Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 6: 8759, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26508274

RESUMEN

Patterning of the Drosophila embryonic termini is achieved by localized activation of the Torso receptor by the growth factor Trunk. Governing this event is the perforin-like protein Torso-like, which is localized to the extracellular space at the embryo poles and has long been proposed to control localized proteolytic activation of Trunk. However, a protease involved in terminal patterning remains to be identified, and the role of Torso-like remains unknown. Here we find that Trunk is cleaved intracellularly by Furin proteases. We further show that Trunk is secreted, and that levels of extracellular Trunk are greatly reduced in torso-like null mutants. On the basis of these and previous findings, we suggest that Torso-like functions to mediate secretion of Trunk, thus providing the mechanism for spatially restricted activation of Torso. Our data represent an alternative mechanism for the spatial control of receptor signalling, and define a different role for perforin-like proteins in eukaryotes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/embriología , Furina/metabolismo , Animales , Tipificación del Cuerpo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Femenino , Furina/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Transducción de Señal
2.
Proc Natl Acad Sci U S A ; 110(36): 14688-92, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959885

RESUMEN

Activation of the Drosophila receptor tyrosine kinase Torso (Tor) only at the termini of the embryo is achieved by the localized expression of the maternal gene Torso-like (Tsl). Tor has a second function in the prothoracic gland as the receptor for prothoracicotropic hormone (PTTH) that initiates metamorphosis. Consistent with the function of Tor in this tissue, Tsl also localizes to the prothoracic gland and influences developmental timing. Despite these commonalities, in our studies of Tsl we unexpectedly found that tsl and tor have opposing effects on body size; tsl null mutants are smaller than normal, rather than larger as would be expected if the PTTH/Tor pathway was disrupted. We further found that whereas both genes regulate developmental timing, tsl does so independently of tor. Although tsl null mutants exhibit a similar length delay in time to pupariation to tor mutants, in tsl:tor double mutants this delay is strikingly enhanced. Thus, loss of tsl is additive rather than epistatic to loss of tor. We also find that phenotypes generated by ectopic PTTH expression are independent of tsl. Finally, we show that a modified form of tsl that can rescue developmental timing cannot rescue terminal patterning, indicating that Tsl can function via distinct mechanisms in different contexts. We conclude that Tsl is not just a specialized cue for Torso signaling but also acts independently of PTTH/Tor in the control of body size and the timing of developmental progression. These data highlight surprisingly diverse developmental functions for this sole Drosophila member of the perforin-like superfamily.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Tirosina Quinasas Receptoras/genética , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Tamaño Corporal/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/crecimiento & desarrollo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Femenino , Hibridación in Situ , Hormonas de Insectos/genética , Hormonas de Insectos/metabolismo , Larva/química , Larva/genética , Larva/crecimiento & desarrollo , Modelos Lineales , Masculino , Metamorfosis Biológica/genética , Mutación , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/genética , Factores de Tiempo
3.
Dev Dyn ; 241(11): 1695-707, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22972499

RESUMEN

BACKGROUND: Microarray studies have shown that the E2F transcription factor influences the expression of many genes but it is unclear how many of these targets are important for E2F-mediated control of cell proliferation. RESULTS: We assembled a collection of mutant alleles of 44 dE2F1-dependent genes and tested whether these could modify visible phenotypes caused by the tissue-specific depletion of dE2F1. More than half of the mutant alleles dominantly enhanced de2f1-dsRNA phenotypes suggesting that the in vivo functions of dE2F1 can be limited by the reduction in the level of expression of many different targets. Unexpectedly, several mutant alleles suppressed de2f1-dsRNA phenotypes. One of the strongest of these suppressors was Orc5. Depletion of ORC5 increased proliferation in cells with reduced dE2F1 and specifically elevated the expression of dE2F1-regulated genes. Importantly, these effects were independent of dE2F1 protein levels, suggesting that reducing the level of ORC5 did not interfere with the general targeting of dE2F1. CONCLUSIONS: We propose that the interaction between ORC5 and dE2F1 may reflect a feedback mechanism between replication initiation proteins and dE2F1 that ensures that proliferating cells maintain a robust level of replication proteins for the next cell cycle.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factores de Transcripción E2F/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Proliferación Celular , Drosophila , Proteínas de Drosophila/genética , Factores de Transcripción E2F/genética , Inmunohistoquímica , Hibridación in Situ , Modelos Biológicos , Complejo de Reconocimiento del Origen/genética , Complejo de Reconocimiento del Origen/metabolismo , Factores de Transcripción/genética
4.
PLoS One ; 7(4): e35641, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22530061

RESUMEN

BACKGROUND: For insects the sense of smell and associated olfactory-driven behaviours are essential for survival. Insects detect odorants with families of olfactory receptor proteins that are very different to those of mammals, and there are likely to be other unique genes and genetic pathways involved in the function and development of the insect olfactory system. METHODOLOGY/PRINCIPAL FINDINGS: We have performed a genetic screen of a set of 505 Drosophila melanogaster gene trap insertion lines to identify novel genes expressed in the adult olfactory organs. We identified 16 lines with expression in the olfactory organs, many of which exhibited expression of the trapped genes in olfactory receptor neurons. Phenotypic analysis showed that six of the lines have decreased olfactory responses in a behavioural assay, and for one of these we showed that precise excision of the P element reverts the phenotype to wild type, confirming a role for the trapped gene in olfaction. To confirm the identity of the genes trapped in the lines we performed molecular analysis of some of the insertion sites. While for many lines the reported insertion sites were correct, we also demonstrated that for a number of lines the reported location of the element was incorrect, and in three lines there were in fact two pGT element insertions. CONCLUSIONS/SIGNIFICANCE: We identified 16 new genes expressed in the Drosophila olfactory organs, the majority in neurons, and for several of the gene trap lines demonstrated a defect in olfactory-driven behaviour. Further characterisation of these genes and their roles in olfactory system function and development will increase our understanding of how the insect olfactory system has evolved to perform the same essential function to that of mammals, but using very different molecular genetic mechanisms.


Asunto(s)
Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Bulbo Olfatorio/metabolismo , Animales , Conducta Animal , Drosophila melanogaster/fisiología , Femenino , Regulación de la Expresión Génica , Masculino , Trastornos del Olfato/genética , Neuronas Receptoras Olfatorias/metabolismo
5.
Genes Dev ; 26(4): 356-68, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22345517

RESUMEN

E2F transcription factors are important regulators of cell proliferation and are frequently dysregulated in human malignancies. To identify novel regulators of E2F function, we used Drosophila as a model system to screen for mutations that modify phenotypes caused by reduced levels of dE2F1. This screen identified components of the Pumilio translational repressor complex (Pumilio, Nanos, and Brain tumor) as suppressors of dE2F1-RNAi phenotypes. Subsequent experiments provided evidence that Pumilio complexes repress dE2F1 levels and that this mechanism of post-transcriptional regulation is conserved in human cells. The human Pumilio homologs Pum 1 and Pum 2 repress the translation of E2F3 by binding to the E2F3 3' untranslated region (UTR) and also enhance the activity of multiple E2F3 targeting microRNAs (miRNAs). E2F3 is an oncogene with strong proliferative potential and is regularly dysregulated or overexpressed in cancer. Interestingly, Pumilio/miRNA-mediated regulation of E2F3 is circumvented in cancer cells in several different ways. Bladder carcinomas selectively down-regulate miRNAs that cooperate with Pumilio to target E2F3, and multiple tumor cell lines shorten the 3' end of the E2F3 mRNA, removing the Pumilio regulatory elements. These studies suggest that Pumilio-miRNA repression of E2F3 translation provides an important level of E2F regulation that is frequently abrogated in cancer cells.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factores de Transcripción E2F/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Regiones no Traducidas 3'/genética , Animales , Línea Celular Tumoral , Drosophila , Proteínas de Drosophila/genética , Factores de Transcripción E2F/metabolismo , Células HeLa , Humanos , MicroARNs/genética , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética
6.
Dev Biol ; 344(1): 36-51, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20416294

RESUMEN

Geminin was identified in Xenopus as a dual function protein involved in the regulation of DNA replication and neural differentiation. In Xenopus, Geminin acts to antagonize the Brahma (Brm) chromatin-remodeling protein, Brg1, during neural differentiation. Here, we investigate the interaction of Geminin with the Brm complex during Drosophila development. We demonstrate that Drosophila Geminin (Gem) interacts antagonistically with the Brm-BAP complex during wing development. Moreover, we show in vivo during wing development and biochemically that Brm acts to promote EGFR-Ras-MAPK signaling, as indicated by its effects on pERK levels, while Gem opposes this. Furthermore, gem and brm alleles modulate the wing phenotype of a Raf gain-of-function mutant and the eye phenotype of a EGFR gain-of-function mutant. Western analysis revealed that Gem over-expression in a background compromised for Brm function reduces Mek (MAPKK/Sor) protein levels, consistent with the decrease in ERK activation observed. Taken together, our results show that Gem and Brm act antagonistically to modulate the EGFR-Ras-MAPK signaling pathway, by affecting Mek levels during Drosophila development.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Receptores ErbB/metabolismo , Regulación del Desarrollo de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Transactivadores/metabolismo , Proteínas ras/metabolismo , Animales , Animales Modificados Genéticamente , Geminina , Modelos Biológicos , Mutación , Fenotipo , ARN Bicatenario/metabolismo , Transducción de Señal , Alas de Animales
7.
Nature ; 455(7212): 552-6, 2008 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-18794899

RESUMEN

The E2F1 transcription factor can promote proliferation or apoptosis when activated, and is a key downstream target of the retinoblastoma tumour suppressor protein (pRB). Here we show that E2F1 is a potent and specific inhibitor of beta-catenin/T-cell factor (TCF)-dependent transcription, and that this function contributes to E2F1-induced apoptosis. E2F1 deregulation suppresses beta-catenin activity in an adenomatous polyposis coli (APC)/glycogen synthase kinase-3 (GSK3)-independent manner, reducing the expression of key beta-catenin targets including c-MYC. This interaction explains why colorectal tumours, which depend on beta-catenin transcription for their abnormal proliferation, keep RB1 intact. Remarkably, E2F1 activity is also repressed by cyclin-dependent kinase-8 (CDK8), a colorectal oncoprotein. Elevated levels of CDK8 protect beta-catenin/TCF-dependent transcription from inhibition by E2F1. Thus, by retaining RB1 and amplifying CDK8, colorectal tumour cells select conditions that collectively suppress E2F1 and enhance the activity of beta-catenin.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Factor de Transcripción E2F1/antagonistas & inhibidores , Factor de Transcripción E2F1/metabolismo , Proteína de Retinoblastoma/metabolismo , Transcripción Genética , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Apoptosis , Línea Celular , Quinasa 8 Dependiente de Ciclina , Regulación de la Expresión Génica , Genes myc/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Proteína de Retinoblastoma/genética , Transducción de Señal , Factores de Transcripción TCF/metabolismo , Proteínas Wnt/metabolismo
8.
Genes Dev ; 22(8): 1011-24, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18367646

RESUMEN

The Drosophila retinoblastoma family of proteins (RBF1 and RBF2) and their mammalian homologs (pRB, p130, and p107) are best known for their regulation of the G1/S transition via the repression of E2F-dependent transcription. However, RB family members also possess additional functions. Here, we report that rbf1 mutant larvae have extensive defects in chromatin condensation during mitosis. We describe a novel interaction between RBF1 and dCAP-D3, a non-SMC component of the Condensin II complex that links RBF1 to the regulation of chromosome structure. RBF1 physically interacts with dCAP-D3, RBF1 and dCAP-D3 partially colocalize on polytene chromosomes, and RBF1 is required for efficient association of dCAP-D3 with chromatin. dCap-D3 mutants also exhibit chromatin condensation defects, and mutant alleles of dCap-D3 suppress cellular and developmental phenotypes induced by the overexpression of RBF1. Interestingly, this interaction is conserved between flies and humans. The re-expression of pRB into a pRB-deficient human tumor cell line promotes chromatin association of hCAP-D3 in a manner that depends on the LXCXE-binding cleft of pRB. These results uncover an unexpected link between pRB/RBF1 and chromatin condensation, providing a mechanism by which the functional inactivation of RB family members in human tumor cells may contribute to genome instability.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Complejos Multiproteicos/metabolismo , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Humanos , Larva/metabolismo , Microscopía Fluorescente , Modelos Biológicos , Complejos Multiproteicos/genética , Neoplasias/metabolismo , Proteína de Retinoblastoma , Factores de Transcripción/genética
9.
Development ; 135(8): 1451-61, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18321983

RESUMEN

The endocycle is a commonly observed variant cell cycle in which cells undergo repeated rounds of DNA replication with no intervening mitosis. How the cell cycle machinery is modified to transform a mitotic cycle into endocycle has long been a matter of interest. In both plants and animals, the transition from the mitotic cycle to the endocycle requires Fzr/Cdh1, a positive regulator of the Anaphase-Promoting Complex/Cyclosome (APC/C). However, because many of its targets are transcriptionally downregulated upon entry into the endocycle, it remains unclear whether the APC/C functions beyond the mitotic/endocycle boundary. Here, we report that APC/C Fzr/Cdh1 activity is required to promote the G/S oscillation of the Drosophila endocycle. We demonstrate that compromising APC/C activity, after cells have entered the endocycle, inhibits DNA replication and results in the accumulation of multiple APC/C targets, including the mitotic cyclins and Geminin. Notably, our data suggest that the activity of APC/C Fzr/Cdh1 during the endocycle is not continuous but is cyclic, as demonstrated by the APC/C-dependent oscillation of the pre-replication complex component Orc1. Taken together, our data suggest a model in which the cyclic activity of APC/C Fzr/Cdh1 during the Drosophila endocycle is driven by the periodic inhibition of Fzr/Cdh1 by Cyclin E/Cdk2. We propose that, as is observed in mitotic cycles, during endocycles, APC/C Fzr/Cdh1 functions to reduce the levels of the mitotic cyclins and Geminin in order to facilitate the relicensing of DNA replication origins and cell cycle progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/citología , Drosophila/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Animales Modificados Genéticamente , Proteínas Cdh1 , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Replicación del ADN , Regulación hacia Abajo , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Genes de Insecto , Mitosis , Modelos Biológicos , Complejo de Reconocimiento del Origen/genética , Complejo de Reconocimiento del Origen/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/genética
10.
Curr Biol ; 17(9): 808-12, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17462898

RESUMEN

Histone-tail modifications play a fundamental role in the processes that establish chromatin structure and determine gene expression. One such modification, histone methylation, was considered irreversible until the recent discovery of histone demethylases. Lsd1 was the first histone demethylase to be identified. Lsd1 is highly conserved in many species, from yeast to humans, but its function has primarily been studied through biochemical approaches. The mammalian ortholog has been shown to demethylate monomethyl- and dimethyl-K4 and -K9 residues of histone H3. Here we describe the effects of Lsd1 mutation in Drosophila. The inactivation of dLsd1 strongly affects the global level of monomethyl- and dimethyl-H3-K4 methylation and results in elevated expression of a subset of genes. dLsd1 is not an essential gene, but animal viability is strongly reduced in mutant animals in a gender-specific manner. Interestingly, dLsd1 mutants are sterile and possess defects in ovary development, indicating that dLsd1 has tissue-specific functions. Mutant alleles of dLsd1 suppress positional-effect variegation, suggesting a disruption of the balance between euchromatin and heterochromatin. Taken together, these results show that dLsd1-mediated H3-K4 demethylation has a significant and specific role in Drosophila development.


Asunto(s)
Metilación de ADN , Proteínas de Drosophila/metabolismo , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Fenotipo , Animales , Western Blotting , Drosophila/embriología , Proteínas de Drosophila/genética , Mutación/genética , Oxidorreductasas N-Desmetilantes/genética
11.
Genes Dev ; 19(14): 1723-34, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024661

RESUMEN

Precise control of cell proliferation and differentiation is critical for organogenesis. Geminin (Gem) has been proposed to link cell cycle exit and differentiation as a prodifferentiation factor and plays a role in neural cell fate acquisition. Here, we identified the SWI/SNF chromatin-remodeling protein Brg1 as an interacting partner of Gem. Brg1 has been implicated in cell cycle withdrawal and cellular differentiation. Surprisingly, we discovered that Gem antagonizes Brg1 activity during neurogenesis to maintain the undifferentiated cell state. Down-regulation of Gem expression normally precedes neuronal differentiation, and gain- and loss-of-function experiments in Xenopus embryos and mouse P19 cells demonstrated that Gem was essential to prevent premature neurogenesis. Misexpression of Gem also suppressed ectopic neurogenesis driven by Ngn and NeuroD. Gem's activity to block differentiation depended upon its ability to bind Brg1 and could be mediated by Gem's inhibition of proneural basic helix-loop-helix (bHLH)-Brg1 interactions required for bHLH target gene activation. Our data demonstrate a novel mechanism of Gem activity, through regulation of SWI/SNF chromatin-remodeling proteins, and indicate that Gem is an essential regulator of neurogenesis that can control the timing of neural progenitor differentiation and maintain the undifferentiated cell state.


Asunto(s)
Proteínas de Drosophila/fisiología , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Secuencia de Bases , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Diferenciación Celular , Línea Celular , ADN/genética , ADN Helicasas , Drosophila/citología , Drosophila/embriología , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Geminina , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/química , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/fisiología , Ratones , Modelos Neurológicos , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/fisiología , Mutagénesis , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Activación Transcripcional , Xenopus , Proteínas de Xenopus/química , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA