Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Cell ; 78(5): 862-875.e8, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32348780

ABSTRACT

Nuclear RNA interference (RNAi) pathways work together with histone modifications to regulate gene expression and enact an adaptive response to transposable RNA elements. In the germline, nuclear RNAi can lead to trans-generational epigenetic inheritance (TEI) of gene silencing. We identified and characterized a family of nuclear Argonaute-interacting proteins (ENRIs) that control the strength and target specificity of nuclear RNAi in C. elegans, ensuring faithful inheritance of epigenetic memories. ENRI-1/2 prevent misloading of the nuclear Argonaute NRDE-3 with small RNAs that normally effect maternal piRNAs, which prevents precocious nuclear translocation of NRDE-3 in the early embryo. Additionally, they are negative regulators of nuclear RNAi triggered from exogenous sources. Loss of ENRI-3, an unstable protein expressed mostly in the male germline, misdirects the RNAi response to transposable elements and impairs TEI. The ENRIs determine the potency and specificity of nuclear RNAi responses by gating small RNAs into specific nuclear Argonautes.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Silencing/physiology , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Nucleus/metabolism , Germ Cells/metabolism , Nuclear Proteins/metabolism , RNA Interference/physiology , RNA, Double-Stranded/metabolism , RNA, Nuclear/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics
2.
Mol Cell ; 75(2): 340-356.e10, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31253575

ABSTRACT

The microRNAs encoded by the miR-17∼92 polycistron are commonly overexpressed in cancer and orchestrate a wide range of oncogenic functions. Here, we identify a mechanism for miR-17∼92 oncogenic function through the disruption of endogenous microRNA (miRNA) processing. We show that, upon oncogenic overexpression of the miR-17∼92 primary transcript (pri-miR-17∼92), the microprocessor complex remains associated with partially processed intermediates that aberrantly accumulate. These intermediates reflect a series of hierarchical and conserved steps in the early processing of the pri-miR-17∼92 transcript. Encumbrance of the microprocessor by miR-17∼92 intermediates leads to the broad but selective downregulation of co-expressed polycistronic miRNAs, including miRNAs derived from tumor-suppressive miR-34b/c and from the Dlk1-Dio3 polycistrons. We propose that the identified steps of polycistronic miR-17∼92 biogenesis contribute to the oncogenic re-wiring of gene regulation networks. Our results reveal previously unappreciated functional paradigms for polycistronic miRNAs in cancer.


Subject(s)
Carcinogenesis/genetics , MicroRNAs/genetics , RNA Processing, Post-Transcriptional/genetics , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Iodide Peroxidase/genetics , Membrane Proteins/genetics , MicroRNAs/biosynthesis , Nucleic Acid Conformation
3.
Nucleic Acids Res ; 50(16): 9397-9412, 2022 09 09.
Article in English | MEDLINE | ID: mdl-35993810

ABSTRACT

Precise maintenance of PTEN dosage is crucial for tumor suppression across a wide variety of cancers. Post-transcriptional regulation of Pten heavily relies on regulatory elements encoded by its 3'UTR. We previously reported the important diversity of 3'UTR isoforms of Pten mRNAs produced through alternative polyadenylation (APA). Here, we reveal the direct regulation of Pten APA by the mammalian cleavage factor I (CFIm) complex, which in turn contributes to PTEN protein dosage. CFIm consists of the UGUA-binding CFIm25 and APA regulatory subunits CFIm59 or CFIm68. Deep sequencing analyses of perturbed (KO and KD) cell lines uncovered the differential regulation of Pten APA by CFIm59 and CFIm68 and further revealed that their divergent functions have widespread impact for APA in transcriptomes. Differentially regulated genes include numerous factors within the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signalling pathway that PTEN counter-regulates. We further reveal a stratification of APA dysregulation among a subset of PTEN-driven cancers, with recurrent alterations among PI3K/Akt pathway genes regulated by CFIm. Our results refine the transcriptome selectivity of the CFIm complex in APA regulation, and the breadth of its impact in PTEN-driven cancers.


Subject(s)
Polyadenylation , Proto-Oncogene Proteins c-akt , Animals , Proto-Oncogene Proteins c-akt/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , 3' Untranslated Regions/genetics , Phosphatidylinositol 3-Kinase/genetics , Mammals/genetics
4.
Nucleic Acids Res ; 49(9): 4803-4815, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33758928

ABSTRACT

microRNA (miRNA)-mediated gene silencing is enacted through the recruitment of effector proteins that direct translational repression or degradation of mRNA targets, but the relative importance of their activities for animal development remains unknown. Our concerted proteomic surveys identified the uncharacterized GYF-domain encoding protein GYF-1 and its direct interaction with IFE-4, the ortholog of the mammalian translation repressor 4EHP, as key miRNA effector proteins in Caenorhabditis elegans. Recruitment of GYF-1 protein to mRNA reporters in vitro or in vivo leads to potent translation repression without affecting the poly(A) tail or impinging on mRNA stability. Loss of gyf-1 is synthetic lethal with hypomorphic alleles of embryonic miR-35-42 and larval (L4) let-7 miRNAs, which is phenocopied through engineered mutations in gyf-1 that abolish interaction with IFE-4. GYF-1/4EHP function is cascade-specific, as loss of gyf-1 had no noticeable impact on the functions of other miRNAs, including lin-4 and lsy-6. Overall, our findings reveal the first direct effector of miRNA-mediated translational repression in C. elegans and its physiological importance for the function of several, but likely not all miRNAs.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , MicroRNAs/genetics , Protein Biosynthesis , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Eukaryotic Initiation Factor-4E/metabolism , Genes, Lethal , MicroRNAs/metabolism , Protein Domains , Proteomics , RNA-Induced Silencing Complex/metabolism
7.
Nucleic Acids Res ; 46(19): 10340-10352, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30053103

ABSTRACT

Fine regulation of the phosphatase and tensin homologue (PTEN) phosphatase dosage is critical for homeostasis and tumour suppression. The 3'-untranslated region (3'-UTR) of Pten mRNA was extensively linked to post-transcriptional regulation by microRNAs (miRNAs). In spite of this critical regulatory role, alternative 3'-UTRs of Pten have not been systematically characterized. Here, we reveal an important diversity of Pten mRNA isoforms generated by alternative polyadenylation sites. Several 3'-UTRs are co-expressed and their relative expression is dynamically regulated. In spite of encoding multiple validated miRNA-binding sites, longer isoforms are largely refractory to miRNA-mediated silencing, are more stable and contribute to the bulk of PTEN protein and signalling functions. Taken together, our results warrant a mechanistic re-interpretation of the post-transcriptional mechanisms involving Pten mRNAs and raise concerns on how miRNA-binding sites are being validated.


Subject(s)
MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Polyadenylation/genetics , RNA Isoforms/genetics , 3' Untranslated Regions/genetics , Animals , Homeostasis , Mice , NIH 3T3 Cells , RNA Stability/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
8.
Proc Natl Acad Sci U S A ; 114(21): 5425-5430, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28487484

ABSTRACT

MicroRNAs (miRNAs) play critical roles in a broad variety of biological processes by inhibiting translation initiation and by destabilizing target mRNAs. The CCR4-NOT complex effects miRNA-mediated silencing, at least in part through interactions with 4E-T (eIF4E transporter) protein, but the precise mechanism is unknown. Here we show that the cap-binding eIF4E-homologous protein 4EHP is an integral component of the miRNA-mediated silencing machinery. We demonstrate that the cap-binding activity of 4EHP contributes to the translational silencing by miRNAs through the CCR4-NOT complex. Our results show that 4EHP competes with eIF4E for binding to 4E-T, and this interaction increases the affinity of 4EHP for the cap. We propose a model wherein the 4E-T/4EHP interaction engenders a closed-loop mRNA conformation that blocks translational initiation of miRNA targets.


Subject(s)
MicroRNAs/metabolism , RNA Cap-Binding Proteins/metabolism , RNA Interference , RNA-Induced Silencing Complex/metabolism , Eukaryotic Initiation Factor-4E , HEK293 Cells , HeLa Cells , Humans , Nucleocytoplasmic Transport Proteins/metabolism
9.
Nucleic Acids Res ; 45(12): 7212-7225, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28482037

ABSTRACT

Although strong evidence supports the importance of their cooperative interactions, microRNA (miRNA)-binding sites are still largely investigated as functionally independent regulatory units. Here, a survey of alternative 3΄UTR isoforms implicates a non-canonical seedless site in cooperative miRNA-mediated silencing. While required for target mRNA deadenylation and silencing, this site is not sufficient on its own to physically recruit miRISC. Instead, it relies on facilitating interactions with a nearby canonical seed-pairing site to recruit the Argonaute complexes. We further show that cooperation between miRNA target sites is necessary for silencing in vivo in the C. elegans embryo, and for the recruitment of the Ccr4-Not effector complex. Using a structural model of cooperating miRISCs, we identified allosteric determinants of cooperative miRNA-mediated silencing that are required for both embryonic and larval miRNA functions. Our results delineate multiple cooperative mechanisms in miRNA-mediated silencing and further support the consideration of target site cooperation as a fundamental characteristic of miRNA function.


Subject(s)
Caenorhabditis elegans/genetics , Gene Silencing , MicroRNAs/genetics , RNA-Induced Silencing Complex/chemistry , Transcription Factors/chemistry , 3' Untranslated Regions , Alternative Splicing , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Sequence , Binding Sites , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Embryo, Nonmammalian , MicroRNAs/metabolism , Models, Molecular , Nucleic Acid Conformation , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Nucleic Acids Res ; 45(4): 2081-2098, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28204614

ABSTRACT

MicroRNAs (miRNAs) impinge on the translation and stability of their target mRNAs, and play key roles in development, homeostasis and disease. The gene regulation mechanisms they instigate are largely mediated through the CCR4­NOT deadenylase complex, but the molecular events that occur on target mRNAs are poorly resolved. We observed a broad convergence of interactions of germ granule and P body mRNP components on AIN-1/GW182 and NTL-1/CNOT1 in Caenorhabditis elegans embryos. We show that the miRISC progressively matures on the target mRNA from a scanning form into an effector mRNP particle by sequentially recruiting the CCR4­NOT complex, decapping and decay, or germ granule proteins. Finally, we implicate intrinsically disordered proteins, key components in mRNP architectures, in the embryonic function of lsy-6 miRNA. Our findings define dynamic steps of effector mRNP assembly in miRNA-mediated silencing, and identify a functional continuum between germ granules and P bodies in the C. elegans embryo.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , RNA Interference , Ribonucleoproteins/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cytoplasmic Granules/metabolism , Embryo, Nonmammalian/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intrinsically Disordered Proteins/metabolism , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/metabolism , Ribonucleases/metabolism
11.
Mol Cell ; 40(4): 558-70, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21095586

ABSTRACT

To understand how miRNA-mediated silencing impacts on embryonic mRNAs, we conducted a functional survey of abundant maternal and zygotic miRNA families in the C. elegans embryo. We show that the miR-35-42 and the miR-51-56 miRNA families define maternal and zygotic miRNA-induced silencing complexes (miRISCs), respectively, that share a large number of components. Using a cell-free C. elegans embryonic extract, we demonstrate that the miRISC directs the rapid deadenylation of reporter mRNAs with natural 3'UTRs. The deadenylated targets are translationally suppressed and remarkably stable. Sampling of the predicted miR-35-42 targets reveals that roughly half are deadenylated in a miRNA-dependent manner, but with each target displaying a distinct efficiency and pattern of deadenylation. Finally, we demonstrate that functional cooperation between distinct miRISCs within 3'UTRs is required to potentiate deadenylation. With this report, we reveal the extensive and direct impact of miRNA-mediated deadenylation on embryonic mRNAs.


Subject(s)
3' Untranslated Regions/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Embryo, Nonmammalian/metabolism , MicroRNAs/genetics , RNA 3' End Processing , Animals , Base Sequence , Cell-Free System , Female , Gene Silencing , MicroRNAs/metabolism , Models, Biological , Molecular Sequence Data , Proteomics , RNA-Induced Silencing Complex/metabolism , Zygote/metabolism
12.
Nucleic Acids Res ; 44(12): 5924-35, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27095199

ABSTRACT

Cytoplasmic poly(A)-binding proteins (PABPs) link mRNA 3' termini to translation initiation factors, but they also play key roles in mRNA regulation and decay. Reports from mice, zebrafish and Drosophila further involved PABPs in microRNA (miRNA)-mediated silencing, but through seemingly distinct mechanisms. Here, we implicate the two Caenorhabditis elegans PABPs (PAB-1 and PAB-2) in miRNA-mediated silencing, and elucidate their mechanisms of action using concerted genetics, protein interaction analyses, and cell-free assays. We find that C. elegans PABPs are required for miRNA-mediated silencing in embryonic and larval developmental stages, where they act through a multi-faceted mechanism. Depletion of PAB-1 and PAB-2 results in loss of both poly(A)-dependent and -independent translational silencing. PABPs accelerate miRNA-mediated deadenylation, but this contribution can be modulated by 3'UTR sequences. While greater distances with the poly(A) tail exacerbate dependency on PABP for deadenylation, more potent miRNA-binding sites partially suppress this effect. Our results refine the roles of PABPs in miRNA-mediated silencing and support a model wherein they enable miRNA-binding sites by looping the 3'UTR poly(A) tail to the bound miRISC and deadenylase.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Larva/genetics , MicroRNAs/genetics , Poly A/genetics , Poly(A)-Binding Protein II/genetics , Poly(A)-Binding Protein I/genetics , 3' Untranslated Regions , Adenosine Monophosphate/metabolism , Animals , Binding Sites , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Embryo, Nonmammalian , Gene Silencing , Larva/growth & development , Larva/metabolism , MicroRNAs/metabolism , Poly A/metabolism , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Protein II/metabolism , Protein Binding , Protein Biosynthesis , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism
13.
Mol Cell ; 35(6): 868-80, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19716330

ABSTRACT

MicroRNAs (miRNAs) inhibit mRNA expression in general by base pairing to the 3'UTR of target mRNAs and consequently inhibiting translation and/or initiating poly(A) tail deadenylation and mRNA destabilization. Here we examine the mechanism and kinetics of miRNA-mediated deadenylation in mouse Krebs-2 ascites extract. We demonstrate that miRNA-mediated mRNA deadenylation occurs subsequent to initial translational inhibition, indicating a two-step mechanism of miRNA action, which serves to consolidate repression. We show that a let-7 miRNA-loaded RNA-induced silencing complex (miRISC) interacts with the poly(A)-binding protein (PABP) and the CAF1 and CCR4 deadenylases. In addition, we demonstrate that miRNA-mediated deadenylation is dependent upon CAF1 activity and PABP, which serves as a bona fide miRNA coactivator. Importantly, we present evidence that GW182, a core component of the miRISC, directly interacts with PABP via its C-terminal region and that this interaction is required for miRNA-mediated deadenylation.


Subject(s)
Gene Silencing , MicroRNAs/metabolism , Poly(A)-Binding Proteins/metabolism , Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/metabolism , Animals , Argonaute Proteins , Ascites/genetics , Ascites/metabolism , Autoantigens/metabolism , Binding Sites , Carcinoma, Krebs 2/genetics , Carcinoma, Krebs 2/metabolism , Cell-Free System , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Exoribonucleases , HeLa Cells , Humans , Kinetics , Mice , Poly(A)-Binding Proteins/genetics , Protein Biosynthesis , Protein Structure, Tertiary , Proteins/genetics , RNA Stability , RNA-Induced Silencing Complex/genetics , Receptors, CCR4/metabolism , Repressor Proteins , Ribonucleases , Transfection
14.
Nucleic Acids Res ; 43(15): 7556-65, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26227970

ABSTRACT

Several authors have suggested or inferred that modest changes in microRNA expression can potentiate or impinge on their capacity to mediate gene repression, and that doing so could play a significant role in diseases. Such interpretations are based on several assumptions, namely: (i) changes in microRNA expression correlate with changes in the availability of mature, functional miRISC, (ii) changes in microRNA expression can significantly alter the stoichiometry of miRISC populations with their cognate targets, (iii) and this, in turn, can result in changes in miRISC silencing output. Here, we experimentally challenge those assumptions by quantifying and altering the availability of miRISC across several families of microRNAs. Doing so revealed a surprising fragmentation in the miRISC functional pool, striking differences in the availability of miRNA families and saturability of miRNA-mediated silencing. Furthermore, we provide direct experimental evidence that only a limited subset of miRNAs, defined by a conjuncture of expression threshold, miRISC availability and low target site abundance, is susceptible to competitive effects through microRNA-binding sites.


Subject(s)
Gene Silencing , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism , Binding Sites , HEK293 Cells , Humans
15.
RNA ; 20(9): 1398-409, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25035296

ABSTRACT

MicroRNAs (miRNAs) play critical roles in a variety of biological processes through widespread effects on protein synthesis. Upon association with the miRNA-induced silencing complex (miRISC), miRNAs repress target mRNA translation and accelerate mRNA decay. Degradation of the mRNA is initiated by shortening of the poly(A) tail by the CCR4-NOT deadenylase complex followed by the removal of the 5' cap structure and exonucleolytic decay of the mRNA. Here, we report a direct interaction between the large scaffolding subunit of CCR4-NOT, CNOT1, with the translational repressor and decapping activator protein, DDX6. DDX6 binds to a conserved CNOT1 subdomain in a manner resembling the interaction of the translation initiation factor eIF4A with eIF4G. Importantly, mutations that disrupt the DDX6-CNOT1 interaction impair miRISC-mediated gene silencing in human cells. Thus, CNOT1 facilitates recruitment of DDX6 to miRNA-targeted mRNAs, placing DDX6 as a downstream effector in the miRNA silencing pathway.


Subject(s)
DEAD-box RNA Helicases/metabolism , MicroRNAs/metabolism , Proto-Oncogene Proteins/metabolism , RNA Interference , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites , DEAD-box RNA Helicases/genetics , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Protein Binding , Proto-Oncogene Proteins/genetics , Sequence Homology, Amino Acid
16.
Proc Natl Acad Sci U S A ; 107(8): 3582-7, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133583

ABSTRACT

Argonaute (AGO) proteins interact with distinct classes of small RNAs to direct multiple regulatory outcomes. In many organisms, including plants, fungi, and nematodes, cellular RNA-dependent RNA polymerases (RdRPs) use AGO targets as templates for amplification of silencing signals. Here, we show that distinct RdRPs function sequentially to produce small RNAs that target endogenous loci in Caenorhabditis elegans. We show that DCR-1, the RdRP RRF-3, and the dsRNA-binding protein RDE-4 are required for the biogenesis of 26-nt small RNAs with a 5' guanine (26G-RNAs) and that 26G-RNAs engage the Piwi-clade AGO, ERGO-1. Our findings support a model in which targeting by ERGO-1 recruits a second RdRP (RRF-1 or EGO-1), which in turn transcribes 22G-RNAs that interact with worm-specific AGOs (WAGOs) to direct gene silencing. ERGO-1 targets exhibit a nonrandom distribution in the genome and appear to include many gene duplications, suggesting that this pathway may control overexpression resulting from gene expansion.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , RNA Interference , RNA, Small Interfering/biosynthesis , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Endoribonucleases/metabolism , Gene Silencing , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Ribonuclease III , Transcription, Genetic
17.
RNA ; 16(5): 893-903, 2010 May.
Article in English | MEDLINE | ID: mdl-20354150

ABSTRACT

Years after the discovery that Dicer is a key enzyme in gene silencing, the role of its helicase domain remains enigmatic. Here we show that this domain is critical for accumulation of certain endogenous small interfering RNAs (endo-siRNAs) in Caenorhabditis elegans. The domain is required for the production of the direct products of Dicer, or primary endo-siRNAs, and consequently affects levels of downstream intermediates, the secondary endo-siRNAs. Consistent with the role of endo-siRNAs in silencing, their loss correlates with an increase in cognate mRNA levels. We find that the helicase domain of Dicer is not necessary for microRNA (miRNA) processing, or RNA interference following exposure to exogenous double-stranded RNA. Comparisons of wild-type and helicase-defective strains using deep-sequencing analyses show that the helicase domain is required by a subset of annotated endo-siRNAs, in particular, those associated with the slightly longer 26-nucleotide small RNA species containing a 5' guanosine.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , RNA, Helminth/genetics , RNA, Helminth/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Gene Deletion , Genes, Helminth , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Point Mutation , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Interference , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease III/genetics , Sequence Homology, Amino Acid
18.
Front Genet ; 13: 832547, 2022.
Article in English | MEDLINE | ID: mdl-35401681

ABSTRACT

The 5'-terminal cap is a fundamental determinant of eukaryotic gene expression which facilitates cap-dependent translation and protects mRNAs from exonucleolytic degradation. Enzyme-directed hydrolysis of the cap (decapping) decisively affects mRNA expression and turnover, and is a heavily regulated event. Following the identification of the decapping holoenzyme (Dcp1/2) over two decades ago, numerous studies revealed the complexity of decapping regulation across species and cell types. A conserved set of Dcp1/2-associated proteins, implicated in decapping activation and molecular scaffolding, were identified through genetic and molecular interaction studies, and yet their exact mechanisms of action are only emerging. In this review, we discuss the prevailing models on the roles and assembly of decapping co-factors, with considerations of conservation across species and comparison across physiological contexts. We next discuss the functional convergences of decapping machineries with other RNA-protein complexes in cytoplasmic P bodies and compare current views on their impact on mRNA stability and translation. Lastly, we review the current models of decapping activation and highlight important gaps in our current understanding.

19.
Elife ; 102021 07 05.
Article in English | MEDLINE | ID: mdl-34223818

ABSTRACT

We describe MIP-1 and MIP-2, novel paralogous C. elegans germ granule components that interact with the intrinsically disordered MEG-3 protein. These proteins promote P granule condensation, form granules independently of MEG-3 in the postembryonic germ line, and balance each other in regulating P granule growth and localization. MIP-1 and MIP-2 each contain two LOTUS domains and intrinsically disordered regions and form homo- and heterodimers. They bind and anchor the Vasa homolog GLH-1 within P granules and are jointly required for coalescence of MEG-3, GLH-1, and PGL proteins. Animals lacking MIP-1 and MIP-2 show temperature-sensitive embryonic lethality, sterility, and mortal germ lines. Germline phenotypes include defects in stem cell self-renewal, meiotic progression, and gamete differentiation. We propose that these proteins serve as scaffolds and organizing centers for ribonucleoprotein networks within P granules that help recruit and balance essential RNA processing machinery to regulate key developmental transitions in the germ line.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Germ Cells/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/genetics
20.
Stem Cell Reports ; 15(1): 198-213, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32619492

ABSTRACT

Human embryonic stem cells (hESCs) readily differentiate to somatic or germ lineages but have impaired ability to form extra-embryonic lineages such as placenta or yolk sac. Here, we demonstrate that naive hESCs can be converted into cells that exhibit the cellular and molecular phenotypes of human trophoblast stem cells (hTSCs) derived from human placenta or blastocyst. The resulting "transdifferentiated" hTSCs show reactivation of core placental genes, acquisition of a placenta-like methylome, and the ability to differentiate to extravillous trophoblasts and syncytiotrophoblasts. Modest differences are observed between transdifferentiated and placental hTSCs, most notably in the expression of certain imprinted loci. These results suggest that naive hESCs can differentiate to extra-embryonic lineage and demonstrate a new way of modeling human trophoblast specification and placental methylome establishment.


Subject(s)
DNA Methylation/genetics , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Transcriptome/genetics , Trophoblasts/cytology , Cell Transdifferentiation/genetics , Epithelial Cell Adhesion Molecule/metabolism , Female , Genomic Imprinting , Humans , Integrin alpha2/metabolism , Placenta/cytology , Pregnancy , Pregnancy Trimester, First/physiology , Reproducibility of Results , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL