Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Rep ; 34(13): 108923, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33789115

RESUMO

Despite decades of work, much remains elusive about molecular events at the interplay between physiological and structural changes underlying neuronal plasticity. Here, we combined repetitive live imaging and expansion microscopy in organotypic brain slice cultures to quantitatively characterize the dynamic changes of the intracellular versus surface pools of GluA2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) across the different dendritic spine types and the shaft during hippocampal homeostatic plasticity. Mechanistically, we identify ephrinB2 and glutamate receptor interacting protein (GRIP) 1 as mediating AMPAR relocation to the mushroom spine surface following lesion-induced denervation. Moreover, stimulation with the ephrinB2 specific receptor EphB4 not only prevents the lesion-induced disappearance of mushroom spines but is also sufficient to shift AMPARs to the surface and rescue spine recovery in a GRIP1 dominant-negative background. Thus, our results unravel a crucial role for ephrinB2 during homeostatic plasticity and identify a potential pharmacological target to improve dendritic spine plasticity upon injury.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Espinhas Dendríticas/metabolismo , Efrina-B2/metabolismo , Homeostase , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Animais , Membrana Celular/metabolismo , Denervação , Camundongos Endogâmicos C57BL , Receptor EphB4/metabolismo , Receptores de AMPA/metabolismo
2.
Cell Rep ; 31(4): 107568, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32348765

RESUMO

Anti-angiogenic treatment targeting vascular endothelial growth factor (VEGF)-VEGFR2 signaling has shown limited efficacy in lung cancer patients. Here, we demonstrate that inhibition of VEGFR2 in tumor cells, expressed in ∼20% of non-squamous non-small cell lung cancer (NSCLC) patients, leads to a pro-invasive phenotype. Drug-induced inhibition of tumor VEGFR2 interferes with the formation of the EphA2/VEGFR2 heterocomplex, thereby allowing RSK to interact with Serine 897 of EphA2. Inhibition of RSK decreases phosphorylation of Serine 897 EphA2. Selective genetic modeling of Serine 897 of EphA2 or inhibition of EphA2 abrogates the formation of metastases in vivo upon VEGFR2 inhibition. In summary, these findings demonstrate that VEGFR2-targeted therapy conditions VEGFR2-positive NSCLC to Serine 897 EphA2-dependent aggressive tumor growth and metastasis. These data shed light on the molecular mechanisms explaining the limited efficacy of VEGFR2-targeted anti-angiogenic treatment in lung cancer patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Receptor EphA2/metabolismo , Serina/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Invasividade Neoplásica , Metástase Neoplásica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
3.
Nat Cell Biol ; 22(1): 97-107, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31907411

RESUMO

Diffuse brain infiltration by glioma cells causes detrimental disease progression, but its multicellular coordination is poorly understood. We show here that glioma cells infiltrate the brain collectively as multicellular networks. Contacts between moving glioma cells are adaptive epithelial-like or filamentous junctions stabilized by N-cadherin, ß-catenin and p120-catenin, which undergo kinetic turnover, transmit intercellular calcium transients and mediate directional persistence. Downregulation of p120-catenin compromises cell-cell interaction and communication, disrupts collective networks, and both the cadherin and RhoA binding domains of p120-catenin are required for network formation and migration. Deregulating p120-catenin further prevents diffuse glioma cell infiltration of the mouse brain with marginalized microlesions as the outcome. Transcriptomics analysis has identified p120-catenin as an upstream regulator of neurogenesis and cell cycle pathways and a predictor of poor clinical outcome in glioma patients. Collective glioma networks infiltrating the brain thus depend on adherens junctions dynamics, the targeting of which may offer an unanticipated strategy to halt glioma progression.


Assuntos
Junções Aderentes/metabolismo , Cateninas/metabolismo , Adesão Celular/fisiologia , Glioma/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Caderinas/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo/fisiologia , Glioma/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , delta Catenina
4.
Elife ; 82019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31868583

RESUMO

Axon branching is crucial for proper formation of neuronal networks. Although originally identified as an angiogenic factor, VEGF also signals directly to neurons to regulate their development and function. Here we show that VEGF and its receptor VEGFR2 (also known as KDR or FLK1) are expressed in mouse hippocampal neurons during development, with VEGFR2 locally expressed in the CA3 region. Activation of VEGF/VEGFR2 signaling in isolated hippocampal neurons results in increased axon branching. Remarkably, inactivation of VEGFR2 also results in increased axon branching in vitro and in vivo. The increased CA3 axon branching is not productive as these axons are less mature and form less functional synapses with CA1 neurons. Mechanistically, while VEGF promotes the growth of formed branches without affecting filopodia formation, loss of VEGFR2 increases the number of filopodia and enhances the growth rate of new branches. Thus, a controlled VEGF/VEGFR2 signaling is required for proper CA3 hippocampal axon branching during mouse hippocampus development.


Assuntos
Axônios/fisiologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Efrina-B2/genética , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Pseudópodes/metabolismo , Transdução de Sinais/genética , Sinapses/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
5.
Cell ; 179(7): 1661-1676.e19, 2019 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-31835038

RESUMO

Reliable detection of disseminated tumor cells and of the biodistribution of tumor-targeting therapeutic antibodies within the entire body has long been needed to better understand and treat cancer metastasis. Here, we developed an integrated pipeline for automated quantification of cancer metastases and therapeutic antibody targeting, named DeepMACT. First, we enhanced the fluorescent signal of cancer cells more than 100-fold by applying the vDISCO method to image metastasis in transparent mice. Second, we developed deep learning algorithms for automated quantification of metastases with an accuracy matching human expert manual annotation. Deep learning-based quantification in 5 different metastatic cancer models including breast, lung, and pancreatic cancer with distinct organotropisms allowed us to systematically analyze features such as size, shape, spatial distribution, and the degree to which metastases are targeted by a therapeutic monoclonal antibody in entire mice. DeepMACT can thus considerably improve the discovery of effective antibody-based therapeutics at the pre-clinical stage. VIDEO ABSTRACT.


Assuntos
Anticorpos/uso terapêutico , Aprendizado Profundo , Diagnóstico por Computador/métodos , Quimioterapia Assistida por Computador/métodos , Neoplasias/patologia , Animais , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Metástase Neoplásica , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Software , Microambiente Tumoral
6.
Mol Autism ; 9: 56, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30443311

RESUMO

Background: Altered neuronal development is discussed as the underlying pathogenic mechanism of autism spectrum disorders (ASD). Copy number variations of 16p11.2 have recurrently been identified in individuals with ASD. Of the 29 genes within this region, quinolinate phosphoribosyltransferase (QPRT) showed the strongest regulation during neuronal differentiation of SH-SY5Y neuroblastoma cells. We hypothesized a causal relation between this tryptophan metabolism-related enzyme and neuronal differentiation. We thus analyzed the effect of QPRT on the differentiation of SH-SY5Y and specifically focused on neuronal morphology, metabolites of the tryptophan pathway, and the neurodevelopmental transcriptome. Methods: The gene dosage-dependent change of QPRT expression following Chr16p11.2 deletion was investigated in a lymphoblastoid cell line (LCL) of a deletion carrier and compared to his non-carrier parents. Expression of QPRT was tested for correlation with neuromorphology in SH-SY5Y cells. QPRT function was inhibited in SH-SY5Y neuroblastoma cells using (i) siRNA knockdown (KD), (ii) chemical mimicking of loss of QPRT, and (iii) complete CRISPR/Cas9-mediated knock out (KO). QPRT-KD cells underwent morphological analysis. Chemically inhibited and QPRT-KO cells were characterized using viability assays. Additionally, QPRT-KO cells underwent metabolite and whole transcriptome analyses. Genes differentially expressed upon KO of QPRT were tested for enrichment in biological processes and co-regulated gene-networks of the human brain. Results: QPRT expression was reduced in the LCL of the deletion carrier and significantly correlated with the neuritic complexity of SH-SY5Y. The reduction of QPRT altered neuronal morphology of differentiated SH-SY5Y cells. Chemical inhibition as well as complete KO of the gene were lethal upon induction of neuronal differentiation, but not proliferation. The QPRT-associated tryptophan pathway was not affected by KO. At the transcriptome level, genes linked to neurodevelopmental processes and synaptic structures were affected. Differentially regulated genes were enriched for ASD candidates, and co-regulated gene networks were implicated in the development of the dorsolateral prefrontal cortex, the hippocampus, and the amygdala. Conclusions: In this study, QPRT was causally related to in vitro neuronal differentiation of SH-SY5Y cells and affected the regulation of genes and gene networks previously implicated in ASD. Thus, our data suggest that QPRT may play an important role in the pathogenesis of ASD in Chr16p11.2 deletion carriers.


Assuntos
Transtorno do Espectro Autista/genética , Diferenciação Celular/genética , Neurônios/citologia , Pentosiltransferases/genética , Linhagem Celular Tumoral , Deleção Cromossômica , Cromossomos Humanos Par 16 , Variações do Número de Cópias de DNA , Humanos
7.
Cancer Res ; 78(7): 1805-1819, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29339541

RESUMO

Lung cancer is the leading cause of cancer-related death worldwide, in large part due to its high propensity to metastasize and to develop therapy resistance. Adaptive responses to hypoxia and epithelial-mesenchymal transition (EMT) are linked to tumor metastasis and drug resistance, but little is known about how oxygen sensing and EMT intersect to control these hallmarks of cancer. Here, we show that the oxygen sensor PHD3 links hypoxic signaling and EMT regulation in the lung tumor microenvironment. PHD3 was repressed by signals that induce EMT and acted as a negative regulator of EMT, metastasis, and therapeutic resistance. PHD3 depletion in tumors, which can be caused by the EMT inducer TGFß or by promoter methylation, enhanced EMT and spontaneous metastasis via HIF-dependent upregulation of the EGFR ligand TGFα. In turn, TGFα stimulated EGFR, which potentiated SMAD signaling, reinforcing EMT and metastasis. In clinical specimens of lung cancer, reduced PHD3 expression was linked to poor prognosis and to therapeutic resistance against EGFR inhibitors such as erlotinib. Reexpression of PHD3 in lung cancer cells suppressed EMT and metastasis and restored sensitivity to erlotinib. Taken together, our results establish a key function for PHD3 in metastasis and drug resistance and suggest opportunities to improve patient treatment by interfering with the feedforward signaling mechanisms activated by PHD3 silencing.Significance: This study links the oxygen sensor PHD3 to metastasis and drug resistance in cancer, with implications for therapeutic improvement by targeting this system. Cancer Res; 78(7); 1805-19. ©2018 AACR.


Assuntos
Antineoplásicos/uso terapêutico , Transição Epitelial-Mesenquimal/genética , Cloridrato de Erlotinib/uso terapêutico , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Fator de Crescimento Transformador alfa/metabolismo , Células A549 , Animais , Proteínas Reguladoras de Apoptose , Hipóxia Celular/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Feminino , Células HCT116 , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/genética , Camundongos , Camundongos Nus , Proteínas Mitocondriais/metabolismo , Metástase Neoplásica/genética , Microambiente Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Acta Neuropathol ; 131(2): 281-298, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26687980

RESUMO

Remyelination in multiple sclerosis (MS) lesions often remains incomplete despite the presence of oligodendrocyte progenitor cells (OPCs). Amongst other factors, successful remyelination depends on the phagocytic clearance of myelin debris. However, the proteins in myelin debris that act as potent and selective inhibitors on OPC differentiation and inhibit CNS remyelination remain unknown. Here, we identify the transmembrane signalling protein EphrinB3 as important mediator of this inhibition, using a protein analytical approach in combination with a primary rodent OPC assay. In the presence of EphrinB3, OPCs fail to differentiate. In a rat model of remyelination, infusion of EphrinB3 inhibits remyelination. In contrast, masking EphrinB3 epitopes using antibodies promotes remyelination. Finally, we identify EphrinB3 in MS lesions and demonstrate that MS lesion extracts inhibit OPC differentiation while antibody-mediated masking of EphrinB3 epitopes promotes it. Our findings suggest that EphrinB3 could be a target for therapies aiming at promoting remyelination in demyelinating disease.


Assuntos
Efrina-B3/metabolismo , Esclerose Múltipla/metabolismo , Bainha de Mielina/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Efrina-B3/genética , Epitopos/metabolismo , Feminino , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Knockout , Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/patologia , Neurogênese/fisiologia , Oligodendroglia/patologia , Distribuição Aleatória , Ratos Sprague-Dawley , Receptor EphA4/metabolismo
9.
Mech Dev ; 138 Pt 1: 17-25, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26192337

RESUMO

During CNS development and adult neurogenesis, immature neurons travel from the germinal zones towards their final destination using cellular substrates for their migration. Classically, radial glia and neuronal axons have been shown to act as physical scaffolds to support neuroblast locomotion in processes known as gliophilic and neurophilic migration, respectively (Hatten, 1999; Marin and Rubenstein, 2003; Rakic, 2003). In adulthood, long distance neuronal migration occurs in a glial-independent manner since radial glia cells differentiate into astrocytes after birth. A series of studies highlight a novel mode of neuronal migration that uses blood vessels as scaffolds, the so-called vasophilic migration. This migration mode allows neuroblast navigation in physiological and also pathological conditions, such as neuronal precursor migration after ischemic stroke or cerebral invasion of glioma tumor cells. Here we review the current knowledge about how vessels pave the path for migrating neurons and how trophic factors derived by glio-vascular structures guide neuronal migration both during physiological as well as pathological processes.


Assuntos
Encéfalo/irrigação sanguínea , Movimento Celular/fisiologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios/citologia , Animais , Humanos , Neurogênese/fisiologia
10.
Nat Commun ; 6: 7671, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26220403

RESUMO

The proteins that coordinate complex adipogenic transcriptional networks are poorly understood. 14-3-3ζ is a molecular adaptor protein that regulates insulin signalling and transcription factor networks. Here we report that 14-3-3ζ-knockout mice are strikingly lean from birth with specific reductions in visceral fat depots. Conversely, transgenic 14-3-3ζ overexpression potentiates obesity, without exacerbating metabolic complications. Only the 14-3-3ζ isoform is essential for adipogenesis based on isoform-specific RNAi. Mechanistic studies show that 14-3-3ζ depletion promotes autophagy-dependent degradation of C/EBP-δ, preventing induction of the master adipogenic factors, Pparγ and C/EBP-α. Transcriptomic data indicate that 14-3-3ζ acts upstream of hedgehog signalling-dependent upregulation of Cdkn1b/p27(Kip1). Indeed, concomitant knockdown of p27(Kip1) or Gli3 rescues the early block in adipogenesis induced by 14-3-3ζ knockdown in vitro. Adipocyte precursors in 14-3-3ζKO embryos also appear to have greater Gli3 and p27(Kip1) abundance. Together, our in vivo and in vitro findings demonstrate that 14-3-3ζ is a critical upstream driver of adipogenesis.


Assuntos
Proteínas 14-3-3/genética , Adipogenia/genética , Gordura Intra-Abdominal/metabolismo , Obesidade/genética , Proteínas 14-3-3/metabolismo , Células 3T3-L1 , Animais , Autofagia/genética , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Immunoblotting , Técnicas In Vitro , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Obesidade/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Proteína Gli3 com Dedos de Zinco
11.
Nat Commun ; 6: 6576, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25807892

RESUMO

Angiogenesis produces primitive vascular networks that need pruning to yield hierarchically organized and functional vessels. Despite the critical importance of vessel pruning to vessel patterning and function, the mechanisms regulating this process are not clear. Here we show that EphrinB2, a well-known player in angiogenesis, is an essential regulator of endothelial cell death and vessel pruning. This regulation depends upon phosphotyrosine-EphrinB2 signalling repressing c-jun N-terminal kinase 3 activity via STAT1. JNK3 activation causes endothelial cell death. In the absence of JNK3, hyaloid vessel physiological pruning is impaired, associated with abnormal persistence of hyaloid vessels, defective retinal vasculature and microphthalmia. This syndrome closely resembles human persistent hyperplastic primary vitreus (PHPV), attributed to failed involution of hyaloid vessels. Our results provide evidence that EphrinB2/STAT1/JNK3 signalling is essential for vessel pruning, and that defects in this pathway may contribute to PHPV.


Assuntos
Células Endoteliais/metabolismo , Efrina-B2/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Neovascularização Fisiológica/genética , Vasos Retinianos/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Imunoprecipitação da Cromatina , Citometria de Fluxo , Imunofluorescência , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Immunoblotting , Imunoprecipitação , Técnicas In Vitro , Camundongos , Camundongos Knockout , Neovascularização Patológica/genética , Vítreo Primário Hiperplásico Persistente/genética , Transdução de Sinais
12.
Stem Cells ; 33(1): 253-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25205248

RESUMO

Nerve cells are continuously generated from stem cells in the adult mammalian subventricular zone (SVZ) and hippocampal dentate gyrus. We have previously noted that stem/progenitor cells in the SVZ and the subgranular layer (SGL) of the dentate gyrus express high levels of plasma membrane-bound nucleoside triphosphate diphosphohydrolase 2 (NTPDase2), an ectoenzyme that hydrolyzes extracellular nucleoside diphosphates and triphosphates. We inferred that deletion of NTPDase2 would increase local extracellular nucleoside triphosphate concentrations perturbing purinergic signaling and boosting progenitor cell proliferation and neurogenesis. Using newly generated mice globally null for Entpd2, we demonstrate that NTPDase2 is the major ectonucleotidase in these progenitor cell-rich areas. Using BrdU-labeling protocols, we have measured stem cell proliferation and determined long-term survival of cell progeny under basal conditions. Brains of Entpd2 null mice revealed increased progenitor cell proliferation in both the SVZ and the SGL. However, this occurred without noteworthy alterations in long-term progeny survival. The hippocampal stem cell pool and the pool of the intermediate progenitor type-2 cells clearly expanded. However, substantive proportions of these proliferating cells were lost during expansion at around type-3 stage. Cell loss was paralleled by decreases in cAMP response element-binding protein phosphorylation in the doublecortin-positive progenitor cell population and by an increase in labeling for activated caspase-3 levels. We propose that NTPDase2 has functionality in scavenging mitogenic extracellular nucleoside triphosphates in neurogenic niches of the adult brain, thereby acting as a homeostatic regulator of nucleotide-mediated neural progenitor cell proliferation and expansion.


Assuntos
Adenosina Trifosfatases/metabolismo , Encéfalo/citologia , Células-Tronco Neurais/citologia , Nicho de Células-Tronco/fisiologia , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Proliferação de Células/fisiologia , Imuno-Histoquímica , Camundongos , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Transdução de Sinais
13.
Purinergic Signal ; 11(1): 155-60, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25504514

RESUMO

Eye formation in vertebrates is controlled by a conserved pattern of molecular networks. Homeobox transcription factors are crucially involved in the establishment and maintenance of the retina. A previous study of Massé et al. (Nature, 449: 1058-62, 2007) using morpholino knockdown identified the ectonucleotidase NTPDase2 and the P2Y1 receptor as essential elements for eye formation in embryos of the clawed frog Xenopus laevis. In order to investigate whether a similarly essential mechanism would be active in mammalian eye development, we analyzed mice KO for Entpd2 or P2ry1 as well as double KO for Entpd2/P2ry1. These mice developed normal eyes. In order to identify potential deficits in the molecular identity or in the arrangement of the cellular elements of the retina, we performed an immunohistological analysis using a variety of retinal markers. The analysis of single and double KO mice demonstrated that NTPDase2 and P2Y1 receptors are not required for murine eye formation, as previously shown for eye development in Xenopus laevis.


Assuntos
Adenosina Trifosfatases/metabolismo , Olho/embriologia , Organogênese/genética , Receptores Purinérgicos P2Y1/metabolismo , Retina/embriologia , Adenosina Trifosfatases/genética , Animais , Olho/metabolismo , Camundongos , Camundongos Knockout , Receptores Purinérgicos P2Y1/genética , Retina/metabolismo
14.
Nat Commun ; 5: 5577, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25420589

RESUMO

Tumours exploit their hypoxic microenvironment to induce a more aggressive phenotype, while curtailing the growth-inhibitory effects of hypoxia through mechanisms that are poorly understood. The prolyl hydroxylase PHD3 is regulated by hypoxia and plays an important role in tumour progression. Here we identify PHD3 as a central regulator of epidermal growth factor receptor (EGFR) activity through the control of EGFR internalization to restrain tumour growth. PHD3 controls EGFR activity by acting as a scaffolding protein that associates with the endocytic adaptor Eps15 and promotes the internalization of EGFR. In consequence, loss of PHD3 in tumour cells suppresses EGFR internalization and hyperactivates EGFR signalling to enhance cell proliferation and survival. Our findings reveal that PHD3 inactivation provides a novel route of EGFR activation to sustain proliferative signalling in the hypoxic microenvironment.


Assuntos
Endocitose , Receptores ErbB/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Neoplasias/enzimologia , Transdução de Sinais , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Neoplasias/genética , Neoplasias/fisiopatologia , Ligação Proteica
15.
Nat Commun ; 5: 5582, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25420773

RESUMO

Solid tumours are exposed to microenvironmental factors such as hypoxia that normally inhibit cell growth. However, tumour cells are capable of counteracting these signals through mechanisms that are largely unknown. Here we show that the prolyl hydroxylase PHD3 restrains tumour growth in response to microenvironmental cues through the control of EGFR. PHD3 silencing in human gliomas or genetic deletion in a murine high-grade astrocytoma model markedly promotes tumour growth and the ability of tumours to continue growing under unfavourable conditions. The growth-suppressive function of PHD3 is independent of the established PHD3 targets HIF and NF-κB and its hydroxylase activity. Instead, loss of PHD3 results in hyperphosphorylation of epidermal growth factor receptor (EGFR). Importantly, epigenetic/genetic silencing of PHD3 preferentially occurs in gliomas without EGFR amplification. Our findings reveal that PHD3 inactivation provides an alternative route of EGFR activation through which tumour cells sustain proliferative signalling even under conditions of limited oxygen availability.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Glioblastoma/fisiopatologia , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Hipóxia/fisiopatologia , Pró-Colágeno-Prolina Dioxigenase/genética , Animais , Apoptose , Linhagem Celular Tumoral , Receptores ErbB/genética , Feminino , Técnicas de Inativação de Genes , Glioblastoma/enzimologia , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Hipóxia/enzimologia , Hipóxia/genética , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/deficiência , Masculino , Camundongos Knockout , Oxigênio/metabolismo , Pró-Colágeno-Prolina Dioxigenase/deficiência
16.
Cell ; 144(4): 566-76, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21335238

RESUMO

TAp63α, a homolog of the p53 tumor suppressor, is a quality control factor in the female germline. Remarkably, already undamaged oocytes express high levels of the protein, suggesting that TAp63α's activity is under tight control of an inhibitory mechanism. Biochemical studies have proposed that inhibition requires the C-terminal transactivation inhibitory domain. However, the structural mechanism of TAp63α inhibition remains unknown. Here, we show that TAp63α is kept in an inactive dimeric state. We reveal that relief of inhibition leads to tetramer formation with ∼20-fold higher DNA affinity. In vivo, phosphorylation-triggered tetramerization of TAp63α is not reversible by dephosphorylation. Furthermore, we show that a helix in the oligomerization domain of p63 is crucial for tetramer stabilization and competes with the transactivation domain for the same binding site. Our results demonstrate how TAp63α is inhibited by complex domain-domain interactions that provide the basis for regulating quality control in oocytes.


Assuntos
Oócitos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Transativadores/química , Transativadores/metabolismo , Animais , DNA/metabolismo , Dimerização , Feminino , Raios gama , Camundongos , Modelos Moleculares , Fosforilação , Multimerização Proteica , Proteína Supressora de Tumor p53/metabolismo
17.
Spermatogenesis ; 1(4): 354-365, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22332119

RESUMO

The 14-3-3 family of proteins interacts with various cellular phosphoproteins and regulates multiple cell signaling cascades. Identification of 14-3-3 interactors is important to define 14-3-3 functions in various biological pathways. The binding partners of protein 14-3-3 in testis are not known. The main goal of this study was to identify the 14-3-3 interactome in testis to determine the 14-3-3 regulated cellular processes in testis. We used transgenic mice expressing tandem affinity tagged 14-3-3ζ (TAP-14-3-3ζ) driven by the ubiquitin promoter to isolate 14-3-3 binding proteins. The 14-3-3 complexes in testis were isolated using a two-step tandem affinity purification (TAP) followed by identification with liquid chromatography/tandem mass spectrometry (LC-MS/MS). A total of 135 proteins were found to be associated with 14-3-3 in vivo in testis. Comparison of the testis 14-3-3 proteome with known 14-3-3 binding proteins showed that 71 of the proteins identified in this study are novel 14-3-3 interactors. Eight of these novel 14-3-3 interacting proteins are predominantly expressed in testis. The 14-3-3 interactors predominant in testis are: protein phosphatase1γ2 (PP1γ2), spermatogenesis associated 18 (SPATA18), phosphoglycerate kinase-2 (PGK2), testis specific gene A-2 (TSGA-2), dead box polypeptide 4 (DDX4), piwi homolog 1, protein kinase NYD-SP25 and EAN57. The fact that some of these proteins are indispensable for spermatogenesis suggests that their binding to 14-3-3 may be important for their function in germ cell division and maturation. These findings are discussed in context of the putative functions of 14-3-3 in spermatogenesis.

18.
Nature ; 465(7297): 487-91, 2010 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-20445540

RESUMO

The formation and guidance of specialized endothelial tip cells is essential for both developmental and pathological angiogenesis. Notch-1 signalling regulates the generation of tip cells, which respond to gradients of vascular endothelial growth factor (VEGF-A). The molecular cues and signalling pathways that control the guidance of tip cells are poorly understood. Bidirectional signalling by Eph receptors and ephrin ligands represents one of the most important guidance cues involved in axon path finding. Here we show that ephrin-B2 reverse signalling involving PDZ interactions regulates endothelial tip cell guidance to control angiogenic sprouting and branching in physiological and pathological angiogenesis. In vivo, ephrin-B2 PDZ-signalling-deficient mice (ephrin-B2DeltaV) exhibit a reduced number of tip cells with fewer filopodial extensions at the vascular front in the mouse retina. In pathological settings, impaired PDZ signalling decreases tumour vascularization and growth. Mechanistically, we show that ephrin-B2 controls VEGF receptor (VEGFR)-2 internalization and signalling. Importantly, internalization of VEGFR2 is necessary for activation and downstream signalling of the receptor and is required for VEGF-induced tip cell filopodial extension. Together, our results suggest that ephrin-B2 at the tip cell filopodia regulates the proper spatial activation of VEGFR2 endocytosis and signalling to direct filopodial extension. Blocking ephrin-B2 reverse signalling may be an attractive alternative or combinatorial anti-angiogenic therapy strategy to disrupt VEGFR2 function in tumour angiogenesis.


Assuntos
Astrocitoma/irrigação sanguínea , Astrocitoma/metabolismo , Efrina-B2/metabolismo , Neovascularização Patológica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Astrocitoma/patologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Endocitose , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Efrina-B2/deficiência , Efrina-B2/genética , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neovascularização Fisiológica , Pseudópodes/metabolismo , Retina , Vasos Retinianos/citologia , Vasos Retinianos/fisiologia , Transdução de Sinais
19.
Nat Neurosci ; 10(3): 301-10, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17310244

RESUMO

Dendritic spines are small protrusions emerging from dendrites that receive excitatory input. The process of spine morphogenesis occurs both in the developing brain and during synaptic plasticity. Molecules regulating the cytoskeleton are involved in spine formation and maintenance. Here we show that reverse signaling by the transmembrane ligands for Eph receptors, ephrinBs, is required for correct spine morphogenesis. The molecular mechanism underlying this function of ephrinBs involves the SH2 and SH3 domain-containing adaptor protein Grb4 and the G protein-coupled receptor kinase-interacting protein (GIT) 1. Grb4 binds by its SH2 domain to Tyr392 in the synaptic localization domain of GIT1. Phosphorylation of Tyr392 and the recruitment of GIT1 to synapses are regulated by ephrinB activation. Disruption of this pathway in cultured rat hippocampal neurons impairs spine morphogenesis and synapse formation. We thus show an important role for ephrinB reverse signaling in spine formation and have mapped the downstream pathway involved in this process.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ciclo Celular/fisiologia , Espinhas Dendríticas/fisiologia , Proteínas Oncogênicas/fisiologia , Receptores da Família Eph/fisiologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Hipocampo/citologia , Humanos , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/metabolismo , Mutagênese/fisiologia , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção
20.
Circ Res ; 94(6): 743-51, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14764452

RESUMO

Homeobox genes (Hox) encode for transcription factors, which regulate cell proliferation and migration and play an important role in the development of the cardiovascular system during embryogenesis. In this study, we investigated the role of HoxA9 for endothelial cell migration and angiogenesis in vitro and identified a novel target gene, the EphB4 receptor. Inhibition of HoxA9 expression decreased endothelial cell tube formation and inhibited endothelial cell migration, suggesting that HoxA9 regulates angiogenesis. Because Eph receptor tyrosine kinases importantly contribute to angiogenesis, we examined whether HoxA9 may transcriptionally regulate the expression of EphB4. Downregulation of HoxA9 reduced the expression of EphB4. Chromatin-immunoprecipitation revealed that HoxA9 interacted with the EphB4 promoter, whereas a deletion construct of HoxA9 without DNA-binding motif (Delta(aa) 206-272) did not bind. Consistently, HoxA9 wild-type overexpression activated the EphB4 promoter as determined by reporter gene expression. HoxA9 binds to the EphB4 promoter and stimulates its expression resulting in an increase of endothelial cell migration and tube forming activity. Thus, modulation of EphB4 expression may contribute to the proangiogenic effect of HoxA9 in endothelial cells.


Assuntos
Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Neovascularização Fisiológica/fisiologia , Receptor EphB4/fisiologia , Transcrição Gênica , Movimento Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Humanos , Morfogênese , Mutagênese Sítio-Dirigida , Neovascularização Fisiológica/genética , Oligonucleotídeos Antissenso/farmacologia , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptor EphB4/biossíntese , Receptor EphB4/genética , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA