Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Neuroscience ; 448: 287-298, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-32905841

RESUMO

The integrity of the perirhinal cortex (PRh) is essential for object recognition memory (ORM) function, and damage to this brain area in animals and humans induces irreversible ORM deficits. Here, we show that activation of area V2, a brain area interconnected with brain circuits of ventral stream and medial temporal lobe that sustain ORM, by expression of regulator of G-protein signaling 14 of 414 amino acids (RGS14414) restored ORM in memory-deficient PRh-lesioned rats and nonhuman primates. Furthermore, this treatment was sufficient for full recovery of ORM in rodent models of aging and Alzheimer's disease, conditions thought to affect multiple brain areas. Thus, RGS14414-mediated activation of area V2 has therapeutic relevance in the recovery of recognition memory, a type of memory that is primarily affected in patients or individuals with symptoms of memory dysfunction. These findings suggest that area V2 modulates the processing of memory-related information through activation of interconnected brain circuits formed by the participation of distinct brain areas.


Assuntos
Doença de Alzheimer , Córtex Perirrinal , Proteínas RGS , Envelhecimento , Animais , Humanos , Transtornos da Memória , Primatas , Ratos , Roedores
2.
J Alzheimers Dis ; 28(3): 567-78, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22045482

RESUMO

The etiology of the more common (sporadic) forms of Alzheimer's disease (AD) remains unknown, although age is the most important risk factor. Nevertheless, interactions between environmental risk factors and genetic background may also influence the onset and progression of sporadic AD. Chronic stress, associated with altered memory and other neurological processes, is thought to influence the pathogenesis of AD. Hence, we evaluated the effect of unpredictable and consecutive chronic mild stressors on the onset of an AD-related pathology in the Tg2576 mouse line that overexpresses the human amyloid-ß protein precursor with the Swedish mutation (hAßPP(Swe)). Two months after exposure to chronic mild stress, 4 month-old animals that normally display no pathological features of AD, not only expressed pathological markers but also experienced cognitive dysfunction in the Morris water maze test. These findings suggest that chronic mild stress accelerates the onset of cognitive impairment and produces an increase in hippocampal amyloid-ß and phospho-tau levels on a background of AD susceptibility.


Assuntos
Doença de Alzheimer/complicações , Transtornos Cognitivos/etiologia , Fenótipo , Estresse Psicológico/complicações , Estresse Psicológico/etiologia , Fatores Etários , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Doença Crônica , Transtornos Cognitivos/genética , Corticosterona/sangue , Modelos Animais de Doenças , Progressão da Doença , Feminino , Hipocampo/metabolismo , Humanos , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Mutação/genética , Fosforilação/genética , Estatísticas não Paramétricas , Estresse Psicológico/sangue , Proteínas tau/metabolismo
3.
J Med Chem ; 54(23): 7986-99, 2011 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-22029386

RESUMO

We report the synthesis of new compounds 4-35 based on structural modifications of different moieties of previously described lead UCM-2550. The new nonpiperazine derivatives, representing second-generation agonists, were assessed for binding affinity, selectivity, and functional activity at the 5-HT(1A) receptor (5-HT(1A)R). Computational ß(2)-based homology models of the ligand-receptor complexes were used to explain the observed structure-affinity relationships. Selected candidates were also evaluated for their potential in vitro and in vivo neuroprotective properties. Interestingly, compound 26 (2-{6-[(3,4-dihydro-2H-chromen-2-ylmethyl)amino]hexyl}tetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione) has been characterized as a high-affinity and potent 5-HT(1A)R agonist (K(i) = 5.9 nM, EC(50) = 21.8 nM) and exhibits neuroprotective effect in neurotoxicity assays in primary cell cultures from rat hippocampus and in the MCAO model of focal cerebral ischemia in rats.


Assuntos
Benzopiranos/síntese química , Compostos Bicíclicos Heterocíclicos com Pontes/síntese química , Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/síntese química , Receptor 5-HT1A de Serotonina/metabolismo , Agonistas do Receptor 5-HT1 de Serotonina/síntese química , Sequência de Aminoácidos , Animais , Benzopiranos/química , Benzopiranos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Morte Celular , Células Cultivadas , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Hipocampo/citologia , Ataque Isquêmico Transitório/patologia , Modelos Moleculares , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Ensaio Radioligante , Ratos , Agonistas do Receptor 5-HT1 de Serotonina/química , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Relação Estrutura-Atividade
4.
Behav Brain Res ; 220(2): 338-43, 2011 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-21238494

RESUMO

This study was undertaken to know whether cognition deficits produced by chronic mild stress (CMS) were associated with pathological markers of Alzheimer's disease (AD). The results show that the impairment in the Morris water maze test induced by CMS correlated with an increase in CDK5-dependent phospho-tau levels and with an increase in APP processing. Mice exposed to CMS may then constitute a non-transgenic model for sporadic forms of AD.


Assuntos
Transtornos Cognitivos/etiologia , Quinase 5 Dependente de Ciclina/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo , Proteínas tau/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Aprendizagem da Esquiva/fisiologia , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFI/metabolismo , Fosforilação/fisiologia , Proteínas do Grupo Polycomb , Tempo de Reação/fisiologia , Serina/metabolismo , Estresse Psicológico/patologia , Fatores de Transcrição/metabolismo
5.
J Alzheimers Dis ; 23(2): 195-206, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20930307

RESUMO

The mitochondrial voltage-dependent anion channel 1 (VDAC1) is involved in the release of apoptotic proteins with possible relevance in Alzheimer's disease (AD) neuropathology. Through proteomic analysis followed by Western blotting and immunohistochemical techniques, we have found that VDAC1 is overexpressed in the hippocampus from amyloidogenic AD transgenic mice models. VDAC1 was also overexpressed in postmortem brain tissue from AD patients at an advanced stage of the disease. Interestingly, amyloid-ß (Aß) soluble oligomers were able to induce upregulation of VDAC1 in a human neuroblastoma cell line, further supporting a correlation between Aß levels and VDAC1 expression. In hippocampal extracts from transgenic mice, a significant increase was observed in the levels of VDAC1 phosphorylated at an epitope that is susceptible to phosphorylation by glycogen synthase kinase-3ß, whose activity was also increased. The levels of hexokinase I (HXKI), which interacts with VDAC1 and affects its function, were decreased in mitochondrial samples from AD models. Since phospho-VDAC and reduced HXKI levels favors a VDAC1 conformational state more prone to the release proapoptotic factors, regulation of the function of this channel may be a promising therapeutic approach to combat AD.


Assuntos
Doença de Alzheimer/metabolismo , Hipocampo/metabolismo , Mitocôndrias/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting , Eletroforese em Gel Bidimensional , Ensaio de Imunoadsorção Enzimática , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hexoquinase/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Fosforilação
6.
Rev Neurol ; 50(11): 667-75, 2010 Jun 01.
Artigo em Espanhol | MEDLINE | ID: mdl-20514639

RESUMO

The amyloid-beta peptide cascade hypothesis has provided a useful framework for the research on Alzheimer's disease for nearly 20 years. According to this hypothesis, an increase in amyloid-beta levels triggers all of the pathological features of the disease, including tau hyperphosphorylation, appearance of neurofibrillary tangles, synaptic dysfunction and neuronal cell death. Even though amyloid-beta peptide has an important role in the neurodegenerative process, different findings, such as the presence of abundant plaques in old cognitively normal individuals or the limited success of therapeutical approaches targeting only amyloid-beta, cast some doubt on a unique role for this peptide. At present, it is rather accepted that amyloid-beta peptide acts in parallel with other factors causing Alzheimer's disease that should be also considered at the time of designing useful therapeutic strategies.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Animais , Animais Geneticamente Modificados , Apolipoproteína E4/genética , Morte Celular , Humanos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Neurônios/patologia , Fatores de Risco , Proteínas tau/metabolismo
7.
Neuropsychopharmacology ; 35(7): 1593-604, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20336061

RESUMO

Clinical studies suggest that agonists at peroxisome proliferator-activated receptor gamma (PPARgamma) may exert beneficial effects in patients with mild-to-moderate Alzheimer's disease (AD), but the mechanism for the potential therapeutic interest of this class of drugs has not yet been elucidated. Here, in mice overexpressing mutant human amyloid precursor protein, we found that chronic treatment with rosiglitazone, a high-affinity agonist at PPARgamma, facilitated beta-amyloid peptide (Abeta) clearance. Rosiglitazone not only reduced Abeta burden in the brain but, importantly, almost completely removed the abundant amyloid plaques observed in the hippocampus and entorhinal cortex of 13-month-old transgenic mice. In the hippocampus, neuropil threads containing phosphorylated tau, probably corresponding to dystrophic neurites, were also decreased by the drug. Rosiglitazone switched on the activated microglial phenotype, promoting its phagocytic ability, reducing the expression of proinflammatory markers and inducing factors for alternative differentiation. The decreased amyloid pathology may account for the reduction of p-tau-containing neuropil threads and for the rescue of impaired recognition and spatial memory in the transgenic mice. This study provides further insights into the mechanisms for the beneficial effect of rosiglitazone in AD patients.


Assuntos
Doença de Alzheimer/complicações , Hipoglicemiantes/uso terapêutico , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Tiazolidinedionas/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Antígeno CD11b/metabolismo , Antígenos CD36/metabolismo , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Testes Neuropsicológicos , Fragmentos de Peptídeos/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Rosiglitazona , Proteínas tau/metabolismo
8.
J Alzheimers Dis ; 17(4): 773-86, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19542617

RESUMO

Synapse loss occurs early in Alzheimer's disease (AD) and is considered the best pathological correlate of cognitive decline. Ephrins and Eph receptors are involved in regulation of excitatory neurotransmission and play a role in cytoskeleton remodeling. We asked whether alterations in Eph receptors could underlie cognitive impairment in an AD mouse model overexpressing human amyloid-beta protein precursor (hA beta PP) with familial mutations (hA beta PP swe-ind mice). We found that EphA4 and EphB2 receptors were reduced in the hippocampus before the development of impaired object recognition and spatial memory. Similar results were obtained in another line of transgenic A beta PP mice, Tg2576. A reduction in Eph receptor levels was also found in postmortem hippocampal tissue from patients with incipient AD. At the time of onset of memory decline inhA beta PP swe-ind mice, no change in surface expression of AMPA or NMDA receptor subunits was apparent, but we found changes in Eph-receptor downstream signaling, in particular a decrease in membrane-associated phosho-cofilin levels that may cause cytoskeletal changes and disrupted synaptic activity. Consistent with this finding, Eph receptor activation in cell culture increased phosho-cofilin levels. The results suggest that alterations in Eph receptors may play a role in synaptic dysfunction in the hippocampus leading to cognitive impairment in a model of AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cognição , Hipocampo/metabolismo , Memória , Receptor EphA4/metabolismo , Receptor EphB2/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Testes Neuropsicológicos , Mudanças Depois da Morte , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reconhecimento Psicológico , Percepção Espacial , Fatores de Tempo
9.
J Neurochem ; 109(2): 360-70, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19220704

RESUMO

The protective effects of insulin-like growth factor I on the somatostatin (SRIF) system in the temporal cortex after beta-amyloid (Abeta) injury may be mediated through its N-terminal tripeptide glycine-proline-glutamate (GPE). GPE is cleaved to cyclo[Pro-Gly] (cPG), a metabolite suggested to mediate in neuroprotective actions. We evaluated the effects of GPE and cPG in the temporal cortex of Abeta25-35-treated rats on SRIF and SRIF receptor protein and mRNA levels, adenylyl cyclase activity, cell death, Abeta25-35 accumulation, cytosolic calcium levels ([Ca(2+)](c)) and the intracellular signaling mechanisms involved. GPE and cPG did not change Abeta25-35 levels, but GPE partially restored SRIF and SRIF receptor 2 protein content and mRNA levels and protected against cell death after Abeta25-35 insult, which was coincident with Akt activation and glycogen synthase kinase 3beta inhibition. In addition, GPE displaced glutamate from NMDA receptors and blocked the glutamate induced rise in cytosolic calcium in isolated rat neurons and moderately increased Ca(2+) influx per se. Our findings suggest that GPE, but not its metabolite, mimics insulin-like growth factor I effects on the SRIF system through a mechanism independent of Abeta clearance that involves modulation of calcium and glycogen synthase kinase 3beta signaling.


Assuntos
Sinalização do Cálcio/fisiologia , Quinase 3 da Glicogênio Sintase/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Oligopeptídeos/fisiologia , Somatostatina/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Células Cultivadas , Feminino , Glicogênio Sintase Quinase 3 beta , Fator de Crescimento Insulin-Like I/química , Fragmentos de Peptídeos/toxicidade , Gravidez , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia
10.
Neuropsychopharmacology ; 34(7): 1721-32, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19145227

RESUMO

Chromatin modification through histone acetylation is a molecular pathway involved in the regulation of transcription underlying memory storage. Sodium 4-phenylbutyrate (4-PBA) is a well-known histone deacetylase inhibitor, which increases gene transcription of a number of genes, and also exerts neuroprotective effects. In this study, we report that administration of 4-PBA reversed spatial learning and memory deficits in an established mouse model of Alzheimer's disease (AD) without altering beta-amyloid burden. We also observed that the phosphorylated form of tau was decreased in the AD mouse brain after 4-PBA treatment, an effect probably due to an increase in the inactive form of the glycogen synthase kinase 3beta (GSK3beta). Interestingly, we found a dramatic decrease in brain histone acetylation in the transgenic mice that may reflect an indirect transcriptional repression underlying memory impairment. The administration of 4-PBA restored brain histone acetylation levels and, as a most likely consequence, activated the transcription of synaptic plasticity markers such as the GluR1 subunit of the AMPA receptor, PSD95, and microtubule-associated protein-2. The results suggest that 4-PBA, a drug already approved for clinical use, may provide a novel approach for the treatment of AD.


Assuntos
Doença de Alzheimer/complicações , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Fenilbutiratos/uso terapêutico , Tauopatias/tratamento farmacológico , Tauopatias/etiologia , Acetilação/efeitos dos fármacos , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Modelos Animais de Doenças , Embrião de Mamíferos , Hipocampo/patologia , Histonas/metabolismo , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo
11.
Neurobiol Dis ; 33(3): 369-78, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19101630

RESUMO

Transgenic mice expressing mutant human amyloid precursor protein (APP) develop an age-dependent amyloid pathology and memory deficits, but no overt neuronal loss. Here, in mice overexpressing wild-type human APP (hAPP(wt)) we found an early memory impairment, particularly in the water maze and to a lesser extent in the object recognition task, but beta-amyloid peptide (Abeta(42)) was barely detectable in the hippocampus. In these mice, hAPP processing was basically non-amyloidogenic, with high levels of APP carboxy-terminal fragments, C83 and APP intracellular domain. A tau pathology with an early increase in the levels of phosphorylated tau in the hippocampus, a likely consequence of enhanced ERK1/2 activation, was also observed. Furthermore, these mice presented a loss of synapse-associated proteins: PSD95, AMPA and NMDA receptor subunits and phosphorylated CaMKII. Importantly, signs of neurodegeneration were found in the hippocampal CA1 subfield and in the entorhinal cortex that were associated to a marked loss of MAP2 immunoreactivity. Conversely, in mice expressing mutant hAPP, high levels of Abeta(42) were found in the hippocampus, but no signs of neurodegeneration were apparent. The results support the notion of Abeta-independent pathogenic pathways in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cognição/fisiologia , Hipocampo/metabolismo , Hipocampo/patologia , Aprendizagem em Labirinto , Memória , Fragmentos de Peptídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Reconhecimento Psicológico , Precursor de Proteína beta-Amiloide/genética , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Degeneração Neural/genética , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Fosforilação , Nexinas de Proteases , Receptores de AMPA/metabolismo , Receptores de Superfície Celular/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/fisiologia , Proteínas tau/metabolismo
12.
Biochem Biophys Res Commun ; 379(2): 406-10, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19109927

RESUMO

Clinical trials with rosiglitazone, a potent agonist at peroxisome proliferator-activated receptor gamma (PPARgamma) suggest an improvement of cognitive function in Alzheimer's disease (AD) patients. The mechanisms mediating this potential beneficial effect remain to be fully elucidated. In mice overexpressing mutant human amyloid precursor protein (hAPP), a model of AD, we found that memory impairment in the object recognition test was prevented and also reversed by chronic rosiglitazone treatment. Given the possible involvement of glucocorticoid receptors (GR) in the actions of PPARgamma-ligands, we studied the effect of chronic rosiglitazone treatment on GR levels in the hippocampus of hAPP mice. An early down-regulation of GR, not related to elevated plasma corticosterone levels, was found in different hippocampal subfields of the transgenic mice and this decrease was prevented by rosiglitazone. In parallel with behavioural studies, rosiglitazone also normalized GR levels in older animals. This effect may contribute to explain the attenuation of memory decline by PPARgamma activation in an AD mouse model.


Assuntos
Doença de Alzheimer/complicações , Hipocampo/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , PPAR gama/agonistas , Receptores de Glucocorticoides/metabolismo , Tiazolidinedionas/uso terapêutico , Administração Oral , Doença de Alzheimer/sangue , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Regulação para Baixo , Hipocampo/metabolismo , Humanos , Transtornos da Memória/sangue , Transtornos da Memória/genética , Camundongos , Camundongos Transgênicos , PPAR gama/metabolismo , Receptores de Glucocorticoides/sangue , Rosiglitazona , Tiazolidinedionas/administração & dosagem
13.
Int J Dev Neurosci ; 26(6): 541-50, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18573330

RESUMO

The involvement of vascular wall in response to neuronal death was challenged here using a transient forebrain ischemia model in gerbil, which causes CA1 neuronal death and trigger neurogenesis in hippocampus. We found an important vascular reaction in CA1 5 days after ischemia evaluated by Von Willebrand factor and Vimentin immunoreactivity, as well as increased expression of angiogenic and neurogenic regulators: Vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF-2). Analysing the morphology and cell phenotype by confocal microscopy, we confirmed the colocalization of the neurogenic markers (bromodeoxyuridine-neuronal nuclei-TOPRO-3) in newborn cells associated to vascular walls in CA1 and dentate gyrus of hippocampus 32 days after ischemia. The results indicate that vascular tissues may participate in neurogenesis after brain ischemia, reinforce the notion that blood vessels represent a source of neuronal progenitor cells in damaged brain areas and suggest that molecular and cellular manipulation of the vascular wall may expand the possibilities of novel regenerative therapies.


Assuntos
Células-Tronco Adultas/fisiologia , Proliferação de Células , Artérias Cerebrais/patologia , Hipocampo/patologia , Ataque Isquêmico Transitório/patologia , Neurônios/fisiologia , Animais , Antígenos/metabolismo , Bromodesoxiuridina/metabolismo , Morte Celular , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/metabolismo , Gerbillinae , Ataque Isquêmico Transitório/fisiopatologia , Masculino , Microscopia Confocal , Proteínas do Tecido Nervoso/metabolismo , Quinolinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vimentina/metabolismo , Fator de von Willebrand/imunologia
14.
Neurosci Res ; 61(1): 27-37, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18329120

RESUMO

We studied hippocampal cellular proliferation and neurogenesis processes in a model of transient global cerebral ischemia in gerbils by labelling dividing cells with 5'-Bromo-2'-deoxyuridine (BrdU). Surrounding the region of selective neuronal death (CA1 pyramidal layer of the hippocampus), an important increase in reactive astrocytes and BrdU-labelled cells was detected 5 days after ischemia. A similar result was found in the dentate gyrus (DG) 12 days after ischemia. The differentiation of the BrdU+ cells was investigated 28 days after BrdU administration by analyzing the morphology, anatomic localization and cell phenotype by triple fluorescent labelling (BrdU, adult neural marker NeuN and DNA marker TOPRO-3) using confocal laser-scanning microscopy. This analysis showed increased neurogenesis in the DG in case of ischemia and triple positive labelling in some newborn cells in CA1. Seven brain hemispheres from gerbils subjected to ischemia did not develop CA1 neuronal death; hippocampus from these hemispheres did not show any of the above mentioned findings. Our results indicate that ischemia triggers proliferation in CA1 and neurogenesis in the DG in response to CA1 pyramidal neuronal death, independently of the reduced cerebral blood flow or the cell migration from subventricular zone (SVZ).


Assuntos
Hipocampo/irrigação sanguínea , Hipocampo/patologia , Ataque Isquêmico Transitório/patologia , Neurônios/fisiologia , Animais , Antimetabólitos , Bromodesoxiuridina , Morte Celular/fisiologia , Proliferação de Células , Artérias Cerebrais/patologia , Imunofluorescência , Gerbillinae , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Imuno-Histoquímica , Masculino , Microscopia Confocal , Células Piramidais/fisiologia , Fixação de Tecidos
15.
Brain Res ; 1199: 159-66, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18269931

RESUMO

It is known that the activation of 5-hydroxytryptamine receptor type 1A (5HT(1A) receptor) may protect against brain damage induced by transient global ischemia. The biochemical mechanisms that underlie this neuroprotective effect remain however to be fully elucidated. Given that serotonergic drugs may regulate N-methyl-d-aspartate (NMDA) receptor function, which is implicated in events leading to ischemia-induced neuronal cell death, and also stimulate the expression of brain-derived neurotrophic factor (BDNF), which is down-regulated in cerebral ischemia, we sought to determine the effects of the selective 5-HT1A receptor agonist, 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT), on the levels of NMDA receptor NR1 subunit and BDNF in gerbil hippocampus after transient global cerebral ischemia. Pretreatment with 8-OH-DPAT (1 mg/kg) prevented the neuronal loss in CA1 subfield 72 h after ischemia and also the dramatic decrease in BDNF immunoreactivity observed in this area at an earlier time. NMDA receptor NR1 levels in whole hippocampus were not affected 24 h after ischemia, but the levels of the subunit phosphorylated at the protein kinase A (PKA) site, pNR1(Ser897), were significantly increased, and this increase was prevented by the same 8-OH-DPAT dose, a probable consequence of the increased phosphatase 1 (PP1) enzyme activity found in ischemic gerbils pretreated with the 5-HT(1A) receptor agonist. The results indicate that both NR1 subunit phosphorylation and the neurotrophin BDNF account, at least in part, for the neuroprotective effect of 8-OH-DPAT on cell damage induced by global ischemia in the gerbil hippocampus and support the potential interest of 5-HT1A receptor activation in the search for neuroprotective strategies.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/patologia , Ataque Isquêmico Transitório/patologia , Neurônios/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralina/administração & dosagem , Análise de Variância , Animais , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Esquema de Medicação , Regulação da Expressão Gênica/efeitos dos fármacos , Gerbillinae , Ataque Isquêmico Transitório/prevenção & controle , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fosfopiruvato Hidratase/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Agonistas do Receptor de Serotonina/administração & dosagem , Fatores de Tempo
16.
Bioorg Med Chem Lett ; 18(6): 2078-82, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18262786

RESUMO

A series of constrained pentapeptide analogues of the fragment Abeta(31-35) has been prepared using solid phase synthesis protocols. The results of conformational studies and surface plasmon resonance (SPR) experiments seem to indicate that the affinity of these constrained analogues for immobilized Abeta(25-35) peptide could be related to their ability to adopt a Leu34N-Ile31O beta-turn-like folded conformation.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/síntese química , Peptídeos beta-Amiloides/farmacologia , Animais , Western Blotting , Sobrevivência Celular , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Imobilização , Estrutura Molecular , Neurônios/citologia , Neurônios/metabolismo , Estrutura Secundária de Proteína , Ratos , Ratos Wistar , Ressonância de Plasmônio de Superfície
17.
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
19.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA