Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nucleic Acids Res ; 51(17): 9279-9293, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37602378

ABSTRACT

Proteins containing a RNB domain, originally identified in Escherichia coli RNase II, are widely present throughout the tree of life. Many RNB proteins have 3'-5' exoribonucleolytic activity but some have lost catalytic activity during evolution. Database searches identified a new RNB domain-containing protein in human: HELZ2. Analysis of genomic and expression data combined with evolutionary information suggested that the human HELZ2 protein is produced from an unforeseen non-canonical initiation codon in Hominidae. This unusual property was confirmed experimentally, extending the human protein by 247 residues. Human HELZ2 was further shown to be an active ribonuclease despite the substitution of a key residue in its catalytic center. HELZ2 RNase activity is lost in cells from some cancer patients as a result of somatic mutations. HELZ2 harbors also two RNA helicase domains and several zinc fingers and its expression is induced by interferon treatment. We demonstrate that HELZ2 is able to degrade structured RNAs through the coordinated ATP-dependent displacement of duplex RNA mediated by its RNA helicase domains and its 3'-5' ribonucleolytic action. The expression characteristics and biochemical properties of HELZ2 support a role for this factor in response to viruses and/or mobile elements.


Subject(s)
RNA Helicases , Humans , Codon, Initiator , Exoribonucleases/metabolism , Interferons/genetics , RNA/metabolism , RNA Helicases/chemistry , RNA Helicases/genetics
2.
Nat Rev Mol Cell Biol ; 11(5): 379-84, 2010 05.
Article in English | MEDLINE | ID: mdl-20379206

ABSTRACT

GW182 proteins have emerged as key components of microRNA (miRNA) silencing complexes in animals. Although the precise molecular function of GW182 proteins is not fully understood, new findings indicate that they act as poly(A)-binding protein (PABP)-interacting proteins (PAIPs) that promote gene silencing, at least in part, by interfering with cytoplasmic PABP1 (PABPC1) function during translation and mRNA stabilization. This recent discovery paves the way for future studies of miRNA silencing mechanisms.


Subject(s)
Autoantigens/metabolism , MicroRNAs/metabolism , Poly(A)-Binding Protein I/metabolism , Amino Acid Sequence , Animals , Autoantigens/chemistry , Binding Sites , Gene Silencing , Molecular Sequence Data , Poly(A)-Binding Protein I/antagonists & inhibitors , Protein Binding , Protein Structure, Tertiary
3.
PLoS Genet ; 15(2): e1007917, 2019 02.
Article in English | MEDLINE | ID: mdl-30707697

ABSTRACT

Hbs1 has been established as a central component of the cell's translational quality control pathways in both yeast and prokaryotic models; however, the functional characteristics of its human ortholog (Hbs1L) have not been well-defined. We recently reported a novel human phenotype resulting from a mutation in the critical coding region of the HBS1L gene characterized by facial dysmorphism, severe growth restriction, axial hypotonia, global developmental delay and retinal pigmentary deposits. Here we further characterize downstream effects of the human HBS1L mutation. HBS1L has three transcripts in humans, and RT-PCR demonstrated reduced mRNA levels corresponding with transcripts V1 and V2 whereas V3 expression was unchanged. Western blot analyses revealed Hbs1L protein was absent in the patient cells. Additionally, polysome profiling revealed an abnormal aggregation of 80S monosomes in patient cells under baseline conditions. RNA and ribosomal sequencing demonstrated an increased translation efficiency of ribosomal RNA in Hbs1L-deficient fibroblasts, suggesting that there may be a compensatory increase in ribosome translation to accommodate the increased 80S monosome levels. This enhanced translation was accompanied by upregulation of mTOR and 4-EBP protein expression, suggesting an mTOR-dependent phenomenon. Furthermore, lack of Hbs1L caused depletion of Pelota protein in both patient cells and mouse tissues, while PELO mRNA levels were unaffected. Inhibition of proteasomal function partially restored Pelota expression in human Hbs1L-deficient cells. We also describe a mouse model harboring a knockdown mutation in the murine Hbs1l gene that shared several of the phenotypic elements observed in the Hbs1L-deficient human including facial dysmorphism, growth restriction and retinal deposits. The Hbs1lKO mice similarly demonstrate diminished Pelota levels that were rescued by proteasome inhibition.


Subject(s)
GTP-Binding Proteins/genetics , Mammals/genetics , Microfilament Proteins/genetics , Monosomy/genetics , Animals , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mutation/genetics , Phenotype , Polyribosomes/genetics , Proteasome Endopeptidase Complex/genetics , RNA/genetics , RNA, Messenger/genetics , Ribosomes/genetics , TOR Serine-Threonine Kinases/genetics , Up-Regulation/genetics
4.
Mol Cell ; 51(3): 360-73, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23932717

ABSTRACT

The PAN2-PAN3 deadenylase complex functions in general and miRNA-mediated mRNA degradation and is specifically recruited to miRNA targets by GW182/TNRC6 proteins. We describe the PAN3 adaptor protein crystal structure that, unexpectedly, forms intertwined and asymmetric homodimers. Dimerization is mediated by a coiled coil that links an N-terminal pseudokinase to a C-terminal knob domain. The PAN3 pseudokinase binds ATP, and this function is required for mRNA degradation in vivo. We further identified conserved surfaces required for mRNA degradation, including the binding surface for the PAN2 deadenylase on the knob domain. The most remarkable structural feature is the presence of a tryptophan-binding pocket at the dimer interface, which mediates binding to TNRC6C in human cells. Together, our data reveal the structural basis for the interaction of PAN3 with PAN2 and the recruitment of the PAN2-PAN3 complex to miRNA targets by TNRC6 proteins.


Subject(s)
Autoantigens/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Exoribonucleases/metabolism , RNA-Binding Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Autoantigens/chemistry , Binding Sites , Crystallography, X-Ray , Humans , MicroRNAs/genetics , Molecular Sequence Data , Protein Multimerization , Protein Structure, Tertiary , RNA/metabolism , RNA Stability , RNA-Binding Proteins/chemistry
5.
EMBO J ; 35(11): 1186-203, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009120

ABSTRACT

miRNAs associate with Argonaute (AGO) proteins to silence the expression of mRNA targets by inhibiting translation and promoting deadenylation, decapping, and mRNA degradation. A current model for silencing suggests that AGOs mediate these effects through the sequential recruitment of GW182 proteins, the CCR4-NOT deadenylase complex and the translational repressor and decapping activator DDX6. An alternative model posits that AGOs repress translation by interfering with eIF4A function during 43S ribosomal scanning and that this mechanism is independent of GW182 and the CCR4-NOT complex in Drosophila melanogaster Here, we show that miRNAs, AGOs, GW182, the CCR4-NOT complex, and DDX6/Me31B repress and degrade polyadenylated mRNA targets that are translated via scanning-independent mechanisms in both human and Dm cells. This and additional observations indicate a common mechanism used by these proteins and miRNAs to mediate silencing. This mechanism does not require eIF4A function during ribosomal scanning.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , Transcription Factors/metabolism , Argonaute Proteins/genetics , Autoantigens/metabolism , Cell Line , DEAD-box RNA Helicases/metabolism , Drosophila Proteins/metabolism , Humans , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribosomes
6.
Mol Cell ; 44(1): 120-33, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21981923

ABSTRACT

miRNAs are posttranscriptional regulators of gene expression that associate with Argonaute and GW182 proteins to repress translation and/or promote mRNA degradation. miRNA-mediated mRNA degradation is initiated by deadenylation, although it is not known whether deadenylases are recruited to the mRNA target directly or by default, as a consequence of a translational block. To answer this question, we performed a screen for potential interactions between the Argonaute and GW182 proteins and subunits of the two cytoplasmic deadenylase complexes. We found that human GW182 proteins recruit the PAN2-PAN3 and CCR4-CAF1-NOT deadenylase complexes through direct interactions with PAN3 and NOT1, respectively. These interactions are critical for silencing and are conserved in D. melanogaster. Our findings reveal that GW182 proteins provide a docking platform through which deadenylase complexes gain access to the poly(A) tail of miRNA targets to promote their deadenylation, and they further indicate that deadenylation is a direct effect of miRNA regulation.


Subject(s)
Cytoplasm/metabolism , Drosophila Proteins/metabolism , MicroRNAs/metabolism , Animals , Carrier Proteins/metabolism , Drosophila melanogaster , Exoribonucleases/metabolism , Gene Silencing , Genetic Complementation Test , HeLa Cells , Humans , Poly A/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary , RNA-Binding Proteins , Receptors, CCR4/metabolism , Retinoblastoma-Binding Protein 4/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism
7.
Nat Rev Genet ; 12(2): 99-110, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21245828

ABSTRACT

Despite their widespread roles as regulators of gene expression, important questions remain about target regulation by microRNAs. Animal microRNAs were originally thought to repress target translation, with little or no influence on mRNA abundance, whereas the reverse was thought to be true in plants. Now, however, it is clear that microRNAs can induce mRNA degradation in animals and, conversely, translational repression in plants. Recent studies have made important advances in elucidating the relative contributions of these two different modes of target regulation by microRNAs. They have also shed light on the specific mechanisms of target silencing, which, although it differs fundamentally between plants and animals, shares some common features between the two kingdoms.


Subject(s)
Gene Silencing , MicroRNAs/genetics , Protein Biosynthesis , RNA Stability , Animals , Humans , Models, Genetic
8.
Nucleic Acids Res ; 41(2): 978-94, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23172285

ABSTRACT

Animal miRNAs silence the expression of mRNA targets through translational repression, deadenylation and subsequent mRNA degradation. Silencing requires association of miRNAs with an Argonaute protein and a GW182 family protein. In turn, GW182 proteins interact with poly(A)-binding protein (PABP) and the PAN2-PAN3 and CCR4-NOT deadenylase complexes. These interactions are required for the deadenylation and decay of miRNA targets. Recent studies have indicated that miRNAs repress translation before inducing target deadenylation and decay; however, whether translational repression and deadenylation are coupled or represent independent repressive mechanisms is unclear. Another remaining question is whether translational repression also requires GW182 proteins to interact with both PABP and deadenylases. To address these questions, we characterized the interaction of Drosophila melanogaster GW182 with deadenylases and defined the minimal requirements for a functional GW182 protein. Functional assays in D. melanogaster and human cells indicate that miRNA-mediated translational repression and degradation are mechanistically linked and are triggered through the interactions of GW182 proteins with PABP and deadenylases.


Subject(s)
Drosophila Proteins/metabolism , MicroRNAs/metabolism , Poly(A)-Binding Proteins/metabolism , RNA Interference , Ribonucleases/metabolism , Animals , Carrier Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HeLa Cells , Humans , Protein Biosynthesis , Protein Interaction Domains and Motifs , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , Transcription Factors/chemistry
9.
EMBO J ; 29(24): 4146-60, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21063388

ABSTRACT

miRNA-mediated gene silencing requires the GW182 proteins, which are characterized by an N-terminal domain that interacts with Argonaute proteins (AGOs), and a C-terminal silencing domain (SD). In Drosophila melanogaster (Dm) GW182 and a human (Hs) orthologue, TNRC6C, the SD was previously shown to interact with the cytoplasmic poly(A)-binding protein (PABPC1). Here, we show that two regions of GW182 proteins interact with PABPC1: the first contains a PABP-interacting motif 2 (PAM2; as shown before for TNRC6C) and the second contains the M2 and C-terminal sequences in the SD. The latter mediates indirect binding to the PABPC1 N-terminal domain. In D. melanogaster cells, the second binding site dominates; however, in HsTNRC6A-C the PAM2 motif is essential for binding to both Hs and DmPABPC1. Accordingly, a single amino acid substitution in the TNRC6A-C PAM2 motif abolishes the interaction with PABPC1. This mutation also impairs TNRC6s silencing activity. Our findings reveal that despite species-specific differences in the relative strength of the PABPC1-binding sites, the interaction between GW182 proteins and PABPC1 is critical for miRNA-mediated silencing in animal cells.


Subject(s)
Drosophila Proteins/metabolism , Gene Silencing , MicroRNAs/metabolism , Poly(A)-Binding Protein I/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Substitution/genetics , Animals , Binding Sites , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster , Genes, Reporter , Humans , Immunoprecipitation , Luciferases/genetics , Luciferases/metabolism , Mutagenesis, Site-Directed , Poly(A)-Binding Protein I/genetics , Protein Interaction Mapping , RNA-Binding Proteins/genetics
10.
Nucleic Acids Res ; 40(12): 5651-65, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22402495

ABSTRACT

GW182 family proteins are essential for miRNA-mediated gene silencing in animal cells. They are recruited to miRNA targets via interactions with Argonaute proteins and then promote translational repression and degradation of the miRNA targets. The human and Drosophila melanogaster GW182 proteins share a similar domain organization and interact with PABPC1 as well as with subunits of the PAN2-PAN3 and CCR4-NOT deadenylase complexes. The homologous proteins in Caenorhabditis elegans, AIN-1 and AIN-2, lack most of the domains present in the vertebrate and insect proteins, raising the question as to how AIN-1 and AIN-2 contribute to silencing. Here, we show that both AIN-1 and AIN-2 interact with Argonaute proteins through GW repeats in the middle region of the AIN proteins. However, only AIN-1 interacts with C. elegans and D. melanogaster PABPC1, PAN3, NOT1 and NOT2, suggesting that AIN-1 and AIN-2 are functionally distinct. Our findings reveal a surprising evolutionary plasticity of the GW182 protein interaction network and demonstrate that binding to PABPC1 and deadenylase complexes has been maintained throughout evolution, highlighting the significance of these interactions for silencing.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/metabolism , Poly(A)-Binding Protein I/metabolism , Ribonucleases/metabolism , Animals , Argonaute Proteins/antagonists & inhibitors , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Carrier Proteins/chemistry , Cell Line , Drosophila/genetics , Drosophila Proteins/antagonists & inhibitors , Gene Silencing , Poly(A)-Binding Protein I/chemistry , Protein Interaction Domains and Motifs , Protein Subunits/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Repetitive Sequences, Amino Acid
11.
Nucleic Acids Res ; 40(21): 11058-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22977175

ABSTRACT

The CCR4-NOT complex plays a crucial role in post-transcriptional mRNA regulation in eukaryotic cells. It catalyzes the removal of mRNA poly(A) tails, thereby repressing translation and committing mRNAs to decay. The conserved core of the complex consists of a catalytic module comprising two deadenylases (CAF1/POP2 and CCR4a/b) and the NOT module, which contains at least NOT1, NOT2 and NOT3. NOT1 bridges the interaction between the two modules and therefore, acts as a scaffold protein for the assembly of the complex. Here, we present the crystal structures of the CAF1-binding domain of human NOT1 alone and in complex with CAF1. The NOT1 domain comprises five helical hairpins that adopt an MIF4G (middle portion of eIF4G) fold. This NOT1 MIF4G domain binds CAF1 through a pre-formed interface and leaves the CAF1 catalytic site fully accessible to RNA substrates. The conservation of critical structural and interface residues suggests that the NOT1 MIF4G domain adopts a similar fold and interacts with CAF1 in a similar manner in all eukaryotes. Our findings shed light on the assembly of the CCR4-NOT complex and provide the basis for dissecting the role of the NOT module in mRNA deadenylation.


Subject(s)
Ribonucleases/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Drosophila Proteins/metabolism , Drosophila melanogaster , Exoribonucleases/metabolism , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Ribonucleases/metabolism , Sequence Alignment , Transcription Factors/metabolism
12.
Proc Natl Acad Sci U S A ; 108(26): 10466-71, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21646546

ABSTRACT

Argonaute proteins (AGOs) are essential effectors in RNA-mediated gene silencing pathways. They are characterized by a bilobal architecture, in which one lobe contains the N-terminal and PAZ domains and the other contains the MID and PIWI domains. Here, we present the first crystal structure of the MID-PIWI lobe from a eukaryotic AGO, the Neurospora crassa QDE-2 protein. Compared to prokaryotic AGOs, the domain orientation is conserved, indicating a conserved mode of nucleic acid binding. The PIWI domain shows an adaptable surface loop next to a eukaryote-specific α-helical insertion, which are both likely to contact the PAZ domain in a conformation-dependent manner to sense the functional state of the protein. The MID-PIWI interface is hydrophilic and buries residues that were previously thought to participate directly in the allosteric regulation of guide RNA binding. The interface includes the binding pocket for the guide RNA 5' end, and residues from both domains contribute to binding. Accordingly, micro-RNA (miRNA) binding is particularly sensitive to alteration in the MID-PIWI interface in Drosophila melanogaster AGO1 in vivo. The structure of the QDE-2 MID-PIWI lobe provides molecular and mechanistic insight into eukaryotic AGOs and has significant implications for understanding the role of these proteins in silencing.


Subject(s)
Fungal Proteins/chemistry , Crystallography, X-Ray , Fungal Proteins/metabolism , Models, Molecular , Neurospora crassa/chemistry , Nucleic Acids/metabolism , Protein Binding , Protein Conformation
13.
Adv Exp Med Biol ; 768: 147-63, 2013.
Article in English | MEDLINE | ID: mdl-23224969

ABSTRACT

GW182 family proteins are essential for microRNA-mediated gene silencing in animal cells. They are recruited to miRNA targets through direct interactions with Argonaute proteins and promote target silencing. They do so by repressing translation and enhancing mRNA turnover. Although the precise mechanism of action of GW182 proteins is not fully understood, these proteins have been shown to interact with the cytoplasmic poly(A)-binding protein (PABP) and with the PAN2-PAN3 and CCR4-NOT deadenylase complexes. These findings suggest that GW182 proteins function as scaffold proteins for the assembly of the multiprotein complex that silences miRNA targets.


Subject(s)
Argonaute Proteins/genetics , Autoantigens/genetics , MicroRNAs/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Animals , Argonaute Proteins/metabolism , Autoantigens/chemistry , Autoantigens/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , MicroRNAs/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Protein Binding , Protein Biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism
14.
RNA ; 15(1): 21-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029310

ABSTRACT

miRNAs silence gene expression by repressing translation and/or by promoting mRNA decay. In animal cells, degradation of partially complementary miRNA targets occurs via deadenylation by the CAF1-CCR4-NOT1 deadenylase complex, followed by decapping and subsequent exonucleolytic digestion. To determine how generally miRNAs trigger deadenylation, we compared mRNA expression profiles in D. melanogaster cells depleted of AGO1, CAF1, or NOT1. We show that approximately 60% of AGO1 targets are regulated by CAF1 and/or NOT1, indicating that deadenylation is a widespread effect of miRNA regulation. However, neither a poly(A) tail nor mRNA circularization are required for silencing, because mRNAs whose 3' ends are generated by a self-cleaving ribozyme are also silenced in vivo. We show further that miRNAs trigger mRNA degradation, even when binding by 40S ribosomal subunits is inhibited in cis. These results indicate that miRNAs promote mRNA decay by altering mRNP composition and/or conformation, rather than by directly interfering with the binding and function of ribosomal subunits.


Subject(s)
MicroRNAs/metabolism , RNA Stability/physiology , Animals , Argonaute Proteins , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eukaryotic Initiation Factors , Gene Silencing , MicroRNAs/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Poly A/genetics , Poly A/metabolism , Protein Biosynthesis , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinoblastoma-Binding Protein 4 , Ribonucleases/genetics , Ribonucleases/metabolism , Ribonucleoproteins/metabolism
15.
RNA ; 14(12): 2609-17, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18974281

ABSTRACT

Messenger RNAs harboring nonsense codons (or premature translation termination codons [PTCs]) are degraded by a conserved quality-control mechanism known as nonsense-mediated mRNA decay (NMD), which prevents the accumulation of truncated and potentially harmful proteins. In Drosophila melanogaster, degradation of PTC-containing messages is initiated by endonucleolytic cleavage in the vicinity of the nonsense codon. The endonuclease responsible for this cleavage has not been identified. Here, we show that SMG6 is the long sought NMD endonuclease. First, cells expressing an SMG6 protein mutated at catalytic residues fail to degrade PTC-containing messages. Moreover, the SMG6-PIN domain can be replaced with the active PIN domain of an unrelated protein, indicating that its sole function is to provide endonuclease activity for NMD. Unexpectedly, we found that the catalytic activity of SMG6 contributes to the degradation of PTC-containing mRNAs in human cells. Thus, SMG6 is a conserved endonuclease that degrades mRNAs terminating translation prematurely in metazoa.


Subject(s)
Codon, Nonsense , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Endoribonucleases/metabolism , Animals , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Endoribonucleases/chemistry , Protein Structure, Tertiary
16.
Nucleic Acids Res ; 31(24): 7302-10, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14654705

ABSTRACT

The Hfq protein, which shares sequence and structural homology with the Sm and Lsm proteins, binds to various RNAs, primarily recognizing AU-rich single-stranded regions. In this paper, we study the ability of the Escherichia coli Hfq protein to bind to a polyadenylated fragment of rpsO mRNA. Hfq exhibits a high specificity for a 100-nucleotide RNA harboring 18 3'-terminal A-residues. Structural analysis of the adenylated RNA-Hfq complex and gel shift assays revealed the presence of two Hfq binding sites. Hfq binds primarily to the poly(A) tail, and to a lesser extent a U-rich sequence in a single-stranded region located between two hairpin structures. The oligo(A) tail and the interhelical region are sensitive to 3'-5' exoribonucleases and RNase E hydrolysis, respectively, in vivo. In vitro assays demonstrate that Hfq protects poly(A) tails from exonucleolytic degradation by both PNPase and RNase II. In addition, RNase E processing, which occurred close to the U-rich sequence, is impaired by the presence of Hfq. These data suggest that Hfq modulates the sensitivity of RNA to ribonucleases in the cell.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/metabolism , Poly A/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Base Sequence , Binding Sites , Electrophoretic Mobility Shift Assay , Exoribonucleases/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Poly A/chemistry , Poly A/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomal Proteins/genetics , Substrate Specificity , Thermodynamics
17.
Nat Struct Mol Biol ; 21(7): 599-608, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880343

ABSTRACT

The PAN2-PAN3 complex functions in general and microRNA-mediated mRNA deadenylation. However, mechanistic insight into PAN2 and its complex with the asymmetric PAN3 dimer is lacking. Here, we describe crystal structures that show that Neurospora crassa PAN2 comprises two independent structural units: a C-terminal catalytic unit and an N-terminal assembly unit that engages in a bipartite interaction with PAN3 dimers. The catalytic unit contains the exonuclease domain in an intimate complex with a potentially modulatory ubiquitin-protease-like domain. The assembly unit contains a WD40 propeller connected to an adaptable linker. The propeller contacts the PAN3 C-terminal domain, whereas the linker reinforces the asymmetry of the PAN3 dimer and prevents the recruitment of a second PAN2 molecule. Functional data indicate an essential role for PAN3 in coordinating PAN2-mediated deadenylation with subsequent steps in mRNA decay, which lead to complete mRNA degradation.


Subject(s)
Exoribonucleases/physiology , Fungal Proteins/physiology , RNA Stability , RNA, Messenger/metabolism , Binding Sites , Crystallography, X-Ray , Dimerization , Exoribonucleases/chemistry , Exoribonucleases/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Neurospora crassa , Protein Interaction Mapping , Protein Structure, Tertiary
18.
Article in English | MEDLINE | ID: mdl-23209154

ABSTRACT

MicroRNAs (miRNAs) are a large family of endogenous noncoding RNAs that, together with the Argonaute family of proteins (AGOs), silence the expression of complementary mRNA targets posttranscriptionally. Perfectly complementary targets are cleaved within the base-paired region by catalytically active AGOs. In the case of partially complementary targets, however, AGOs are insufficient for silencing and need to recruit a protein of the GW182 family. GW182 proteins induce translational repression, mRNA deadenylation and exonucleolytic target degradation. Recent work has revealed a direct molecular link between GW182 proteins and cellular deadenylase complexes. These findings shed light on how miRNAs bring about target mRNA degradation and promise to further our understanding of the mechanism of miRNA-mediated repression.


Subject(s)
Argonaute Proteins/metabolism , Autoantigens/metabolism , Exoribonucleases/metabolism , Gene Silencing/physiology , MicroRNAs/metabolism , Models, Biological , Poly(A)-Binding Proteins/metabolism , Protein Interaction Maps/physiology , RNA-Binding Proteins/metabolism , Autoantigens/genetics , Protein Interaction Maps/genetics , RNA-Binding Proteins/genetics
19.
Nat Struct Mol Biol ; 19(12): 1324-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142987

ABSTRACT

The removal of the mRNA 5' cap structure by the decapping enzyme DCP2 leads to rapid 5'→3' mRNA degradation by XRN1, suggesting that the two processes are coordinated, but the coupling mechanism is unknown. DCP2 associates with the decapping activators EDC4 and DCP1. Here we show that XRN1 directly interacts with EDC4 and DCP1 in human and Drosophila melanogaster cells, respectively. In D. melanogaster cells, this interaction is mediated by the DCP1 EVH1 domain and a DCP1-binding motif (DBM) in the XRN1 C-terminal region. The NMR structure of the DCP1 EVH1 domain bound to the DBM reveals that the peptide docks at a conserved aromatic cleft, which is used by EVH1 domains to recognize proline-rich ligands. Our findings reveal a role for XRN1 in decapping and provide a molecular basis for the coupling of decapping to 5'→3' mRNA degradation.


Subject(s)
Endopeptidases/metabolism , Exoribonucleases/metabolism , Microtubule-Associated Proteins/metabolism , RNA, Messenger/metabolism , Amino Acid Sequence , Animals , Drosophila melanogaster , Endopeptidases/chemistry , Exoribonucleases/chemistry , Humans , Microtubule-Associated Proteins/chemistry , Molecular Sequence Data , Proteolysis , Sequence Homology, Amino Acid
20.
Mol Cell Biol ; 29(23): 6220-31, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19797087

ABSTRACT

GW182 family proteins are essential in animal cells for microRNA (miRNA)-mediated gene silencing, yet the molecular mechanism that allows GW182 to promote translational repression and mRNA decay remains largely unknown. Previous studies showed that while the GW182 N-terminal domain interacts with Argonaute proteins, translational repression and degradation of miRNA targets are promoted by a bipartite silencing domain comprising the GW182 middle and C-terminal regions. Here we show that the GW182 C-terminal region is required for GW182 to release silenced mRNPs; moreover, GW182 dissociates from miRNA targets at a step of silencing downstream of deadenylation, indicating that GW182 is required to initiate but not to maintain silencing. In addition, we show that the GW182 bipartite silencing domain competes with eukaryotic initiation factor 4G for binding to PABPC1. The GW182-PABPC1 interaction is also required for miRNA target degradation; accordingly, we observed that PABPC1 associates with components of the CCR4-NOT deadenylase complex. Finally, we show that PABPC1 overexpression suppresses the silencing of miRNA targets. We propose a model in which the GW182 silencing domain promotes translational repression, at least in part, by interfering with mRNA circularization and also recruits the deadenylase complex through the interaction with PABPC1.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Silencing , MicroRNAs/genetics , Poly(A)-Binding Protein I/metabolism , Protein Biosynthesis , Animals , Argonaute Proteins , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Poly(A)-Binding Protein I/genetics , Protein Binding , Ribonucleases/genetics , Ribonucleases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL