras Musgosas Hipocampais/patologia , Substrato Quinase C Rico em Alanina Miristoilada , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogranina , Proteínas/genética , Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Proteínas de Transporte Vesicular
5.
Biol Psychiatry ; 48(6): 518-30, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11018224

RESUMO

Clinical studies over the years have provided evidence that monoamine signaling and hypothalamic-pituitary-adrenal axis disruption are integral to the pathophysiology of bipolar disorder. A full understanding of the pathophysiology from a molecular to a systems level must await the identification of the susceptibility and protective genes driving the underlying neurobiology of bipolar disorder. Furthermore, the complexity of the unique biology of this affective disorder, which includes the predisposition to episodic and often progressive mood disturbance, and the dynamic nature of compensatory processes in the brain, coupled with limitations in experimental design, have hindered our progress to date. Imaging studies in patient populations have provided evidence of a role for anterior cingulate, amygdala, and prefrontal cortex in the pathophysiology of bipolar disorder. More recent research strategies designed to uncover the molecular mechanisms underlying our pharmacologic treatments and their interaction in the regulation of signal transduction as well as more advanced brain imaging studies remain promising approaches. This experimental strategy provides data derived from the physiologic response of the system in affected individuals and addresses the critical dynamic interaction with pharmacologic agents that effectively modify the clinical expression of the pathophysiology.


Assuntos
Antipsicóticos/farmacologia , Transtorno Bipolar/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/fisiopatologia , Encéfalo/fisiopatologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Bombas de Íon/metabolismo , Lítio/farmacologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Proteína Quinase C/metabolismo
6.
Biochem Biophys Res Commun ; 276(1): 183-8, 2000 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-11006104

RESUMO

The myristoylated alanine-rich C kinase substrate (MARCKS) is a prominent PKC-substrate in the brain, which has been implicated in brain development, cytoskeletal remodeling, calcium/calmodulin signaling, and neuroplasticity. The sequence of the Macs gene codes for a protein that has three highly conserved domains including a 5' myristoylation region and a 25-amino-acid phosphorylation site domain (PSD), which are involved in anchoring MARCKS to the cellular membrane. In this study, we examined the role of the myristoylation signal in the regulation of MARCKS in transfected rat hippocampal cells (H19-7) following retinoic acid (RA) treatment. A mutant MARCKS lacking the myristoylation signal was engineered by substitution of alanine for glycine at position 2 of the Macs gene and was found to be exclusively expressed in the cytosol fraction of transfected cells. Exposure of the wild-type MARCKS-transfected cells to RA resulted in an apparent shift of MARCKS from the membrane to the cytosol, while the total protein of wild-type MARCKS was not significantly changed. In contrast, RA-exposed cells transfected with the mutant MARCKS revealed a dramatic reduction of expression of MARCKS protein in both cytosol and total protein fractions. These data suggest that the absence of the myristoyl moiety may not only alter the anchoring of the protein to the membrane but also play a novel role in modulating cellular levels of MARCKS protein in response to RA.


Assuntos
Antineoplásicos/farmacologia , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Proteínas/metabolismo , Tretinoína/farmacologia , Animais , Linhagem Celular Transformada , Regulação para Baixo/efeitos dos fármacos , Mutação , Substrato Quinase C Rico em Alanina Miristoilada , Fosforilação , Proteína Quinase C/metabolismo , Proteínas/genética , Ratos
7.
Neuroscience ; 97(3): 581-9, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10828540

RESUMO

We have previously shown that the myristoylated alanine-rich C kinase substrate, a primary protein kinase C substrate in brain that binds and cross-links filamentous actin, is enriched in neuronal growth cones and is developmentally regulated in brain. Here we examined myristoylated alanine-rich C kinase substrate expression in the facial motor nucleus during axonal regeneration following facial nerve axotomy or facial nerve resection lesions, which impede regeneration, or following motor neuron degeneration induced by the retrograde neurotoxin ricin. For comparative purposes, the protein kinase C substrates myristoylated alanine-rich C kinase substrate-like protein and growth-associated protein-43 were examined in parallel. Myristoylated alanine-rich C kinase substrate messenger RNA exhibited a robust increase in both neurons and non-neuronal cells in the facial motor nucleus beginning four days after axotomy, peaked at seven days (2.5-fold), and declined back to baseline levels by 40 days. Myristoylated alanine-rich C kinase substrate protein similarly exhibited a twofold elevation in the facial motor nucleus determined four and 14 days post-axotomy. Following nerve resection, myristoylated alanine-rich C kinase substrate messenger RNA levels increased at seven days and returned to baseline levels by 40 days. Unlike myristoylated alanine-rich C kinase substrate messenger RNA, myristoylated alanine-rich C kinase substrate-like messenger RNA levels did not increase in the facial motor nucleus at any time point following nerve axotomy or resection, whereas growth-associated protein-43 messenger RNA exhibited a rapid (one day) and prolonged (40 days) elevation in facial motor nucleus neurons following either nerve axotomy or resection. Ricin-induced degeneration of facial motor neurons elevated myristoylated alanine-rich C kinase substrate and myristoylated alanine-rich C kinase substrate-like messenger RNAs in both microglia (lectin-positive) and astrocytes (glial fibrillary acidic protein-positive).Collectively, these data demonstrate that myristoylated alanine-rich C kinase substrate exhibits a unique expression profile in the facial motor nucleus following facial nerve lesions, and it is proposed that myristoylated alanine-rich C kinase substrate may serve to mediate actin-membrane cytoskeletal plasticity in both neurons and glial cells in response to protein kinaseC-mediated signaling during nerve regeneration and degeneration.


Assuntos
Nervo Facial/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neurônios Motores/metabolismo , Regeneração Nervosa/fisiologia , Proteínas/genética , Animais , Axotomia , Nervo Facial/citologia , Proteína GAP-43/genética , Masculino , Proteínas de Membrana/genética , Neurônios Motores/citologia , Substrato Quinase C Rico em Alanina Miristoilada , Degeneração Neural/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , RNA Mensageiro/metabolismo , Ratos , Regulação para Cima/fisiologia , Proteínas de Transporte Vesicular
8.
J Clin Psychiatry ; 61 Suppl 9: 5-15, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10826655

RESUMO

Since its discovery, lithium has been shown to act upon various neurotransmitter systems at multiple levels of signaling in the brain. Lithium, affecting each neurotransmitter system within complex interactive neuronal networks, is suggested to restore the balance among aberrant signaling pathways in critical regions of the brain. Recent molecular studies have revealed the action of lithium on signal transduction mechanisms, such as phosphoinositide hydrolysis, adenylyl cyclase, G protein, glycogen synthase kinase-3beta, protein kinase C, and its substrate myristoylated alanine-rich C kinase substrate. Such effects are thought to trigger long-term changes in neuronal signaling patterns that account for the prophylactic properties of lithium in the treatment of bipolar disorder. Through its effects on glycogen synthase kinase-3beta and protein kinase C, lithium may alter the level of phosphorylation of cytoskeletal proteins, which leads to neuroplastic changes associated with mood stabilization. Chronic lithium regulates transcriptional factors, which in turn may modulate the expression of a variety of genes that compensate for aberrant signaling associated with the pathophysiology of bipolar disorder. Future studies on long-term neuroplastic changes caused by lithium in the brain will set the stage for new drug-discovery opportunities.


Assuntos
Encéfalo/efeitos dos fármacos , Lítio/farmacologia , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/metabolismo , Transtorno Bipolar/fisiopatologia , Química Encefálica/efeitos dos fármacos , Proteínas do Citoesqueleto/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Transporte de Íons/efeitos dos fármacos , Lítio/farmacocinética , Lítio/uso terapêutico , Plasticidade Neuronal/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/fisiologia , Neurotransmissores/fisiologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
10.
J Clin Psychiatry ; 61 Supp 13: 42-57, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11153812

RESUMO

The underlying pathophysiology of bipolar disorder is a continually evolving complexity of multilayer interacting and independent systems. The dearth of adequate preclinical or clinical models that incorporate the various features of the illness, i.e., acute and chronic, recurrent and episodic, and time-course and treatment-related variables, has made the consistency and interpretation of data difficult. Newer technologies and the availability of structurally and mechanistically distinct pharmacologic agents have expanded opportunities for experimental study. In addition to the well-known neurotransmitter systems that are disrupted in mood disorders, critical guanine nucleotide-binding protein (G protein)-coupled signaling pathways are implicated in modulating mood state. Regulation of gene expression and identification of factors regulating neuroplasticity and neurotrophic events in the central nervous system in bipolar disorder are 2 of the more recent approaches contributing to clarification of the pathophysiology and potential treatment options.


Assuntos
Transtorno Bipolar/fisiopatologia , Transtorno Bipolar/tratamento farmacológico , Morte Celular/fisiologia , Ritmo Circadiano/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Modelos Animais de Doenças , Dopamina/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Humanos , Lítio/farmacologia , Lítio/uso terapêutico , Neurotransmissores/fisiologia , Norepinefrina/fisiologia , Fosfatidilinositóis/fisiologia , Proteína Quinase C/fisiologia , Serotonina/fisiologia , Transdução de Sinais/fisiologia , Ácido Valproico/farmacologia , Ácido Valproico/uso terapêutico
11.
J Biol Chem ; 274(51): 36472-8, 1999 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-10593944

RESUMO

Phosphorylation of myristoylated alanine-rich protein kinase C substrate (MARCKS) by protein kinase C eliminates actin filament cross-linking activity, but residual filament binding activity docks phosphorylated MARCKS on filamentous actin. The postulated actin-binding region of MARCKS, which includes a Ca(2+)-calmodulin-binding site, has been portrayed with alpha-helical structure, analogous to other calmodulin-binding domains. Previous speculation suggested that MARCKS may dimerize to form the two functional actin-binding sites requisite for cross-linking activity. Contrary to these hypotheses, we show that MARCKS peptide with actin-cross-linking activity has an extended structure in aqueous solution but assumes a more compact structure upon phosphorylation. We hypothesize that structural changes in the MARCKS peptide induced by phosphorylation create a dynamic structure that, on average, has only one actin-binding site. Moreover, independent of the state of phosphorylation, this peptide is monomeric rather than dimeric, implying that two distinct actin-binding sites are responsible for the actin-cross-linking activity of unphosphorylated MARCKS. These studies uniquely elucidate the mechanism by which phosphorylation of MARCKS induces structural changes and suggest how these structural changes determine biological activity.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Conformação Proteica , Proteína Quinase C/metabolismo , Proteínas/química , Proteínas/metabolismo , Sequência de Aminoácidos , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Substrato Quinase C Rico em Alanina Miristoilada , Fosforilação , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato
12.
Biol Psychiatry ; 46(10): 1328-51, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10578449

RESUMO

Understanding the biology of the pharmacological stabilization of mood will undoubtedly serve to provide significant insight into the pathophysiology of manic-depressive illness (MDI). Accumulating evidence from our laboratories and those of other researchers has identified the family of protein kinase C isozymes as a shared target in the brain for the long-term action of both lithium and valproate. In rats chronically treated with lithium, there is a reduction in the hippocampus of the expression of two protein kinase isozymes, alpha and epsilon, as well as a reduction in the expression of a major PKC substrate, MARCKS, which has been implicated in long-term neuroplastic events in the developing and adult brain. In addition, we have been investigating the down-stream impact of these mood stabilizers on another kinase system, GSK-3 beta and on the AP-1 family of transcription factors. Further studies have generated promising preliminary data in support of the antimanic action of tamoxifen, and antiestrogen that is also a PKC inhibitor. Future studies must address the therapeutic relevance of these protein targets in the brain using innovative strategies in both animal and clinical investigations to ultimately create opportunities for the discovery of the next generations of mood stabilizers for the treatment of MDI.


Assuntos
Distinções e Prêmios , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/enzimologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/uso terapêutico , Ativadores de Enzimas/uso terapêutico , Proteína Quinase C/metabolismo , Pesquisa , Transdução de Sinais/efeitos dos fármacos , Animais , Antimaníacos/farmacologia , Antimaníacos/uso terapêutico , Ligação Competitiva , Transtorno Bipolar/genética , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Modelos Animais de Doenças , Ativadores de Enzimas/farmacologia , Quinase 3 da Glicogênio Sintase , Humanos , Lítio/farmacologia , Lítio/uso terapêutico , Masculino , Proteína Quinase C/genética , Ratos , Ratos Sprague-Dawley , Fator de Transcrição AP-1/efeitos dos fármacos , Fator de Transcrição AP-1/genética , Transcrição Gênica/genética , Ácido Valproico/farmacologia , Ácido Valproico/uso terapêutico
13.
J Neurochem ; 73(5): 2175-83, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10537078

RESUMO

Activation of protein kinase C (PKC) is one of the biochemical pathways thought to be activated during activity-dependent synaptic plasticity in the brain, and long-term potentiation (LTP) and long-term depression (LTD) are two of the most extensively studied models of synaptic plasticity. Here we have examined changes in the in situ phosphorylation level of two major PKC substrates, myristoylated alanine-rich C kinase substrate (MARCKS) and growth-associated protein (GAP)-43/B-50, after pharmacological stimulation or induction of LTP or LTD in the CA1 field of the hippocampus. We find that direct PKC activation with phorbol esters, K+-induced depolarization, and activation of metabotropic glutamate receptors increase the in situ phosphorylation of both MARCKS and GAP-43/B-50. The induction of LTP increased the in situ phosphorylation of both MARCKS and GAP-43/B-50 at 10 min following high-frequency stimulation, but only GAP-43/B-50 phosphorylation remained elevated 60 min after LTP induction. Furthermore, blockade of LTP induction with the NMDA receptor antagonist D-2-amino-5-phosphonopentanoic acid prevented elevations in GAP-43/B-50 phosphorylation but did not prevent the elevation in MARCKS phosphorylation 10 min following LTP induction. The induction of LTD resulted in a reduction in GAP-43/B-50 phosphorylation but did not affect MARCKS phosphorylation. Together these findings show that activity-dependent synaptic plasticity elicits PKC-mediated phosphorylation of substrate proteins in a highly selective and coordinated manner and demonstrate the compartmentalization of PKC-substrate interactions. Key Words: Protein kinase C-Myristoylated alanine-rich C kinase substrate-Growth-associated protein-43-Long-term potentiation-Long-term depression-(RS)-alpha-Methyl-4-carboxyphenylglycine-D-2-Amino-5-ph osphonopentanoic acid-Glutamate.


Assuntos
Proteína GAP-43/metabolismo , Hipocampo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Potenciação de Longa Duração , Proteínas de Membrana , Proteína Quinase C/metabolismo , Proteínas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Técnicas de Imunoadsorção , Substrato Quinase C Rico em Alanina Miristoilada , Ésteres de Forbol/farmacologia , Fosforilação , Transmissão Sináptica
14.
Brain Res ; 832(1-2): 136-44, 1999 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-10375659

RESUMO

The expression of GAP-43 was modulated genetically in the adult rat nigrostriatal or septohippocampal pathway using recombinant adeno-associated virus (rAAV) vectors incorporating the neuron specific enolase (NSE) promoter and either a rat GAP-43 cDNA or the corresponding antisense sequence. Bicistronic expression of green fluorescent protein (GFP) enabled us to evaluate transduced neurons selectively. Single injections of rAAV into the substantia nigra pars compacta (SNc) transduced both dopaminergic and non-dopaminergic neurons stably for the 3-month duration of the study. Transduction with the GAP-43 vector in this region: (1) increased GAP-43 mRNA levels 2-fold compared to controls; (2) led to GAP-43 immunoreactivity in neuronal perikarya, axons, and dendrites that was not observed otherwise; and (3) resulted in GAP-43/ GFP-positive axons that were traced to the striatum where they formed clusters of aberrant nets. The GAP-43 antisense vector, in contrast, decreased neuropil GAP-43 immunoreactivity compared to controls in the SNc. In septum, injections of the GAP-43 expressing vector also caused aberrant clusters of GAP-43 labelled fibers in terminal fields, i.e., fornix and hippocampus, that were not observed in control tissues. It therefore appears that rAAV vectors provide a novel approach for modulating intraneuronal GAP-43 expression in the adult brain.


Assuntos
Encéfalo/fisiologia , Proteína GAP-43/biossíntese , Técnicas de Transferência de Genes , Degeneração Neural , Regeneração Nervosa , Neurônios/fisiologia , Vias Aferentes/fisiologia , Animais , Encéfalo/metabolismo , Corpo Estriado/fisiologia , Proteínas de Fluorescência Verde , Hipocampo/fisiologia , Indicadores e Reagentes , Proteínas Luminescentes/análise , Ratos , Ratos Sprague-Dawley , Septo Pelúcido/fisiologia , Substância Negra/fisiologia
16.
J Neurochem ; 72(4): 1735-43, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10098884

RESUMO

Protein kinase C (PKC) consists of a family of Ca2+/phospholipid-dependent isozymes that has been implicated in the delayed neurotoxic effects of glutamate in vitro. In the present study, we assessed the effect of the glutamate analogue kainic acid (KA) on the subcellular expression of PKC isozymes in the hippocampus (HPC) in the period preceding (0.5, 1.5, 12, and 24 h) and during (120 h) hippocampal necrosis using western blot analysis and PKC isozyme-specific antibodies. Before subcellular fractionation (cytosol + membrane), hippocampi were microdissected into "HPC" (fields CA1-CA3) and "dentate gyrus" (DG; granule cells + hilus) regions. Four general patterns of alterations in PKC isozyme expression/distribution were observed following KA treatment. The first pattern was a relative stability in expression following KA treatment and was most apparent for cytosol PKCalpha (HPC + DG) and membrane (HPC) and cytosol (DG) PKCbetaII. The second pattern, observed with PKCgamma and PKCepsilon, was characterized by an initial increase in expression in both membrane and cytosolic fractions before seizure activity (0.5 h) followed by a gradual decrease until significant reductions are observed by 120 h. The third pattern, exhibited by PKCdelta, involved an apparent translocation, increasing in the membrane and decreasing in the cytosol, followed by down-regulation in both fractions and subsequent recovery. The fourth pattern was observed with PKCzeta only and entailed a significant reduction in expression before and during limbic motor seizures followed by a dramatic fivefold increase in the membrane fraction during the period of hippocampal necrosis (120 h). Although these patterns did not segregate according to conventional PKC isozyme classifications, they do indicate dynamic isozyme-specific regulation by KA. The subcellular redistribution of PKC isozymes may contribute to the histopathological sequelae produced by KA in the hippocampus and may model the pathogenesis associated with diseases involving glutamate-induced neurotoxicity.


Assuntos
Giro Denteado/enzimologia , Agonistas de Aminoácidos Excitatórios , Ácido Caínico , Proteína Quinase C/análise , Animais , Química Encefálica/efeitos dos fármacos , Citosol/enzimologia , Giro Denteado/citologia , Isoenzimas/análise , Masculino , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurotoxinas/farmacologia , Proteína Quinase C beta , Proteína Quinase C-alfa , Proteína Quinase C-delta , Proteína Quinase C-épsilon , Ratos , Ratos Sprague-Dawley , Frações Subcelulares/enzimologia
17.
J Clin Psychiatry ; 60 Suppl 2: 21-6; discussion 40-1, 113-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10073384

RESUMO

BACKGROUND: Although suicide is a leading cause of death in the United States and represents a significant public health threat, little is known about the neurobiological or molecular factors that contribute to its pathophysiology. A number of studies now indicate that lithium has considerable efficacy in the prevention of suicide in patients with affective disorders, and accumulating evidence indicates that protein kinase C (PKC) and its substrates, in particular the myristoylated alanine-rich C kinase substrate (MARCKS), are primary targets of chronic lithium treatment. We therefore hypothesized that a dysregulation in MARCKS expression in key brain regions could contribute to the pathophysiology associated with suicide. To address this, we examined MARCKS, as well as the closely related MARCKS-related protein (MRP), mRNA expression in the hippocampus and dorsolateral prefrontal cortex of suicide victims and normal controls. METHOD: MARCKS and MRP mRNA expression was assessed by quantitative in situ hybridization histochemistry performed on postmortem hippocampal and dorsolateral prefrontal cortex sections from suicide (N = 9) and normal control (N = 10) brains. RESULTS: In the normal hippocampus, both MARCKS and MRP mRNA expression were highest in the granule cell layer and low-moderate in CA1, CA3, and hilus. A high level of MRP mRNA expression was also observed in the white matter of the fimbria/fornix. Neither MARCKS nor MRP mRNA expression levels differed significantly in the granule cell layer, CA3, hilus, or CA1 in suicide victims relative to normal controls (1-way ANOVA, p > .05). In the normal prefrontal cortex, MARCKS was expressed exclusively in gray matter (layers I-VI), whereas MRP was expressed in both gray and white matter. Neither MARCKS nor MRP mRNA expression levels in the gray and white matter regions of the dorsal prefrontal cortex differed between suicides and normal controls (1-way ANOVA, p > .05). CONCLUSION: The present findings are the first to demonstrate the expression and distribution of MARCKS and MRP in the human hippocampus and dorsolateral prefrontal cortex, and their expression pattern within these regions bears strong resemblance to those observed in the adult rat brain. Comparison of MARCKS and MRP mRNA expression in the hippocampus and prefrontal cortex of suicide victims and normal controls indicates that these 2 mRNAs are not differentially regulated in these regions. However, differences in MARCKS and MRP protein expression and function cannot be ruled out by the present findings.


Assuntos
Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Córtex Pré-Frontal/metabolismo , Proteína Quinase C/química , Proteínas/química , Suicídio/estatística & dados numéricos , Adulto , Animais , Autorradiografia , Feminino , Hipocampo/química , Hipocampo/enzimologia , Histocitoquímica , Humanos , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Substrato Quinase C Rico em Alanina Miristoilada , Córtex Pré-Frontal/química , Córtex Pré-Frontal/enzimologia , Biossíntese de Proteínas , Proteína Quinase C/biossíntese , Proteína Quinase C/metabolismo , Proteínas/metabolismo , Sondas RNA , RNA Mensageiro/metabolismo , Ratos
18.
Aust N Z J Psychiatry ; 33 Suppl: S65-83, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10622182

RESUMO

The long-term treatment of manic-depressive illness (MDI) likely involves the strategic regulation of signalling pathways and gene expression in critical neuronal circuits. Accumulated evidence has identified signalling pathways, in particular the family of protein kinase C (PKC) isozymes, as targets for the long-term action of lithium. Chronic lithium administration produces a reduction in the expression of PKC alpha and epsilon, as well as a major PKC substrate, MARCKS, which has been implicated in long-term neuroplastic events in the developing and adult brain. More recently, studies have demonstrated robust effects of lithium on another kinase system, GSK-3beta, and on neuroprotective/neurotrophic proteins in the brain. Given the key roles of these signalling cascades in the amplification and integration of signals in the central nervous system, these findings have clear implications not only for research into the neurobiology of MDI, but also for the future development of novel and innovative treatment strategies.


Assuntos
Transtorno Bipolar/fisiopatologia , Proteína Quinase C/metabolismo , Transdução de Sinais , Adulto , Antimaníacos/farmacologia , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/genética , Encéfalo/fisiologia , Humanos , Isoenzimas , Carbonato de Lítio/farmacologia
19.
Proc Natl Acad Sci U S A ; 95(24): 14517-22, 1998 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-9826732

RESUMO

The myristoylated alanine-rich C kinase substrate (MARCKS) is a prominent protein kinase C (PKC) substrate in brain that is expressed highly in hippocampal granule cells and their axons, the mossy fibers. Here, we examined hippocampal infrapyramidal mossy fiber (IP-MF) limb length and spatial learning in heterozygous Macs mutant mice that exhibit an approximately 50% reduction in MARCKS expression relative to wild-type controls. On a 129B6(N3) background, the Macs mutation produced IP-MF hyperplasia, a significant increase in hippocampal PKCepsilon expression, and proficient spatial learning relative to wild-type controls. However, wild-type 129B6(N3) mice exhibited phenotypic characteristics resembling inbred 129Sv mice, including IP-MF hypoplasia relative to inbred C57BL/6J mice and impaired spatial-reversal learning, suggesting a significant contribution of 129Sv background genes to wild-type and possibly mutant phenotypes. Indeed, when these mice were backcrossed with inbred C57BL/6J mice for nine generations to reduce 129Sv background genes, the Macs mutation did not effect IP-MF length or hippocampal PKCepsilon expression and impaired spatial learning relative to wild-type controls, which now showed proficient spatial learning. Moreover, in a different strain (B6SJL(N1), the Macs mutation also produced a significant impairment in spatial learning that was reversed by transgenic expression of MARCKS. Collectively, these data indicate that the heterozygous Macs mutation modifies the expression of linked 129Sv gene(s), affecting hippocampal mossy fiber development and spatial learning performance, and that MARCKS plays a significant role in spatial learning processes.


Assuntos
Hipocampo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana , Fibras Nervosas/fisiologia , Proteína Quinase C/genética , Proteínas/genética , Proteínas/fisiologia , Animais , Encéfalo/patologia , Córtex Cerebral/metabolismo , Quimera , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Hiperplasia , Isoenzimas/genética , Deficiências da Aprendizagem/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Substrato Quinase C Rico em Alanina Miristoilada , Fibras Nervosas/patologia , Proteína Quinase C-épsilon , Células Piramidais/fisiologia , Percepção Espacial , Transcrição Gênica
20.
Life Sci ; 63(19): 1715-24, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9806227

RESUMO

The Xenopus oocyte has been shown to be a useful model for the study of signal transduction pathways. The present study investigated whether or not the oocyte could be used to study the effects of lithium on signal transduction mechanisms by comparing the dynamics of lithium homeostasis in the oocyte and a human immortalized hippocampal cell line using Flame Atomic Emission Spectroscopy (FAES). A biphasic pattern of lithium uptake was observed in the oocyte in the presence of 5 mM extracellular lithium. The late phase of lithium uptake, which started after 30 minutes of incubation time, was sensitive to phloretin, an inhibitor of Na+/Li+ counter-transport. Differences in lithium efflux kinetics further characterized the two observed phases of accumulation and also suggested that lithium might be distributed in different pools within the oocyte, including one sequestered in organelles or associated with cytosolic proteins. An analogous sequestered pool was not, however, observed in the hippocampal cell line indicating that lithium is distributed differently in these cell types. This suggests that the Xenopus oocyte might not be a suitable model for evaluating the effects of lithium on signal transduction pathways because of the unknown contribution of the sequestered pool on predicting relevant physiological effects.


Assuntos
Homeostase/fisiologia , Lítio/metabolismo , Transdução de Sinais/fisiologia , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Linhagem Celular , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Hipocampo/citologia , Hipocampo/metabolismo , Oócitos/metabolismo , Ouabaína/farmacologia , Floretina/farmacologia , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...