Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Commun Signal ; 18(1): 121, 2020 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-32771000

RESUMO

BACKGROUND: The PIM family kinases promote cancer cell survival and motility as well as metastatic growth in various types of cancer. We have previously identified several PIM substrates, which support cancer cell migration and invasiveness. However, none of them are known to regulate cellular movements by directly interacting with the actin cytoskeleton. Here we have studied the phosphorylation-dependent effects of PIM1 on actin capping proteins, which bind as heterodimers to the fast-growing actin filament ends and stabilize them. METHODS: Based on a phosphoproteomics screen for novel PIM substrates, we have used kinase assays and fluorescence-based imaging techniques to validate actin capping proteins as PIM1 substrates and interaction partners. We have analysed the functional consequences of capping protein phosphorylation on cell migration and adhesion by using wound healing and real-time impedance-based assays. We have also investigated phosphorylation-dependent effects on actin polymerization by analysing the protective role of capping protein phosphomutants in actin disassembly assays. RESULTS: We have identified capping proteins CAPZA1 and CAPZB2 as PIM1 substrates, and shown that phosphorylation of either of them leads to increased adhesion and migration of human prostate cancer cells. Phosphorylation also reduces the ability of the capping proteins to protect polymerized actin from disassembly. CONCLUSIONS: Our data suggest that PIM kinases are able to induce changes in actin dynamics to support cell adhesion and movement. Thus, we have identified a novel mechanism through which PIM kinases enhance motility and metastatic behaviour of cancer cells. Video abstract.


Assuntos
Proteínas de Capeamento de Actina/metabolismo , Movimento Celular , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Actinas/metabolismo , Animais , Adesão Celular , Linhagem Celular Tumoral , Extensões da Superfície Celular/metabolismo , Citoplasma/metabolismo , Humanos , Masculino , Camundongos , Fosforilação , Multimerização Proteica , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores
2.
Front Cell Neurosci ; 8: 272, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25309320

RESUMO

Genetic anomalies on the JNK pathway confer susceptibility to autism spectrum disorders, schizophrenia, and intellectual disability. The mechanism whereby a gain or loss of function in JNK signaling predisposes to these prevalent dendrite disorders, with associated motor dysfunction, remains unclear. Here we find that JNK1 regulates the dendritic field of L2/3 and L5 pyramidal neurons of the mouse motor cortex (M1), the main excitatory pathway controlling voluntary movement. In Jnk1-/- mice, basal dendrite branching of L5 pyramidal neurons is increased in M1, as is cell soma size, whereas in L2/3, dendritic arborization is decreased. We show that JNK1 phosphorylates rat HMW-MAP2 on T1619, T1622, and T1625 (Uniprot P15146) corresponding to mouse T1617, T1620, T1623, to create a binding motif, that is critical for MAP2 interaction with and stabilization of microtubules, and dendrite growth control. Targeted expression in M1 of GFP-HMW-MAP2 that is pseudo-phosphorylated on T1619, T1622, and T1625 increases dendrite complexity in L2/3 indicating that JNK1 phosphorylation of HMW-MAP2 regulates the dendritic field. Consistent with the morphological changes observed in L2/3 and L5, Jnk1-/- mice exhibit deficits in limb placement and motor coordination, while stride length is reduced in older animals. In summary, JNK1 phosphorylates HMW-MAP2 to increase its stabilization of microtubules while at the same time controlling dendritic fields in the main excitatory pathway of M1. Moreover, JNK1 contributes to normal functioning of fine motor coordination. We report for the first time, a quantitative Sholl analysis of dendrite architecture, and of motor behavior in Jnk1-/- mice. Our results illustrate the molecular and behavioral consequences of interrupted JNK1 signaling and provide new ground for mechanistic understanding of those prevalent neuropyschiatric disorders where genetic disruption of the JNK pathway is central.

3.
Adv Exp Med Biol ; 800: 37-57, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24243099

RESUMO

Incorrect placement of nerve cells during brain development leaves us at risk of diseases and conditions ranging from epilepsy and mental retardation to schizophrenia and dyslexia. The developing brain produces cells at an impressive rate, with up to 250,000 new cells generated every minute. These newborn cells migrate long distances in sequential waves to settle in the layers that make up the cerebral cortex. If a nerve cell moves too fast or too slow during this journey, it may not take the correct route or reach its appropriate destination. Much knowledge has been accumulated on molecular cues and transcriptional programs regulating cortical development. More recently, components of the c-Jun N-terminal signaling cascade have been brought to light as important intracellular regulators of nerve cell motility. In this chapter, we focus on this family of protein kinases, their upstream activators and downstream targets in the context of neuronal migration. We first present basic information on these molecules, much of which derives from studies outside the nervous system. We then highlight key findings on JNK signaling in brain where it phosphorylates brain-specific proteins that influence microtubule homeostasis. Finally, we summarize recent findings from transgenic mice on the regulation of neuronal migration by JNK cascade components and by JNK substrates.


Assuntos
Movimento Celular , Córtex Cerebral/enzimologia , Dislexia/enzimologia , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases , Neurônios/enzimologia , Esquizofrenia/enzimologia , Animais , Córtex Cerebral/patologia , Dislexia/genética , Dislexia/patologia , Humanos , MAP Quinase Quinase 4/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Esquizofrenia/genética , Esquizofrenia/patologia
4.
Nat Neurosci ; 14(3): 305-13, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21297631

RESUMO

Cell migration is the consequence of the sum of positive and negative regulatory mechanisms. Although appropriate migration of neurons is a principal feature of brain development, the negative regulatory mechanisms remain obscure. We found that JNK1 was highly active in developing cortex and that selective inhibition of JNK in the cytoplasm markedly increased both the frequency of exit from the multipolar stage and radial migration rate and ultimately led to an ill-defined cellular organization. Moreover, regulation of multipolar-stage exit and radial migration in Jnk1(-/-) (also known as Mapk8) mice, resulted from consequential changes in phosphorylation of the microtubule regulator SCG10 (also called stathmin-2). Expression of an SCG10 mutant that mimics the JNK1-phosphorylated form restored normal migration in the brains of Jnk1(-/-) mouse embryos. These findings indicate that the phosphorylation of SCG10 by JNK1 is a fundamental mechanism that governs the transition from the multipolar stage and the rate of neuronal cell movement during cortical development.


Assuntos
Movimento Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Estatmina , Tubulina (Proteína)/metabolismo
5.
Expert Opin Ther Targets ; 12(1): 31-43, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18076368

RESUMO

BACKGROUND: Cell stress and tissue injury lead to c-Jun N-terminal kinase (JNK) activation, which is known to contribute to cell death. Paradoxically, strong evidence supports an important role for JNK in the regeneration of neuronal processes, subsequent to injury. OBJECTIVE: Recent research revealed the growth cone-associated protein superior cervical ganglion-10 protein as a candidate effector for the regeneration pathway mediated by JNK1. This implies that neuroprotective strategies targeting JNK may have negative effects on neuronal regeneration, unless JNK1 function is spared, and that the mechanistic relationships between JNK1 and neuronal regeneration deserve increased attention. RESULTS: This review proposes a model reconciling the microtubule regulatory properties of superior cervical ganglion protein 10 with its role as a JNK effector of regeneration and highlight remaining issues to be resolved.


Assuntos
Proteínas de Membrana/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Regeneração Nervosa/fisiologia , Gânglio Cervical Superior/metabolismo , Animais , Sistemas de Liberação de Medicamentos/métodos , Humanos , Proteínas de Membrana/genética , Microtúbulos/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/biossíntese , Proteína Quinase 8 Ativada por Mitógeno/genética , Estatmina
6.
J Cell Biol ; 173(2): 265-77, 2006 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-16618812

RESUMO

c-Jun NH(2)-terminal kinases (JNKs) are essential during brain development, when they regulate morphogenic changes involving cell movement and migration. In the adult, JNK determines neuronal cytoarchitecture. To help uncover the molecular effectors for JNKs in these events, we affinity purified JNK-interacting proteins from brain. This revealed that the stathmin family microtubule-destabilizing proteins SCG10, SCLIP, RB3, and RB3' interact tightly with JNK. Furthermore, SCG10 is also phosphorylated by JNK in vivo on sites that regulate its microtubule depolymerizing activity, serines 62 and 73. SCG10-S73 phosphorylation is significantly decreased in JNK1-/- cortex, indicating that JNK1 phosphorylates SCG10 in developing forebrain. JNK phosphorylation of SCG10 determines axodendritic length in cerebrocortical cultures, and JNK site-phosphorylated SCG10 colocalizes with active JNK in embryonic brain regions undergoing neurite elongation and migration. We demonstrate that inhibition of cytoplasmic JNK and expression of SCG10-62A/73A both inhibited fluorescent tubulin recovery after photobleaching. These data suggest that JNK1 is responsible for regulation of SCG10 depolymerizing activity and neurite elongation during brain development.


Assuntos
Axônios/fisiologia , Dendritos/fisiologia , Microtúbulos/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fatores de Crescimento Neural/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Proteínas de Ligação ao Cálcio , Proteínas de Transporte , Linhagem Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Camundongos , Proteínas dos Microtúbulos , Proteína Quinase 8 Ativada por Mitógeno/análise , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Fosforilação , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estatmina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...