Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS Genet ; 14(10): e1007688, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30325918

RESUMO

Oncogenic mutations in the small GTPase Ras contribute to ~30% of human cancers. However, Ras mutations alone are insufficient for tumorigenesis, therefore it is paramount to identify cooperating cancer-relevant signaling pathways. We devised an in vivo near genome-wide, functional screen in Drosophila and discovered multiple novel, evolutionarily-conserved pathways controlling Ras-driven epithelial tumorigenesis. Human gene orthologs of the fly hits were significantly downregulated in thousands of primary tumors, revealing novel prognostic markers for human epithelial tumors. Of the top 100 candidate tumor suppressor genes, 80 were validated in secondary Drosophila assays, identifying many known cancer genes and multiple novel candidate genes that cooperate with Ras-driven tumorigenesis. Low expression of the confirmed hits significantly correlated with the KRASG12 mutation status and poor prognosis in pancreatic cancer. Among the novel top 80 candidate cancer genes, we mechanistically characterized the function of the top hit, the Tetraspanin family member Tsp29Fb, revealing that Tsp29Fb regulates EGFR signaling, epithelial architecture and restrains tumor growth and invasion. Our functional Drosophila screen uncovers multiple novel and evolutionarily conserved epithelial cancer genes, and experimentally confirmed Tsp29Fb as a key regulator of EGFR/Ras induced epithelial tumor growth and invasion.


Assuntos
Proteínas de Drosophila/genética , IMP Desidrogenase/genética , Neoplasias/genética , Tetraspanina 29/genética , Animais , Animais Geneticamente Modificados , Carcinogênese/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Genes ras , Testes Genéticos/métodos , Humanos , IMP Desidrogenase/metabolismo , Masculino , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Oncogenes , Transdução de Sinais , Tetraspanina 29/metabolismo , Proteínas Supressoras de Tumor/genética
2.
Genome Res ; 25(11): 1692-702, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26294687

RESUMO

In eukaryotic cells, RNAs exist as ribonucleoprotein particles (RNPs). Despite the importance of these complexes in many biological processes, including splicing, polyadenylation, stability, transportation, localization, and translation, their compositions are largely unknown. We affinity-purified 20 distinct RNA-binding proteins (RBPs) from cultured Drosophila melanogaster cells under native conditions and identified both the RNA and protein compositions of these RNP complexes. We identified "high occupancy target" (HOT) RNAs that interact with the majority of the RBPs we surveyed. HOT RNAs encode components of the nonsense-mediated decay and splicing machinery, as well as RNA-binding and translation initiation proteins. The RNP complexes contain proteins and mRNAs involved in RNA binding and post-transcriptional regulation. Genes with the capacity to produce hundreds of mRNA isoforms, ultracomplex genes, interact extensively with heterogeneous nuclear ribonuclear proteins (hnRNPs). Our data are consistent with a model in which subsets of RNPs include mRNA and protein products from the same gene, indicating the widespread existence of auto-regulatory RNPs. From the simultaneous acquisition and integrative analysis of protein and RNA constituents of RNPs, we identify extensive cross-regulatory and hierarchical interactions in post-transcriptional control.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas de Drosophila/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Splicing de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Análise de Sequência de RNA , Transfecção
3.
Proc Natl Acad Sci U S A ; 108(21): E128-35, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21555590

RESUMO

The most common monogenic cause of small-vessel disease leading to ischemic stroke and vascular dementia is the neurodegenerative syndrome cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), which is associated with mutations in the Notch 3 receptor. CADASIL pathology is characterized by vascular smooth muscle cell degeneration and accumulation of diagnostic granular osmiophilic material (GOM) in vessels. The functional nature of the Notch 3 mutations causing CADASIL and their mechanistic connection to small-vessel disease and GOM accumulation remain enigmatic. To gain insight into how Notch 3 function is linked to CADASIL pathophysiology, we studied two phenotypically distinct mutations, C455R and R1031C, respectively associated with early and late onset of stroke, by using hemodynamic analyses in transgenic mouse models, receptor activity assays in cell culture, and proteomic examination of postmortem human tissue. We demonstrate that the C455R and R1031C mutations define different hypomorphic activity states of Notch 3, a property linked to ischemic stroke susceptibility in mouse models we generated. Importantly, these mice develop osmiophilic deposits and other age-dependent phenotypes that parallel remarkably the human condition. Proteomic analysis of human brain vessels, carrying the same CADASIL mutations, identified clusterin and collagen 18 α1/endostatin as GOM components. Our findings link loss of Notch signaling with ischemic cerebral small-vessel disease, a prevalent human condition. We determine that CADASIL pathophysiology is associated with hypomorphic Notch 3 function in vascular smooth muscle cells and implicate the accumulation of clusterin and collagen 18 α1/endostatin in brain vessel pathology.


Assuntos
Alelos , Arteríolas/patologia , Transtornos Cerebrovasculares/etiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Encéfalo/irrigação sanguínea , Modelos Animais de Doenças , Humanos , Isquemia , Camundongos , Mutação de Sentido Incorreto , Receptor Notch3 , Receptores Notch/genética , Transgenes
4.
Nat Commun ; 13(1): 243, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017475

RESUMO

The vertebral column of individual mammalian species often exhibits remarkable robustness in the number and identity of vertebral elements that form (known as axial formulae). The genetic mechanism(s) underlying this constraint however remain ill-defined. Here, we reveal the interplay of three regulatory pathways (Gdf11, miR-196 and Retinoic acid) is essential in constraining total vertebral number and regional axial identity in the mouse, from cervical through to tail vertebrae. All three pathways have differing control over Hox cluster expression, with heterochronic and quantitative changes found to parallel changes in axial identity. However, our work reveals an additional role for Hox genes in supporting axial elongation within the tail region, providing important support for an emerging view that mammalian Hox function is not limited to imparting positional identity as the mammalian body plan is laid down. More broadly, this work provides a molecular framework to interrogate mechanisms of evolutionary change and congenital anomalies of the vertebral column.


Assuntos
Padronização Corporal/fisiologia , Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Diferenciação de Crescimento/metabolismo , MicroRNAs/metabolismo , Coluna Vertebral/metabolismo , Tretinoína/metabolismo , Animais , Evolução Biológica , Padronização Corporal/genética , Proteínas Morfogenéticas Ósseas/genética , Genes Homeobox , Fatores de Diferenciação de Crescimento/genética , Proteínas de Homeodomínio , Mamíferos , Camundongos , MicroRNAs/genética , Cauda/metabolismo , Transcriptoma
5.
Nat Commun ; 13(1): 7766, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36522318

RESUMO

The vertebrate main-body axis is laid down during embryonic stages in an anterior-to-posterior (head-to-tail) direction, driven and supplied by posteriorly located progenitors. Whilst posterior expansion and segmentation appears broadly uniform along the axis, there is developmental and evolutionary support for at least two discrete modules controlling processes within different axial regions: a trunk and a tail module. Here, we identify Nuclear receptor subfamily 6 group A member 1 (Nr6a1) as a master regulator of trunk development in the mouse. Specifically, Nr6a1 was found to control vertebral number and segmentation of the trunk region, autonomously from other axial regions. Moreover, Nr6a1 was essential for the timely progression of Hox signatures, and neural versus mesodermal cell fate choice, within axial progenitors. Collectively, Nr6a1 has an axially-restricted role in all major cellular and tissue-level events required for vertebral column formation, supporting the view that changes in Nr6a1 levels may underlie evolutionary changes in axial formulae.


Assuntos
Mesoderma , Vertebrados , Animais , Camundongos , Vertebrados/genética , Coluna Vertebral , Regulação da Expressão Gênica no Desenvolvimento , Padronização Corporal/genética
6.
Glia ; 57(13): 1450-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19243017

RESUMO

Neural crest (NC) cells are a multipotent, highly migratory cell population that generates most of the components of the peripheral nervous system (PNS), including the glial Schwann cells (SC) and boundary cap (BC) cells. These latter cells are located at the interface between the central nervous system and PNS, at the exit/entry points of ventral motor/dorsal sensory axons and give rise to all SC in the nerve roots and to a subset of nociceptive neurons and satellite cells in the dorsal root ganglia. In the present study we have compared BC cells with two closely related cell types, NC and Schwann cell precursors (SCpr), by RNA profiling. This led to the definition of a set of 10 genes that show specific expression in BC cells and/or in their derivatives along the nerve roots. Analysis of the expression of these genes during mouse development revealed novel features, of those most important are: (i) dorsal and ventral nerve root BC cell derivatives express different sets of genes, suggesting that they have distinct properties; (ii) these cells undergo major modifications in their gene expression pattern between embryonic days 14.5 and 17.5, possibly linked to the SCpr-immature Schwann cell transition; (iii) nerve roots SC differ from more distal SC not only in their origins and locations, but also in their gene expression patterns. In conclusion, the identification of these novel makers opens the way for a detailed characterization of BC cells in both mouse and man.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/embriologia , Crista Neural/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA/metabolismo , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Células de Schwann/fisiologia , Raízes Nervosas Espinhais/embriologia , Raízes Nervosas Espinhais/fisiologia , Células-Tronco/fisiologia
7.
Cancer Res ; 67(2): 520-7, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234759

RESUMO

The small G-protein Ras is a tightly controlled regulator of cell fate. Prolonged or persistent arrest in the activated GTP-loaded state by mutation of Ras as in lung cancer or in a Ras-GTPase-activating protein as in neurofibromatosis type 1 promotes tumorigenesis. We now show that the tumor-suppressor protein merlin (mutated in neurofibromatosis type 2) also controls Ras activity. Systematic analysis of growth factor signaling located the step of merlin interference to the activation of Ras and Rac. Merlin independently uncouples both Ras and Rac from growth factor signals. In the case of Ras, merlin acts downstream of the receptor tyrosine kinase-growth factor receptor binding protein 2 (Grb2)-SOS complex. However, merlin does not bind either SOS or Ras, but it counteracts the ERM (ezrin, radixin, moesin)-dependent activation of Ras, which correlates with the formation of a complex comprising ERM proteins, Grb2, SOS, Ras, and filamentous actin. Because efficient signaling from Ras requires Rac-p21-activated kinase-dependent phosphorylations of Raf and mitogen-activated protein/extracellular signal-regulated kinase kinase, merlin can also inhibit signal transfer from dominantly active Ras mutants. We propose that the interference of merlin with Ras- and Rac-dependent signal transfer represents part of the tumor-suppressive action of merlin.


Assuntos
Neurofibromina 2/metabolismo , Proteínas rac de Ligação ao GTP/antagonistas & inibidores , Proteínas ras/antagonistas & inibidores , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Células NIH 3T3 , Neurilemoma , Neurofibromina 2/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos , Proteínas Son Of Sevenless/metabolismo , Transfecção , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo
8.
Methods Mol Biol ; 1940: 47-61, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30788817

RESUMO

Lentiviral gene transfer technologies exploit the natural efficiency of viral transduction to integrate exogenous genes into mammalian cells. This provides a simple research tool for inducing transgene expression or endogenous gene knockdown in both dividing and nondividing cells. This chapter describes an improved protocol for polyethylenimine (PEI)-mediated multi-plasmid transfection and polyethylene glycol (PEG) precipitation to generate and concentrate lentiviral vectors.


Assuntos
Fibroblastos/virologia , Técnicas de Transferência de Genes , Lentivirus/crescimento & desenvolvimento , Lentivirus/genética , Plasmídeos/genética , Animais , Linhagem Celular , Vetores Genéticos/genética , Células HEK293 , Humanos , Camundongos , Polietilenoglicóis/química , Polietilenoimina/química
9.
Clin Exp Metastasis ; 25(1): 53-64, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17952617

RESUMO

The integrin alpha(v)beta(3) is involved in multiple aspects of malignant cancer, including tumor angiogenesis and metastasis, which makes the receptor a key target for the development of anti-cancer therapies. We report here on the production, the characterization and the in vivo anti-angiogenic and anti-metastatic properties of a novel alpha(v)beta(3)-binding disintegrin, DisBa-01, isolated from a cDNA library made with RNAs from the venom gland of Bothrops alternatus. The 11,637 Da-recombinant monomeric form of DisBa-01 displayed an RGD motif and interacted with purified alpha(v)beta(3) integrin in surface plasmon resonance studies, in a dose-dependent and cation sensitive manner. A three-dimensional molecular model of DisBa-01 in complex with alpha(v)beta(3) predicted a large surface of contacts with the beta(3) subunit. DisBa-01 inhibited the adhesion of alpha(v)beta(3)-expressing human microvascular endothelial cell line-1 (HMEC-1) and murine melanoma cell line B16F10 to vitronectin (IC(50) = 555 nM and 225 nM, respectively), and transiently inhibited their proliferation without direct cell toxicity, but did not affect the binding nor the proliferation of a human breast cancer-derived cell line (MDA-MB-231) not expressing alpha(v)beta(3). In vivo, DisBa-01 dose-dependently decreased bFGF-induced angiogenesis in a matrigel plug assay in athymic nude mice (IC(50) = 83 nM). When injected intravenously to C57BL/6 mice together with B16F10 melanoma cells, DisBa-01 time- and dose-dependently inhibited lung metastasis monitored by bioluminescent imaging. We conclude that DisBa-01 is a potent new inhibitor of alpha(v)beta(3)-dependent adherence mechanisms involved in neo-vascularization and tumor metastasis processes.


Assuntos
Venenos de Crotalídeos/farmacologia , Desintegrinas/farmacologia , Integrina alfaVbeta3/antagonistas & inibidores , Melanoma Experimental/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Sequência de Bases , Bothrops , Adesão Celular/efeitos dos fármacos , Clonagem Molecular , Venenos de Crotalídeos/química , Desintegrinas/química , Desintegrinas/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Integrina alfaVbeta3/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
10.
FEBS J ; 284(14): 2231-2250, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28544778

RESUMO

Tyrosine phosphorylation-dependent signalling is coordinated by the opposing actions of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). There is a growing list of adaptor proteins that interact with PTPs and facilitate the dephosphorylation of substrates. The extent to which any given adaptor confers selectivity for any given substrate in vivo remains unclear. Here we have taken advantage of Drosophila melanogaster as a model organism to explore the influence of the SH3/SH2 adaptor protein Dock on the abilities of the membrane (PTP61Fm)- and nuclear (PTP61Fn)-targeted variants of PTP61F (the Drosophila othologue of the mammalian enzymes PTP1B and TCPTP respectively) to repress PTK signalling pathways in vivo. PTP61Fn effectively repressed the eye overgrowth associated with activation of the epidermal growth factor receptor (EGFR), PTK, or the expression of the platelet-derived growth factor/vascular endothelial growth factor receptor (PVR) or insulin receptor (InR) PTKs. PTP61Fn repressed EGFR and PVR-induced mitogen-activated protein kinase signalling and attenuated PVR-induced STAT92E signalling. By contrast, PTP61Fm effectively repressed EGFR- and PVR-, but not InR-induced tissue overgrowth. Importantly, coexpression of Dock with PTP61F allowed for the efficient repression of the InR-induced eye overgrowth, but did not enhance the PTP61Fm-mediated inhibition of EGFR and PVR-induced signalling. Instead, Dock expression increased, and PTP61Fm coexpression further exacerbated the PVR-induced eye overgrowth. These results demonstrate that Dock selectively enhances the PTP61Fm-mediated attenuation of InR signalling and underscores the specificity of PTPs and the importance of adaptor proteins in regulating PTP function in vivo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Masculino , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Receptores Proteína Tirosina Quinases/genética , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo
11.
Brain Res ; 1644: 118-26, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27174004

RESUMO

Mutations in NOTCH 3 are the cause of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a neurological disorder characterized by stroke, and vascular cognitive impairment and dementia. Loss of vascular smooth muscle cells (VSMC) and accumulation of granular osmiophilic material (GOM) deposits are hallmarks of CADASIL. There are no therapies for CADASIL and experimental endpoints to examine the preclinical efficacy of potential drugs are lacking. This study aims to use a mouse carrying the C455R mutation in Notch 3 to identify biomarkers associated with CADASIL. Mass spectrometry and antibody arrays were used to explore the aorta and blood proteomes of CADASIL mice, ELISA assays were utilized for biomarker validation, a ligand-dependent assay was applied to examine the relationship between Notch signaling and biomarker expression, and retinal histology was performed for quantification of VSMC loss in arteries. Two-hundred day-old mice with the C455R CADASIL mutation in Notch 3 mice display robust VSMC loss in retinal arteries and had increased plasma levels of collagen18α1/endostatin (col18α1) and high-temperature requirement A serine peptidase 1 (HTRA1) and reduced levels of Notch 3 extracellular domain (N3ECD), compared to control wild type mice. Measurements of plasma endostatin, HTRA1 and N3ECD, along with VSMC quantification in retinal arteries, may serve as surrogate endpoints for assessing efficacy in preclinical therapeutic studies of CADASIL using mice.


Assuntos
CADASIL/sangue , CADASIL/diagnóstico , Receptor Notch3/genética , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Endostatinas/sangue , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/patologia , Fenótipo , Proteômica , Artéria Retiniana/patologia , Serina Endopeptidases/sangue
12.
J Neurosci Methods ; 123(2): 167-73, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12606065

RESUMO

We have devised a simple method to purify mitotically active Schwann cells (SC) from peripheral nerves of adult mice. Nerves were predegenerated in vitro for 7 days and after dissociation cells were plated on poly-L-lysine/laminin coated dishes in N2 serum-free culture medium supplemented with forskolin and heregulin-beta1. Primary cultures were purified from contaminating fibroblasts by magnetic cell sorting (MACS) based on SC membrane specific expression of p75(NGFR) and enriched to about 99% of SC after MACS from 34 to 91% before sorting. After sorting, purified adult mouse SC were propagated for three passages until confluent to a total surface of 160 cm(2) per mouse (two sciatic and two trigeminal nerves). In addition, we show that this method can be used to purify tumoral SC from mouse NF2-related schwannomas.


Assuntos
Citometria de Fluxo/métodos , Magnetismo/instrumentação , Nervos Periféricos/citologia , Células de Schwann/citologia , Animais , Divisão Celular/fisiologia , Células Cultivadas , Camundongos , Nervos Periféricos/metabolismo , Células de Schwann/metabolismo
13.
Invest Ophthalmol Vis Sci ; 55(8): 5191-9, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-25015359

RESUMO

PURPOSE: Pericytes, the vascular cells that constitute the outer layer of capillaries, have been shown to have a crucial role in vascular development and stability. Loss of pericytes precedes endothelial cell dysfunction and vascular degeneration in small-vessel diseases, including diabetic retinopathy. Despite their clinical relevance, the cellular pathways controlling survival of retinal pericytes remain largely uncharacterized. Therefore, we investigated the role of Notch signaling, a master regulator of cell fate decisions, in retinal pericyte survival. METHODS: A coculture system of ligand-dependent Notch signaling was developed using primary cultured retinal pericytes and a mesenchymal cell line derived from an inducible mouse model expressing the Delta-like 1 Notch ligand. This model was used to examine the effect of Notch activity on pericyte survival using quantitative PCR (qPCR) and a light-induced cell death assay. The effect of Notch gain- and loss-of-function was analyzed in monocultures of retinal pericytes using antibody arrays to interrogate the expression of apoptosis-related proteins. RESULTS: Primary cultured retinal pericytes differentially expressed key molecules of the Notch pathway and displayed strong expression of canonical Notch/RBPJK (recombination signal-binding protein 1 for J-kappa) downstream targets. A gene expression screen using gain- and loss-of-function approaches identified genes relevant to cell survival as downstream targets of Notch activity in retinal pericytes. Ligand-mediated Notch activity protected retinal pericytes from light-induced cell death. CONCLUSIONS: Our results have identified signature genes downstream of Notch activity in retinal pericytes and suggest that tight regulation of Notch signaling is crucial for pericyte survival.


Assuntos
Sobrevivência Celular/fisiologia , Pericitos/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Notch/fisiologia , Retina/citologia , Animais , Bovinos , Técnicas de Cocultura , Células-Tronco Mesenquimais/fisiologia , Modelos Animais , Reação em Cadeia da Polimerase/métodos , Retina/metabolismo , Transdução de Sinais/fisiologia
14.
Clin Cancer Res ; 19(14): 3856-70, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23714726

RESUMO

PURPOSE: The growth and survival of neurofibromatosis type 2 (NF2)-deficient cells are enhanced by the activation of multiple signaling pathways including ErbBs/IGF-1R/Met, PI3K/Akt, and Ras/Raf/Mek/Erk1/2. The chaperone protein HSP90 is essential for the stabilization of these signaling molecules. The aim of the study was to characterize the effect of HSP90 inhibition in various NF2-deficient models. EXPERIMENTAL DESIGN: We tested efficacy of the small-molecule NXD30001, which has been shown to be a potent HSP90 inhibitor. The antiproliferative activity of NXD30001 was tested in NF2-deficient cell lines and in human primary schwannoma and meningioma cultures in vitro. The antitumor efficacy of HSP90 inhibition in vivo was verified in two allograft models and in one NF2 transgenic model. The underlying molecular alteration was further characterized by a global transcriptome approach. RESULTS: NXD30001 induced degradation of client proteins in and suppressed proliferation of NF2-deficient cells. Differential expression analysis identified subsets of genes implicated in cell proliferation, cell survival, vascularization, and Schwann cell differentiation whose expression was altered by NXD30001 treatment. The results showed that NXD30001 in NF2-deficient schwannoma suppressed multiple pathways necessary for tumorigenesis. CONCLUSIONS: HSP90 inhibition showing significant antitumor activity against NF2-related tumor cells in vitro and in vivo represents a promising option for novel NF2 therapies.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Lactonas/farmacologia , Neurofibromatose 2/tratamento farmacológico , Oximas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Neurofibromatose 2/metabolismo , Proteólise , Transcriptoma/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Cell Biol ; 192(3): 481-95, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21300852

RESUMO

Spinal muscular atrophy (SMA), a devastating neurodegenerative disorder characterized by motor neuron loss and muscle atrophy, has been linked to mutations in the Survival Motor Neuron (SMN) gene. Based on an SMA model we developed in Drosophila, which displays features that are analogous to the human pathology and vertebrate SMA models, we functionally linked the fibroblast growth factor (FGF) signaling pathway to the Drosophila homologue of SMN, Smn. Here, we characterize this relationship and demonstrate that Smn activity regulates the expression of FGF signaling components and thus FGF signaling. Furthermore, we show that alterations in FGF signaling activity are able to modify the neuromuscular junction defects caused by loss of Smn function and that muscle-specific activation of FGF is sufficient to rescue Smn-associated abnormalities.


Assuntos
Proteínas de Drosophila/genética , Drosophila/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Atrofia Muscular Espinal/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Animais , Drosophila/genética , Proteínas de Drosophila/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , Proteínas de Ligação a RNA/metabolismo
16.
J Soc Biol ; 201(3): 223-8, 2007.
Artigo em Francês | MEDLINE | ID: mdl-18157073

RESUMO

Animal models allowing more sensitive and early detection of tumorigenesis and metastasis are instrumental in the fight for developing effective therapies against aggressive forms of cancer. In the present chapter, the advantages and limitations of the bioluminescent imaging (BLI) approach are discussed. Although BLI provides rapid, highly sensitive, noninvasive and quantitative detection of small tumors and micrometastases, several issues like the low anatomic resolution or the attenuation of the luminescent signal with tissue depth must be considered when using this technology.


Assuntos
Medições Luminescentes/métodos , Neoplasias/fisiopatologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Metástase Neoplásica , Neoplasias/patologia , Sensibilidade e Especificidade
17.
J Biol Chem ; 277(35): 31279-82, 2002 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-12130630

RESUMO

Neurofibromatosis type 2 (NF2), a syndrome associated with multiple tumors of the nervous system, mostly schwannomas, is caused by mutations in the NF2 tumor suppressor gene that encodes schwannomin (Sch). Here we examined NF2 pathogenetic mutations that result in misfolding of the FERM domain. We found that these mutant forms of Sch were efficiently degraded by the ubiquitin-proteasome pathway. In transfected cells, Sch Delta F118 was 3-fold more efficiently degraded than the related molecule ezrin bearing the equivalent mutation. In heterozygous Nf2 knock-out mouse fibroblasts, endogenous mutant Sch Delta 81-121, but not wild type Sch, was also degraded by proteasomes. We further show that this degradation pathway is functional in primary Schwann cells. We analyzed Sch Delta 39-121 expressed in a transgenic mouse model of NF2 and found that Sch Delta 39-121, but not the endogenous wild type Sch, was unstable due to proteasome-mediated degradation. Altogether these results suggest that degradation of mutant Sch mediated by the ubiquitin-proteasome pathway is a physiopathological pathway contributing to the loss of Sch function in NF2 patients.


Assuntos
Adenosina Trifosfatases/administração & dosagem , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Ubiquitina/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Linhagem Celular , Fibroblastos/metabolismo , Genes da Neurofibromatose 2 , Cinética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurofibromina 2/deficiência , Complexo de Endopeptidases do Proteassoma , Proteínas Recombinantes/metabolismo , Células de Schwann/metabolismo , Deleção de Sequência , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA