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








Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 299(8): 105038, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37442231

RESUMO

Covalent amino acid modification significantly expands protein functional capability in regulating biological processes. Tyrosine residues can undergo phosphorylation, sulfation, adenylation, halogenation, and nitration. These posttranslational modifications (PTMs) result from the actions of specific enzymes: tyrosine kinases, tyrosyl-protein sulfotransferase(s), adenylate transferase(s), oxidoreductases, peroxidases, and metal-heme containing proteins. Whereas phosphorylation, sulfation, and adenylation modify the hydroxyl group of tyrosine, tyrosine halogenation and nitration target the adjacent carbon residues. Because aberrant tyrosine nitration has been associated with human disorders and with animal models of disease, we have created an updated and curated database of 908 human nitrated proteins. We have also analyzed this new resource to provide insight into the role of tyrosine nitration in cancer biology, an area that has not previously been considered in detail. Unexpectedly, we have found that 879 of the 1971 known sites of tyrosine nitration are also sites of phosphorylation suggesting an extensive role for nitration in cell signaling. Overall, the review offers several forward-looking opportunities for future research and new perspectives for understanding the role of tyrosine nitration in cancer biology.


Assuntos
Neoplasias , Proteínas , Tirosina , Animais , Humanos , Fosforilação , Proteínas/metabolismo , Transdução de Sinais , Tirosina/metabolismo
2.
Hum Mol Genet ; 28(19): 3282-3292, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31272106

RESUMO

A pathological hallmark of spinal muscular atrophy (SMA) is severe motor neuron (MN) loss, which results in muscle weakness and often infantile or childhood mortality. Although it is well established that deficient expression of survival motor neuron (SMN) protein causes SMA, the molecular pathways that execute MN cell death are poorly defined. The c-Jun NH2-terminal kinases (JNKs) are stress-activated kinases with multiple substrates including c-Jun, which can be activated during neuronal injury and neurodegenerative disease leading to neuronal apoptosis. Recently, increased JNK-c-Jun signaling was reported in SMA raising the possibility that JNK inhibitors could be a novel treatment for this disease. We examined JNK-c-Jun activity in SMA mouse and human cultured cells and tissues. Anisomycin treatment of human SMA fibroblasts and sciatic nerve ligation in SMA mice provoked robust phosphorylated-c-Jun (p-c-Jun) expression indicating that SMN-deficiency does not prevent activation of the stress-induced JNK-c-Jun signaling pathway. Despite retained capacity to activate JNK-c-Jun, we observed no basal increase of p-c-Jun levels in SMA compared to control cultured cells, human or mouse spinal cord tissues, or mouse MNs during the period of MN loss in severe SMA model mice. In both controls and SMA, ~50% of α-MN nuclei express p-c-Jun with decreasing expression during the early postnatal period. Together these studies reveal no evidence of stress-activated JNK-c-Jun signaling in MNs of SMA mice or human tissues, but do highlight the important role of JNK-c-Jun activity during normal MN development raising caution about JNK antagonism in this pediatric neuromuscular disease.


Assuntos
Anisomicina/farmacologia , Atrofia Muscular Espinal/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Medula Espinal/citologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Fosforilação , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
3.
J Alzheimers Dis ; 54(4): 1629-1647, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27636841

RESUMO

Alzheimer's disease (AD) is characterized by neuronal death, neurofibrillary tangles, and senile plaques. Amyloid-beta (Aß) is the major component of plaques and consists of two prominent isoforms, Aß40 and Aß42. As many risk factors for AD are vascular in origin and blood vessel defects in clearing Aß from the brain are a potential key component of AD pathology, we have focused on the neuron-blood vessel interface, and in particular, the vascular basement membrane, which coats blood vessels and physically separates them from neurons. A prominent component of the vascular basement membrane is the extracellular matrix proteoglycan perlecan. Domain V (DV) is the C-terminal domain and is generated by perlecan proteolysis. DV interacts with the α2 integrin and Aß is a ligand for both α2ß1 and αvß1. Due to the known interaction of DV with α2ß1 and α2ß1's requirement for Aß deposition and neurotoxicity, we hypothesized that DV and/or its C-terminal domain, LG3, might alter neurotoxic signaling pathways by directly blocking or otherwise interfering with α2ß1 binding by Aß. Our study suggests that α2ß1 mediates Aß-induced activation of c-Jun and caspase-3, key components of the neurotoxic pathway, in primary cortical and hippocampal neurons. We further demonstrate that DV and/or LG3 may therapeutically modulate these α2ß1 mediated neurotoxic effects suggesting that they or other α2ß1 integrin modulators could represent a novel approach to treat AD. Finally, our results suggest different neurotoxicity susceptibilities between cortical and hippocampal neurons to Aß40 and Aß42 as further underscored by differing neuroprotective potencies of LG3 in each cell type.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Integrina alfa2beta1/antagonistas & inibidores , Integrina alfa2beta1/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Gravidez , Transdução de Sinais/fisiologia
4.
Neurobiol Aging ; 36(2): 693-709, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25442111

RESUMO

The interaction of amyloid-beta (Aß) and tau in the pathogenesis of Alzheimer's disease is a subject of intense inquiry, with the bulk of evidence indicating that changes in tau are downstream of Aß. It has been shown however, that human tau overexpression in amyloid precursor protein transgenic mice increases Aß plaque deposition. Here, we confirm that human tau increases Aß levels. To determine if the observed changes in Aß levels were because of intracellular or extracellular secreted tau (eTau for extracellular tau), we affinity purified secreted tau from Alzheimer's disease patient-derived cortical neuron conditioned media and analyzed it by liquid chromatography-mass spectrometry. We found the extracellular species to be composed predominantly of a series of N-terminal fragments of tau, with no evidence of C-terminal tau fragments. We characterized a subset of high affinity tau antibodies, each capable of engaging and neutralizing eTau. We found that neutralizing eTau reduces Aß levels in vitro in primary human cortical neurons where exogenously adding eTau increases Aß levels. In vivo, neutralizing human tau in 2 human tau transgenic models also reduced Aß levels. We show that the human tau insert sequence is sufficient to cause the observed increase in Aß levels. Our data furthermore suggest that neuronal hyperactivity may be the mechanism by which this regulation occurs. We show that neuronal hyperactivity regulates both eTau secretion and Aß production. Electrophysiological analysis shows for the first time that secreted eTau causes neuronal hyperactivity. Its induction of hyperactivity may be the mechanism by which eTau regulates Aß production. Together with previous findings, these data posit a novel connection between tau and Aß, suggesting a dynamic mechanism of positive feed forward regulation. Aß drives the disease pathway through tau, with eTau further increasing Aß levels, perpetuating a destructive cycle.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Neurônios/metabolismo , Proteínas tau/fisiologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Cromatografia Líquida , Humanos , Espectrometria de Massas , Camundongos Transgênicos , Neurônios/fisiologia , Proteínas tau/química , Proteínas tau/isolamento & purificação
5.
Mol Cell Neurosci ; 56: 355-64, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23891805

RESUMO

Development of therapeutics for genetically complex neurodegenerative diseases such as sporadic amyotrophic lateral sclerosis (ALS) has largely been hampered by lack of relevant disease models. Reprogramming of sporadic ALS patients' fibroblasts into induced pluripotent stem cells (iPSC) and differentiation into affected neurons that show a disease phenotype could provide a cellular model for disease mechanism studies and drug discovery. Here we report the reprogramming to pluripotency of fibroblasts from a large cohort of healthy controls and ALS patients and their differentiation into motor neurons. We demonstrate that motor neurons derived from three sALS patients show de novo TDP-43 aggregation and that the aggregates recapitulate pathology in postmortem tissue from one of the same patients from which the iPSC were derived. We configured a high-content chemical screen using the TDP-43 aggregate endpoint both in lower motor neurons and upper motor neuron like cells and identified FDA-approved small molecule modulators including Digoxin demonstrating the feasibility of patient-derived iPSC-based disease modeling for drug screening.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Reprogramação Celular , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios Motores/citologia , Esclerose Lateral Amiotrófica/metabolismo , Estudos de Casos e Controles , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia
6.
Neurobiol Aging ; 33(7): 1379-88, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21126803

RESUMO

Amyloid-ß (Aß) peptide is a key component of amyloid plaques, one of the pathological features of Alzheimer's disease. Another feature is pronounced cell loss in the brain leading to an enlargement of the ventricular area and a decrease in brain weight and volume. Aß plaque deposition and neuronal toxicity can be modeled by treating human cortical neuronal cultures with Aß and showing robust Aß deposition and neurotoxicity that is mediated by α2ß1 and αvß1 integrins. The current study expands on these findings by showing that the domain V of perlecan, a known α2 integrin ligand, inhibits Aß neurotoxicity in an α2 integrin-dependent manner. Additionally, Aß binds more efficiently to cells expressing activated α2 integrin. Finally the inhibition of Aß neurotoxicity with domain V is synergistic with inhibitors of αv integrin and ß1 integrin. We propose that domain V and potentially other α2 integrin ligands could be a new therapeutic approach for inhibiting the Aß plaque deposition and neurotoxicity observed in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Proteoglicanas de Heparan Sulfato/fisiologia , Integrina alfa2/metabolismo , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia
7.
Bioorg Med Chem Lett ; 21(18): 5521-7, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21813278
8.
Bioorg Med Chem Lett ; 21(12): 3726-9, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21570836

RESUMO

In this Letter, we describe the evolution of selective JNK3 inhibitors from 1, that routinely exhibit >10-fold selectivity over JNK1 and >1000-fold selectivity over related MAPKs. Strong SAR was found for substitution of the naphthalene ring, as well as for inhibitors adopting different central scaffolds. Significant potency gains were appreciated by inverting the polarity of the thione of the parent triazolothione 1, resulting in potent compounds with attractive pharmacokinetic profiles.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Naftalenos/síntese química , Tionas/síntese química , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Concentração Inibidora 50 , Camundongos , Microssomos Hepáticos/enzimologia , Estrutura Molecular , Naftalenos/química , Naftalenos/farmacologia , Solubilidade , Relação Estrutura-Atividade , Tionas/química , Tionas/farmacologia
9.
Bioorg Med Chem Lett ; 21(6): 1838-43, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21316234

RESUMO

The SAR of a series of tri-substituted thiophene JNK3 inhibitors is described. By optimizing both the N-aryl acetamide region of the inhibitor and the 4-position of the thiophene we obtained single digit nanomolar compounds, such as 47, which demonstrated an in vivo effect on JNK activity when dosed orally in our kainic acid mouse model as measured by phospho-c-jun reduction.


Assuntos
Encéfalo/metabolismo , MAP Quinase Quinase 4/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Administração Oral , Desenho de Fármacos , Ligação de Hidrogênio , Modelos Moleculares , Inibidores de Proteínas Quinases/síntese química , Relação Estrutura-Atividade
10.
Bioorg Med Chem Lett ; 21(1): 315-9, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21112785

RESUMO

In this Letter, we describe the discovery of selective JNK2 and JNK3 inhibitors, such as 10, that routinely exhibit >10-fold selectivity over JNK1 and >1000-fold selectivity over related MAPKs, p38α and ERK2. Substitution of the naphthalene ring affords an isoform selective JNK3 inhibitor, 30, with approximately 10-fold selectivity over both JNK1 and JNK2. A naphthalene ring penetrates deep into the selectivity pocket accounting for the differentiation amongst the kinases. Interestingly, the gatekeeper Met146 sulfide interacts with the naphthalene ring in a sulfur-π stacking interaction. Compound 38 ameliorates neurotoxicity induced by amyloid-ß in human cortical neurons. Lastly, we demonstrate how to install propitious in vitro CNS-like properties into these selective inhibitors.


Assuntos
Aminopiridinas/química , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/química , Inibidores de Proteínas Quinases/química , Triazinas/química , Aminopiridinas/farmacocinética , Aminopiridinas/uso terapêutico , Animais , Sítios de Ligação , Sistema Nervoso Central/metabolismo , Simulação por Computador , Humanos , Camundongos , Microssomos Hepáticos/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/uso terapêutico , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Relação Estrutura-Atividade , Triazinas/farmacocinética , Triazinas/uso terapêutico
11.
J Biol Chem ; 286(9): 7315-26, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21189253

RESUMO

Transcription factors of the nuclear factor 1 (NFI) family regulate normal brain development in vertebrates. However, multiple splice variants of four NFI isoforms exist, and their biological functions have yet to be elucidated. Here, we cloned and analyzed human NFI-X3, a novel splice variant of the nfix gene, which contains a unique transcriptional activation (TA) domain completely conserved in primates. In contrast to previously cloned NFI-X1, overexpression of NFI-X3 potently activates NFI reporters, including glial fibrillary acidic protein (GFAP) reporter, in astrocytes and glioma cells. The GAL4 fusion protein containing the TA domain of NFI-X3 strongly activates the GAL4 reporter, whereas the TA domain of NFI-X1 is ineffective. The expression of NFI-X3 is dramatically up-regulated during the differentiation of neural progenitors to astrocytes and precedes the expression of astrocyte markers, such as GFAP and SPARCL1 (Secreted Protein, Acidic and Rich in Cysteines-like 1). Overexpression of NFI-X3 dramatically up-regulates GFAP and SPARCL1 expression in glioma cells, whereas the knockdown of NFI-X3 diminishes the expression of both GFAP and SPARCL1 in astrocytes. Although activation of astrocyte-specific genes involves DNA demethylation and subsequent increase of histone acetylation, NFI-X3 activates GFAP expression, in part, by inducing alterations in the nucleosome architecture that lead to the increased recruitment of RNA polymerase II.


Assuntos
Processamento Alternativo/fisiologia , Astrócitos/citologia , Astrócitos/fisiologia , Fatores de Transcrição NFI/genética , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Sequência Conservada , Células-Tronco Embrionárias/citologia , Proteínas da Matriz Extracelular/genética , Fibroblastos/citologia , Marcadores Genéticos , Proteína Glial Fibrilar Ácida/genética , Glioblastoma , Células HEK293 , Humanos , Mamíferos , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição NFI/química , Fatores de Transcrição NFI/metabolismo , Regiões Promotoras Genéticas/fisiologia , Estrutura Terciária de Proteína , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Ativação Transcricional/fisiologia
12.
Bioorg Med Chem Lett ; 20(24): 7303-7, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21071223

RESUMO

From high throughput screening, we discovered compound 1, the prototype for a series of disubstituted thiophene inhibitors of JNK which is selective towards closely related MAP kinases p38 and Erk2. Herein we describe the evolution of these compounds to a novel class of thiophene and thiazole JNK inhibitors that retain favorable solubility, permeability, and P-gp properties for development as CNS agents for treatment of neurodegeneration. Compound 61 demonstrated JNK3 IC(50)=77 nM and retained the excellent broad kinase selectivity observed for the series.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Quinolinas/síntese química , Tiazóis/química , Tiofenos/química , Animais , Desenho de Fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Microssomos Hepáticos/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/química , Quinolinas/farmacologia , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/farmacologia , Tiofenos/síntese química , Tiofenos/farmacologia
13.
J Biol Chem ; 285(5): 3417-27, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-19923222

RESUMO

Immunotherapy targeting of amyloid beta (Abeta) peptide in transgenic mouse models of Alzheimer disease (AD) has been widely demonstrated to resolve amyloid deposition as well as associated neuronal, glial, and inflammatory pathologies. These successes have provided the basis for ongoing clinical trials of immunotherapy for treatment of AD in humans. Acute as well as chronic Abeta-targeted immunotherapy has also been demonstrated to reverse Abeta-related behavioral deficits assessing memory in AD transgenic mouse models. We observe that three antibodies targeting the same linear epitope of Abeta, Abeta(3-7), differ in their ability to reverse contextual fear deficits in Tg2576 mice in an acute testing paradigm. Reversal of contextual fear deficit by the antibodies does not correlate with in vitro recognition of Abeta in a consistent or correlative manner. To better define differences in antigen recognition at the atomic level, we determined crystal structures of Fab fragments in complex with Abeta. The conformation of the Abeta peptide recognized by all three antibodies was highly related and is also remarkably similar to that observed in independently reported Abeta:antibody crystal structures. Sequence and structural differences between the antibodies, particularly in CDR3 of the heavy chain variable region, are proposed to account for differing in vivo properties of the antibodies under study. These findings provide a structural basis for immunotherapeutic strategies targeting Abeta species postulated to underlie cognitive deficits in AD.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/química , Animais , Comportamento Animal , Reagentes de Ligações Cruzadas/farmacologia , Cristalografia por Raios X/métodos , Modelos Animais de Doenças , Epitopos/química , Heterozigoto , Humanos , Cinética , Masculino , Camundongos , Conformação Molecular , Proteínas Recombinantes/química
14.
Stem Cells ; 27(5): 1173-81, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19418463

RESUMO

Even though astrocytes are critical for both normal brain functions and the development and progression of neuropathological states, including neuroinflammation associated with neurodegenerative diseases, the mechanisms controlling gene expression during astrocyte differentiation are poorly understood. Thus far, several signaling pathways were shown to regulate astrocyte differentiation, including JAK-STAT, bone morphogenic protein-2/Smads, and Notch. More recently, a family of nuclear factor-1 (NFI-A, -B, -C, and -X) was implicated in the regulation of vertebral neocortex development, with NFI-A and -B controlling the onset of gliogenesis. Here, we developed an in vitro model of differentiation of stem cells towards neural progenitors (NP) and subsequently astrocytes. The transition from stem cells to progenitors was accompanied by an expected change in the expression profile of markers, including Sox-2, Musashi-1, and Oct4. Subsequently, generated astrocytes were characterized by proper morphology, increased glutamate uptake, and marker gene expression. We used this in vitro differentiation model to study the expression and functions of NFIs. Interestingly, stem cells expressed only background levels of NFIs, while differentiation to NP activated the expression of NFI-A. More importantly, NFI-X expression was induced during the later stages of differentiation towards astrocytes. In addition, NFI-X and -C were required for the expression of glial fibrillary acidic protein and secreted protein acidic and rich in cystein-like protein 1, which are the markers of astrocytes at the later stages of differentiation. We conclude that an expression program of NFIs is executed during the differentiation of astrocytes, with NFI-X and -C controlling the expression of astrocytic markers at late stages of differentiation.


Assuntos
Astrócitos/citologia , Diferenciação Celular/genética , Regulação da Expressão Gênica , Fatores de Transcrição NFI/metabolismo , Neurônios/citologia , Células-Tronco/citologia , Animais , Astrócitos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Proteínas da Matriz Extracelular/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Modelos Biológicos , Fatores de Transcrição NFI/genética , Neurônios/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Células-Tronco/metabolismo
15.
J Biol Chem ; 284(5): 2598-2602, 2009 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-19004816

RESUMO

Several neurological diseases, including Parkinson disease and dementia with Lewy bodies, are characterized by the accumulation of alpha-synuclein phosphorylated at Ser-129 (p-Ser-129). The kinase or kinases responsible for this phosphorylation have been the subject of intense investigation. Here we submit evidence that polo-like kinase 2 (PLK2, also known as serum-inducible kinase or SNK) is a principle contributor to alpha-synuclein phosphorylation at Ser-129 in neurons. PLK2 directly phosphorylates alpha-synuclein at Ser-129 in an in vitro biochemical assay. Inhibitors of PLK kinases inhibited alpha-synuclein phosphorylation both in primary cortical cell cultures and in mouse brain in vivo. Finally, specific knockdown of PLK2 expression by transduction with short hairpin RNA constructs or by knock-out of the plk2 gene reduced p-Ser-129 levels. These results indicate that PLK2 plays a critical role in alpha-synuclein phosphorylation in central nervous system.


Assuntos
Sistema Nervoso Central/metabolismo , Proteínas Quinases/metabolismo , Serina/metabolismo , alfa-Sinucleína/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Sistema Nervoso Central/enzimologia , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases , Interferência de RNA , alfa-Sinucleína/química
16.
J Biol Chem ; 284(6): 3408-17, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19074142

RESUMO

Chronic inflammation and inflammatory cytokines have recently been implicated in the development and progression of various types of cancer. In the brain, neuroinflammatory cytokines affect the growth and differentiation of both normal and malignant glial cells, with interleukin 1 (IL-1) shown to be secreted by the majority of glioblastoma cells. Recently, elevated levels of sphingosine kinase 1 (SphK1), but not SphK2, were correlated with a shorter survival prognosis for patients with glioblastoma multiforme. SphK1 is a lipid kinase that produces the pro-growth, anti-apoptotic sphingosine 1-phosphate, which can induce invasion of glioblastoma cells. Here, we show that the expression of IL-1 correlates with the expression of SphK1 in glioblastoma cells, and neutralizing anti-IL-1 antibodies inhibit both the growth and invasion of glioblastoma cells. Furthermore, IL-1 up-regulates SphK1 mRNA levels, protein expression, and activity in both primary human astrocytes and various glioblastoma cell lines; however, it does not affect SphK2 expression. The IL-1-induced SphK1 up-regulation can be blocked by the inhibition of JNK, the overexpression of the dominant-negative c-Jun(TAM67), and the down-regulation of c-Jun expression by small interference RNA. Activation of SphK1 expression by IL-1 occurs on the level of transcription and is mediated via a novel AP-1 element located within the first intron of the sphk1 gene. In summary, our results suggest that SphK1 expression is transcriptionally regulated by IL-1 in glioblastoma cells, and this pathway may be important in regulating survival and invasiveness of glioblastoma cells.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Interleucina-1/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Elementos de Resposta , Linhagem Celular Tumoral , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Interleucina-1/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Invasividade Neoplásica , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo
17.
Neurobiol Aging ; 29(10): 1485-93, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17442458

RESUMO

Beta-amyloid (Abeta) is the principal component of the extracellular plaques present in patients with Alzheimer's disease. Several studies have recently shown that acutely applied Abeta inhibits the induction of LTP in the hippocampus. In the present studies, we have investigated the role of integrins in such Abeta-mediated block of LTP in the dentate gyrus in vitro and in the CA1 in vivo. Selective antibodies to the alpha v integrin subunit were found to prevent the Abeta inhibition of LTP, both in the dentate gyrus in vitro and in the CA1 in vivo. In contrast, two control antibodies did not prevent such action of Abeta. In addition, a small molecule nonpeptide antagonist of alpha v-containing integrins and two other antagonistic ligands of integrins, superfibronectin and the disintegrin echistatin, also prevented the Abeta inhibition of LTP. These studies indicate that alpha v integrins may be important mediators of synaptic dysfunction prior to neurodegeneration in Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Integrina alfaV/metabolismo , Potenciação de Longa Duração/fisiologia , Inibição Neural/fisiologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/toxicidade , Animais , Anticorpos/farmacologia , Giro Denteado/metabolismo , Giro Denteado/fisiopatologia , Hipocampo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Inibição Neural/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Peptídeos/farmacologia , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
18.
Neurobiol Aging ; 28(2): 226-37, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16448724

RESUMO

Pathological hallmarks of Alzheimer's disease are the presence of extracellular amyloid plaques, intracellular neurofibrillary tangles, and neurodegeneration. The principal component of amyloid plaques is the amyloid-beta peptide (Abeta). Accumulating evidence indicates that Abeta may play a causal role in Alzheimer's disease. In this report, we demonstrate that Abeta deposition and neurotoxicity in human cortical primary neurons are mediated through alpha2beta1 and alphaVbeta1 integrins using specific integrin-blocking antibodies. An aberrant integrin signaling pathway causing the neurotoxicity is mediated through Pyk2. The role of alpha2beta1 and alphaVbeta1 integrins can be extended to another amyloidosis using an amylin in vitro neurotoxicity model. These results indicate that the alpha2beta1 and alphaVbeta1 integrin signaling pathway may be critical components of neurodegeneration in Alzheimer's disease and that integrins may recognize and be activated by a shared structural motif of polymerizing amyloidogenic proteins.


Assuntos
Peptídeos beta-Amiloides/administração & dosagem , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Integrina alfa2beta1/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Apoptose/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Neurônios/efeitos dos fármacos , Neurotoxinas/administração & dosagem
19.
J Biol Chem ; 281(46): 34955-64, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-17012236

RESUMO

Reactive astrogliosis is the gliotic response to brain injury with activated astrocytes and microglia being the major effector cells. These cells secrete inflammatory cytokines, proteinases, and proteinase inhibitors that influence extracellular matrix (ECM) remodeling. In astrocytes, the expression of tissue inhibitor of metalloproteinases-1 (TIMP-1) is up-regulated by interleukin-1 (IL-1), which is a major neuroinflammatory cytokine. We report that IL-1 activates TIMP-1 expression via both the IKK/NF-kappaB and MEK3/6/p38/ATF-2 pathways in astrocytes. The activation of the TIMP-1 gene can be blocked by using pharmacological inhibitors, including BAY11-7082 and SB202190, overexpression of the dominant-negative inhibitor of NF-kappaB (IkappaBalphaSR), or by the knock-down of p65 subunit of NF-kappaB. Binding of activated NF-kappaB (p50/p65 heterodimer) and ATF-2 (homodimer) to two novel regulatory elements located -2.7 and -2.2 kb upstream of the TIMP-1 transcription start site, respectively, is required for full IL-1-responsiveness. Mutational analysis of these regulatory elements and their weak activity when linked to the minimal tk promoter suggest that cooperative binding is required to activate transcription. In contrast to astrocytes, we observed that TIMP-1 is expressed at lower levels in gliomas and is not regulated by IL-1. We provide evidence that the lack of TIMP-1 activation in gliomas results from either dysfunctional IKK/NF-kappaB or MEK3/6/p38/ATF-2 activation by IL-1. In summary, we propose a novel mechanism of TIMP-1 regulation, which ensures an increased supply of the inhibitor after brain injury, and limits ECM degradation. This mechanism does not function in gliomas, and may in part explain the increased invasiveness of glioma cells.


Assuntos
Astrócitos/metabolismo , Interleucina-1/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator 2 Ativador da Transcrição/metabolismo , Regulação da Expressão Gênica , Glioma/metabolismo , Humanos , NF-kappa B/metabolismo , RNA Mensageiro , Transdução de Sinais
20.
Proc Natl Acad Sci U S A ; 102(8): 3058-63, 2005 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-15710903

RESUMO

Based on a variety of genetic, biochemical, and neuropathological evidence, amyloid-beta peptide (Abeta) has been suggested to be causal in Alzheimer's disease (AD). Abeta has been shown to mediate neurodegenerative and inflammatory changes associated with amyloid plaques, as well as exert direct neurotoxicity through oligomeric forms of Abeta. The mechanism of Abeta toxicity, however, remains largely unknown. In this work, we show that an early event after exposure of postmitotic neurons to Abeta is tyrosine phosphorylation of FISH adapter protein. FISH binds to and potentially regulates certain ADAM family members. We present evidence that FISH and ADAM12 mediate the neurotoxic effect of Abeta. Expression of an ADAM12 protease-deficient mutant (ADAM12DeltaMP) blocks Abeta-induced neuronal death, and expression of an N-terminal fragment of FISH reduces Abeta toxicity. The C-terminal fragment of FISH containing the ADAMs binding region is found to be sufficient for induction of neuronal death, which is prevented by coexpression of the ADAM12DeltaMP. Abeta treatment, as well as expression of the C-terminal toxic FISH fragment, induces accumulation of ADAM12 N-terminal cleavage product in conditioned medium, demonstrating activation of the ADAM metalloprotease/sheddase activity. ADAM12 protein is reduced in AD brains, pointing to a possible increase in ADAM12 proteolytic activity. These data suggest that Abeta toxicity is mediated by FISH and ADAM12 and may provide insights into therapeutic strategies for AD treatment.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Peptídeos beta-Amiloides/toxicidade , Proteínas de Membrana/fisiologia , Metaloendopeptidases/fisiologia , Neurônios/efeitos dos fármacos , Proteínas ADAM , Proteína ADAM12 , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Humanos , Proteínas de Membrana/análise , Metaloendopeptidases/análise , Neurônios/patologia , Fosforilação , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA