Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biomed Microdevices ; 20(1): 2, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29159519

RESUMO

Integration of microelectronics with microfluidics enables sophisticated lab-on-a-chip devices for sensing and actuation. In this paper, we investigate a novel method for in-situ microfluidics fabrication and packaging on wafer level. Two novel photo-patternable adhesive polymers were tested and compared, PA-S500H and DXL-009. The microfluidics fabrication method employs photo lithographical patterning of spin coated polymer films of PA or DXL and direct bonding of formed microfluidics to a top glass cover using die-to-wafer level bonding. These new adhesive materials remove the need for additional gluing layers. With this approach, we fabricated disposable microfluidic flow cytometers and evaluated the performance of those materials in the context of this application. DXL-009 exhibits lower autofluorescence compared to PA-S500H which improves detection sensitivity of fluorescently stained cells. Results obtained from the cytotoxicity test reveals that both materials are biocompatible. The functionality of these materials was demonstrated by detection of immunostained monocytes in microfluidic flow cytometers. The flexible, fully CMOS compatible fabrication process of these photo-patternable adhesive materials will simplify prototyping and mass manufacturing of sophisticated microfluidic devices with integrated microelectronics.


Assuntos
Adesivos/química , Citometria de Fluxo/instrumentação , Dispositivos Lab-On-A-Chip , Animais , Fibroblastos , Citometria de Fluxo/métodos , Humanos , Teste de Materiais , Camundongos , Polímeros/química , Razão Sinal-Ruído
2.
Neuron ; 37(6): 911-24, 2003 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-12670421

RESUMO

Parkin, an E3 ubiquitin ligase that degrades proteins with aberrant conformations, is associated with autosomal recessive juvenile Parkinsonism (AR-JP). The molecular basis of selective neuronal death in AR-JP is unknown. Here we show in an organismal system that panneuronal expression of Parkin substrate Pael-R causes age-dependent selective degeneration of Drosophila dopaminergic (DA) neurons. Coexpression of Parkin degrades Pael-R and suppresses its toxicity, whereas interfering with endogenous Drosophila Parkin function promotes Pael-R accumulation and augments its toxicity. Furthermore, overexpression of Parkin can mitigate alpha-Synuclein-induced neuritic pathology and suppress its toxicity. Our study implicates Parkin as a central player in the molecular pathway of Parkinson's disease (PD) and suggests that manipulating Parkin expression may provide a novel avenue of PD therapy.


Assuntos
Dopamina/fisiologia , Proteínas de Drosophila/toxicidade , Drosophila , Ligases/fisiologia , Neurônios/fisiologia , Ubiquitina-Proteína Ligases , Envelhecimento , Animais , Animais Geneticamente Modificados , Western Blotting , Encéfalo/metabolismo , Morte Celular , Células Cultivadas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Expressão Gênica , Imuno-Histoquímica , Ligases/genética , Proteínas do Tecido Nervoso/toxicidade , Doença de Parkinson , Interferência de RNA , Receptores de Endotelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinucleínas , alfa-Sinucleína
3.
J Neurosci ; 26(20): 5383-92, 2006 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-16707790

RESUMO

Necdin, a member of the MAGE (melanoma antigen) protein family, is expressed predominantly in terminally differentiated neurons. The necdin gene NDN is maternally imprinted and expressed only from the paternal allele, the deficiency of which is implicated in the pathogenesis of the neurodevelopmental disorder Prader-Willi syndrome. Necdin binds to its homologous MAGE protein MAGE-D1 (also known as NRAGE or Dlxin-1), which interacts with Msx (msh homeobox) and Dlx (distal-less homeobox) family homeodomain transcription factors. Members of the Dlx homeobox gene family are involved in the differentiation and specification of forebrain GABAergic neurons. Here we demonstrate that necdin associates with Dlx homeodomain proteins via MAGE-D1 to promote the differentiation of GABAergic neurons in mouse embryonic forebrain. Immunohistochemical analysis revealed that necdin was coexpressed with Dlx2, Dlx5, or MAGE-D1 in a subpopulation of embryonic forebrain cells. Necdin bound to Dlx2 and Dlx5 via MAGE-D1 and enhanced Dlx2-dependent activation of the Wnt1 (wingless-type MMTV integration site family) promoter. Necdin significantly increased the populations of cells expressing the GABAergic neuron markers calbindin D-28k and glutamic acid decarboxylase when overexpressed by electroporation in cultured forebrain slices. In this assay, Dlx5N, a truncated Dlx5 mutant that competes with Dlx2 to bind MAGE-D1, diminished the effect of necdin on GABAergic neuron differentiation. Furthermore, mutant mice lacking the paternal necdin allele showed a significant reduction in the differentiation of forebrain GABAergic neurons in vivo and in vitro. These results suggest that paternally expressed necdin facilitates the differentiation and specification of GABAergic neurons in cooperation with Dlx homeodomain proteins.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Prosencéfalo/embriologia , Fatores de Transcrição/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Calbindinas , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , Glutamato Descarboxilase/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Mutação/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Técnicas de Cultura de Órgãos , Regiões Promotoras Genéticas/genética , Prosencéfalo/metabolismo , Ligação Proteica/fisiologia , Proteína G de Ligação ao Cálcio S100 , Fatores de Transcrição/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
4.
J Neurosci ; 26(46): 12003-13, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17108174

RESUMO

The cell cycle-regulatory transcription factor E2F1 induces apoptosis of postmitotic neurons in developmental and pathological situations. E2F1 transcriptionally activates many proapoptotic genes including the cyclin-dependent protein kinase cell division cycle 2 (Cdc2). Necdin is a potent mitotic suppressor expressed predominantly in postmitotic neurons and interacts with E2F1 to suppress E2F1-mediated gene transcription. The necdin gene NDN is maternally imprinted and expressed only from the paternal allele. Deletion of the paternal NDN is implicated in the pathogenesis of Prader-Willi syndrome, a genomic imprinting-associated neurodevelopmental disorder. Here, we show that paternally expressed necdin represses E2F1-dependent cdc2 gene transcription and attenuates apoptosis of postmitotic neurons. Necdin was abundantly expressed in differentiated cerebellar granule neurons (CGNs). Neuronal activity deprivation elevated the expression of both E2F1 and Cdc2 in primary CGNs prepared from mice at postnatal day 6, whereas the necdin levels remained unchanged. In chromatin immunoprecipitation analysis, endogenous necdin was associated with the cdc2 promoter containing an E2F-binding site in activity-deprived CGNs. After activity deprivation, CGNs underwent apoptosis, which was augmented in those prepared from mice defective in the paternal Ndn allele (Ndn(+m/-p)). The levels of cdc2 mRNA, protein, and kinase activity were significantly higher in Ndn(+m/-p) CGNs than in wild-type CGNs under activity-deprived conditions. Furthermore, the populations of Cdc2-immunoreactive and apoptotic cells were increased in the cerebellum in vivo of Ndn(+m/-p) mice. These results suggest that endogenous necdin attenuates neuronal apoptosis by suppressing the E2F1-Cdc2 system.


Assuntos
Apoptose/genética , Proteína Quinase CDC2/biossíntese , Córtex Cerebelar/metabolismo , Regulação para Baixo/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Animais , Animais Recém-Nascidos , Sítios de Ligação/genética , Proteína Quinase CDC2/genética , Contagem de Células , Células Cultivadas , Córtex Cerebelar/citologia , Córtex Cerebelar/crescimento & desenvolvimento , Fator de Transcrição E2F1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/metabolismo , Elementos Reguladores de Transcrição/genética
5.
Gene Expr Patterns ; 7(3): 244-51, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17084677

