Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38746105

RESUMO

Complexes that control mRNA stability and translation promote timely cell-state transitions during differentiation by ensuring appropriate expression patterns of key developmental regulators. The Drosophila RNA-binding protein Brain tumor (Brat) promotes degradation of target transcripts during the maternal-to-zygotic transition in syncytial embryos and in uncommitted intermediate neural progenitors (immature INPs). We identified Ubiquitin-specific protease 5 (Usp5) as a Brat interactor essential for the degradation of Brat target mRNAs in both cell types. Usp5 promotes Brat-dedadenylase pre-complex assembly in mitotic neural stem cells (neuroblasts) by bridging Brat and the scaffolding components of deadenylase complexes lacking their catalytic subunits. The adaptor protein Miranda binds the RNA-binding domain of Brat, limiting its ability to bind target mRNAs in mitotic neuroblasts. Cortical displacement of Miranda activates Brat-mediated mRNA decay in immature INPs. We propose that the assembly of an enzymatically inactive and RNA-binding-deficient pre-complex poises mRNA degradation machineries for rapid activation driving timely developmental transitions.

2.
G3 (Bethesda) ; 6(11): 3541-3551, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27591754

RESUMO

Metazoan embryos undergo a maternal-to-zygotic transition (MZT) during which maternal gene products are eliminated and the zygotic genome becomes transcriptionally active. During this process, RNA-binding proteins (RBPs) and the microRNA-induced silencing complex (miRISC) target maternal mRNAs for degradation. In Drosophila, the Smaug (SMG), Brain tumor (BRAT), and Pumilio (PUM) RBPs bind to and direct the degradation of largely distinct subsets of maternal mRNAs. SMG has also been shown to be required for zygotic synthesis of mRNAs and several members of the miR-309 family of microRNAs (miRNAs) during the MZT. Here, we have carried out global analysis of small RNAs both in wild-type and in smg mutants. Our results show that 85% of all miRNA species encoded by the genome are present during the MZT. Whereas loss of SMG has no detectable effect on Piwi-interacting RNAs (piRNAs) or small interfering RNAs (siRNAs), zygotic production of more than 70 species of miRNAs fails or is delayed in smg mutants. SMG is also required for the synthesis and stability of a key miRISC component, Argonaute 1 (AGO1), but plays no role in accumulation of the Argonaute family proteins associated with piRNAs or siRNAs. In smg mutants, maternal mRNAs that are predicted targets of the SMG-dependent zygotic miRNAs fail to be cleared. BRAT and PUM share target mRNAs with these miRNAs but not with SMG itself. We hypothesize that SMG controls the MZT, not only through direct targeting of a subset of maternal mRNAs for degradation but, indirectly, through production and function of miRNAs and miRISC, which act together with BRAT and/or PUM to control clearance of a distinct subset of maternal mRNAs.

3.
Mol Cell ; 63(1): 97-109, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27292797

RESUMO

Small non-coding RNAs called piRNAs serve as guides for an adaptable immune system that represses transposable elements in germ cells of Metazoa. In Drosophila the RDC complex, composed of Rhino, Deadlock and Cutoff (Cuff) bind chromatin of dual-strand piRNA clusters, special genomic regions, which encode piRNA precursors. The RDC complex is required for transcription of piRNA precursors, though the mechanism by which it licenses transcription remained unknown. Here, we show that Cuff prevents premature termination of RNA polymerase II. Cuff prevents cleavage of nascent RNA at poly(A) sites by interfering with recruitment of the cleavage and polyadenylation specificity factor (CPSF) complex. Cuff also protects processed transcripts from degradation by the exonuclease Rat1. Our work reveals a conceptually different mechanism of transcriptional enhancement. In contrast to other factors that regulate termination by binding to specific signals on nascent RNA, the RDC complex inhibits termination in a chromatin-dependent and sequence-independent manner.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/biossíntese , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Adenosina/metabolismo , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Biologia Computacional , Bases de Dados Genéticas , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Exorribonucleases/metabolismo , Genes Reporter , Proteínas Associadas aos Microtúbulos/metabolismo , Complexos Multiproteicos , Polímeros/metabolismo , Ligação Proteica , Estabilidade de RNA , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Terminação da Transcrição Genética
4.
Curr Top Dev Biol ; 113: 43-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26358870

RESUMO

Drosophila late-stage oocytes and early embryos are transcriptionally silent. Thus, control of gene expression during these developmental periods is posttranscriptional and posttranslational. Global changes in the transcriptome and proteome occur during oocyte maturation, after egg activation and fertilization, and upon zygotic genome activation. We review the scale, content, and dynamics of these global changes; the factors that regulate these changes; and the mechanisms by which they are accomplished. We highlight the intimate relationship between the clearance of maternal gene products and the activation of the embryo's own genome, and discuss the fact that each of these complementary components of the maternal-to-zygotic transition can be subdivided into several phases that serve different biological roles and are regulated by distinct factors.


Assuntos
Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Zigoto/fisiologia , Animais , Drosophila melanogaster/embriologia , Desenvolvimento Embrionário/genética , Feminino , Oócitos/citologia , Oogênese/genética
5.
Genome Biol ; 16: 94, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25962635

RESUMO

BACKGROUND: Brain tumor (BRAT) is a Drosophila member of the TRIM-NHL protein family. This family is conserved among metazoans and its members function as post-transcriptional regulators. BRAT was thought to be recruited to mRNAs indirectly through interaction with the RNA-binding protein Pumilio (PUM). However, it has recently been demonstrated that BRAT directly binds to RNA. The precise sequence recognized by BRAT, the extent of BRAT-mediated regulation, and the exact roles of PUM and BRAT in post-transcriptional regulation are unknown. RESULTS: Genome-wide identification of transcripts associated with BRAT or with PUM in Drosophila embryos shows that they bind largely non-overlapping sets of mRNAs. BRAT binds mRNAs that encode proteins associated with a variety of functions, many of which are distinct from those implemented by PUM-associated transcripts. Computational analysis of in vitro and in vivo data identified a novel RNA motif recognized by BRAT that confers BRAT-mediated regulation in tissue culture cells. The regulatory status of BRAT-associated mRNAs suggests a prominent role for BRAT in post-transcriptional regulation, including a previously unidentified role in transcript degradation. Transcriptomic analysis of embryos lacking functional BRAT reveals an important role in mediating the decay of hundreds of maternal mRNAs during the maternal-to-zygotic transition. CONCLUSIONS: Our results represent the first genome-wide analysis of the mRNAs associated with a TRIM-NHL protein and the first identification of an RNA motif bound by this protein family. BRAT is a prominent post-transcriptional regulator in the early embryo through mechanisms that are largely independent of PUM.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila/genética , RNA Mensageiro Estocado/genética , Proteínas de Ligação a RNA/genética , Animais , Sítios de Ligação , Neoplasias Encefálicas/diagnóstico , Proteínas de Ligação a DNA/metabolismo , Drosophila/embriologia , Proteínas de Drosophila/metabolismo , Repressão Epigenética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Estudos de Associação Genética , Mutação , Proteínas Nucleares , RNA Mensageiro Estocado/metabolismo , Proteínas de Ligação a RNA/metabolismo , Técnicas de Cultura de Tecidos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
J Immunol ; 191(2): 640-9, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23772036

RESUMO

IL-17, a major inflammatory cytokine plays a critical role in the pathogenesis of many autoimmune inflammatory diseases. In this study, we report a new function of RNA-binding protein HuR in IL-17-induced Act1-mediated chemokine mRNA stabilization. HuR deficiency markedly reduced IL-17-induced chemokine expression due to increased mRNA decay. Act1-mediated HuR polyubiquitination was required for the binding of HuR to CXCL1 mRNA, leading to mRNA stabilization. Although IL-17 induced the coshift of Act1 and HuR to the polysomal fractions in a sucrose gradient, HuR deficiency reduced the ratio of translation-active/translation-inactive IL-17-induced chemokine mRNAs. Furthermore, HuR deletion in distal lung epithelium attenuated IL-17-induced neutrophilia. In summary, HuR functions to couple receptor-proximal signaling to posttranscriptional machinery, contributing to IL-17-induced inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL5/genética , Proteínas ELAV/metabolismo , Interleucina-17/metabolismo , Estabilidade de RNA , Animais , Linhagem Celular , Proteínas ELAV/genética , Células HeLa , Humanos , Inflamação/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica , RNA Mensageiro/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais , Ubiquitinação
7.
Differentiation ; 86(4-5): 159-70, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24418439

RESUMO

The Drosophila Hindsight (hnt) gene encodes a C2H2-type Zinc-finger protein, HNT, that plays multiple developmental roles including control of embryonic germ band retraction and regulation of retinal cell fate and morphogenesis. While the developmental functions of the human HNT homolog, RREB-1, are unknown, it has been shown to function as a transcriptional modulator of several tumor suppressor genes. Here we investigate HNT's functional motifs, target genes and its regulatory abilities. We show that the C-terminal region of HNT, containing the last five of its 14 Zinc fingers, binds in vitro to DNA elements very similar to those identified for RREB-1. We map HNT's in vivo binding sites on salivary gland polytene chromosomes and define, at high resolution, where HNT is bound to two target genes, hnt itself and nervy (nvy). Data from both loss-of-function and over-expression experiments show that HNT attenuates the transcription of these two targets in a tissue-specific manner. RREB-1, when expressed in Drosophila, binds to the same polytene chromosome sites as HNT, attenuates expression of the hnt and nvy genes, and rescues the germ band retraction phenotype. HNT's ninth Zinc finger has degenerated or been lost in the vertebrate lineage. We show that a HNT protein mutant for this finger can also attenuate target gene expression and rescue germ band retraction. Thus HNT and RREB-1 are functional homologs at the level of DNA binding, transcriptional regulation and developmental control.


Assuntos
Sequência Conservada/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Animais , Sítios de Ligação , Proteínas de Ligação a DNA/metabolismo , Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mamíferos , Morfogênese/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo
8.
Cell ; 138(5): 885-97, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19737517

RESUMO

Chromatin remodeling by Polycomb group (PcG) and trithorax group (trxG) proteins regulates gene expression in all metazoans. Two major complexes, Polycomb repressive complexes 1 and 2 (PRC1 and PRC2), are thought to mediate PcG-dependent repression in flies and mammals. In Drosophila, PcG/trxG protein complexes are recruited by PcG/trxG response elements (PREs). However, it has been unclear how PcG/trxG are recruited in vertebrates. Here we have identified a vertebrate PRE, PRE-kr, that regulates expression of the mouse MafB/Kreisler gene. PRE-kr recruits PcG proteins in flies and mouse F9 cells and represses gene expression in a PcG/trxG-dependent manner. PRC1 and 2 bind to a minimal PRE-kr region, which can recruit stable PRC1 binding but only weak PRC2 binding when introduced ectopically, suggesting that PRC1 and 2 have different binding requirements. Thus, we provide evidence that similar to invertebrates, PREs act as entry sites for PcG/trxG chromatin remodeling in vertebrates.


Assuntos
Expressão Gênica , Proteínas Repressoras/metabolismo , Elementos de Resposta , Rombencéfalo/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Galinhas , Montagem e Desmontagem da Cromatina , Inversão Cromossômica , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Fator de Transcrição MafB/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Proteínas Repressoras/química , Proteínas Repressoras/genética
10.
Curr Biol ; 14(5): 372-80, 2004 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-15028211

RESUMO

BACKGROUND: Two extraembryonic tissues form early in Drosophila development. One, the amnioserosa, has been implicated in the morphogenetic processes of germ band retraction and dorsal closure. The developmental role of the other, the yolk sac, is obscure. RESULTS: By using live-imaging techniques, we report intimate interactions between the amnioserosa and the yolk sac during germ band retraction and dorsal closure. These tissue interactions fail in a subset of myospheroid (mys: betaPS integrin) mutant embryos, leading to failure of germ band retraction and dorsal closure. The Drosophila homolog of mammalian basigin (EMMPRIN, CD147)-an integrin-associated transmembrane glycoprotein-is highly enriched in the extraembryonic tissues. Strong dominant genetic interactions between basigin and mys mutations cause severe defects in dorsal closure, consistent with basigin functioning together with betaPS integrin in extraembryonic membrane apposition. During normal development, JNK signaling is upregulated in the amnioserosa, as midgut closure disrupts contact with the yolk sac. Subsequently, the amnioserosal epithelium degenerates in a process that is independent of the reaper, hid, and grim cell death genes. In mys mutants that fail to establish contact between the extraembryonic membranes, the amnioserosa undergoes premature disintegration and death. CONCLUSIONS: Intimate apposition of the amnioserosa and yolk sac prevents anoikis of the amnioserosa. Survival of the amnioserosa is essential for germ band retraction and dorsal closure. We hypothesize that during normal development, loss of integrin-dependent contact between the extraembryonic tissues results in JNK-dependent amnioserosal disintegration and death, thus representing an example of developmentally programmed anoikis.


Assuntos
Anoikis/fisiologia , Antígenos CD , Antígenos de Neoplasias , Drosophila/embriologia , Glicoproteínas de Membrana/metabolismo , Saco Vitelino/metabolismo , Sequência de Aminoácidos , Animais , Basigina , Sequência Conservada , Drosophila/genética , Drosophila/metabolismo , Embrião não Mamífero/metabolismo , Embrião não Mamífero/ultraestrutura , Imuno-Histoquímica , Glicoproteínas de Membrana/genética , Membranas/embriologia , Membranas/metabolismo , Microscopia Eletrônica , Dados de Sequência Molecular , Morfogênese/fisiologia , Mutação/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA