Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38746105

ABSTRACT

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.
RNA ; 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627019

ABSTRACT

The sequence-specific RNA-binding protein Pumilio controls Drosophila development; however, the network of mRNAs that it regulates remains incompletely characterized. In this study, we utilize knockdown and knockout approaches coupled with RNA-Seq to measure the impact of Pumilio on the transcriptome of Drosophila cells in culture. We also use an improved RNA co-immunoprecipitation method to identify Pumilio-bound mRNAs in Drosophila embryos. Integration of these datasets with the locations of Pumilio binding motifs across the transcriptome reveal novel direct Pumilio target genes involved in neural, muscle, wing, and germ cell development, and cellular proliferation. These genes include components of Wnt, TGF-beta, MAPK/ERK, and Notch signaling pathways, DNA replication, and lipid metabolism. We identify the mRNAs regulated by the CCR4-NOT deadenylase complex, a key factor in Pumilio-mediated repression, and observe concordant regulation of Pumilio:CCR4-NOT target mRNAs. Computational modeling reveals that Pumilio binding, binding site number, clustering, and sequence context are important determinants of regulation. In contrast, we show that the responses of direct mRNA targets to Pumilio-mediated repression are not influenced by their content of optimal synonymous codons. Moreover, contrary to a prevailing model, we do not detect a role for CCR4-NOT in the degradation of mRNAs with low codon optimality. Together, the results of this work provide new insights into the Pumilio regulatory network and mechanisms, and the parameters that influence the efficacy of Pumilio-mediated regulation.

3.
Sci Adv ; 10(15): eadg7894, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38608012

ABSTRACT

During Drosophila oogenesis, the Oskar (OSK) RNA binding protein (RBP) determines the amount of germ plasm that assembles at the posterior pole of the oocyte. Here, we identify mechanisms that subsequently regulate germ plasm assembly in the early embryo. We show that the Smaug (SMG) RBP is transported into the germ plasm of the early embryo where it accumulates in the germ granules. SMG binds to and represses translation of the osk messenger RNA (mRNA) as well as the bruno 1 (bru1) mRNA, which encodes an RBP that we show promotes germ plasm production. Loss of SMG or mutation of SMG's binding sites in the osk or bru1 mRNA results in excess translation of these transcripts in the germ plasm, accumulation of excess germ plasm, and budding of excess primordial germ cells (PGCs). Therefore, SMG triggers a posttranscriptional regulatory pathway that attenuates the amount of germ plasm in embryos to modulate the number of PGCs.


Subject(s)
Drosophila , Lizards , Animals , Cytoplasm , Germ Cells , RNA, Messenger/genetics , Cell Count
4.
bioRxiv ; 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37693497

ABSTRACT

The sequence-specific RNA-binding protein Pumilio controls development of Drosophila; however, the network of mRNAs that it regulates remains incompletely characterized. In this study, we utilize knockdown and knockout approaches coupled with RNA-Seq to measure the impact of Pumilio on the transcriptome of Drosophila cells. We also used an improved RNA co-immunoprecipitation method to identify Pumilio bound mRNAs in Drosophila embryos. Integration of these datasets with the content of Pumilio binding motifs across the transcriptome revealed novel direct Pumilio target genes involved in neural, muscle, wing, and germ cell development, and cellular proliferation. These genes include components of Wnt, TGF-beta, MAPK/ERK, and Notch signaling pathways, DNA replication, and lipid metabolism. Additionally, we identified the mRNAs regulated by the CCR4-NOT deadenylase complex, a key factor in Pumilio-mediated repression, and observed concordant regulation of Pumilio:CCR4-NOT target mRNAs. Computational modeling revealed that Pumilio binding, binding site number, density, and sequence context are important determinants of regulation. Moreover, the content of optimal synonymous codons in target mRNAs exhibits a striking functional relationship to Pumilio and CCR4-NOT regulation, indicating that the inherent translation efficiency and stability of the mRNA modulates their response to these trans-acting regulatory factors. Together, the results of this work provide new insights into the Pumilio regulatory network and mechanisms, and the parameters that influence the efficacy of Pumilio-mediated regulation.

5.
bioRxiv ; 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36909513

ABSTRACT

During Drosophila oogenesis, the Oskar (OSK) RNA-binding protein (RBP) determines the amount of germ plasm that assembles at the posterior pole of the oocyte. Here we identify the mechanisms that regulate the osk mRNA in the early embryo. We show that the Smaug (SMG) RBP is transported into the germ plasm of the early embryo where it accumulates in the germ granules. SMG binds to and represses translation of the osk mRNA itself as well as the bruno 1 (bru1) mRNA, which encodes an RBP that we show promotes germ plasm production. Loss of SMG or mutation of SMG's binding sites in the osk or bru1 mRNAs results in ectopic translation of these transcripts in the germ plasm and excess PGCs. SMG therefore triggers a post-transcriptional regulatory pathway that attenuates germ plasm synthesis in embryos, thus modulating the number of PGCs.

6.
PLoS Genet ; 17(4): e1009500, 2021 04.
Article in English | MEDLINE | ID: mdl-33798193

ABSTRACT

Localization of oskar mRNA includes two distinct phases: transport from nurse cells to the oocyte, a process typically accompanied by cortical anchoring in the oocyte, followed by posterior localization within the oocyte. Signals within the oskar 3' UTR directing transport are individually weak, a feature previously hypothesized to facilitate exchange between the different localization machineries. We show that alteration of the SL2a stem-loop structure containing the oskar transport and anchoring signal (TAS) removes an inhibitory effect such that in vitro binding by the RNA transport factor, Egalitarian, is elevated as is in vivo transport from the nurse cells into the oocyte. Cortical anchoring within the oocyte is also enhanced, interfering with posterior localization. We also show that mutation of Staufen recognized structures (SRSs), predicted binding sites for Staufen, disrupts posterior localization of oskar mRNA just as in staufen mutants. Two SRSs in SL2a, one overlapping the Egalitarian binding site, are inferred to mediate Staufen-dependent inhibition of TAS anchoring activity, thereby promoting posterior localization. The other three SRSs in the oskar 3' UTR are also required for posterior localization, including two located distant from any known transport signal. Staufen, thus, plays multiple roles in localization of oskar mRNA.


Subject(s)
Drosophila Proteins/genetics , Oocytes/growth & development , RNA-Binding Proteins/genetics , Animals , Drosophila Proteins/ultrastructure , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Inverted Repeat Sequences/genetics , Mutation/genetics , RNA-Binding Proteins/ultrastructure
7.
PLoS Genet ; 17(2): e1009378, 2021 02.
Article in English | MEDLINE | ID: mdl-33600493

ABSTRACT

Mutations in RanBP2 (also known as Nup358), one of the main components of the cytoplasmic filaments of the nuclear pore complex, contribute to the overproduction of acute necrotizing encephalopathy (ANE1)-associated cytokines. Here we report that RanBP2 represses the translation of the interleukin 6 (IL6) mRNA, which encodes a cytokine that is aberrantly up-regulated in ANE1. Our data indicates that soon after its production, the IL6 messenger ribonucleoprotein (mRNP) recruits Argonautes bound to let-7 microRNA. After this mRNP is exported to the cytosol, RanBP2 sumoylates mRNP-associated Argonautes, thereby stabilizing them and enforcing mRNA silencing. Collectively, these results support a model whereby RanBP2 promotes an mRNP remodelling event that is critical for the miRNA-mediated suppression of clinically relevant mRNAs, such as IL6.


Subject(s)
Argonaute Proteins/genetics , Eukaryotic Initiation Factors/genetics , Gene Expression Regulation , MicroRNAs/genetics , Molecular Chaperones/genetics , Nuclear Pore Complex Proteins/genetics , 3' Untranslated Regions/genetics , Argonaute Proteins/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , MicroRNAs/metabolism , Molecular Chaperones/metabolism , Mutation , Nuclear Pore Complex Proteins/metabolism , Pancreatitis, Acute Necrotizing/genetics , Pancreatitis, Acute Necrotizing/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sumoylation
8.
Nat Commun ; 11(1): 4608, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929085

ABSTRACT

Actinobacteria produce antibacterial and antifungal specialized metabolites. Many insects harbour actinobacteria on their bodies or in their nests and use these metabolites for protection. However, some actinobacteria produce metabolites that are toxic to insects and the evolutionary relevance of this toxicity is unknown. Here we explore chemical interactions between streptomycetes and the fruit fly Drosophila melanogaster. We find that many streptomycetes produce specialized metabolites that have potent larvicidal effects against the fly; larvae that ingest spores of these species die. The mechanism of toxicity is specific to the bacterium's chemical arsenal: cosmomycin D producing bacteria induce a cell death-like response in the larval digestive tract; avermectin producing bacteria induce paralysis. Furthermore, low concentrations of volatile terpenes like 2-methylisoborneol that are produced by streptomycetes attract fruit flies such that they preferentially deposit their eggs on contaminated food sources. The resulting larvae are killed during growth and development. The phenomenon of volatile-mediated attraction and specialized metabolite toxicity suggests that some streptomycetes pose an evolutionary risk to insects in nature.


Subject(s)
Bacteria/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/microbiology , Actinobacteria/physiology , Animals , Anthracyclines/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Camphanes/toxicity , Cell Death/drug effects , Drosophila melanogaster/drug effects , Larva/drug effects , Larva/microbiology , Metabolome , Spores, Bacterial/metabolism , Spores, Bacterial/physiology , Streptomyces/physiology , Survival Analysis , Volatile Organic Compounds/pharmacology
9.
Cell Rep ; 31(12): 107783, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579915

ABSTRACT

In animal embryos, the maternal-to-zygotic transition (MZT) hands developmental control from maternal to zygotic gene products. We show that the maternal proteome represents more than half of the protein-coding capacity of Drosophila melanogaster's genome, and that 2% of this proteome is rapidly degraded during the MZT. Cleared proteins include the post-transcriptional repressors Cup, Trailer hitch (TRAL), Maternal expression at 31B (ME31B), and Smaug (SMG). Although the ubiquitin-proteasome system is necessary for clearance of these repressors, distinct E3 ligase complexes target them: the C-terminal to Lis1 Homology (CTLH) complex targets Cup, TRAL, and ME31B for degradation early in the MZT and the Skp/Cullin/F-box-containing (SCF) complex targets SMG at the end of the MZT. Deleting the C-terminal 233 amino acids of SMG abrogates F-box protein interaction and confers immunity to degradation. Persistent SMG downregulates zygotic re-expression of mRNAs whose maternal contribution is degraded by SMG. Thus, clearance of SMG permits an orderly MZT.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Repressor Proteins/genetics , Transcription, Genetic , Zygote/metabolism , Animals , Down-Regulation/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Female , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Biosynthesis/genetics , Protein Subunits/metabolism , Proteolysis , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Ribonucleoproteins/metabolism , Time Factors , Transcriptome/genetics , Ubiquitin/metabolism
10.
EMBO Rep ; 21(7): e48425, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32383557

ABSTRACT

From fly to mammals, the Smaug/Samd4 family of prion-like RNA-binding proteins control gene expression by destabilizing and/or repressing the translation of numerous target transcripts. However, the regulation of its activity remains poorly understood. We show that Smaug's protein levels and mRNA repressive activity are downregulated by Hedgehog signaling in tissue culture cells. These effects rely on the interaction of Smaug with the G-protein coupled receptor Smoothened, which promotes the phosphorylation of Smaug by recruiting the kinase Fused. The activation of Fused and its binding to Smaug are sufficient to suppress its ability to form cytosolic bodies and to antagonize its negative effects on endogenous targets. Importantly, we demonstrate in vivo that HH reduces the levels of smaug mRNA and increases the level of several mRNAs downregulated by Smaug. Finally, we show that Smaug acts as a positive regulator of Hedgehog signaling during wing morphogenesis. These data constitute the first evidence for a post-translational regulation of Smaug and reveal that the fate of several mRNAs bound to Smaug is modulated by a major signaling pathway.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , RNA-Binding Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Repressor Proteins/metabolism , Smoothened Receptor/genetics
11.
Cell Rep ; 30(10): 3353-3367.e7, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160542

ABSTRACT

G3BP RNA-binding proteins are important components of stress granules (SGs). Here, we analyze the role of the Drosophila G3BP Rasputin (RIN) in unstressed cells, where RIN is not SG associated. Immunoprecipitation followed by microarray analysis identifies over 550 mRNAs that copurify with RIN. The mRNAs found in SGs are long and translationally silent. In contrast, we find that RIN-bound mRNAs, which encode core components of the transcription, splicing, and translation machinery, are short, stable, and highly translated. We show that RIN is associated with polysomes and provide evidence for a direct role for RIN and its human homologs in stabilizing and upregulating the translation of their target mRNAs. We propose that when cells are stressed, the resulting incorporation of RIN/G3BPs into SGs sequesters them away from their short target mRNAs. This would downregulate the expression of these transcripts, even though they are not incorporated into stress granules.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Protein Biosynthesis , RNA Stability/genetics , RNA-Binding Proteins/metabolism , Animals , Base Sequence , Carrier Proteins/genetics , Cytoplasmic Granules/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Gene Ontology , Humans , Mice , Mitochondria/metabolism , Mutation/genetics , NIH 3T3 Cells , Polyribosomes/metabolism , RNA Recognition Motif/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Transcriptome/genetics , Zygote/metabolism
12.
Genome Biol ; 18(1): 211, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089021

ABSTRACT

BACKGROUND: All mRNAs are bound in vivo by proteins to form mRNA-protein complexes (mRNPs), but changes in the composition of mRNPs during posttranscriptional regulation remain largely unexplored. Here, we have analyzed, on a transcriptome-wide scale, how microRNA-mediated repression modulates the associations of the core mRNP components eIF4E, eIF4G, and PABP and of the decay factor DDX6 in human cells. RESULTS: Despite the transient nature of repressed intermediates, we detect significant changes in mRNP composition, marked by dissociation of eIF4G and PABP, and by recruitment of DDX6. Furthermore, although poly(A)-tail length has been considered critical in post-transcriptional regulation, differences in steady-state tail length explain little of the variation in either PABP association or mRNP organization more generally. Instead, relative occupancy of core components correlates best with gene expression. CONCLUSIONS: These results indicate that posttranscriptional regulatory factors, such as microRNAs, influence the associations of PABP and other core factors, and do so without substantially affecting steady-state tail length.


Subject(s)
MicroRNAs/metabolism , Poly A/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Animals , Drosophila , HEK293 Cells , Humans , MicroRNAs/genetics , Polyadenylation , Protein Binding , Protein Biosynthesis , RNA Stability , RNA, Messenger/genetics , Saccharomyces cerevisiae/metabolism
13.
Elife ; 62017 09 06.
Article in English | MEDLINE | ID: mdl-28875934

ABSTRACT

In animal embryos, control of development is passed from exclusively maternal gene products to those encoded by the embryonic genome in a process referred to as the maternal-to-zygotic transition (MZT). We show that the RNA-binding protein, ME31B, binds to and represses the expression of thousands of maternal mRNAs during the Drosophila MZT. However, ME31B carries out repression in different ways during different phases of the MZT. Early, it represses translation while, later, its binding leads to mRNA destruction, most likely as a consequence of translational repression in the context of robust mRNA decay. In a process dependent on the PNG kinase, levels of ME31B and its partners, Cup and Trailer Hitch (TRAL), decrease by over 10-fold during the MZT, leading to a change in the composition of mRNA-protein complexes. We propose that ME31B is a global repressor whose regulatory impact changes based on its biological context.


Subject(s)
DEAD-box RNA Helicases/metabolism , Down-Regulation , Drosophila Proteins/metabolism , Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation , RNA, Messenger, Stored/metabolism , Animals , Protein Serine-Threonine Kinases/metabolism , Ribonucleoproteins/metabolism
15.
Mol Cell ; 63(1): 97-109, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27292797

ABSTRACT

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.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , RNA Polymerase II/metabolism , RNA, Small Interfering/biosynthesis , RNA-Binding Proteins/metabolism , Transcription, Genetic , Adenosine/metabolism , Animals , Animals, Genetically Modified , Binding Sites , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cleavage And Polyadenylation Specificity Factor/metabolism , Computational Biology , Databases, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Exoribonucleases/metabolism , Genes, Reporter , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes , Polymers/metabolism , Protein Binding , RNA Stability , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Transcription Termination, Genetic
16.
RNA ; 22(4): 636-55, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26847261

ABSTRACT

Post-transcriptional regulation of mRNAs plays an essential role in the control of gene expression. mRNAs are regulated in ribonucleoprotein (RNP) complexes by RNA-binding proteins (RBPs) along with associated protein and noncoding RNA (ncRNA) cofactors. A global understanding of post-transcriptional control in any cell type requires identification of the components of all of its RNP complexes. We have previously shown that these complexes can be purified by immunoprecipitation using anti-RBP synthetic antibodies produced by phage display. To develop the large number of synthetic antibodies required for a global analysis of RNP complex composition, we have established a pipeline that combines (i) a computationally aided strategy for design of antigens located outside of annotated domains, (ii) high-throughput antigen expression and purification in Escherichia coli, and (iii) high-throughput antibody selection and screening. Using this pipeline, we have produced 279 antibodies against 61 different protein components of Drosophila melanogaster RNPs. Together with those produced in our low-throughput efforts, we have a panel of 311 antibodies for 67 RNP complex proteins. Tests of a subset of our antibodies demonstrated that 89% immunoprecipitate their endogenous target from embryo lysate. This panel of antibodies will serve as a resource for global studies of RNP complexes in Drosophila. Furthermore, our high-throughput pipeline permits efficient production of synthetic antibodies against any large set of proteins.


Subject(s)
Antibodies/chemistry , Drosophila Proteins/immunology , Ribonucleoproteins/immunology , Amino Acid Sequence , Animals , Antibodies/metabolism , Antigens/immunology , Antigens/isolation & purification , Blotting, Western , Complementarity Determining Regions , Drosophila Proteins/isolation & purification , Drosophila melanogaster , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Immunoprecipitation , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Ribonucleoproteins/isolation & purification
17.
J Neurosci ; 35(47): 15666-81, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26609159

ABSTRACT

Here, we have asked about post-transcriptional mechanisms regulating murine developmental neurogenesis, focusing upon the RNA-binding proteins Smaug2 and Nanos1. We identify, in embryonic neural precursors of the murine cortex, a Smaug2 protein/nanos1 mRNA complex that is present in cytoplasmic granules with the translational repression proteins Dcp1 and 4E-T. We show that Smaug2 inhibits and Nanos1 promotes neurogenesis, with Smaug2 knockdown enhancing neurogenesis and depleting precursors, and Nanos1 knockdown inhibiting neurogenesis and maintaining precursors. Moreover, we show that Smaug2 likely regulates neurogenesis by silencing nanos1 mRNA. Specifically, Smaug2 knockdown inappropriately increases Nanos1 protein, and the Smaug2 knockdown-mediated neurogenesis is rescued by preventing this increase. Thus, Smaug2 and Nanos1 function as a bimodal translational repression switch to control neurogenesis, with Smaug2 acting in transcriptionally primed precursors to silence mRNAs important for neurogenesis, including nanos1 mRNA, and Nanos1 acting during the transition to neurons to repress the precursor state. SIGNIFICANCE STATEMENT: The mechanisms instructing neural stem cells to generate the appropriate progeny are still poorly understood. Here, we show that the RNA-binding proteins Smaug2 and Nanos1 are critical regulators of this balance and provide evidence supporting the idea that neural precursors are transcriptionally primed to generate neurons but translational regulation maintains these precursors in a stem cell state until the appropriate developmental time.


Subject(s)
Cell Differentiation/physiology , Cerebral Cortex/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , RNA-Binding Proteins/physiology , Repressor Proteins/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Female , Male , Mammals , Mice , Protein Biosynthesis/physiology
18.
Curr Top Dev Biol ; 113: 43-84, 2015.
Article in English | MEDLINE | ID: mdl-26358870

ABSTRACT

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.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Zygote/physiology , Animals , Drosophila melanogaster/embryology , Embryonic Development/genetics , Female , Oocytes/cytology , Oogenesis/genetics
19.
Genome Biol ; 16: 94, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25962635

ABSTRACT

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.


Subject(s)
Brain Neoplasms/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/genetics , RNA, Messenger, Stored/genetics , RNA-Binding Proteins/genetics , Animals , Binding Sites , Brain Neoplasms/diagnosis , DNA-Binding Proteins/metabolism , Drosophila/embryology , Drosophila Proteins/metabolism , Epigenetic Repression , Female , Gene Expression Regulation, Developmental , Genetic Association Studies , Mutation , Nuclear Proteins , RNA, Messenger, Stored/metabolism , RNA-Binding Proteins/metabolism , Tissue Culture Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Neuron ; 84(4): 723-39, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25456498

ABSTRACT

Here, we have addressed the mechanisms that determine genesis of the correct numbers of neurons during development, focusing on the embryonic cortex. We identify in neural precursors a repressive complex involving eIF4E1 and its binding partner 4E-T that coordinately represses translation of proteins that determine neurogenesis. This eIF4E1/4E-T complex is present in granules with the processing body proteins Lsm1 and Rck, and disruption of this complex causes premature and enhanced neurogenesis and neural precursor depletion. Analysis of the 4E-T complex shows that it is highly enriched in mRNAs encoding transcription factors and differentiation-related proteins. These include the proneurogenic bHLH mRNAs, which colocalize with 4E-T in granules and whose protein products are aberrantly upregulated following knockdown of eIF4E, 4E-T, or processing body proteins. Thus, neural precursors are transcriptionally primed to generate neurons, but an eIF4E/4E-T complex sequesters and represses translation of proneurogenic proteins to determine appropriate neurogenesis.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Neurogenesis/genetics , Neurons/physiology , Nucleocytoplasmic Transport Proteins/genetics , Transcription, Genetic , Animals , HEK293 Cells , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...