RESUMO

The MAGE (melanoma antigen) family is characterized by a large conserved domain termed MAGE homology domain. Originally identified MAGE genes encoding tumor rejection antigens are expressed only in cancers and male germ cells. Necdin, which contains the MAGE homology domain, is highly expressed in postmitotic cells such as neurons and skeletal muscle cells. The human necdin gene NDN is transcribed only from the paternal allele through genomic imprinting, and its deficiency is implicated in the pathogenesis of the neurodevelopmental disorder Prader-Willi syndrome. Although over 30 MAGE genes have been identified in humans, fruit fly (Drosophila melanogaster) has only a single MAGE gene that encodes a protein similar to necdin homologous MAGE proteins. In this study, we analyzed the spatiotemporal expression patterns of MAGE mRNA and the encoded protein during fly development. Whole-mount embryo in situ hybridization analysis revealed that MAGE mRNA was highly expressed at the syncytial blastoderm stage and in the ventral and procephalic neurogenic regions of the ectoderm during gastrulation. In contrast, MAGE expression was nearly undetectable in postmitotic neurons of the central nervous system at late embryonic stages. During postembryonic neurogenesis, MAGE was highly expressed in neural stem cells (neuroblasts) and their progeny (ganglion mother cells and postmitotic neurons) at larval and pupal stages. MAGE was also expressed in postmitotic neurons including mushroom body neurons and retinal photoreceptors in adulthood. These results indicate that MAGE expression lasts throughout the postembryonic neurogenesis in Drosophila.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/genética , Masculino , Sistema Nervoso/crescimento & desenvolvimento , Neurônios/metabolismo , Proteínas Recombinantes/genética
6.
Sci Rep ; 7(1): 12731, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28986553

RESUMO

Psychological stress induces many diseases including post-traumatic stress disorder (PTSD); however, the causal relationship between stress and brain atrophy has not been clarified. Applying single-prolonged stress (SPS) to explore the global effect of severe stress, we performed brain magnetic resonance imaging (MRI) acquisition and Voxel-based morphometry (VBM). Significant atrophy was detected in the bilateral thalamus and right visual cortex. Fluorescent immunohistochemistry for Iba-1 as the marker of activated microglia indicates regional microglial activation as stress-reaction in these atrophic areas. These data certify the impact of severe psychological stress on the atrophy of the visual cortex and the thalamus. Unexpectedly, these results are similar to chronic neuropathic pain rather than PTSD clinical research. We believe that some sensitisation mechanism from severe stress-induced atrophy in the visual cortex and thalamus, and the functional defect of the visual system may be a potential therapeutic target for stress-related diseases.


Assuntos
Estresse Psicológico/patologia , Tálamo/patologia , Córtex Visual/patologia , Animais , Atrofia , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Masculino , Proteínas dos Microfilamentos/metabolismo , Microglia/metabolismo , Ratos Sprague-Dawley
7.
J Neurosci ; 25(30): 7090-9, 2005 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-16049186

RESUMO

Necdin is a multifunctional signaling protein that stabilizes terminal differentiation of postmitotic neurons. The human necdin gene in chromosome 15q11-q12 is maternally imprinted, paternally transcribed, and not expressed in Prader-Willi syndrome, a human genomic imprinting-associated neurodevelopmental disorder. Although necdin-deficient mice display several abnormal phenotypes reminiscent of this syndrome, little is known about molecular mechanisms that lead to the neurodevelopmental defects. Here, we demonstrate that paternally expressed necdin is required for physiological development of nerve growth factor (NGF)-dependent sensory neurons. Mouse embryos defective in the paternal necdin allele displayed absent necdin expression in the dorsal root ganglia, in which the tropomyosin-related kinase A (TrkA) receptor tyrosine kinase and the p75 neurotrophin receptor were expressed in a normal manner. Necdin interacted with both TrkA and p75 to facilitate the association between these receptors. NGF-induced phosphorylation of TrkA and mitogen-activated protein kinase was significantly diminished in the necdin-null sensory ganglia. Furthermore, the mice lacking the paternal necdin allele displayed augmented apoptosis in the sensory ganglia in vivo and had a reduced population of substance P-containing neurons. These mutant mice showed significantly high tolerance to thermal pain, which is often seen in individuals with Prader-Willi syndrome. These results suggest that paternally expressed necdin facilitates TrkA signaling to promote the survival of NGF-dependent nociceptive neurons.


Assuntos
Impressão Genômica/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios Aferentes/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptor trkA/metabolismo , Animais , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Pai , Feminino , Gânglios Espinais/citologia , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Neurônios Aferentes/fisiologia , Nociceptores/citologia , Nociceptores/fisiologia , Células PC12 , Síndrome de Prader-Willi/genética , Gravidez , Ratos , Receptor de Fator de Crescimento Neural/metabolismo , Transdução de Sinais/fisiologia
8.
Brain Res Mol Brain Res ; 107(2): 166-75, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12425945

RESUMO

We have previously reported that overexpression of wild-type amyloid precursor protein (APP) in postmitotic neurons induces cleavage-dependent activation of caspase-3 both in vivo and in vitro. In this study, we investigated the mechanism underlying APP-induced caspase-3 activation using adenovirus-mediated gene transfer into postmitotic neurons derived from human embryonal carcinoma NT2 cells. Overexpression of wild-type APP significantly increased intracellular (45)Ca(2+) content prior to the activation of caspase-3 in NT2-derived neurons. Chelation of intracellular Ca(2+) markedly suppressed APP-induced activation of caspase-3. Furthermore, calpain, a Ca(2+)-dependent cysteine protease, was activated in neurons overexpressing APP as assessed by increased levels of calpain-cleaved alpha-fodrin and autolytic mu-calpain fragments. Neither calpain nor caspase-3 was activated in neurons expressing an APP mutant defective in the Abeta(1-20) domain. Calpain inhibitors almost completely suppressed APP-induced activation of neuronal caspase-3. E64d, a membrane permeable inhibitor of calpain, significantly suppressed APP-induced neuronal death. These results suggest that overexpression of wild-type APP activates calpain that mediates caspase-3 activation in postmitotic neurons.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Calpaína/metabolismo , Caspases/metabolismo , Morte Celular/fisiologia , Neurônios/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Encéfalo/fisiopatologia , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Calpaína/antagonistas & inibidores , Caspase 3 , Quelantes/farmacologia , Inibidores Enzimáticos/farmacologia , Expressão Gênica/fisiologia , Vetores Genéticos , Humanos , Imuno-Histoquímica , Ionóforos/farmacologia , Mutação/fisiologia , Transfecção , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
9.
Brain Nerve ; 64(2): 163-73, 2012 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-22308261

RESUMO

In this paper, we have outlined the neurobiological basis of alcohol and drug dependence. The prevalence of drug dependence is a serious social problem in many countries, including Japan. This problem involves many background factors, including those pertaining to medical sciences, socio economics, and politics. First, we briefly describe the findings pertaining to psychotomimetic drugs as a model of schizophrenia. The biological pathogenesis of schizophrenic disorders is still unknown. The symptoms of methamphetamine (MAP) and phencyclidine (PCP) psychoses are very similar to those of schizophrenic disorders involving hallucination or delusion. PCP causes not only positive symptoms but also negative symptoms. Therefore, it has been considered as a more comprehensive model of schizophrenia than other drugs. Furthermore, amotivational syndrome, which is observed in patients with chronic cannabis and organic solvent dependence, is similar to the negative symptoms of schizophrenia. Understanding the neurobiological basis of drug dependence by using the molecular biological approach will provide an important clue for elucidating the mechanisms underlying schizophrenia and endogenous psychiatric disorders. Next, we discuss account for the neurobiological mechanisms underlying drug dependence. The reward system in the brain, which is common for all dependent drugs, has been explained, and the stages of addiction corresponding to the development of drug dependence have been discussed followed. In addition, we have discussed the epigenetics aspects of substance dependence, which is one of the hottest topics in psychiatric genetics. We expect that further studies of the mechanisms underlying drug dependence will aid in elucidating of the pathophysiology of various psychiatric diseases.


Assuntos
Alcoolismo/genética , Transtornos Relacionados ao Uso de Substâncias/genética , Alcoolismo/fisiopatologia , Humanos , Esquizofrenia/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia
11.
Proc Natl Acad Sci U S A ; 102(38): 13670-5, 2005 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-16155123

RESUMO

Parkinson's disease (PD) is the most common movement disorder characterized by dopaminergic dysfunction and degeneration. The cause of most PD cases is unknown, although postmortem studies have implicated the involvement of oxidative stress. The identification of familial PD-associated genes offers the opportunity to study mechanisms of PD pathogenesis in model organisms. Here, we show that DJ-1A, a Drosophila homologue of the familial PD-associated gene DJ-1, plays an essential role in oxidative stress response and neuronal maintenance. Inhibition of DJ-1A function through RNA interference (RNAi) results in cellular accumulation of reactive oxygen species, organismal hypersensitivity to oxidative stress, and dysfunction and degeneration of dopaminergic and photoreceptor neurons. To identify other genes that may interact with DJ-1A in regulating cell survival, we performed genetic interaction studies and identified components of the phosphatidylinositol 3-kinase (PI3K)/Akt-signaling pathway as specific modulators of DJ-1A RNAi-induced neurodegeneration. PI3K signaling suppresses DJ-1A RNAi phenotypes at least in part by reducing cellular reactive oxygen species levels. Consistent with the genetic interaction results, we also found reduced phosphorylation of Akt in DJ-1A RNAi animals, indicating an impairment of PI3K/Akt signaling by DJ-1A down-regulation. Together with recent findings in mammalian systems, these results implicate impairments of PI3K/Akt signaling and oxidative stress response in DJ-1-associated disease pathogenesis. We also observed impairment of PI3K/Akt signaling in the fly parkin model of PD, hinting at a common molecular event in the pathogenesis of PD. Manipulation of PI3K/Akt signaling may therefore offer therapeutic benefits for the treatment of PD.


Assuntos
Proteínas de Drosophila/metabolismo , Estresse Oxidativo/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Drosophila , Proteínas de Drosophila/genética , Ativação Enzimática/genética , Humanos , Estresse Oxidativo/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteínas Proto-Oncogênicas c-akt , Interferência de RNA/fisiologia , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética
12.
Exp Cell Res ; 277(2): 220-32, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12083804

RESUMO

Necdin is expressed predominantly in postmitotic neurons and serves as a growth suppressor that is functionally similar to the retinoblastoma tumor suppressor protein. Using primary cultures of dorsal root ganglion (DRG) of mouse embryos, we investigated the involvement of necdin in the terminal differentiation of neurons. DRG cells were prepared from mouse embryos at 12.5 days of gestation and cultured in the presence of nerve growth factor (NGF). Immunocytochemistry revealed that necdin accumulated in the nucleus of differentiated neurons that showed neurite extension and expressed the neuronal markers microtubule-associated protein 2 and synaptophysin. Suppression of necdin expression in DRG cultures treated with antisense oligonucleotides led to a marked reduction in the number of terminally differentiated neurons. The antisense oligonucleotide-treated cells did not attempt to reenter the cell cycle, but underwent death with characteristics of apoptosis such as caspase-3 activation, nuclear condensation, and chromosomal DNA fragmentation. Furthermore, a caspase-3 inhibitor rescued antisense oligonucleotide-treated cells from apoptosis and significantly increased the population of terminally differentiated neurons. These results suggest that necdin mediates the terminal differentiation and survival of NGF-dependent DRG neurons and that necdin-deficient nascent neurons are destined to caspase-3-dependent apoptosis.


Assuntos
Gânglios Espinais/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas Nucleares/metabolismo , Animais , Apoptose , Caspase 3 , Caspases/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Sobrevivência Celular , Células Cultivadas , Gânglios Espinais/embriologia , Camundongos , Camundongos Endogâmicos ICR , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Nucleares/genética , Oligodesoxirribonucleotídeos Antissenso
13.
Cell ; 116(5): 671-82, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-15006350

RESUMO

Multisite hyperphosphorylation of tau has been implicated in the pathogenesis of neurodegenerative diseases including Alzheimer's disease (AD). However, the phosphorylation events critical for tau toxicity and mechanisms regulating these events are largely unknown. Here we show that Drosophila PAR-1 kinase initiates tau toxicity by triggering a temporally ordered phosphorylation process. PAR-1 directly phosphorylates tau at S262 and S356. This phosphorylation event is a prerequisite for the action of downstream kinases, including glycogen synthase kinase 3 (GSK-3) and cyclin-dependent kinase-5 (Cdk5), to phosphorylate several other sites and generate disease-associated phospho-epitopes. The initiator role of PAR-1 is further underscored by the fact that mutating PAR-1 phosphorylation sites causes a much greater reduction of overall tau phosphorylation and toxicity than mutating S202, one of the downstream sites whose phosphorylation depends on prior PAR-1 action. These findings begin to differentiate the effects of various phosphorylation events on tau toxicity and provide potential therapeutic targets.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Proteínas Quinases/metabolismo , Proteínas tau/metabolismo , Proteínas tau/toxicidade , Animais , Animais Geneticamente Modificados , Quinase 5 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Mutagênese Sítio-Dirigida , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Fenótipo , Fosforilação , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/ultraestrutura , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Transdução de Sinais/fisiologia , Proteínas tau/genética
14.
J Biol Chem ; 279(39): 40484-93, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15272023

RESUMO

Necdin is a potent growth suppressor that is expressed predominantly in postmitotic cells such as neurons and skeletal muscle cells. Necdin shows a significant homology to MAGE (melanoma antigen) family proteins, all of which contain a large homology domain. MAGE-D1 (NRAGE, Dlxin-1) interacts with the Dlx/Msx family homeodomain proteins via an interspersed hexapeptide repeat domain distinct from the homology domain. Here we report that necdin associates with the Msx homeodomain proteins via MAGE-D1 to modulate their function. In vitro binding and co-immunoprecipitation analyses revealed that MAGE-D1 directly interacted with necdin via the homology domain and Msx1 (or Msx2) via the repeat domain. A ternary complex of necdin, MAGE-D1, and Msx2 was formed in vitro, and an endogenous complex containing these three proteins was detected in differentiating embryonal carcinoma cells. Co-expression of necdin and MAGE-D1 released Msx-dependent transcriptional repression. C2C12 myoblast cells that were stably transfected with Msx2 cDNA showed a marked reduction in myogenic differentiation, and co-expression of necdin and MAGE-D1 canceled the Msx2-dependent repression. These results suggest that necdin and MAGE-D1 cooperate to modulate the function of Dlx/Msx homeodomain proteins in cellular differentiation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Animais , Western Blotting , Células COS , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , DNA Complementar/metabolismo , Deleção de Genes , Vetores Genéticos , Proteínas de Homeodomínio , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Microscopia de Fluorescência , Mitose , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Neurônios/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Distribuição Tecidual , Transcrição Gênica
15.
Exp Cell Res ; 286(2): 241-51, 2003 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12749853

RESUMO

The amyloid precursor protein (APP) is a transmembrane protein whose abnormal processing is associated with the pathogenesis of Alzheimer's disease. In this study, we examined the expression and role of cell-associated APP in primary dorsal root ganglion (DRG) neurons. When dissociated DRG cells prepared from mouse embryos were treated with nerve growth factor (NGF), neuronal APP levels were transiently elevated. DRG neurons treated with an antibody against cell surface APP failed to mature and underwent apoptosis. When NGF was withdrawn from the cultures after a 36-h NGF treatment, virtually all neurons underwent apoptosis by 48 h. During the course of apoptosis, some neurons with intact morphology contained increased levels of APP immunoreactivity, whereas the APP levels were greatly reduced in apoptotic neurons. Furthermore, affected neurons contained immunoreactivities for activated caspase-3, a caspase-cleaved APP fragment (APPDeltaC31), and Abeta. Downregulation of endogenous APP expression by treatment with an APP antisense oligodeoxynucleotide significantly increased the number of apoptotic neurons in NGF-deprived DRG cultures. Furthermore, overexpression of APP by adenovirus vector-mediated gene transfer reduced the number of apoptotic neurons deprived of NGF. These results suggest that endogenous APP is upregulated to exert an antiapoptotic effect on neurotrophin-deprived DRG neurons and subsequently undergoes caspase-dependent proteolysis.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Apoptose/genética , Sobrevivência Celular/genética , Gânglios Espinais/metabolismo , Neurônios Aferentes/metabolismo , Regulação para Cima/genética , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/agonistas , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Animais , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3 , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feto , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Fator de Crescimento Neural/farmacologia , Neurônios Aferentes/citologia , Neurônios Aferentes/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA