Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 161: 105548, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34752923

RESUMO

TDP-43 pathology is a hallmark of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal lobar degeneration (FTLD). Namely, both diseases feature aggregated and phosphorylated TDP-43 containing inclusions in the cytoplasm and a loss of nuclear TDP-43 in affected neurons. It has been reported that tau tubulin kinase (TTBK)1/2 phosphorylate TDP-43 and TTBK1/2 overexpression induced neuronal loss and behavioral deficits in a C. elegans model of ALS. Here we aimed to elucidate the molecular mechanisms of TTBK1 in TDP-43 pathology. TTBK1 levels were observed to be elevated in ALS patients' post-mortem motor cortex. Also, TTBK1 was found to phosphorylate TDP-43 at disease-relevant sites in vitro directly, and this phosphorylation accelerated TDP-43 formation of high molecular species. Overexpression of TTBK1 in mammalian cells induced TDP-43 phosphorylation and the construction of high molecular species, concurrent with TDP-43 mis-localization and cytoplasmic inclusions. In addition, when TTBK1 was knocked down or pharmacologically inhibited, TDP-43 phosphorylation and aggregation were significantly alleviated. Functionally, TTBK1 knockdown could rescue TDP-43 overexpression-induced neurite and neuronal loss in iPSC-derived GABAergic neurons. These findings suggest that phosphorylation plays a critical role in the pathogenesis of TDP-43 pathology and that TTBK1 inhibition may have therapeutic potential for the treatment of ALS and FTLD.


Assuntos
Esclerose Lateral Amiotrófica , Degeneração Lobar Frontotemporal , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Caenorhabditis elegans , Proteínas de Ligação a DNA/genética , Degeneração Lobar Frontotemporal/genética , Degeneração Lobar Frontotemporal/patologia , Humanos , Mamíferos , Proteínas Serina-Treonina Quinases/genética
2.
Sci Rep ; 11(1): 3438, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33564035

RESUMO

ApoEε4 is a major genetic risk factor for Alzheimer's disease (AD), a disease hallmarked by extracellular amyloid-beta (Aß) plaques and intracellular neurofibrillary tangles (NFTs). The presence of the ApoEε4 allele is associated with increased Aß deposition and a role for ApoEε4 in the potentiation of tau pathology has recently emerged. This study focused on comparing the effects of adeno-associated virus (AAV)-mediated overexpression of the three predominant human ApoE isoforms within astrocytes. The isoform-specific effects of human ApoE were evaluated within in vitro models of tau pathology within neuron/astrocyte co-cultures, as well as in a transgenic tau mouse model. Tau aggregation, accumulation, and phosphorylation were measured to determine if the three isoforms of human ApoE had differential effects on tau. Astrocytic overexpression of the human ApoEε4 allele increased phosphorylation and misfolding of overexpressed neuronal tau in multiple models, including the aggregation and accumulation of added tau oligomers, in an isoform-specific manner. The ability of ApoEε4 to increase tau aggregation could be inhibited by an ApoEε4-specific antibody. This study indicates that astrocytic expression of ApoEε4 can potentiate tau aggregation and phosphorylation within neurons and supports a gain of toxic function hypothesis for the effect of hApoEε4 on tau.


Assuntos
Alelos , Doença de Alzheimer/metabolismo , Apolipoproteína E4/biossíntese , Astrócitos/metabolismo , Regulação da Expressão Gênica , Agregados Proteicos , Proteínas tau , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/genética , Animais , Apolipoproteína E4/genética , Astrócitos/patologia , Modelos Animais de Doenças , Ratos , Ratos Sprague-Dawley , Proteínas tau/genética , Proteínas tau/metabolismo
3.
ACS Chem Biol ; 12(2): 519-527, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28032990

RESUMO

N-methyl-d-aspartate receptors (NMDARs) mediate glutamatergic signaling that is critical to cognitive processes in the central nervous system, and NMDAR hypofunction is thought to contribute to cognitive impairment observed in both schizophrenia and Alzheimer's disease. One approach to enhance the function of NMDAR is to increase the concentration of an NMDAR coagonist, such as glycine or d-serine, in the synaptic cleft. Inhibition of alanine-serine-cysteine transporter-1 (Asc-1), the primary transporter of d-serine, is attractive because the transporter is localized to neurons in brain regions critical to cognitive function, including the hippocampus and cortical layers III and IV, and is colocalized with d-serine and NMDARs. To identify novel Asc-1 inhibitors, two different screening approaches were performed with whole-cell amino acid uptake in heterologous cells stably expressing human Asc-1: (1) a high-throughput screen (HTS) of 3 M compounds measuring 35S l-cysteine uptake into cells attached to scintillation proximity assay beads in a 1536 well format and (2) an iterative focused screen (IFS) of a 45 000 compound diversity set using a 3H d-serine uptake assay with a liquid scintillation plate reader in a 384 well format. Critically important for both screening approaches was the implementation of counter screens to remove nonspecific inhibitors of radioactive amino acid uptake. Furthermore, a 15 000 compound expansion step incorporating both on- and off-target data into chemical and biological fingerprint-based models for selection of additional hits enabled the identification of novel Asc-1-selective chemical matter from the IFS that was not identified in the full-collection HTS.


Assuntos
Sistema y+ de Transporte de Aminoácidos/antagonistas & inibidores , Ensaios de Triagem em Larga Escala , Animais , Teorema de Bayes , Células CHO , Cricetinae , Cricetulus , Humanos , Aprendizado de Máquina
4.
J Neurosci ; 35(42): 14234-50, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26490863

RESUMO

Neuronal inclusions of hyperphosphorylated and aggregated tau protein are a pathological hallmark of several neurodegenerative tauopathies, including Alzheimer's disease (AD). The hypothesis of tau transmission in AD has emerged from histopathological studies of the spatial and temporal progression of tau pathology in postmortem patient brains. Increasing evidence in cellular and animal models supports the phenomenon of intercellular spreading of tau. However, the molecular and cellular mechanisms of pathogenic tau transmission remain unknown. The studies described herein investigate tau pathology propagation using human neurons derived from induced pluripotent stem cells. Neurons were seeded with full-length human tau monomers and oligomers and chronic effects on neuronal viability and function were examined over time. Tau oligomer-treated neurons exhibited an increase in aggregated and phosphorylated pathological tau. These effects were associated with neurite retraction, loss of synapses, aberrant calcium homeostasis, and imbalanced neurotransmitter release. In contrast, tau monomer treatment did not produce any measureable changes. This work supports the hypothesis that tau oligomers are toxic species that can drive the spread of tau pathology and neurodegeneration. SIGNIFICANCE STATEMENT: Several independent studies have implicated tau protein as central to Alzheimer's disease progression and cell-to-cell pathology propagation. In this study, we investigated the ability of different tau species to propagate pathology in human neurons derived from induced pluripotent stem cells, which to date has not been shown. We demonstrated that tau oligomers, but not monomers, induce accumulation of pathological, hyperphosphorylated tau. This effect was accompanied with neurite degeneration, loss of synapses, aberrant calcium homeostasis, imbalanced neurotransmitter release, and ultimately with neuronal death. This study bridges various tau pathological phenotypes into a single and relevant induced pluripotent stem cell neuronal model of human disease that can be applied to the discovery of the mechanisms of tau-induced neurodegeneration.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Proteínas tau/metabolismo , Proteínas tau/toxicidade , Análise de Variância , Cálcio/metabolismo , Sobrevivência Celular , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Microfluídica , Microscopia de Força Atômica , Neurotransmissores/metabolismo , Fosforilação , Transporte Proteico/fisiologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Proteínas tau/química
5.
Neuropharmacology ; 82: 161-73, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23291536

RESUMO

Previous work has suggested that activation of mGlu5 receptor augments NMDA receptor function and thereby may constitute a rational approach addressing glutamate hypofunction in schizophrenia and a target for novel antipsychotic drug development. Here, we report the in vitro activity, in vivo efficacy and safety profile of 5PAM523 (4-Fluorophenyl){(2R,5S)-5-[5-(5-fluoropyridin-2-yl)-1,2,4-oxadiazol-3-yl]-2-methylpiperidin-1-yl}methanone), a structurally novel positive allosteric modulator selective of mGlu5. In cells expressing human mGlu5 receptor, 5PAM523 potentiated threshold responses to glutamate in fluorometric calcium assays, but does not have any intrinsic agonist activity. 5PAM523 acts as an allosteric modulator as suggested by the binding studies showing that 5PAM523 did not displace the binding of the orthosteric ligand quisqualic acid, but did partially compete with the negative allosteric modulator, MPyEP. In vivo, 5PAM523 reversed amphetamine-induced locomotor activity in rats. Therefore, both the in vitro and in vivo data demonstrate that 5PAM523 acts as a selective mGlu5 PAM and exhibits anti-psychotic like activity. To study the potential for adverse effects and particularly neurotoxicity, brain histopathological exams were performed in rats treated for 4 days with 5PAM523 or vehicle. The brain exam revealed moderate to severe neuronal necrosis in the rats treated with the doses of 30 and 50 mg/kg, particularly in the auditory cortex and hippocampus. To investigate whether this neurotoxicity is mechanism specific to 5PAM523, similar safety studies were carried out with three other structurally distinct selective mGlu5 PAMs. Results revealed a comparable pattern of neuronal cell death. Finally, 5PAM523 was tested in mGlu5 knock-out (KO) and wild type (WT) mice. mGlu5 WT mice treated with 5PAM523 for 4 days at 100 mg/kg presented significant neuronal death in the auditory cortex and hippocampus. Conversely, mGlu5 KO mice did not show any neuronal loss by histopathology, suggesting that enhancement of mGlu5 function is responsible for the toxicity of 5PAM523. This study reveals for the first time that augmentation of mGlu5 function with selective allosteric modulators results in neurotoxicity.


Assuntos
Antipsicóticos/toxicidade , Benzamidas/toxicidade , Encéfalo/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Fármacos Atuantes sobre Aminoácidos Excitatórios/toxicidade , Oxidiazóis/toxicidade , Receptor de Glutamato Metabotrópico 5/metabolismo , Regulação Alostérica , Animais , Antipsicóticos/química , Antipsicóticos/farmacocinética , Benzamidas/química , Benzamidas/farmacocinética , Encéfalo/patologia , Encéfalo/fisiopatologia , Células CHO , Morte Celular/fisiologia , Células Cultivadas , Cricetulus , Fármacos Atuantes sobre Aminoácidos Excitatórios/química , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacocinética , Feminino , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Necrose/patologia , Necrose/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/fisiologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/fisiopatologia , Oxidiazóis/química , Oxidiazóis/farmacocinética , Ratos Sprague-Dawley , Ratos Wistar , Receptor de Glutamato Metabotrópico 5/genética
6.
Neuropharmacology ; 62(3): 1453-60, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21112344

RESUMO

The glutamatergic hypofunction hypothesis of schizophrenia has led to the development of novel therapeutic strategies modulating NMDA receptor function. One of these strategies targets the activation of the metabotropic glutamate receptor 5 (mGlu5 receptor) using positive allosteric modulators (PAMs). Our goal was to evaluate the potential for repeated administration of the mGlu5 receptor PAM, CDPPB (3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide) (30 mg/kg) to induce tolerance to the anti-psychotic like effect using the amphetamine-induced hyperlocomotion rat model, and to produce receptor desensitization in mGlu5 receptor-enriched brain regions. CDPPB dose dependently reduced the locomotor response to amphetamine when administered acutely, and the same effect was observed following 7-day pre-treatment regime. In addition, 7-day dosing of CDPPB did not affect mGlu5 receptor density in the striatum, nor did it change mGlu5 receptor PAM-induced phosphorylation of NMDA, GluN1 and GluN2b, receptor subunits in striatum compared to the levels measured acutely. In contrast, in the frontal cortex, repeated administration of CDPPB decreased mGlu5 receptor density and resulted in a loss of its ability to increase GluN1 and GluN2b levels. Consistent with a reduction of cortical mGlu5 receptor density and phosphorylation, CDPPB (30 mg/kg) significantly affected sleep architecture as determined by cortical EEG at day one however by the seventh day of dosing all sleep changes were absent. Together these results suggest that the development of tolerance induced by the repeated treatment with the mGlu5 receptor PAM, CDPPB, may depend not only on the system being measured (sleep architecture vs psychostimulant induced hyperactivity), but also on the brain region involved with frontal cortex being a more susceptible region to receptor desensitization and internalization than striatum.


Assuntos
Benzamidas/administração & dosagem , Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Pirazóis/administração & dosagem , Receptores de Glutamato Metabotrópico/fisiologia , Regulação Alostérica/efeitos dos fármacos , Animais , Córtex Cerebral/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Masculino , Agitação Psicomotora/fisiopatologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor de Glutamato Metabotrópico 5 , Sono/efeitos dos fármacos
7.
J Neurochem ; 118(6): 1016-31, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21736568

RESUMO

The EphA4 receptor and its ephrin ligands are involved in astrocytic gliosis following CNS injury. Therefore, a strategy aimed at the blockade of EphA4 signaling could have broad therapeutic interest in brain disorders. We have identified novel small molecule inhibitors of EphA4 kinase in specific enzymatic and cell-based assays. In addition, we have demonstrated in two in vitro models of scratch injury that EphA4 receptor kinase is activated through phosphorylation and is involved in the repopulation of the wound after the scratch. A potent EphA4 kinase inhibitor significantly inhibited wound closure and reduced the accumulation of the reactive astrocytes inside the scratch. We have also shown that after the transient focal cerebral ischemia in rats, a large glial scar is formed by the accumulation of astrocytes and chondroitin sulfate proteoglycan surrounding the infarcted tissue at 7 days and 14 days of reperfusion. EphA4 protein expression is highly up-regulated in the same areas at these time points, supporting its potential role in the glial scar formation and maintenance. Taken together, these results suggest that EphA4 kinase inhibitors might interfere with the astrogliosis reaction and thereby lead to improved neurological outcome after ischemic injury.


Assuntos
Gliose/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Receptor EphA4/antagonistas & inibidores , Ferimentos e Lesões/patologia , Animais , Astrócitos/patologia , Western Blotting , Células CHO , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Gliose/patologia , Humanos , Imuno-Histoquímica , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley , Bibliotecas de Moléculas Pequenas , Cicatrização/efeitos dos fármacos
8.
Neuropharmacology ; 57(5-6): 531-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19627999

RESUMO

In the search for strategies to treat schizophrenia, attention has focused on enhancing NMDA receptor function. In vitro experiments show that metabotropic glutamate 5 receptor (mGluR5) activation enhances NMDA receptor activity, and in vivo experiments indicate that mGluR5 positive allosteric modulators (PAMs) are effective in preclinical assays measuring antipsychotic potential and cognition. Here we characterized the dose-effect function of CDPPB (3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide), an mGluR5 PAM, on novel object recognition memory in unimpaired Wistar Hannover rats (0, 10 or 30 mg/kg CDPPB) and animals with an MK-801-induced deficit (0, 3, 10, or 30 mg/kg CDPPB). In each experiment compound was given 30 min prior to the first exposure in order to affect acquisition/consolidation of the memory. In both cases, an inverted-U-shaped dose-effect function was observed, with lower doses improving recognition but higher doses having no effect. We then examined the effects of CDPPB (0, 3, 10, or 30 mg/kg) on markers of synaptic plasticity in prefrontal cortex and hippocampus, focusing on the expression and phosphorylation status of proteins involved in NMDA related signaling, including the NMDA receptor subunits NR1 and NR2B, the AMPA receptor subunit GluR1, alphaCa((2+))/CaM dependent Ser-Thr kinases II (alphaCaMKII), and the transcription factor CREB. Expression and phosphorylation of many of these proteins, particularly in the prefrontal cortex, were also characterized by an inverted-U-shaped dose-effect function. Taken together, these findings show that mGluR5 activation enhances NMDA receptor function and markers of neuronal plasticity commensurate with improvements in recognition memory. However, the effects of CDPPB are heavily dependent on dose, with higher doses being ineffective in improving recognition memory and producing downstream effects consistent with heightened NMDA receptor activation. These findings may have important implications for the development of mGluR5 PAMs to treat schizophrenia.


Assuntos
Benzamidas/farmacologia , Encéfalo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Pirazóis/farmacologia , Receptores de AMPA/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Animais , Benzamidas/administração & dosagem , Encéfalo/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fármacos do Sistema Nervoso Central/administração & dosagem , Maleato de Dizocilpina , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Fosforilação/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Pirazóis/administração & dosagem , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Reconhecimento Psicológico/fisiologia
9.
Mol Pharmacol ; 66(2): 204-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15266010

RESUMO

Pharmacological studies suggest a role for CB1 cannabinoid receptors (CB1R) in regulating neurogenesis in the adult brain. To investigate this possibility, we measured neurogenesis by intraperitoneal injection of bromodeoxyuridine (BrdU), which labels newborn neurons, in wild-type and CB1R-knockout (CB1R-KO) mice. CB1R-KO mice showed reductions in the number of BrdU-labeled cells to approximately 50% of wild-type (WT) levels in dentate gyrus and subventricular zone (SVZ), suggesting that CB1R activation promotes neurogenesis. To test this further, WT mice were given the CB1R antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboximide hydrochloride (SR141716A) before measuring neurogenesis with BrdU. SR141716A paradoxically increased the number of BrdU-labeled cells by approximately 50% in SVZ; another CB1R antagonist, 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide (AM251), had a similar effect. To investigate this discrepancy, SR141716A was given to CB1R-KO mice, in which it still stimulated neurogenesis, indicating involvement of a non-CB1 receptor. Action at one such non-CB1, SR141716A-sensitive site, the VR1 vanilloid receptor, was tested by administering SR141716A to VR1-KO mice, in which the ability of SR141716A to enhance neurogenesis was abolished. Thus, CB1 and VR1 receptors both seem to have roles in regulating adult neurogenesis.


Assuntos
Receptor CB1 de Canabinoide/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Receptor CB1 de Canabinoide/deficiência , Receptor CB1 de Canabinoide/genética
10.
J Neurosci ; 23(21): 7767-75, 2003 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-12944505

RESUMO

Focal cerebral ischemia (FCI) induces rapid neuronal death in the ischemic core, which gradually expands toward the penumbra, partly as the result of a neuroinflammatory response. It is known that propagation of neuroinflammation involves microglial cells, the resident macrophages of the brain, which are highly motile when activated by specific signals. However, the signals that increase microglial cell motility in response to FCI remain mostly elusive. Here, we tested the hypothesis that endocannabinoids mediate neuroinflammation propagation by increasing microglial cell motility. We found that, in mouse cerebral cortex, FCI greatly increases palmitoylethanolamide (PEA), only moderately increases anandamide [arachidonylethanolamide (AEA)], and does not affect 2-arachidonoylglycerol levels. We also found that PEA potentiates AEA-induced microglial cell migration, without affecting other steps of microglial activation, such as proliferation, particle engulfment, and nitric oxide production. This potentiation of microglial cell migration by PEA involves reduction in cAMP levels. In line with this, we provide evidence that PEA acts through Gi/o-coupled receptors. Interestingly, these receptors engaged by PEA are pharmacologically distinct from CB1 and CB2 cannabinoid receptors, as well as from the WIN and abn-CBD (abnormal-cannabidiol) receptors, two recently identified cannabinoid receptors. Our results show that PEA and AEA increase after FCI and synergistically enhance microglial cell motility. Because such a response could participate in the propagation of the FCI-induced neuroinflammation within the CNS, and because PEA is likely to act through its own receptor, a better understanding of the receptor engaged by PEA may help guide the search for improved therapies against neuroinflammation.


Assuntos
Isquemia Encefálica/metabolismo , Movimento Celular , Microglia/fisiologia , Ácidos Palmíticos/metabolismo , Ácidos Palmíticos/farmacologia , Amidas , Animais , Ácidos Araquidônicos/metabolismo , Ácidos Araquidônicos/farmacologia , Moduladores de Receptores de Canabinoides , Canabinoides/metabolismo , Canabinoides/farmacologia , Divisão Celular , Linhagem Celular , Córtex Cerebral/metabolismo , Endocanabinoides , Etanolaminas , Ácidos Graxos Insaturados/farmacologia , Glicerídeos/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Óxido Nítrico/biossíntese , Fagocitose , Alcamidas Poli-Insaturadas , Receptores de Canabinoides , Receptores de Droga/metabolismo
11.
J Neurosci ; 22(22): 9771-5, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12427832

RESUMO

Endogenous cannabinoid signaling pathways have been implicated in protection of the brain from hypoxia, ischemia, and trauma, but the mechanism for these protective effects is uncertain. We found that in CB1 cannabinoid receptor knock-out mice, mortality from permanent focal cerebral ischemia was increased, infarct size and neurological deficits after transient focal cerebral ischemia were more severe, cerebral blood flow in the ischemic penumbra during reperfusion was reduced, and NMDA neurotoxicity was increased compared with wild-type littermates. These findings indicate that endogenous cannabinoid signaling pathways protect mice from ischemic stroke by a mechanism that involves CB1 receptors, and suggest that both blood vessels and neurons may be targets of this protective effect.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/patologia , Receptores de Droga/deficiência , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Velocidade do Fluxo Sanguíneo/genética , Pressão Sanguínea , Encéfalo/efeitos dos fármacos , Circulação Cerebrovascular/genética , Modelos Animais de Doenças , Genótipo , Heterozigoto , Homozigoto , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , N-Metilaspartato/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Receptores de Canabinoides , Receptores de Droga/genética , Reperfusão , Taxa de Sobrevida
12.
Mol Pharmacol ; 62(4): 828-35, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12237329

RESUMO

To identify novel genes involved in cannabinoid receptor-mediated signaling, we used cDNA microarrays to detect changes in mRNA expression in the forebrains of mice 12 h after they were given a single intraperitoneal dose of the naturally-occurring Cannabis sativa alkaloid Delta(9)-tetrahydrocannabinol (Delta(9)-THC) or the synthetic cannabinoid receptor agonist (R)-(+)-2,3-dihydro-5-methyl-3-[(morpholinyl)methyl] pyrrolo[1,2,3-de]-1,4-benzoxazin-yl-1-naphtalenylmethanone mesylate [R(+)-WIN 55,212-2]. Of approximately 11,000 genes from a mouse brain cDNA library that were probed, 65 showed altered (increased or decreased at least 2-fold) expression after exposure to Delta(9)-THC, 41 after exposure to R(+)-WIN 55,212-2, and 20 genes after exposure to both drugs. Genes affected similarly by Delta(9)-THC and R(+)-WIN 55,212-2 were considered likely to reflect cannabinoid receptor activation, and expression of the protein products of two such genes not previously implicated in cannabinoid signaling-melanocyte-specific gene-related gene 1 (MRG1) and hexokinase 4 (glucokinase, GK)-was measured by Western blotting and immunohistochemistry. Western blots showed approximately 2-fold increases in the levels of both proteins in mouse forebrain. Immunohistochemistry revealed preferential localization of MRG1 to cerebral blood vessels and of GK to hypothalamic neurons. These findings suggest that MRG1 and GK are cannabinoid-regulated genes and that they may be involved in the vascular and hypothalamic effects of cannabinoids, respectively.


Assuntos
Encéfalo/fisiologia , Canabinoides/metabolismo , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Glucoquinase/genética , Proteínas Repressoras , Transdução de Sinais/fisiologia , Transativadores/genética , Analgésicos/farmacologia , Animais , Benzoxazinas , Encéfalo/efeitos dos fármacos , Dronabinol/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Alucinógenos/farmacologia , Masculino , Camundongos , Morfolinas/farmacologia , Naftalenos/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos
13.
J Neurochem ; 81(2): 355-64, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12064483

RESUMO

Geldanamycin (GA), a benzoquinone ansamycin, binds Hsp90 in vitro, releases heat shock factor (HSF1) and induces heat shock proteins (Hsps). Because viral and transgenic overexpression of Hsps protects cells against ischemia in vitro, we hypothesized that GA would protect brain from focal ischemia by inducing Hsps in vivo. Adult male Sprague-Dawley rats were subjected to 2-hour middle cerebral artery occlusions (MCAO) using the suture technique followed by 22-h reperfusions. GA or vehicle was injected into the lateral cerebral ventricles (i.c.v) 24 h before ischemia. Geldanamycin at 1 microg/kg decreased infarct volumes by 55.7% (p < 0.01) and TUNEL-positive cells by 30% in cerebral cortex. GA also improved behavioral outcomes (p < 0.01) and reduced brain edema (p < 0.05). Western blots showed that the 1 microg/kg GA dose induced Hsp70 and Hsp25 protein 8.2-fold and 2.7-fold, respectively, by 48 h following administration. Immunocytochemistry showed that GA induced Hsp70 in neurons and Hsp25 in glia and arteries in cortex, hippocampus, hypothalamus, and other brain regions. GA reduced co-immunoprecipitation of HSF1 with Hsp90 in brain tissue homogenates, promoted HSE-binding of HSF in brain nuclear extracts using gel shift assays, and increased luciferase reporter gene transcription for the Hsp70 promoter in PC12 cells. The data show that geldanamycin protects brain from focal ischemia and that this may be due, at least in part, to geldanamycin stimulation of heat shock gene transcription.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Quinonas/farmacologia , Animais , Benzoquinonas , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico HSP70/metabolismo , Fatores de Transcrição de Choque Térmico , Imuno-Histoquímica , Injeções Intraventriculares , Lactamas Macrocíclicas , Masculino , Proteínas de Neoplasias/metabolismo , Ligação Proteica/efeitos dos fármacos , Ratos , Fatores de Transcrição , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...