Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Cell ; 57(8): 1024-1036.e5, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35472319

RESUMO

The transcription factor Myc drives cell growth across animal phyla and is activated in most forms of human cancer. However, it is unclear which Myc target genes need to be regulated to induce growth and whether multiple targets act additively or if induction of each target is individually necessary. Here, we identified Myc target genes whose regulation is conserved between humans and flies and deleted Myc-binding sites (E-boxes) in the promoters of fourteen of these genes in Drosophila. E-box mutants of essential genes were homozygous viable, indicating that the E-boxes are not required for basal expression. Eight E-box mutations led to Myc-like phenotypes; the strongest mutant, ppanEbox-/-, also made the flies resistant to Myc-induced cell growth without affecting Myc-induced apoptosis. The ppanEbox-/- flies are healthy and display only a minor developmental delay, suggesting that it may be possible to treat or prevent tumorigenesis by targeting individual downstream targets of Myc.


Assuntos
Proteínas Proto-Oncogênicas c-myc , Ribossomos , Animais , Proliferação de Células/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ribossomos/metabolismo , Ativação Transcricional , Regulação para Cima
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35074910

RESUMO

E2F transcription factors are master regulators of the eukaryotic cell cycle. In Drosophila, the sole activating E2F, E2F1, is both required for and sufficient to promote G1→S progression. E2F1 activity is regulated both by binding to RB Family repressors and by posttranscriptional control of E2F1 protein levels by the EGFR and TOR signaling pathways. Here, we investigate cis-regulatory elements in the E2f1 messenger RNA (mRNA) that enable E2f1 translation to respond to these signals and promote mitotic proliferation of wing imaginal disc and intestinal stem cells. We show that small upstream open reading frames (uORFs) in the 5' untranslated region (UTR) of the E2f1 mRNA limit its translation, impacting rates of cell proliferation. E2f1 transgenes lacking these 5'UTR uORFs caused TOR-independent expression and excess cell proliferation, suggesting that TOR activity can bypass uORF-mediated translational repression. EGFR signaling also enhanced translation but through a mechanism less dependent on 5'UTR uORFs. Further, we mapped a region in the E2f1 mRNA that contains a translational enhancer, which may also be targeted by TOR signaling. This study reveals translational control mechanisms through which growth signaling regulates cell cycle progression.


Assuntos
Ciclo Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Regulação da Expressão Gênica , Biossíntese de Proteínas , Fatores de Transcrição/metabolismo , Animais , Biomarcadores , Proliferação de Células , Imunofluorescência , Mitose , Fases de Leitura Aberta , Processamento Pós-Transcricional do RNA , Estresse Fisiológico/genética , Regiões não Traduzidas , Asas de Animais/metabolismo
3.
Cell Rep ; 7(2): 588-598, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24726363

RESUMO

One promising approach for in vivo studies of cell proliferation is the FUCCI system (fluorescent ubiquitination-based cell cycle indicator). Here, we report the development of a Drosophila-specific FUCCI system (Fly-FUCCI) that allows one to distinguish G1, S, and G2 phases of interphase. Fly-FUCCI relies on fluorochrome-tagged degrons from the Cyclin B and E2F1 proteins, which are degraded by the ubiquitin E3-ligases APC/C and CRL4(Cdt2), during mitosis or the onset of S phase, respectively. These probes can track cell-cycle patterns in cultured Drosophila cells, eye and wing imaginal discs, salivary glands, the adult midgut, and probably other tissues. To support a broad range of experimental applications, we have generated a toolkit of transgenic Drosophila lines that express the Fly-FUCCI probes under control of the UASt, UASp, QUAS, and ubiquitin promoters. The Fly-FUCCI system should be a valuable tool for visualizing cell-cycle activity during development, tissue homeostasis, and neoplastic growth.


Assuntos
Ciclo Celular , Proliferação de Células , Drosophila/citologia , Microscopia de Fluorescência/métodos , Ubiquitinação , Ciclossomo-Complexo Promotor de Anáfase/genética , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Animais , Linhagem Celular , Ciclina B/genética , Ciclina B/metabolismo , Drosophila/genética , Drosophila/metabolismo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Especificidade de Órgãos
4.
Nat Rev Mol Cell Biol ; 15(3): 197-210, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24556841

RESUMO

In endoreplication cell cycles, known as endocycles, cells successively replicate their genomes without segregating chromosomes during mitosis and thereby become polyploid. Such cycles, for which there are many variants, are widespread in protozoa, plants and animals. Endocycling cells can achieve ploidies of >200,000 C (chromatin-value); this increase in genomic DNA content allows a higher genomic output, which can facilitate the construction of very large cells or enhance macromolecular secretion. These cells execute normal S phases, using a G1-S regulatory apparatus similar to the one used by mitotic cells, but their capability to segregate chromosomes has been suppressed, typically by downregulation of mitotic cyclin-dependent kinase activity. Endocycles probably evolved many times, and the various endocycle mechanisms found in nature highlight the versatility of the cell cycle control machinery.


Assuntos
Arabidopsis/crescimento & desenvolvimento , Arabidopsis/genética , Ciclo Celular/genética , Processos de Crescimento Celular/genética , Mitose , Ploidias , Animais , Evolução Molecular , Variação Genética
5.
Cold Spring Harb Perspect Biol ; 5(1): a012948, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23284048

RESUMO

Developmentally programmed polyploidy occurs by at least four different mechanisms, two of which (endoreduplication and endomitosis) involve switching from mitotic cell cycles to endocycles by the selective loss of mitotic cyclin-dependent kinase (CDK) activity and bypassing many of the processes of mitosis. Here we review the mechanisms of endoreplication, focusing on recent results from Drosophila and mice.


Assuntos
Drosophila/genética , Endorreduplicação/fisiologia , Animais , Ciclo Celular/fisiologia , Ciclina E/metabolismo , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 2 Dependente de Ciclina/fisiologia , Replicação do DNA , Drosophila/citologia , Camundongos , Poliploidia
6.
Nature ; 480(7375): 123-7, 2011 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-22037307

RESUMO

Endocycles are variant cell cycles comprised of DNA synthesis (S)- and gap (G)-phases but lacking mitosis. Such cycles facilitate post-mitotic growth in many invertebrate and plant cells, and are so ubiquitous that they may account for up to half the world's biomass. DNA replication in endocycling Drosophila cells is triggered by cyclin E/cyclin dependent kinase 2 (CYCE/CDK2), but this kinase must be inactivated during each G-phase to allow the assembly of pre-Replication Complexes (preRCs) for the next S-phase. How CYCE/CDK2 is periodically silenced to allow re-replication has not been established. Here, using genetic tests in parallel with computational modelling, we show that the endocycles of Drosophila are driven by a molecular oscillator in which the E2F1 transcription factor promotes CycE expression and S-phase initiation, S-phase then activates the CRL4(CDT2) ubiquitin ligase, and this in turn mediates the destruction of E2F1 (ref. 7). We propose that it is the transient loss of E2F1 during S phases that creates the window of low Cdk activity required for preRC formation. In support of this model overexpressed E2F1 accelerated endocycling, whereas a stabilized variant of E2F1 blocked endocycling by deregulating target genes, including CycE, as well as Cdk1 and mitotic cyclins. Moreover, we find that altering cell growth by changing nutrition or target of rapamycin (TOR) signalling impacts E2F1 translation, thereby making endocycle progression growth-dependent. Many of the regulatory interactions essential to this novel cell cycle oscillator are conserved in animals and plants, indicating that elements of this mechanism act in most growth-dependent cell cycles.


Assuntos
Ciclo Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Fatores de Transcrição E2F/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Feminino , Masculino , Fase S/fisiologia , Glândulas Salivares/citologia , Fatores de Transcrição , Complexos Ubiquitina-Proteína Ligase
7.
Genes Dev ; 22(12): 1690-703, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559483

RESUMO

Endoreplicating cells undergo multiple rounds of DNA replication leading to polyploidy or polyteny. Oscillation of Cyclin E (CycE)-dependent kinase activity is the main driving force in Drosophila endocycles. High levels of CycE-Cdk2 activity trigger S phase, while down-regulation of CycE-Cdk2 activity is crucial to allow licensing of replication origins. In mitotic cells relicensing in S phase is prevented by Geminin. Here we show that Geminin protein oscillates in endoreplicating salivary glands of Drosophila. Geminin levels are high in S phase, but drop once DNA replication has been completed. DNA licensing is coupled to mitosis through the action of the anaphase-promoting complex/cyclosome (APC/C). We demonstrate that, even though endoreplicating cells never enter mitosis, APC/C activity is required in endoreplicating cells to mediate Geminin oscillation. Down-regulation of APC/C activity results in stabilization of Geminin protein and blocks endocycle progression. Geminin is only abundant in cells with high CycE-Cdk2 activity, suggesting that APC/C-Fzr activity is periodically inhibited by CycE-Cdk2, to prevent relicensing in S-phase cells.


Assuntos
Ciclo Celular/fisiologia , Período de Replicação do DNA/fisiologia , Replicação do DNA/fisiologia , Complexos Ubiquitina-Proteína Ligase/fisiologia , Ciclossomo-Complexo Promotor de Anáfase , Animais , Animais Geneticamente Modificados , Proteínas Cdh1 , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Células Cultivadas , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/fisiologia , Replicação do DNA/genética , Período de Replicação do DNA/genética , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Geminina , Regulação da Expressão Gênica , Mitose/genética , Modelos Biológicos , Origem de Replicação/fisiologia , Glândulas Salivares/metabolismo , Transfecção
8.
EMBO Rep ; 7(12): 1266-72, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17099689

RESUMO

Rca1 (regulator of Cyclin A)/Emi (early mitotic inhibitor) proteins are essential inhibitors of the anaphase-promoting complex/cyclosome (APC/C). In Drosophila, Rca1 is required during G2 to prevent premature cyclin degradation by the Fizzy-related (Fzr)-dependent APC/C activity. Here, we present a structure and function analysis of Rca1 showing that a carboxy-terminal fragment is sufficient for APC/C inhibition. Rca1/Emi proteins contain a conserved F-box and interact with components of the Skp-Cullin-F-box (SCF) complex. So far, no function has been ascribed to this domain. We find that the F-box of Rca1 is dispensable for APC/C-Fzr inhibition during G2. Nevertheless, we show that Rca1 has an additional function at the G1-S transition, which requires the F-box. Overexpression of Rca1 accelerates the G1-S transition in an F-box-dependent manner. Conversely, S-phase entry is delayed in cells in which endogenous Rca1 is replaced by a transgene lacking the F-box. We propose that Rca1 acts as an F-box protein in an as yet uncharacterized SCF complex, which promotes S-phase entry.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Drosophila/genética , Drosophila/embriologia , Motivos F-Box/fisiologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteínas de Ciclo Celular/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Olho/embriologia , Olho/metabolismo , Fase S , Complexos Ubiquitina-Proteína Ligase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...