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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 131(18)2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30158176

RESUMO

Mammalian members of the ErbB family, including the epidermal growth factor receptor (EGFR), can regulate transcription, DNA replication and repair through nuclear entry of either the full-length proteins or their cleaved cytoplasmic domains. In cancer cells, these nuclear functions contribute to tumor progression and drug resistance. Here, we examined whether the single Drosophila EGFR can also localize to the nucleus. A chimeric EGFR protein fused at its cytoplasmic C-terminus to DNA-binding and transcriptional activation domains strongly activated transcriptional reporters when overexpressed in cultured cells or in vivo However, this activity was independent of cleavage and endocytosis. Without an exogenous activation domain, EGFR fused to a DNA-binding domain did not activate or repress transcription. Addition of the same DNA-binding and transcriptional activation domains to the endogenous Egfr locus through genome editing led to no detectable reporter expression in wild-type or oncogenic contexts. These results show that, when expressed at physiological levels, the cytoplasmic domain of the Drosophila EGFR does not have access to the nucleus. Therefore, nuclear EGFR functions are likely to have evolved after vertebrates and invertebrates diverged.


Assuntos
Núcleo Celular/metabolismo , Drosophila/metabolismo , Receptores ErbB/metabolismo , Animais
2.
Development ; 144(14): 2673-2682, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28619822

RESUMO

The COP9 signalosome removes Nedd8 modifications from the Cullin subunits of ubiquitin ligase complexes, reducing their activity. Here, we show that mutations in the Drosophila COP9 signalosome subunit 1b (CSN1b) gene increase the activity of ubiquitin ligases that contain Cullin 1. Analysis of CSN1b mutant phenotypes revealed a requirement for the COP9 signalosome to prevent ectopic expression of Epidermal growth factor receptor (EGFR) target genes. It does so by protecting Capicua, a transcriptional repressor of EGFR target genes, from EGFR pathway-dependent ubiquitylation by a Cullin 1/SKP1-related A/Archipelago E3 ligase and subsequent proteasomal degradation. The CSN1b subunit also maintains basal Capicua levels by protecting it from a separate mechanism of degradation that is independent of EGFR signaling. As a suppressor of tumor growth and metastasis, Capicua may be an important target of the COP9 signalosome in cancer.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas HMGB/metabolismo , Complexos Multiproteicos/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Animais Geneticamente Modificados , Complexo do Signalossomo COP9 , Proteínas Culina/genética , Proteínas Culina/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/crescimento & desenvolvimento , Receptores ErbB/genética , Receptores ErbB/metabolismo , Olho/crescimento & desenvolvimento , Olho/metabolismo , Feminino , Genes de Insetos , Proteínas HMGB/genética , Sistema de Sinalização das MAP Quinases , Masculino , Modelos Biológicos , Complexos Multiproteicos/genética , Mutação , Peptídeo Hidrolases/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteólise , Receptores de Peptídeos de Invertebrados/genética , Receptores de Peptídeos de Invertebrados/metabolismo , Proteínas Repressoras/genética , Ubiquitinação , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo
3.
Development ; 142(8): 1480-91, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25790850

RESUMO

Endocytic trafficking of signaling receptors is an important mechanism for limiting signal duration. Components of the Endosomal Sorting Complexes Required for Transport (ESCRT), which target ubiquitylated receptors to intra-lumenal vesicles (ILVs) of multivesicular bodies, are thought to terminate signaling by the epidermal growth factor receptor (EGFR) and direct it for lysosomal degradation. In a genetic screen for mutations that affect Drosophila eye development, we identified an allele of Vacuolar protein sorting 4 (Vps4), which encodes an AAA ATPase that interacts with the ESCRT-III complex to drive the final step of ILV formation. Photoreceptors are largely absent from Vps4 mutant clones in the eye disc, and even when cell death is genetically prevented, the mutant R8 photoreceptors that develop fail to recruit surrounding cells to differentiate as R1-R7 photoreceptors. This recruitment requires EGFR signaling, suggesting that loss of Vps4 disrupts the EGFR pathway. In imaginal disc cells mutant for Vps4, EGFR and other receptors accumulate in endosomes and EGFR target genes are not expressed; epistasis experiments place the function of Vps4 at the level of the receptor. Surprisingly, Vps4 is required for EGFR signaling even in the absence of Shibire, the Dynamin that internalizes EGFR from the plasma membrane. In ovarian follicle cells, in contrast, Vps4 does not affect EGFR signaling, although it is still essential for receptor degradation. Taken together, these findings indicate that Vps4 can promote EGFR activity through an endocytosis-independent mechanism.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Drosophila/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Receptores ErbB/metabolismo , Adenosina Trifosfatases/genética , Animais , Drosophila , Proteínas de Drosophila/genética , Dinaminas/genética , Dinaminas/metabolismo , Endocitose/genética , Endocitose/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Receptores ErbB/genética , Masculino , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
4.
Development ; 137(2): 273-81, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20040493

RESUMO

Development involves the establishment of boundaries between fields specified to differentiate into distinct tissues. The Drosophila larval eye-antennal imaginal disc must be subdivided into regions that differentiate into the adult eye, antenna and head cuticle. We have found that the transcriptional co-factor Chip is required for cells at the ventral eye-antennal disc border to take on a head cuticle fate; clones of Chip mutant cells in this region instead form outgrowths that differentiate into ectopic eye tissue. Chip acts independently of the transcription factor Homothorax, which was previously shown to promote head cuticle development in the same region. Chip and its vertebrate CLIM homologues have been shown to form complexes with LIM-homeodomain transcription factors, and the domain of Chip that mediates these interactions is required for its ability to suppress the eye fate. We show that two LIM-homeodomain proteins, Arrowhead and Lim1, are expressed in the region of the eye-antennal disc affected in Chip mutants, and that both require Chip for their ability to suppress photoreceptor differentiation when misexpressed in the eye field. Loss-of-function studies support the model that Arrowhead and Lim1 act redundantly, using Chip as a co-factor, to prevent retinal differentiation in regions of the eye disc destined to become ventral head tissue.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Drosophila/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Olho/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Proteínas com Homeodomínio LIM , Proteínas Nucleares/genética , Células Fotorreceptoras/citologia , Células Fotorreceptoras/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
5.
Methods Mol Biol ; 469: 141-61, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19109709

RESUMO

The secreted morphogen Wingless (Wg) has a variety of functions throughout Drosophila eye development, controlling tissue specification, growth, and patterning. Wg plays a critical role in subdividing the eye imaginal disc into separate primordia that will give rise to the adult retina and the surrounding head capsule. During larval development, wg is expressed in the anterior lateral margins of the eye disc, regions that will give rise to head cuticle; Wg signaling promotes the head fate and prevents these marginal regions from initiating ectopic photoreceptor differentiation. Expression of wg at the dorsal margin is earlier and stronger than at the ventral margin, allowing Wg to contribute to specifying the dorsal domain of the eye disc. Finally, during the pupal stages, wg expression surrounds the entire eye and a concentric gradient of Wg establishes several distinct peripheral retinal cell fates. This chapter reviews these aspects of Wg function and describes how to generate clones of cells mutant for genes encoding components of the Wg signaling pathway in the eye disc and examine their effects on photoreceptor differentiation by immunohistochemistry.


Assuntos
Bioensaio/métodos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Morfogênese , Transdução de Sinais/fisiologia , Proteína Wnt1/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Proteína Wnt1/genética
6.
Nat Commun ; 8: 15168, 2017 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-28447612

RESUMO

Controlling nucleus localization is crucial for a variety of cellular functions. In the Drosophila oocyte, nuclear asymmetric positioning is essential for the reorganization of the microtubule (MT) network that controls the polarized transport of axis determinants. A combination of quantitative three-dimensional live imaging and laser ablation-mediated force analysis reveal that nuclear positioning is ensured with an unexpected level of robustness. We show that the nucleus is pushed to the oocyte antero-dorsal cortex by MTs and that its migration can proceed through distinct tracks. Centrosome-associated MTs favour one migratory route. In addition, the MT-associated protein Mud/NuMA that is asymmetrically localized in an Asp-dependent manner at the nuclear envelope hemisphere where MT nucleation is higher promotes a separate route. Our results demonstrate that centrosomes do not provide an obligatory driving force for nuclear movement, but together with Mud, contribute to the mechanisms that ensure the robustness of asymmetric nuclear positioning.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Drosophila/embriologia , Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Oócitos/citologia , Animais , Núcleo Celular/fisiologia , Centrossomo/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo
7.
Methods Mol Biol ; 1328: 99-112, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26324432

RESUMO

The microtubule cytoskeleton is a plastic network of polarized cables. These polymers of tubulin provide orientated routes for the dynamic transport of cytoplasmic molecules and organelles, through which cell polarity is established and maintained. The role of microtubule-mediated transport in the asymmetric localization of axis polarity determinants, in the Drosophila oocyte, has been the subject of extensive studies in the past years. However, imaging the distribution of microtubule fibers in a large cell, where vitellogenesis ensures the uptake of a thick and hazy yolk, presents a series of technical challenges. This chapter briefly reviews some of these aspects and describes two methods designed to circumvent these difficulties. We provide a detailed protocol for the visualization by immunohistochemistry of the three-dimensional organization of tubulin cables in the oocyte. Additionally, we detail the stepwise procedure for the live imaging of microtubule dynamics and network remodeling, using fluorescently labeled microtubule-associated proteins.


Assuntos
Citoesqueleto/ultraestrutura , Microtúbulos/ultraestrutura , Biologia Molecular/métodos , Oogênese/genética , Animais , Citoesqueleto/genética , Drosophila melanogaster/genética , Feminino , Microscopia , Microtúbulos/genética
8.
Curr Biol ; 24(10): 1071-9, 2014 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-24768049

RESUMO

BACKGROUND: The control of apical-basal polarity in epithelial layers is a fundamental event in many processes, ranging from embryonic development to tumor formation. A key feature of polarized epithelial cells is their ability to maintain an asymmetric distribution of specific molecular complexes, including the phosphoinositides PI(4,5)P2 and PI(3,4,5)P3. The spatiotemporal regulation of these phosphoinositides is controlled by the concerted action of phosphoinositide kinases and phosphatases. RESULTS: Using the Drosophila follicular epithelium as a model system in vivo, we show here that PI(4,5)P2 is crucial to maintain apical-basal polarity. PI(4,5)P2 is essentially regulated by the PI4P5 kinase Skittles (SKTL), whereas neither the phosphatase PTEN nor the PI(4,5)P3 kinase DP110 lead to loss of apical-basal polarity. By inactivating SKTL and thereby strongly reducing PI(4,5)P2 levels in a single cell of the epithelium, we observe the disassembly of adherens junctions, actin cytoskeleton reorganization, and apical constriction leading to delamination, a process similar to that observed during epithelial-mesenchymal transition. We provide evidence that PI(4,5)P2 controls the apical targeting of PAR-3/Bazooka to the plasma membrane and that the loss of this polarized distribution is sufficient to induce a similar cell shape change. Finally, we show that PI(4,5)P2 is excluded from the cell apex and that PAR-3 diffuses laterally just prior to the apical constriction in a context of endogenous invagination. CONCLUSIONS: All together, these results indicate that the PIP5 kinase SKTL, by controlling PI(4,5)P2 polarity, regulates PAR-3 localization and thus the size of the apical domain.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Células Epiteliais/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fosfatidilinositóis/metabolismo , Actinas/metabolismo , Junções Aderentes/metabolismo , Animais , Membrana Celular/metabolismo , Polaridade Celular , Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Morfogênese
9.
Genetics ; 190(2): 601-16, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22095083

RESUMO

The Wnt and Hedgehog signaling pathways are essential for normal development and are misregulated in cancer. The casein kinase family of serine/threonine kinases regulates both pathways at multiple levels. However, it has been difficult to determine whether individual members of this family have distinct functions in vivo, due to their overlapping substrate specificities. In Drosophila melanogaster, photoreceptor differentiation is induced by Hedgehog and inhibited by Wingless, providing a sensitive system in which to identify regulators of each pathway. We used a mosaic genetic screen in the Drosophila eye to identify mutations in genes on the X chromosome required for signal transduction. We recovered mutations affecting the transcriptional regulator CREB binding protein, the small GTPase dynamin, the cytoskeletal regulator Actin-related protein 2, and the protein kinase Casein kinase 1α. Consistent with its reported function in the ß-Catenin degradation complex, Casein Kinase 1α mutant cells accumulate ß-Catenin and ectopically induce Wingless target genes. In contrast to previous studies based on RNA interference, we could not detect any effect of the same Casein Kinase 1α mutation on Hedgehog signaling. We thus propose that Casein kinase 1α is essential to allow ß-Catenin degradation and prevent inappropriate Wingless signaling, but its effects on the Hedgehog pathway are redundant with other Casein kinase 1 family members.


Assuntos
Caseína Quinase Ialfa/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Mutação , Células Fotorreceptoras de Invertebrados/citologia , Transdução de Sinais , Proteína Wnt1/metabolismo , Proteína 2 Relacionada a Actina/genética , Animais , Proteína de Ligação a CREB/metabolismo , Caseína Quinase Ialfa/genética , Diferenciação Celular/genética , Dinaminas/genética , Dinaminas/metabolismo , Receptores ErbB/metabolismo , Feminino , Genes Ligados ao Cromossomo X , Proteínas Hedgehog/metabolismo , Masculino , Células Fotorreceptoras de Invertebrados/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores Notch/metabolismo
10.
PLoS One ; 7(9): e45498, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029054

RESUMO

BACKGROUND: TEA domain (TEAD) proteins are highly conserved transcription factors involved in embryonic development and differentiation of various tissues. More recently, emerging evidences for a contribution of these proteins towards apoptosis and cell proliferation regulation have also been proposed. These effects appear to be mediated by the interaction between TEAD and its co-activator Yes-Associated Protein (YAP), the downstream effector of the Hippo tumour suppressor pathway. METHODOLOGY/PRINCIPAL FINDINGS: We further investigated the mechanisms underlying TEAD-mediated apoptosis regulation and showed that overexpression or RNAi-mediated silencing of the TEAD1 protein is sufficient to protect mammalian cell lines from induced apoptosis, suggesting a proapoptotic function for TEAD1 and a non physiological cytoprotective effect for overexpressed TEAD1. Moreover we show that the apoptotic resistance conferred by altered TEAD1 expression is mediated by the transcriptional up-regulation of Livin, a member of the Inhibitor of Apoptosis Protein (IAP) family. In addition, we show that overexpression of a repressive form of TEAD1 can induce Livin up-regulation, indicating that the effect of TEAD1 on Livin expression is indirect and favoring a model in which TEAD1 activates a repressor of Livin by interacting with a limiting cofactor that gets titrated upon TEAD1 up-regulation. Interestingly, we show that overexpression of a mutated form of TEAD1 (Y421H) implicated in Sveinsson's chorioretinal atrophy that strongly reduces its interaction with YAP as well as its activation, can induce Livin expression and protect cells from induced apoptosis, suggesting that YAP is not the cofactor involved in this process. CONCLUSIONS/SIGNIFICANCE: Taken together our data reveal a new, Livin-dependent, apoptotic role for TEAD1 in mammals and provide mechanistic insight downstream of TEAD1 deregulation in cancers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Proteínas Inibidoras de Apoptose/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Transcrição Gênica , Processamento Alternativo , Linhagem Celular , Epistasia Genética , Células HeLa , Humanos , Isoformas de RNA , Fatores de Transcrição de Domínio TEA
11.
Genes Cells ; 11(8): 907-18, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16866874

RESUMO

In Drosophila, the Vestigial-Scalloped (VG-SD) dimeric transcription factor is required for wing cell identity and proliferation. Previous results have shown that VG-SD controls expression of the cell cycle positive regulator dE2F1 during wing development. Since wing disc growth is a homeostatic process, we investigated the possibility that genes involved in cell cycle progression regulate vg and sd expression in feedback loops. We focused our experiments on two major regulators of cell cycle progression: dE2F1 and the antagonist dacapo (dap). Our results reinforce the idea that VG/SD stoichiometry is critical for correct development and that an excess in SD over VG disrupts wing growth. We reveal that transcriptional activity of VG-SD and the VG/SD ratio are both modulated by down-expression of cell cycle genes. We also detected a dap-induced sd up-regulation that disrupts wing growth. Moreover, we observed a rescue of a vg hypomorphic mutant phenotype by dE2F1 that is concomitant with vg and sd induction. This regulation of the VG-SD activity by dE2F1 is dependent on the vg genetic background. Our results support the hypothesis that cell cycle genes fine-tune wing growth and cell proliferation, in part, through control of the VG/SD stoichiometry and activity. This points to a homeostatic feedback regulation between proliferation regulators and the VG-SD wing selector.


Assuntos
Proliferação de Células , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Genes cdc/fisiologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Asas de Animais/embriologia , Animais , Fator de Transcrição E2F1/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Homeostase/genética , Proteínas Mutantes/metabolismo , Ativação Transcricional , Transfecção , Asas de Animais/crescimento & desenvolvimento
12.
Cell Cycle ; 5(7): 740-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16582629

RESUMO

In vitro studies have shown that Drosophila melanogaster has a highly efficient single deoxyribonucleoside kinase (dNK) multisubstrate enzyme. dNK is related to the mammalian Thymidine Kinase 2 (TK2) group involved in the nucleotide synthesis salvage pathway. To study the dNK function in vivo, we constructed transgenic Drosophila strains and impaired the nucleotide de novo synthesis pathway, using antifolates such as aminopterin. Our results show that dNK overexpression rescues both cell death and cell cycle arrest triggered by this anti-cancer drug, and confers global resistance on the fly. Moreover, we show that fly viability and growth depend on the exquisite ratio between dNK expression and its substrate thymidine (dT) in the medium, and that increased dT concentrations trigger apoptosis and a decrease in body mass when dNK is mis-expressed. Finally, dNK expression, unlike that of TK2, is cell cycle dependent and under the control of CyclinE and the dE2F1 transcription factor involved in the G1/S transition. dNK is therefore functionally more closely related to mammalian TK1 than to TK2. This strongly suggests that dNK plays a role in cell proliferation in physiological conditions.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/fisiologia , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Resistencia a Medicamentos Antineoplásicos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Aminopterina/farmacologia , Animais , Proliferação de Células , Sobrevivência Celular , Drosophila melanogaster/efeitos dos fármacos , Fator de Transcrição E2F1/metabolismo , Regulação da Expressão Gênica , Metotrexato/farmacologia , Fenótipo , Timidina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA