Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
RNA ; 29(8): 1140-1165, 2023 08.
Article in English | MEDLINE | ID: mdl-37137667

ABSTRACT

Removal of introns during pre-mRNA splicing, which is central to gene expression, initiates by base pairing of U1 snRNA with a 5' splice site (5'SS). In mammals, many introns contain weak 5'SSs that are not efficiently recognized by the canonical U1 snRNP, suggesting alternative mechanisms exist. Here, we develop a cross-linking immunoprecipitation coupled to a high-throughput sequencing method, BCLIP-seq, to identify NRDE2 (nuclear RNAi-defective 2), and CCDC174 (coiled-coil domain-containing 174) as novel RNA-binding proteins in mouse ES cells that associate with U1 snRNA and 5'SSs. Both proteins bind directly to U1 snRNA independently of canonical U1 snRNP-specific proteins, and they are required for the selection and effective processing of weak 5'SSs. Our results reveal that mammalian cells use noncanonical splicing factors bound directly to U1 snRNA to effectively select suboptimal 5'SS sequences in hundreds of genes, promoting proper splice site choice, and accurate pre-mRNA splicing.


Subject(s)
RNA Precursors , RNA Splice Sites , Animals , Mice , RNA Splice Sites/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , Ribonucleoprotein, U1 Small Nuclear/genetics , RNA Interference , RNA Splicing , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Alternative Splicing , Mammals/genetics
2.
Genes Dev ; 33(17-18): 1221-1235, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31371437

ABSTRACT

TRIM71/LIN-41, a phylogenetically conserved regulator of development, controls stem cell fates. Mammalian TRIM71 exhibits both RNA-binding and protein ubiquitylation activities, but the functional contribution of either activity and relevant primary targets remain poorly understood. Here, we demonstrate that TRIM71 shapes the transcriptome of mouse embryonic stem cells (mESCs) predominantly through its RNA-binding activity. We reveal that TRIM71 binds targets through 3' untranslated region (UTR) hairpin motifs and that it acts predominantly by target degradation. TRIM71 mutations implicated in etiogenesis of human congenital hydrocephalus impair target silencing. We identify a set of primary targets consistently regulated in various human and mouse cell lines, including MBNL1 (Muscleblind-like protein 1). MBNL1 promotes cell differentiation through regulation of alternative splicing, and we demonstrate that TRIM71 promotes embryonic splicing patterns through MBNL1 repression. Hence, repression of MBNL1-dependent alternative splicing may contribute to TRIM71's function in regulating stem cell fates.


Subject(s)
Alternative Splicing/genetics , Gene Expression Regulation/genetics , RNA-Binding Proteins/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Embryonic Stem Cells , Humans , Mice , Mice, Knockout , Mutation , Nucleotide Motifs , Protein Binding , Protein Domains/genetics , RNA Interference , RNA-Binding Proteins/metabolism
3.
Life Sci Alliance ; 2(1)2019 02.
Article in English | MEDLINE | ID: mdl-30808654

ABSTRACT

RNAi is the sequence-specific mRNA degradation guided by siRNAs produced from long dsRNA by RNase Dicer. Proteins executing RNAi are present in mammalian cells but rather sustain the microRNA pathway. Aiming for a systematic analysis of mammalian RNAi, we report here that the main bottleneck for RNAi efficiency is the production of functional siRNAs, which integrates Dicer activity, dsRNA structure, and siRNA targeting efficiency. Unexpectedly, increased expression of Dicer cofactors TARBP2 or PACT reduces RNAi but not microRNA function. Elimination of protein kinase R, a key dsRNA sensor in the interferon response, had minimal positive effects on RNAi activity in fibroblasts. Without high Dicer activity, RNAi can still occur when the initial Dicer cleavage of the substrate yields an efficient siRNA. Efficient mammalian RNAi may use substrates with some features of microRNA precursors, merging both pathways even more than previously suggested. Although optimized endogenous Dicer substrates mimicking miRNA features could evolve for endogenous regulations, the same principles would make antiviral RNAi inefficient as viruses would adapt to avoid efficacy.


Subject(s)
RNA Interference/physiology , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Animals , Base Sequence/genetics , Carrier Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Gene Knockout Techniques , Mice , MicroRNAs/metabolism , NIH 3T3 Cells , Plasmids/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , Transfection , eIF-2 Kinase/genetics
4.
Life Sci Alliance ; 1(4): e201800124, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30456373

ABSTRACT

Eukaryotic genomes produce RNAs lacking protein-coding potential, with enigmatic roles. We integrated three approaches to study large intervening noncoding RNA (lincRNA) gene functions. First, we profiled mouse embryonic stem cells and neural precursor cells at single-cell resolution, revealing lincRNAs expressed in specific cell types, cell subpopulations, or cell cycle stages. Second, we assembled a transcriptome-wide atlas of nuclear lincRNA degradation by identifying targets of the exosome cofactor Mtr4. Third, we developed a reversible depletion system to separate the role of a lincRNA gene from that of its RNA. Our approach distinguished lincRNA loci functioning in trans from those modulating local gene expression. Some genes express stable and/or abundant lincRNAs in single cells, but many prematurely terminate transcription and produce lincRNAs rapidly degraded by the nuclear exosome. This suggests that besides RNA-dependent functions, lincRNA loci act as DNA elements or through transcription. Our integrative approach helps distinguish these mechanisms.

5.
Nature ; 557(7707): 739-743, 2018 05.
Article in English | MEDLINE | ID: mdl-29795351

ABSTRACT

De novo mutations in ADNP, which encodes activity-dependent neuroprotective protein (ADNP), have recently been found to underlie Helsmoortel-Van der Aa syndrome, a complex neurological developmental disorder that also affects several other organ functions 1 . ADNP is a putative transcription factor that is essential for embryonic development 2 . However, its precise roles in transcriptional regulation and development are not understood. Here we show that ADNP interacts with the chromatin remodeller CHD4 and the chromatin architectural protein HP1 to form a stable complex, which we refer to as ChAHP. Besides mediating complex assembly, ADNP recognizes DNA motifs that specify binding of ChAHP to euchromatin. Genetic ablation of ChAHP components in mouse embryonic stem cells results in spontaneous differentiation concomitant with premature activation of lineage-specific genes and in a failure to differentiate towards the neuronal lineage. Molecularly, ChAHP-mediated repression is fundamentally different from canonical HP1-mediated silencing: HP1 proteins, in conjunction with histone H3 lysine 9 trimethylation (H3K9me3), are thought to assemble broad heterochromatin domains that are refractory to transcription. ChAHP-mediated repression, however, acts in a locally restricted manner by establishing inaccessible chromatin around its DNA-binding sites and does not depend on H3K9me3-modified nucleosomes. Together, our results reveal that ADNP, via the recruitment of HP1 and CHD4, regulates the expression of genes that are crucial for maintaining distinct cellular states and assures accurate cell fate decisions upon external cues. Such a general role of ChAHP in governing cell fate plasticity may explain why ADNP mutations affect several organs and body functions and contribute to cancer progression1,3,4. Notably, we found that the integrity of the ChAHP complex is disrupted by nonsense mutations identified in patients with Helsmoortel-Van der Aa syndrome, and this could be rescued by aminoglycosides that suppress translation termination 5 . Therefore, patients might benefit from therapeutic agents that are being developed to promote ribosomal read-through of premature stop codons6,7.


Subject(s)
Cell Lineage/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Chromobox Protein Homolog 5 , Euchromatin/genetics , Euchromatin/metabolism , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Nerve Tissue Proteins/genetics , Neurons/cytology , Nucleosomes/metabolism , Protein Binding , Repressor Proteins/metabolism , Transcription, Genetic
6.
Cell Rep ; 12(4): 709-16, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26190102

ABSTRACT

Induction of double-strand DNA breaks (DSBs) by engineered nucleases, such as CRISPR/Cas9 or transcription activator-like effector nucleases (TALENs), stimulates knockin of exogenous DNA fragments via homologous recombination (HR). However, the knockin efficiencies reported so far have not allowed more complex in vitro genome modifications such as, for instance, simultaneous integration of a DNA fragment at two distinct genomic sites. We developed a reporter system to enrich for cells with engineered nuclease-assisted HR events. Using this system in mouse embryonic stem cells (mESCs), we achieve single-step biallelic and seamless integration of two loxP sites for Cre recombinase-mediated inducible gene knockout, as well as biallelic endogenous gene tagging with high efficiency. Our approach reduces the time and resources required for conditional knockout mESC generation dramatically.


Subject(s)
Embryonic Stem Cells/cytology , Gene Knockout Techniques/methods , Tissue Culture Techniques/methods , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Homologous Recombination , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred C57BL
7.
Biol Reprod ; 90(6): 131, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24829024

ABSTRACT

The oocyte-to-embryo transition (OET) denotes transformation of a highly differentiated oocyte into totipotent blastomeres of the early mammalian embryo. OET depends exclusively on maternal RNAs and proteins accumulated during oocyte growth, which implies importance of post-transcriptional control of gene expression. OET includes replacement of abundant maternal microRNAs (miRNAs), enriched also in differentiated cells and exemplified by the Let-7 family, with embryonic miRNAs common in pluripotent stem cells (the miR-290 family in the mouse). Lin28a and its homolog Lin28b encode RNA-binding proteins, which interfere with Let-7 maturation and facilitate reprogramming of induced pluripotent stem cells. Both Lin28a and Lin28b transcripts are abundant in mouse oocytes. To test the role of maternal expression of Lin28a and Lin28b during oocyte-to-zygote reprogramming, we generated mice with oocyte-specific knockdown of both genes by using transgenic RNA interference. Lin28a and Lin28b are dispensable during oocyte growth because their knockdown has no effect on Let-7a levels in fully grown germinal vesicle (GV)-intact oocytes. Furthermore, transgenic females were fertile and produced healthy offspring, and their overall breeding performance was comparable to that of wild-type mice. At the same time, 2-cell embryos derived from transgenic females showed up-regulation of mature Let-7, suggesting that maternally provided LIN28A and LIN28B function during zygotic genome activation. Consistent with this conclusion is increased translation of Lin28a transcripts upon resumption of meiosis. Our data imply dual repression of Let-7 during OET in the mouse model, the selective suppression of Let-7 biogenesis by Lin28 homologs superimposed on previously reported global suppression of miRNA activity.


Subject(s)
DNA-Binding Proteins/genetics , MicroRNAs/genetics , Oocytes/cytology , RNA, Messenger, Stored/genetics , RNA-Binding Proteins/genetics , Animals , Blastocyst/cytology , Blastomeres/cytology , Cell Differentiation , DNA-Binding Proteins/physiology , Embryo Culture Techniques , Embryo Transfer/methods , Female , Luciferases/genetics , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , RNA Interference/physiology , RNA-Binding Proteins/physiology , Totipotent Stem Cells/cytology , Zygote/cytology
8.
Cell ; 155(4): 807-16, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209619

ABSTRACT

In mammals, a single Dicer participates in biogenesis of small RNAs in microRNA (miRNA) and RNAi pathways. In mice, endogenous RNAi is highly active in oocytes, but not in somatic cells, which we ascribe here to an oocyte-specific Dicer isoform (Dicer(O)). Dicer(O) lacks the N-terminal DExD helicase domain and has higher cleavage activity than the full-length Dicer in somatic cells (Dicer(S)). Unlike Dicer(S), Dicer(O) efficiently produces small RNAs from long double-stranded (dsRNA) substrates. Expression of the Dicer(O) isoform is driven by an intronic MT-C retrotransposon promoter, deletion of which causes loss of Dicer(O) and female sterility. Oocytes from females lacking the MT-C element show meiotic spindle defects and increased levels of endogenous small interfering RNA (endo-siRNA) targets, phenocopying the maternal Dicer null phenotype. The alternative Dicer isoform, whose phylogenetic origin demonstrates evolutionary plasticity of RNA-silencing pathways, is the main determinant of endogenous RNAi activity in the mouse female germline.


Subject(s)
DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Oocytes/metabolism , RNA, Small Interfering/metabolism , Retroelements , Ribonuclease III/genetics , Ribonuclease III/metabolism , Animals , Base Sequence , DEAD-box RNA Helicases/chemistry , Female , Gene Expression , Infertility, Female , Mice , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/chemistry , Ribonuclease III/chemistry
9.
Biol Reprod ; 88(1): 11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23136299

ABSTRACT

The oocyte-to-zygote transition entails transforming a highly differentiated oocyte into totipotent blastomeres and represents one of the earliest obstacles that must be successfully hurdled for continued development. Degradation of maternal mRNAs, which likely lies at the heart of this transition, is characterized by a transition from mRNA stability to instability during oocyte maturation. Although phosphorylation of the oocyte-specific RNA-binding protein MSY2 during maturation is implicated in making maternal mRNAs more susceptible to degradation, mechanisms underlying mRNA degradation during oocyte maturation remain poorly understood. We report that DCP1A and DCP2, proteins responsible for decapping mRNA, are encoded by maternal mRNAs recruited for translation during maturation via cytoplasmic polyadenylation elements located in their 3' untranslated regions. Both DCP1A and DCP2 are phosphorylated during maturation, with CDC2A being the kinase likely responsible for both, although MAPK may be involved in DCP1A phosphorylation. Inhibiting accumulation of DCP1A and DCP2 by RNA interference or morpholinos decreases not only degradation of mRNAs during meiotic maturation but also transcription of the zygotic genome. The results indicate that maternally recruited DCP1A and DCP2 are critical players in the transition from mRNA stability to instability during meiotic maturation and that proper maternal mRNA degradation must be successful to execute the oocyte-to-zygote transition.


Subject(s)
Endoribonucleases/metabolism , Gene Expression Regulation, Developmental/physiology , Oocytes/physiology , RNA Stability/physiology , RNA, Messenger/metabolism , Trans-Activators/metabolism , Animals , Butadienes/pharmacology , Endoribonucleases/genetics , Enzyme Inhibitors/pharmacology , Mice , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Messenger/genetics , Roscovitine , Trans-Activators/genetics
10.
PLoS One ; 7(1): e29919, 2012.
Article in English | MEDLINE | ID: mdl-22235350

ABSTRACT

γ-Tubulin is the key protein for microtubule nucleation. Duplication of the γ-tubulin gene occurred several times during evolution, and in mammals γ-tubulin genes encode proteins which share ∼97% sequence identity. Previous analysis of Tubg1 and Tubg2 knock-out mice has suggested that γ-tubulins are not functionally equivalent. Tubg1 knock-out mice died at the blastocyst stage, whereas Tubg2 knock-out mice developed normally and were fertile. It was proposed that γ-tubulin 1 represents ubiquitous γ-tubulin, while γ-tubulin 2 may have some specific functions and cannot substitute for γ-tubulin 1 deficiency in blastocysts. The molecular basis of the suggested functional difference between γ-tubulins remains unknown. Here we show that exogenous γ-tubulin 2 is targeted to centrosomes and interacts with γ-tubulin complex proteins 2 and 4. Depletion of γ-tubulin 1 by RNAi in U2OS cells causes impaired microtubule nucleation and metaphase arrest. Wild-type phenotype in γ-tubulin 1-depleted cells is restored by expression of exogenous mouse or human γ-tubulin 2. Further, we show at both mRNA and protein levels using RT-qPCR and 2D-PAGE, respectively, that in contrast to Tubg1, the Tubg2 expression is dramatically reduced in mouse blastocysts. This indicates that γ-tubulin 2 cannot rescue γ-tubulin 1 deficiency in knock-out blastocysts, owing to its very low amount. The combined data suggest that γ-tubulin 2 is able to nucleate microtubules and substitute for γ-tubulin 1. We propose that mammalian γ-tubulins are functionally redundant with respect to the nucleation activity.


Subject(s)
Down-Regulation , Embryonic Development/genetics , Microtubules/metabolism , Tubulin/genetics , Tubulin/metabolism , Animals , Cell Line, Tumor , Embryo Implantation , Gene Expression Regulation, Developmental , Humans , Intracellular Space/metabolism , Male , Mice , Mice, Inbred C57BL , Mitosis/genetics , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Time Factors , Tubulin/deficiency
11.
Nucleic Acids Res ; 40(1): 399-413, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21908396

ABSTRACT

Double-stranded RNA (dsRNA) can enter different pathways in mammalian cells, including sequence-specific RNA interference (RNAi), sequence-independent interferon (IFN) response and editing by adenosine deaminases. To study the routing of dsRNA to these pathways in vivo, we used transgenic mice ubiquitously expressing from a strong promoter, an mRNA with a long hairpin in its 3'-UTR. The expressed dsRNA neither caused any developmental defects nor activated the IFN response, which was inducible only at high expression levels in cultured cells. The dsRNA was poorly processed into siRNAs in somatic cells, whereas, robust RNAi effects were found in oocytes, suggesting that somatic cells lack some factor(s) facilitating siRNA biogenesis. Expressed dsRNA did not cause transcriptional silencing in trans. Analysis of RNA editing revealed that a small fraction of long dsRNA is edited. RNA editing neither prevented the cytoplasmic localization nor processing into siRNAs. Thus, a long dsRNA structure is well tolerated in mammalian cells and is mainly causing a robust RNAi response in oocytes.


Subject(s)
Adenosine/metabolism , Oocytes/metabolism , RNA Interference , RNA, Double-Stranded/metabolism , Animals , Cell Line , Cells, Cultured , Deamination , Female , Gene Silencing , Genes, mos , Humans , Interferons/metabolism , Mice , Mice, Transgenic , RNA Editing
12.
Methods Mol Biol ; 820: 133-61, 2012.
Article in English | MEDLINE | ID: mdl-22131030

ABSTRACT

The RNA interference (RNAi) and interferons have been an uneasy marriage. Ever since the discovery of RNAi in mammals, the interferon response has been a feared problem. While RNAi became an efficient and widespread method for gene silencing in mammals, numerous studies recognized several obstacles, including undesirable activation of the interferon response, which need to be overcome to achieve a specific and robust RNAi effect. The aim of this text is to provide theoretical and practical information for scientists who want to control interferon response and other adverse effects in their RNAi experiments.


Subject(s)
Interferons/metabolism , RNA Interference , Amino Acid Sequence , Cloning, Molecular , DNA Primers , Gene Expression Regulation , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , RNA, Double-Stranded , RNA, Small Interfering/genetics
13.
Methods ; 53(2): 136-41, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20708690

ABSTRACT

RNA molecules rarely function alone in cells. For most RNAs, their function requires formation of various ribonucleoprotein (RNP) complexes. For example, mRNP composition can determine mRNA localization, translational repression, level of translation or mRNA stability. RNPs are usually studied by biochemical methods. However, biochemical approaches are unsuitable for some model systems, such as mammalian oocytes and early embryos, due to the small amounts that can be obtained for experimental analysis. In such cases, microscopic techniques are often used to learn about RNPs. Here, we present a review of immunostaining, fluorescence in situ hybridization with subcellular resolution and a combination of both, with emphasis on the mouse oocyte and early embryos models. Application of these techniques to whole-mount fixed oocytes and early embryos can provide information about RNP composition and localization with three-dimensional resolution.


Subject(s)
Immunohistochemistry/methods , Oocytes/chemistry , Ribonucleoproteins/analysis , Animals , Embryo, Mammalian/chemistry , Embryo, Mammalian/pathology , Female , In Situ Hybridization, Fluorescence/methods , Mice , Oocytes/pathology
14.
EMBO Rep ; 11(8): 590-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20651740

ABSTRACT

RNA silencing is a complex of mechanisms that regulate gene expression through small RNA molecules. The microRNA (miRNA) pathway is the most common of these in mammals. Genome-encoded miRNAs suppress translation in a sequence-specific manner and facilitate shifts in gene expression during developmental transitions. Here, we discuss the role of miRNAs in oocyte-to-zygote transition and in the control of pluripotency. Existing data suggest a common principle involving miRNAs in defining pluripotent and differentiated cells. RNA silencing pathways also rapidly evolve, resulting in many unique features of RNA silencing in different taxonomic groups. This is exemplified in the mouse model of oocyte-to-zygote transition, in which the endogenous RNA interference pathway has acquired a novel role in regulating protein-coding genes, while the miRNA pathway has become transiently suppressed.


Subject(s)
MicroRNAs/metabolism , Oocytes/physiology , Pluripotent Stem Cells/physiology , RNA Interference , RNA, Small Interfering/metabolism , Zygote/physiology , Animals , Base Sequence , Humans , MicroRNAs/classification , MicroRNAs/genetics , Molecular Sequence Data , Oocytes/cytology , Phylogeny , Pluripotent Stem Cells/cytology , RNA, Small Interfering/genetics , Sequence Alignment , Zygote/cytology
15.
Curr Biol ; 20(3): 265-70, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20116252

ABSTRACT

MicroRNAs (miRNAs) are small endogenous RNAs that typically imperfectly base pair with 3' untranslated regions (3'UTRs) and mediate translational repression and mRNA degradation. Dicer, which generates small RNAs in the miRNA and RNA interference (RNAi) pathways, is essential for meiotic maturation of mouse oocytes. We found that 3'UTRs of transcripts upregulated in Dicer1(-/-) oocytes are not enriched in miRNA binding sites, implicating a weak impact of miRNAs on the maternal transcriptome. Therefore, we tested the ability of endogenous miRNAs to mediate RNA-like cleavage or translational repression of reporter mRNAs. In contrast to somatic cells, endogenous miRNAs in oocytes poorly repressed translation of mRNA reporters, whereas their RNAi-like activity was much less affected. Reporter mRNA carrying let-7-binding sites failed to localize to P body-like structures in oocytes. Our data suggest that miRNA function is downregulated during oocyte development, an idea supported by normal meiotic maturation of oocytes lacking Dgcr8, which is required for the miRNA but not the RNAi pathway (Suh et al. [1], this issue of Current Biology). Suppressing miRNA function during oocyte growth is likely an early event in reprogramming gene expression during the transition of a differentiated oocyte into pluripotent blastomeres of the embryo.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , Oocytes/metabolism , 3' Untranslated Regions , Animals , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Endoribonucleases/deficiency , Endoribonucleases/genetics , Female , In Vitro Techniques , Mice , Mice, Knockout , Models, Biological , Oocytes/growth & development , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Ribonuclease III
16.
Biol Reprod ; 82(5): 1008-17, 2010 May.
Article in English | MEDLINE | ID: mdl-20075394

ABSTRACT

In mammalian somatic cells, several pathways that converge on deadenylation, decapping, and 5'-3' degradation are found in cytoplasmic foci known as P-bodies. Because controlled mRNA stability is essential for oocyte-to-zygote transition, we examined the dynamics of P-body components in mouse oocytes. We report that oocyte growth is accompanied by loss of P-bodies and a subcortical accumulation of several RNA-binding proteins, including DDX6, CPEB, YBX2 (MSY2), and the exon junction complex. These proteins form transient RNA-containing aggregates in fully grown oocytes with a surrounded nucleolus chromatin configuration. These aggregates disperse during oocyte maturation, consistent with recruitment of maternal mRNAs that occurs during this time. In contrast, levels of DCP1A are low during oocyte growth, and DCP1A does not colocalize with DDX6 in the subcortical aggregates. The amount of DCP1A markedly increases during meiosis, which correlates with the first wave of destabilization of maternal mRNAs. We propose that the cortex of growing oocytes serves as an mRNA storage compartment, which contains a novel type of RNA granule related to P-bodies.


Subject(s)
Cytoplasmic Granules/metabolism , Oocytes/metabolism , RNA, Messenger, Stored/metabolism , Ribonucleoproteins/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation/physiology , Endoribonucleases , Female , Gene Expression Regulation, Developmental/physiology , Intracellular Space , Mice , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Ovary/growth & development , Ovary/metabolism , Protein Conformation , RNA Caps/metabolism , RNA-Binding Proteins/physiology , Structure-Activity Relationship
17.
Plant Physiol ; 150(1): 424-36, 2009 May.
Article in English | MEDLINE | ID: mdl-19304931

ABSTRACT

Salicylic acid (SA) plays a central role in defense against pathogen attack, as well as in germination, flowering, senescence, and the acquisition of thermotolerance. In this report we investigate the involvement of phospholipase D (PLD) in the SA signaling pathway. In presence of exogenous primary alcohols, the production of phosphatidic acid by PLD is diverted toward the formation of phosphatidylalcohols through a reaction called transphosphatidylation. By in vivo metabolic phospholipid labeling with (33)P(i), PLD activity was found to be induced 45 min after addition of SA. We show that incubation of Arabidopsis (Arabidopsis thaliana) cell suspensions with primary alcohols inhibited the induction of two SA-responsive genes, PATHOGENESIS-RELATED1 and WRKY38, in a dose-dependent manner. This inhibitory effect was more pronounced when the primary alcohols were more hydrophobic. Secondary or tertiary alcohols had no inhibitory effect. These results provide compelling arguments for PLD activity being upstream of the induction of these genes by SA. A subsequent study of n-butanol effects on the SA-responsive transcriptome identified 1,327 genes differentially expressed upon SA treatment. Strikingly, the SA response of 380 of these genes was inhibited by n-butanol but not by tert-butanol. A detailed analysis of the regulation of these genes showed that PLD could act both positively and negatively, either on gene induction or gene repression. The overlap with the previously described phosphatidylinositol-4-kinase pathway is discussed.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/enzymology , Phospholipase D/metabolism , Salicylic Acid/metabolism , Signal Transduction , 1-Butanol/pharmacology , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation, Plant , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , tert-Butyl Alcohol/pharmacology
18.
Plant Physiol ; 144(3): 1347-59, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17496105

ABSTRACT

Salicylic acid (SA) has a central role in defense against pathogen attack. In addition, its role in such diverse processes as germination, flowering, senescence, and thermotolerance acquisition has been documented. However, little is known about the early signaling events triggered by SA. Using Arabidopsis (Arabidopsis thaliana) suspension cells as a model, it was possible to show by in vivo metabolic phospholipid labeling with (33)P(i) that SA addition induced a rapid and early (in few minutes) decrease in a pool of phosphatidylinositol (PI). This decrease paralleled an increase in PI 4-phosphate and PI 4,5-bisphosphate. These changes could be inhibited by two different inhibitors of type III PI 4-kinases, phenylarsine oxide and 30 microm wortmannin; no inhibitory effect was seen with 1 microm wortmannin, a concentration inhibiting PI 3-kinases but not PI 4-kinases. We therefore undertook a study of the effects of wortmannin on SA-responsive transcriptomes. Using the Complete Arabidopsis Transcriptome MicroArray chip, we could identify 774 genes differentially expressed upon SA treatment. Strikingly, among these genes, the response to SA of 112 of them was inhibited by 30 microm wortmannin, but not by 1 microm wortmannin.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Arabidopsis/enzymology , Gene Expression Regulation, Plant , Phosphatidylinositols/metabolism , Salicylic Acid/metabolism , Androstadienes , Arabidopsis/genetics , Arabidopsis/metabolism , Cells, Cultured , Gene Expression Profiling , Phosphorus Radioisotopes , Promoter Regions, Genetic , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...