Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Psychiatry ; 22(11): 1562-1575, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-27646263

RESUMO

Neuronal network dysfunction and cognitive decline constitute the most prominent features of Alzheimer's disease (AD), although mechanisms causing such impairments are yet to be determined. Here we report that virus-mediated delivery of the active spliced transcription factor X-Box binding protein 1s (XBP1s) in the hippocampus rescued spine density, synaptic plasticity and memory function in a mouse model of AD. XBP1s transcriptionally activated Kalirin-7 (Kal7), a protein that controls synaptic plasticity. In addition, we found reduced levels of Kal7 in primary neurons exposed to Aß oligomers, transgenic mouse models and human AD brains. Short hairpin RNA-mediated knockdown of Kal7 altered synaptic plasticity and memory formation in naive mice. Further, reduction of endogenous Kal7 compromised the beneficial effects of XBP1s in Alzheimer's model. Hence, our findings reveal that XBP1s is neuroprotective through a mechanism that engages Kal7 pathway with therapeutic implications in AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Animais , Células CHO , Cricetulus , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Plasticidade Neuronal , Neurônios/metabolismo , Cultura Primária de Células , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box/genética , Adulto Jovem
2.
J Cell Biol ; 141(1): 187-97, 1998 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-9531558

RESUMO

Members of the Rho GTPase family regulate the organization of the actin cytoskeleton in response to extracellular growth factors. We have identified three proteins that form a distinct branch of the Rho family: Rnd1, expressed mostly in brain and liver; Rnd2, highly expressed in testis; and Rnd3/RhoE, showing a ubiquitous low expression. At the subcellular level, Rnd1 is concentrated at adherens junctions both in confluent fibroblasts and in epithelial cells. Rnd1 has a low affinity for GDP and spontaneously exchanges nucleotide rapidly in a physiological buffer. Furthermore, Rnd1 lacks intrinsic GTPase activity suggesting that in vivo, it might be constitutively in a GTP-bound form. Expression of Rnd1 or Rnd3/RhoE in fibroblasts inhibits the formation of actin stress fibers, membrane ruffles, and integrin-based focal adhesions and induces loss of cell-substrate adhesion leading to cell rounding (hence Rnd for "round"). We suggest that these proteins control rearrangements of the actin cytoskeleton and changes in cell adhesion.


Assuntos
Actinas/fisiologia , Encéfalo/metabolismo , Adesão Celular/fisiologia , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP , Células 3T3 , Sequência de Aminoácidos , Animais , Bovinos , Mapeamento Cromossômico , Cromossomos Humanos , Cromossomos Humanos Par 12 , Cromossomos Humanos Par 17 , DNA Complementar , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Ligação ao GTP/biossíntese , Proteínas de Ligação ao GTP/genética , Biblioteca Gênica , Humanos , Hibridização In Situ , Cinética , Fígado/metabolismo , Linfócitos/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Família Multigênica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Testículo/metabolismo
3.
Curr Alzheimer Res ; 16(5): 453-457, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30907322

RESUMO

BACKGROUND: Alzheimer's disease (AD) is associated with extracellular accumulation and aggregation of amyloid ß (Aß) peptides ultimately seeding in senile plaques. Recent data show that their direct precursor C99 (ßCTF) also accumulates in AD-affected brain as well as in AD-like mouse models. C99 is consistently detected much earlier than Aß, suggesting that this metabolite could be an early contributor to AD pathology. C99 accumulates principally within endolysosomal and autophagic structures and its accumulation was described as both a consequence and one of the causes of endolysosomalautophagic pathology, the occurrence of which has been documented as an early defect in AD. C99 was also accompanied by C99-derived C83 (αCTF) accumulation occurring within the same intracellular organelles. Both these CTFs were found to dimerize leading to the generation of higher molecular weight CTFs, which were immunohistochemically characterized in situ by means of aggregate-specific antibodies. DISCUSSION: Here, we discuss studies demonstrating a direct link between the accumulation of C99 and C99-derived APP-CTFs and early neurotoxicity. We discuss the role of C99 in endosomal-lysosomalautophagic dysfunction, neuroinflammation, early brain network alterations and synaptic dysfunction as well as in memory-related behavioral alterations, in triple transgenic mice as well as in newly developed AD animal models. CONCLUSION: This review summarizes current evidence suggesting a potential role of the ß -secretasederived APP C-terminal fragment C99 in Alzheimer's disease etiology.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Doença de Alzheimer/metabolismo , Animais , Humanos
4.
Circ Res ; 89(4): 336-42, 2001 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-11509450

RESUMO

Many members of the two-pore-domain potassium (K(+)) channel family have been detected in the mammalian heart but the endogenous correlates of these channels still have to be identified. We investigated whether I(KAA), a background K(+) current activated by negative pressure (stretch) and by arachidonic acid (AA) and sensitive to intracellular acidification, could be the native correlate of TREK-1 in adult rat atrial cells. Using the inside-out configuration of the patch-clamp technique, we found that I(KAA), like TREK-1, was outwardly rectifying in physiological K(+) conditions, with a conductance of 41 pS at +50 mV. Like TREK-1, I(KAA) was reversibly activated by clinical concentrations of volatile anesthetics (in mmol/L, chloroform 0.18, halothane 0.11, and isoflurane 0.69). In cell-attached experiments, I(KAA) was inhibited by chlorophenylthio-cAMP (500 micromol/L) and also by stimulation of beta-adrenergic receptors with isoproterenol (1 micromol/L). In addition, TREK-1 mRNAs were detected in all cardiac tissues, and the TREK-1 protein was immunolocalized in isolated atrial myocytes. Such a background potassium channel might contribute to the positive inotropic effects produced by beta-adrenergic stimulation of the heart. It might also be involved in the regulation of the atrial natriuretic peptide secretion.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Anestésicos Inalatórios/farmacologia , Átrios do Coração/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem , Canais de Potássio/efeitos dos fármacos , Animais , Ácido Araquidônico/farmacologia , Separação Celular , Clorofórmio/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Halotano/farmacologia , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Isoflurano/farmacologia , Isoproterenol/farmacologia , Masculino , Miocárdio/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Potássio/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Estimulação Química
5.
Curr Alzheimer Res ; 13(8): 942-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27040141

RESUMO

Apathy is an early and common neuropsychiatric syndrome in Alzheimer's disease (AD) patients. In clinical trials, apathy is associated with decreased motor activity that can be monitored by actigraphy. The triple transgenic mouse AD model (3xTgAD) has been shown to recapitulate the biochemical lesions as well as many of the synaptic and cognitive alterations associated with AD. In the present work we found that these mice also develop an early and consistent apathy-like behavior as evidenced by a drastic decrease in spontaneous activity measured by actimetry. We recently established that these mice also display an intraneuronal accumulation of the ß-secretase-derived ßAPP fragment (C99) appearing early, in absence of Aß. Interestingly, we found that the apathy-like behavior observed in 3xTgAD mice was temporally associated with C99 accumulation and synaptic alterations. Since it is well known that the genetic background can strongly influence behavior and can induce transcriptional variability in animal models, we decided to determine the influence of genetic background on the above-described alterations. We backcrossed 3xTgAD mice to C57BL/6 and found that the genetic background had no influence on either C99 accumulation or synaptic plasticity alterations, but strongly affected the apathy-like behavior.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Apatia , Camundongos Transgênicos/genética , Atividade Motora/genética , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Envelhecimento/psicologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Patrimônio Genético , Humanos , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Especificidade da Espécie , Sinapses/metabolismo , Sinapses/patologia , Proteínas tau/genética , Proteínas tau/metabolismo
6.
FEBS Lett ; 438(3): 171-6, 1998 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-9827540

RESUMO

Benign familial neonatal convulsions, an autosomal dominant epilepsy of newborns, are linked to mutations affecting two six-transmembrane potassium channels, KCNQ2 and KCNQ3. We isolated four splice variants of KCNQ2 in human brain. Two forms generate, after transient expression in COS cells, a potassium-selective current similar to the KCNQ1 current. L-735,821, a benzodiazepine molecule which inhibits the KCNQ1 channel activity (EC50 = 0.08 microM), also blocks KCNQ2 currents (EC50 = 1.5 microM). Using in situ hybridization, KCNQ2 and KCNQ3 have been localized within the central nervous system, in which they are expressed in the same areas, mainly in the hippocampus, the neocortex and the cerebellar cortex. During brain development, KCNQ3 is expressed later than KCNQ2.


Assuntos
Envelhecimento/metabolismo , Processamento Alternativo , Encéfalo/metabolismo , Regulação da Expressão Gênica , Variação Genética , Canais de Potássio/genética , Transcrição Gênica , Sequência de Aminoácidos , Animais , Benzodiazepinas/farmacologia , Encéfalo/crescimento & desenvolvimento , Células COS , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Canal de Potássio KCNQ2 , Canal de Potássio KCNQ3 , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Canais de Potássio/análise , Canais de Potássio/química , Canais de Potássio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , RNA Mensageiro/análise , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Transfecção
7.
FEBS Lett ; 480(2-3): 137-41, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11034315

RESUMO

KCNQ2 and KCNQ3 subunits belong to the six transmembrane domain K+ channel family and loss of function mutations are associated with benign familial neonatal convulsions. KCNE2 (MirP1) is a single transmembrane domain subunit first described to be a modulator of the HERG potassium channel in the heart. Here, we show that KCNE2 is present in brain, in areas which also express KCNQ2 and KCNQ3 channels. We demonstrate that KCNE2 associates with KCNQ2 and/or KCNQ3 subunits. In transiently transfected COS cells, KCNE2 expression produces an acceleration of deactivation kinetics of KCNQ2 and of the KCNQ2-KCNQ3 complex. Effects of two previously identified arrhythmogenic mutations of KCNE2 have also been analyzed.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células COS , Eletrofisiologia , Humanos , Hibridização In Situ/métodos , Canal de Potássio KCNQ2 , Canal de Potássio KCNQ3 , Dados de Sequência Molecular , Canais de Potássio/genética , Canais de Potássio/fisiologia , Testes de Precipitina , Ratos , Análise de Sequência de Proteína , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
8.
FEBS Lett ; 402(1): 28-32, 1997 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-9013852

RESUMO

The two P domain K+ channel mTWIK-1 has been cloned from mouse brain. In Xenopus oocytes, mTWIK-1 currents are K+-selective, instantaneous, and weakly inward rectifying. These currents are blocked by Ba2+ and quinine, decreased by protein kinase C and increased by internal acidification. The apparent molecular weight of mTWIK-1 in brain is 81 kDa. A 40 kDa form is revealed after treatment with a reducing agent, strongly suggesting that native mTWIK-1 subunits dimerize via a disulfide bridge. TWIK-1 mRNA is expressed abundantly in brain and at lower levels in lung, kidney, and skeletal muscle. In situ hybridization shows that mTWIK-1 expression is restricted to a few brain regions, with the highest levels in cerebellar granule cells, brainstem, hippocampus and cerebral cortex.


Assuntos
Encéfalo/metabolismo , Canais de Potássio de Domínios Poros em Tandem , Canais de Potássio/química , Sequência de Aminoácidos , Animais , Bário/farmacologia , Sequência de Bases , Western Blotting , DNA Complementar/genética , Dimerização , Hibridização In Situ , Potenciais da Membrana , Camundongos , Dados de Sequência Molecular , Peso Molecular , Oócitos , Canais de Potássio/genética , Canais de Potássio/metabolismo , Quinina/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Xenopus
9.
Neuroscience ; 80(2): 345-57, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9284339

RESUMO

G-protein-gated inward rectifier potassium channels mediate the synaptic actions of numerous neurotransmitters in the mammalian brain, and were recently shown to be candidates for genetic mutations leading to neuronal cell death. This report describes the localization of G-protein-gated inward rectifier potassium channel-2 and G-protein-gated inward rectifier potassium channel-4 proteins in the rat brain, as assessed by immunocytochemistry. G-protein-gated inward rectifier potassium channel-2 immunoreactivity was widely distributed throughout the brain, with the strongest staining seen in the hippocampus, septum, granule cell layer of the cerebellum, amygdala and substantia nigra pars compacta. In contrast, G-protein-gated inward rectifier potassium channel-4 immunoreactivity was restricted to some neuronal populations, such as Purkinje cells and neurons of the globus pallidus and the ventral pallidum. The presence of G-protein-gated inward rectifier potassium channel-2 immunoreactivity in substantia nigra pars compacta dopaminergic neurons was confirmed by showing its co-localization with tyrosine hydroxylase by double immunocytochemistry, and also by selectively lesioning dopaminergic neurons with the neurotoxin 6-hydroxydopamine. At the cellular level both proteins were localized in neuronal cell bodies and dendrites, but clear differences were seen in the degree of dendritic staining among neuronal groups. For some neuronal groups the staining of distal dendrites (notably dendritic spines) was strong, while for others the cell body and proximal dendrites were preferentially labelled. In addition, some of the results suggest that G-protein-gated inward rectifier potassium channel-2 protein could be localized in distal axonal terminal fields. A knowledge of the distribution of G-protein-gated inward rectifier potassium channel proteins in the brain could help to elucidate their physiological roles and to evaluate their potential involvement in neurodegenerative processes in animal models and human diseases.


Assuntos
Química Encefálica/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Ativação do Canal Iônico/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/metabolismo , Animais , Encéfalo/citologia , Encéfalo/enzimologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Humanos , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Simpatectomia Química , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Neuroscience ; 95(3): 893-901, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10670456

RESUMO

TRAAK is the sole member of the emerging class of 2P domain K+ channels to be exclusively expressed in neuronal cells. TRAAK produces baseline K+ currents which are strongly stimulated by arachidonic acid and by mechanical stretch, and which are insensitive to the classical K+ channel blockers tetraethylammonium, Ba2+, and Cs+. This report describes the immunolocalization of TRAAK in brain, spinal cord, and retina of the adult mouse. The most striking finding is the widespread distribution of the TRAAK immunoreactivity, with a prominent staining of the cerebellar cortex, neocortex, hippocampus, dentate gyrus, subiculum, the dorsal hippocampal commissure, thalamus, caudate-putamen, olfactory bulb, and several nuclei in the brainstem. Virtually all neurons express TRAAK, and the highest immunoreactivity was seen in soma, and to a lesser degree in axons and/or dendrites in most areas in brain and spinal cord. In the retina, the TRAAK protein is concentrated to the soma of ganglion cells and to the dendrites of all other neurons. Taken together, these results show a wide distribution of TRAAK, a mechanosensitive and arachidonic acid-stimulated neuron-specific baseline K+ channel, in brain, spinal cord and retina.


Assuntos
Ácido Araquidônico/metabolismo , Encéfalo/metabolismo , Canais de Potássio/metabolismo , Retina/metabolismo , Medula Espinal/metabolismo , Animais , Tronco Encefálico/metabolismo , Linhagem Celular , Córtex Cerebelar/metabolismo , Hipocampo/metabolismo , Imuno-Histoquímica , Insetos , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Tecidual
11.
Neuroreport ; 8(4): 969-74, 1997 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-9141074

RESUMO

It has been suggested that a mutation in a G-protein-gated inward rectifier K+ channel (GIRK2) is responsible for inducing cell death in the cerebellum of homozygous weaver (wv/wv) mutant mice. These mice also display a progressive, massive loss of mesencephalic dopaminergic neurones. Using an immunocytochemical method, we detected GIRK2-positive cell bodies and fibres in the substantia nigra pars compacta (SNC) and the ventral tegmental area (VTA) of control (+/+) mice. Cell counts of both GIRK2- and tyrosine hydroxylase (TH)-positive neurones demonstrated a marked loss of SNC cell bodies, especially in 12-month-old (12M) wv/wv mice. A considerable proportion of GIRK2-positive cell bodies were preserved, however. In addition, no loss of GIRK2-positive neurones was observed in the VTA of 12M wv/wv mice, despite of a significant reduction in TH-positive cell bodies. These results suggest that expression of the mutated channel is not a sufficient condition to induce cell death in the ventral mesencephalon of the wv/wv mice.


Assuntos
Mesencéfalo/citologia , Neurônios/citologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/análise , Envelhecimento/fisiologia , Animais , Biomarcadores , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Proteínas de Ligação ao GTP/análise , Imuno-Histoquímica , Masculino , Mesencéfalo/crescimento & desenvolvimento , Camundongos , Camundongos Mutantes Neurológicos , Fibras Nervosas/ultraestrutura , Valores de Referência , Substância Negra/citologia , Substância Negra/crescimento & desenvolvimento , Tegmento Mesencefálico/citologia , Tegmento Mesencefálico/crescimento & desenvolvimento , Tirosina 3-Mono-Oxigenase/análise
12.
Brain Res ; 651(1-2): 353-6, 1994 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-7922587

RESUMO

Secretory phospholipase A2 type II is known to be involved in various inflammatory processes. This paper describes the changes in secretory phospholipase A2 (PLA2) gene expression induced by ischemia and endotoxic shock. Type I PLA2 (pancreatic type) is not expressed in ischemic and endotoxic-shock brains but both ischemia and endotoxin injection induce type II PLA2 expression. The first phase of PLA2 II gene expression following the ischemic insult occurs 1-6 h after ischemia. During that period, PLA2 II gene expression is slightly enhanced and it returns to control levels after 1 day. A second phase corresponding to higher levels of induction of mRNA for PLA2 appears at a later period after ischemia between 7 and 18 days. In situ hybridization shows that PLA2 gene expression in the ischemic brain is localized in regions known to be vulnerable to ischemia (hippocampus and neocortex). Endotoxic shock which leads to a major inflammatory state induces an abundant expression of the PLA2 II mRNA in the brain and this high level of expression appears in a large number of brain structures. The results suggest that the early phase of ischemia-induced PLA2 gene expression could be an additional element in mechanisms leading to neuronal death. The later phase of increased PLA2 mRNA levels is more probably related to the inflammatory response associated to neuronal degeneration.


Assuntos
Expressão Gênica , Isquemia/enzimologia , Fosfolipases A/genética , Prosencéfalo/irrigação sanguínea , Choque Séptico/enzimologia , Animais , Isquemia/genética , Fosfolipases A2 , RNA Mensageiro/metabolismo , Ratos , Choque Séptico/genética
13.
Brain Res ; 659(1-2): 67-74, 1994 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-7820682

RESUMO

Severe forebrain ischemia induces a large increase in expression of NMDA receptor subunits in rat brain. One week after ischemia, levels of NMDA-R1 mRNAs in the CA1 pyramidal cells of hippocampus are 7 times higher than those observed in control rats. At 7 days postischemia, an enhanced immunostaining of the NMDA-R1 subunit was observed in all hippocampal structures indicating that changes in mRNA levels are accompanied by changes in receptor protein level. Riluzole, a potent inhibitor of glutamate release and CNQX, a selective AMPA/kainate antagonist, drastically reduced the ischemia-induced expression of mRNAs for the three NMDA receptor subunits while D-AP5, a selective NMDA antagonist, had essentially no effect. Therefore ischemia-induced expression of NMDA receptor subunits is associated with glutamate release and proceeds via an AMPA/kainate pathway. These results together with those of other groups concerning ischemia effects on AMPA and GABAA receptor levels, suggest an important role of the induced expression of NMDA receptor subunits in the deleterious effects of ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Ácido Glutâmico/fisiologia , Hipocampo/metabolismo , RNA Mensageiro/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Sequência de Bases , Antagonistas de Aminoácidos Excitatórios , Masculino , Dados de Sequência Molecular , Sondas de Oligonucleotídeos/genética , Prosencéfalo/irrigação sanguínea , Ratos , Ratos Wistar , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato/classificação , Riluzol , Tiazóis/farmacologia
14.
Brain Res ; 753(1): 8-17, 1997 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-9125426

RESUMO

The main target for degeneration associated with the weaver mutation is the cerebellum. Expression of the GIRK2 mRNA and protein was studied in cerebellum of 12- and 22-day-old normal and weaver mice. In 12-day-old mice, GIRK2 is expressed at highest levels in the external granule layer (EGL) and in lower levels in the newly forming internal granule layer (IGL). In the weaver cerebellum, a high hybridization signal and dark immunostaining was observed in the EGL due to the higher density of non-migrated cells. In 22-day-old weaver cerebella, there are only few remaining granule cells existing as scattered cells within the IGL and molecular layer. GIRK2 is expressed in these neurons but the majority of cells expressing GIRK2 in these cerebella are Purkinje cells that are also affected by the weaver mutation (position, shape) but have not died. Normal cerebellar granule neurons but not homozygous mutant neurons in primary cultures and cerebellar slices of 8-day-old mice displayed inward rectifier K+ currents. Taken together, these findings suggest that cell loss in the weaver cerebellum is not directly related to a differential content of GIRK2 in the affected neurons during development. The lethal effect of the weaver mutation in specific neurons is probably due to a combination of the abnormal function of the inward rectifier K+ channels and other factors specific to the vulnerable neurons.


Assuntos
Cerebelo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/biossíntese , Animais , Células Cultivadas , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Técnicas de Patch-Clamp , Canais de Potássio/genética , RNA Mensageiro/biossíntese , Valores de Referência
15.
J Neurosci ; 18(9): 3117-23, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9547220

RESUMO

Hypoxia and inhibitors of mitochondrial respiration impair the regulatory volume decrease (RVD) of cerebellar granule neurons after hypotonic swelling. RVD is linked to the opening of volume-regulated anion channels (VRACs). VRACs are outwardly rectifying, inactivate slowly during maintained depolarization, and are permeable to the cellular organic osmolyte taurine. Channel activation requires nonhydrolytic ATP binding and is not modulated by intracellular ADP. VRAC opening is reversibly depressed by hypoxia and by mitochondrial inhibitors such as oligomycin, rotenone, and antimycin A. These results demonstrate that neuronal VRAC activation and swelling are both tightly linked to cellular energy. Moreover, the findings reported in this work may have a particular significance for inherited mitochondrial human diseases, such as mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS), which cause brain swelling and edema.


Assuntos
Hipóxia Celular/fisiologia , Respiração Celular/fisiologia , Cerebelo/fisiologia , Canais de Cloreto/fisiologia , Mitocôndrias/fisiologia , Neurônios/fisiologia , Animais , Ânions , Tamanho Celular/fisiologia , Células Cultivadas , Cerebelo/citologia , Humanos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Xenopus
16.
J Neurochem ; 69(4): 1570-9, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9326286

RESUMO

(-)-Cromakalim, a typical K+-channel opener, prevents neuronal death induced by either glucose and oxygen privation or by high (100 microM) extracellular glutamate in primary cultures of hippocampus. (-)-Cromakalim has no effect on the earliest events associated with exposure to glutamate. It does not prevent the rapid rise of intracellular Ca2+, the initial swelling of neurons, or the induction of c-fos mRNA transcription. (-)-Cromakalim inhibits all delayed effects associated with the excitotoxic effect of glutamate: (a) (-)-cromakalim inhibits the late and major phase of intracellular Ca2+ increase occurring up to hours after glutamate application; and (b) although (-)-cromakalim cannot prevent the initial cellular swelling induced by glutamate, cells that have been pretreated with (-)-cromakalim return to their original size in a few hours, whereas non-(-)-cromakalim-treated cells remain swollen for more prolonged periods. Many neurons surviving the initial necrotic phase of glutamate-induced cell death undergo progressive DNA cleavage leading to apoptosis. This apoptotic process is prevented completely by (-)-cromakalim. Glibenclamide, a potent blocker of the ATP-sensitive K+ channel, abolishes all the beneficial effects of (-)-cromakalim. These findings strongly suggest that (-)-cromakalim has postsynaptic effects that are closely related to the regulation of Ca2+ homeostasis and cell volume.


Assuntos
Cromakalim/farmacologia , Ácido Glutâmico/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cromakalim/antagonistas & inibidores , Glibureto/farmacologia , Hipocampo/citologia , Hipocampo/patologia , Necrose , Neurônios/fisiologia , Concentração Osmolar , Bloqueadores dos Canais de Potássio , Ratos , Ratos Wistar , Estereoisomerismo
17.
Proc Natl Acad Sci U S A ; 92(10): 4666-70, 1995 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-7753861

RESUMO

Preconditioning with sublethal ischemia protects against neuronal damage after subsequent lethal ischemic insults in hippocampal neurons. A pharmacological approach using agonists and antagonists at the adenosine A1 receptor as well as openers and blockers of ATP-sensitive K+ channels has been combined with an analysis of neuronal death and gene expression of subunits of glutamate and gamma-aminobutyric acid receptors, HSP70, c-fos, c-jun, and growth factors. It indicates that the mechanism of ischemic tolerance involves a cascade of events including liberation of adenosine, stimulation of adenosine A1 receptors, and, via these receptors, opening of sulfonylurea-sensitive ATP-sensitive K+ channels.


Assuntos
Adenosina/fisiologia , Hipocampo/fisiopatologia , Ataque Isquêmico Transitório/fisiopatologia , Fatores de Crescimento Neural/biossíntese , Neurônios/fisiologia , Canais de Potássio/fisiologia , Receptores Purinérgicos P1/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/fisiologia , Expressão Gênica , Proteínas de Choque Térmico HSP70/biossíntese , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Injeções Intraventriculares , Ataque Isquêmico Transitório/patologia , Substâncias Macromoleculares , Masculino , Proteínas do Tecido Nervoso/biossíntese , Neurônios/efeitos dos fármacos , Neurônios/patologia , Canais de Potássio/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-jun/biossíntese , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Receptores de GABA/biossíntese , Receptores de Glutamato/biossíntese , Fatores de Tempo
18.
Verh Dtsch Ges Pathol ; 74: 284-8, 1990.
Artigo em Alemão | MEDLINE | ID: mdl-1708604

RESUMO

Surgical specimens of 72 adrenocortical tumours were investigated by conventional histology, immunocytochemistry and DNA-cytophotometry. Histologically, 57 tumours were classified as adenomas and 15 as carcinomas. Nine adenomas weighed more, 2 carcinomas less than 50g. Only in 9 of the latter cases were distant metastases and/or lethal outcome of disease recorded, while the clinical course of the remaining patients was uneventful. No significant differences in DNA content were found between adenomas and carcinomas or between carcinomas with aggressive and indolent behaviour. Neither could immunocytochemistry discriminate between these conditions. Immunostaining with the monoclonal antibody D 11 proved to be the only effective means to definitely type adrenocortical neoplasia. Thirty-one cases exhibited positivity upon immunostaining with a polyclonal antiserum against synaptophysin. This phenomenon has so far not been encountered in non-neuroendocrine neoplasia.


Assuntos
Adenoma/patologia , Neoplasias do Córtex Suprarrenal/patologia , Carcinoma/patologia , DNA de Neoplasias/análise , Adenoma/cirurgia , Neoplasias do Córtex Suprarrenal/cirurgia , Carcinoma/cirurgia , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Ploidias
19.
EMBO J ; 19(8): 1784-93, 2000 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-10775263

RESUMO

Results reported in this work suggest a potential therapeutic value of polyunsaturated fatty acids for cerebral pathologies as previously proposed by others for cardiac diseases. We show that the polyunsaturated fatty acid linolenic acid prevents neuronal death in an animal model of transient global ischemia even when administered after the insult. Linolenic acid also protects animals treated with kainate against seizures and hippocampal lesions. The same effects have been observed in an in vitro model of seizure-like activity using glutamatergic neurons and they have been shown to be associated with blockade of glutamatergic transmission by low concentrations of distinct polyunsaturated fatty acids. Our data suggest that the opening of background K(+) channels, like TREK-1 and TRAAK, which are activated by arachidonic acid and other polyunsaturated fatty acids such as docosahexaenoic acid and linolenic acid, is a significant factor in this neuroprotective effect. These channels are abundant in the brain where they are located both pre- and post-synaptically, and are insensitive to saturated fatty acids, which offer no neuroprotection.


Assuntos
Isquemia Encefálica/prevenção & controle , Ácidos Graxos Insaturados/farmacologia , Neurônios/efeitos dos fármacos , Canais de Potássio , Convulsões/prevenção & controle , Animais , Isquemia Encefálica/patologia , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Ácidos Graxos Insaturados/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas , Ácido Caínico , Magnésio/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/metabolismo , Ácido Palmítico/farmacologia , Canais de Potássio/metabolismo , Convulsões/induzido quimicamente , Convulsões/patologia , Sinapsinas/metabolismo , Fatores de Tempo , Ácido alfa-Linolênico/farmacologia
20.
J Biol Chem ; 271(18): 10433-6, 1996 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-8631835

RESUMO

Mutations of the degenerins (deg-1, mec-4, mec-10) are the major known causes of hereditary neurodegeneration in the nematode Caenorhabditis elegans. We cloned a neuronal degenerin (MDEG) from human and rat brain. MDEG is an amiloride-sensitive cation channel permeable for Na+, K+, and Li+. This channel is activated by the same mutations which cause neurodegeneration in C. elegans. Like the hyperactive C. elegans degenerin mutants, constitutively active mutants of MDEG cause cell death, suggesting that gain of function of this novel neuronal ion channel might be involved in human forms of neurodegeneration.


Assuntos
Amilorida/farmacologia , Caenorhabditis elegans/genética , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/patologia , Canais Iônicos Sensíveis a Ácido , Sequência de Aminoácidos , Animais , Sequência de Bases , Cátions , Canais de Sódio Degenerina , Canais Epiteliais de Sódio , Humanos , Canais Iônicos/genética , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Ratos , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA