Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Front Biosci (Elite Ed) ; 4(6): 2110-20, 2012 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-22202023

RESUMO

Among the numerous intracellular signalling pathways that control brain development and pathogenesis c-Jun N-terminal kinases have a leading role in the Central Nervous System. JNKs regulate a wide range of processes in brain development, plasticity, repair/regeneration, neuronal death and neuroinflammation. Indeed, accumulating evidence underline the potential of JNK targeted molecules towards the treatment of neurodegenerative disorders. The focus of the presenting review is to provide an overview of the reported data linking JNKs to brain function and dysfunction.


Assuntos
Encéfalo/enzimologia , MAP Quinase Quinase 4/metabolismo , Transdução de Sinais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Humanos
3.
PLoS One ; 6(12): e28185, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22164242

RESUMO

BACKGROUND: Oxidative stress is a key feature in the pathogenesis of several neurological disorders. Following oxidative stress stimuli a wide range of pathways are activated and contribute to cellular death. The mechanism that couples c-Jun N-terminal kinase (JNK) signaling, a key pathway in stress conditions, to the small ubiquitin-related modifier (SUMO), an emerging protein in the field, is largely unknown. METHODOLOGY/PRINCIPAL FINDINGS: With this study we investigated if SUMOylation participates in the regulation of JNK activation as well as cellular death in a model of H(2)O(2) induced-oxidative stress. Our data show that H(2)O(2) modulates JNK activation and induces cellular death in neuroblastoma SH-SY5Y cells. Inhibition of JNK's action with the D-JNKI1 peptide rescued cells from death. Following H(2)O(2), SUMO-1 over-expression increased phosphorylation of JNK and exacerbated cell death, although only in conditions of mild oxidative stress. Furthermore inhibition of SUMOylation, following transfection with SENP1, interfered with JNK activation and rescued cells from H(2)O(2) induced death. Importantly, in our model, direct interaction between these proteins can occur. CONCLUSIONS/SIGNIFICANCE: Taken together our results show that SUMOylation may significantly contribute to modulation of JNK activation and contribute to cell death in oxidative stress conditions.


Assuntos
Peróxido de Hidrogênio/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Humanos , Imuno-Histoquímica/métodos , Imunoprecipitação , Estresse Oxidativo , Plasmídeos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo
4.
J Biol Chem ; 286(51): 43871-43880, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22033930

RESUMO

Alzheimer disease (AD) is characterized by cognitive impairment that starts with memory loss to end in dementia. Loss of synapses and synaptic dysfunction are closely associated with cognitive impairment in AD patients. Biochemical and pathological evidence suggests that soluble Aß oligomers correlate with cognitive impairment. Here, we used the TgCRND8 AD mouse model to investigate the role of JNK in long term memory deficits. TgCRND8 mice were chronically treated with the cell-penetrating c-Jun N-terminal kinase inhibitor peptide (D-JNKI1). D-JNKI1, preventing JNK action, completely rescued memory impairments (behavioral studies) as well as the long term potentiation deficits of TgCRND8 mice. Moreover, D-JNKI1 inhibited APP phosphorylation in Thr-668 and reduced the amyloidogenic cleavage of APP and Aß oligomers in brain parenchyma of treated mice. In conclusion, by regulating key pathogenic mechanisms of AD, JNK might hold promise as innovative therapeutic target.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Regulação Enzimológica da Expressão Gênica , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Doença de Alzheimer/genética , Animais , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Eletrofisiologia , Humanos , Aprendizagem em Labirinto , Transtornos da Memória/genética , Camundongos , Modelos Biológicos , Peptídeos/química , Transdução de Sinais , Fatores de Tempo
5.
J Alzheimers Dis ; 26(2): 315-29, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21628793

RESUMO

Alzheimer's disease (AD) is a major clinical concern, and the search for new molecules to combat disease progression remains important. One of the major hallmarks in AD pathogenesis is the hyperphosphorylation of tau and subsequent formation of neurofibrillary tangles. Several kinases are involved in this process. Amongst them, c-Jun N-terminal kinases (JNKs) are activated in AD brains and are also associated with the development of amyloid plaques. This study was designed to investigate the contribution of JNK in tau hyperphosphorylation and whether it may represent a potential therapeutic target for the fight against AD. The specific inhibition of JNK by the cell permeable peptide D-JNKI-1 led to a reduction of p-tau at S202/T205 and S422, two established target sites of JNK, in rat neuronal cultures and in human fibroblasts cultures. Similarly, D-JNKI-1 reduced p-tau at S202/T205 in an in vivo model of AD (TgCRND8 mice). Our findings support the fundamental role of JNK in the regulation of tau hyperphosphorylation and subsequently in AD pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Córtex Cerebral/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/farmacologia , Masculino , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Emaranhados Neurofibrilares/efeitos dos fármacos , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fosforilação , Ratos
6.
J Alzheimers Dis ; 24(4): 633-42, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21321401

RESUMO

c-Jun N-terminal kinases (JNKs) and in particular JNK3 the neuronal specific isoform, have been recognized as important enzymes in the pathology of diverse neurological disorders. Indeed, several efforts have been made to design drugs that inhibit JNK signaling. The success that characterized the new generation of cell permeable peptides raise the hope in the field of neurodegeneration for new therapeutic routes. However, in order to design new and more efficient therapeutical approaches careful re-examination of current knowledge is required. Scaffold proteins are key endogenous regulators of JNK signaling: they can modulate spatial and temporal activation of the JNK signaling and can thus provide the basis for the design of more specific inhibitors. This review focuses on delineating the role of scaffold proteins on the regulation of JNK signaling in neurons. Furthermore the possibility to design a new JNK3 cell permeable peptide inhibitor by targeting the ß-arrestin-JNK3 interaction is discussed.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/enzimologia , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 10 Ativada por Mitógeno/fisiologia , Doenças Neurodegenerativas/patologia , Neurônios/enzimologia , Neurônios/patologia , Proteínas Associadas à Matriz Nuclear/fisiologia
7.
Brain Res ; 1381: 1-10, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20977891

RESUMO

The cyclin dependent kinase 5 (Cdk5)/p35 complex is essential for regulation of cell survival during development and in models of neuronal excitotoxicity. Dysregulation of Cdk5, by cleavage of its neuronal specific activators p35 and p39, has been implicated in various neurodegenerative disorders such as Alzheimer's disease, however targets of the complex that regulate neuronal survival physiologically and/or during pathogenesis are largely unknown. Since hypoxia is a key feature in the pathogenesis of several neuronal disorders we investigated a role for Cdk5/p35 in the neuronal hypoxic response. Our data show that hypoxia modulates the p35/Cdk5 complex in primary cortical neurons at the transcriptional and protein level. Furthermore hypoxic induction of Cdk5 activity correlates with Hif-1α stabilisation, and direct interaction between these proteins can occur. Importantly, we demonstrate that Cdk5-mediated signaling is involved in Hif-1α stabilisation since inhibition of Cdk5 by roscovitine abrogates Hif-1α accumulation and induces cell death. Taken together our results show that the Cdk5/p35 complex may significantly contribute to modulation of Hif-1α stabilisation and impact neuronal survival during oxygen deprivation. Thus this study highlights a new hypoxia-mediated signaling pathway and implicates the cytoskeleton as a potential regulator of Hif-1α.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Trifosfato de Adenosina/metabolismo , Análise de Variância , Animais , Western Blotting , Morte Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Imuno-Histoquímica , Camundongos , Neurônios/citologia , Fosfotransferases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Cerebellum ; 10(2): 281-90, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21191679

RESUMO

Several studies have shown that Purkinje cells die by apoptosis in organotypic slice cultures from postnatal 3-day-old (P3) mice. This cell death is age-dependent and has been proposed as indirect evidence for the programmed Purkinje cell death occurring in in vivo cerebellum. Here, we studied whether c-jun N-terminal kinase (JNK) and p38 kinase pathways contribute to the Purkinje cell death observed in cerebellar slice cultures obtained from P3 mice. Slice culture treatment with D-JNKI1 or SB203580, respectively inhibitors of JNK and p38 MAP kinases, results in a better survival of Purkinje cells. Interestingly, the combined treatment with the two inhibitors potentiated single treatment effects. These results suggest that p38 and JNK pathways might be differently implicated in this Purkinje cell death. Time course experiments found p38 activation immediately post-slicing, whereas JNK activation was detected only 2 h after the culture. We hypothesize that p38 activation might be due to the "sliced condition," and JNK activation might be more specific to P3 age-dependent cell death. The study of JNK and p38 activation in cerebellar lysates from P0 slice culture confirmed JNK activation being specific for the P3 explants, whereas p38 is activated both from P0 and P3 cerebellar slice culture lysates. These results suggest that p38 is activated by the slicing, whereas JNK activation is related to developmental Purkinje cell death.


Assuntos
Apoptose/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células de Purkinje/citologia , Células de Purkinje/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Envelhecimento/fisiologia , Animais , Artefatos , Western Blotting , Ativação Enzimática , Camundongos , Técnicas de Cultura de Órgãos , Manejo de Espécimes/efeitos adversos
9.
Pharmaceuticals (Basel) ; 3(1): 42-58, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-27713242

RESUMO

The phosphorylation of Amyloid Precursor Protein (APP) at Thr668 plays a key role in APP metabolism that is highly relevant to AD. The c-Jun-N-terminal kinase (JNK), glycogen synthase kinase-3ß (GSK-3ß) and cyclin-dependent kinase 5 (Cdk5) can all be responsible for this phosphorylation. These kinases are activated by excitotoxic stimuli fundamental hallmarks of AD. The exposure of cortical neurons to a high dose of NMDA (100 µM) for 30'-45' led to an increase of P-APP Thr668. During NMDA stimulation APP hyperphosphorylation has to be assigned to GSK-3ß activity, since addition of L803-mts, a substrate competitive inhibitor of GSK-3ß reduced APP phosphorylation induced by NMDA. On the contrary, inhibition of JNK and Cdk5 with D-JNKI1 and Roscovitine respectively did not prevent NMDA-induced P-APP increase. These data show a tight connection, in excitotoxic conditions, between APP metabolism and the GSK-3ß signaling pathway.

10.
Pharmaceuticals (Basel) ; 3(2): 379-392, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-27713257

RESUMO

The inability of most drugs to cross the blood-brain barrier and/or plasma membrane limits their use for biomedical applications in the brain. Cell Permeable Peptides (CPPs) overcome this problem and are effective in vivo, crossing the plasma membrane and the blood-brain barrier. CPPs deliver a wide variety of compounds intracellularly in an active form. In fact, many bioactive cargoes have neuroprotective properties, and due to their ability to block protein-protein interactions, offer exciting perspectives in the clinical setting. In this review we give an overview of the Cell Permeable Peptides strategy to deliver neuroprotectants against neurodegeneration in the CNS.

11.
Molecules ; 15(1): 114-27, 2009 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-20110876

RESUMO

Hypoxia is an established factor of neurodegeneration. Nowadays, attention is directed at understanding how alterations in the expression of stress-related signaling proteins contribute to age dependent neuronal vulnerability to injury. The purpose of this study was to investigate how Hif-1alpha, a major neuroprotective factor, and JNK signaling, a key pathway in neurodegeneration, relate to hypoxic injury in young (6DIV) and adult (12DIV) neurons. We could show that in young neurons as compared to mature ones, the protective factor Hif-1alpha is more induced while the stress protein phospho-JNK displays lower basal levels. Indeed, changes in the expression levels of these proteins correlated with increased vulnerability of adult neurons to hypoxic injury. Furthermore, we describe for the first time that treatment with the D-JNKI1, a JNK-inhibiting peptide, rescues adult hypoxic neurons from death and contributes to Hif-1alpha upregulation, probably via a direct interaction with the Hif-1alpha protein.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/enzimologia , Neurônios/patologia , Envelhecimento/patologia , Sequência de Aminoácidos , Animais , Morte Celular , Hipóxia Celular , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Sistema de Sinalização das MAP Quinases , Dados de Sequência Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos
12.
Am J Physiol Regul Integr Comp Physiol ; 295(3): R864-73, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18614764

RESUMO

Hypoxia is linked to changes in blood-brain barrier (BBB) permeability, and loss of BBB integrity is characteristic of many pathological brain diseases including stroke. In particular, astrocytes play a central role in brain homeostasis and BBB function. We investigated how hypoxia affects astrocyte survival and assessed whether VEGF release through hypoxia-inducible factor-1alpha (HIF-1alpha) induction plays a role in tolerance of these cells to insult. Thus primary astrocytes were subjected to normoxic (21% O(2)), hypoxic (1% O(2)), or near-anoxic (<0.1% O(2)) conditions in the presence or absence of glucose. Cell death was significantly initiated after combined oxygen glucose deprivation, and, surprisingly, astrocyte proliferation increased concomitantly. Near anoxic, but not hypoxic, conditions stabilized HIF-1alpha protein and provoked DNA binding activity, whereas oxygen and glucose deprivation accelerated HIF-1alpha accumulation. Unexpectedly, Hif-1alpha knockdown studies showed that elevated VEGF levels following increased insult was only partially due to HIF-1alpha induction, suggesting alternative mechanisms of VEGF regulation. Notably, endogenous VEGF signaling during insult was essential for cell fate since VEGF inhibition appreciably augmented cell death and reduced proliferation. These data suggest Hif-1 only partially contributes to VEGF-mediated astrocyte responses during chronic injury (as occurs in clinical hypoxic/ischemic insults) that may ultimately be responsible for disrupting BBB integrity.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Isquemia Encefálica/patologia , Morte Celular/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Barreira Hematoencefálica/fisiologia , Isquemia Encefálica/metabolismo , Morte Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Hipóxia Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Glucose/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxigênio/farmacologia , RNA Interferente Pequeno , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...