Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
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.
Cell Rep ; 42(1): 111902, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36586408

ABSTRACT

The evolutionary conserved CCR4-NOT complex functions in the cytoplasm as the main mRNA deadenylase in both constitutive mRNA turnover and regulated mRNA decay pathways. The versatility of this complex is underpinned by its modular multi-subunit organization, with distinct structural modules actuating different functions. The structure and function of all modules are known, except for that of the N-terminal module. Using different structural approaches, we obtained high-resolution data revealing the architecture of the human N-terminal module composed of CNOT1, CNOT10, and CNOT11. The structure shows how two helical domains of CNOT1 sandwich CNOT10 and CNOT11, leaving the most conserved domain of CNOT11 protruding into solvent as an antenna. We discovered that GGNBP2, a protein identified as a tumor suppressor and spermatogenic factor, is a conserved interacting partner of the CNOT11 antenna domain. Structural and biochemical analyses thus pinpoint the N-terminal CNOT1-CNOT10-CNOT11 module as a conserved protein-protein interaction platform.


Subject(s)
Transcription Factors , Humans , Transcription Factors/metabolism , Protein Binding
3.
Nucleic Acids Res ; 51(2): 517-535, 2023 01 25.
Article in English | MEDLINE | ID: mdl-35934316

ABSTRACT

N6-Methyladenosine (m6A), one of the most abundant internal modification of eukaryotic mRNAs, participates in the post-transcriptional control of gene expression through recruitment of specific m6A readers. In Saccharomyces cerevisiae, the m6A methyltransferase Ime4 is expressed only during meiosis and its deletion impairs this process. To elucidate how m6A control gene expression, we investigated the function of the budding yeast m6A reader Pho92. We show that Pho92 is an early meiotic factor that promotes timely meiotic progression. High-throughput RNA sequencing and mapping of Pho92-binding sites following UV-crosslinking reveal that Pho92 is recruited to specific mRNAs in an m6A-dependent manner during the meiotic prophase, preceding their down-regulation. Strikingly, point mutations altering m6A sites in mRNAs targeted by Pho92 are sufficient to delay their down-regulation and, in one case, to slow down meiotic progression. Altogether, our results indicate that Pho92 facilitate the meiotic progression by accelerating the down-regulation of timely-regulated mRNAs during meiotic recombination.


mRNAs molecules carry information contained in genes to direct the formation of proteins. In specific circumstances, the cellular machinery modifies some mRNAs through the formation of m6A residues. To understand the function of these m6A marks, the authors used the yeast Saccharomyces cerevisiae in which their formation only occurs during meiosis that leads to spore formation. Characterization of the Pho92 protein that specifically recognizes m6A residues revealed its importance for meiosis. m6A sites bound by Pho92 were identified and shown to be biologically functional. Unexpectedly, Pho92 was found to regulate an early step of meiosis by controlling DNA recombination. Overall, this study provides important clues on the role of m6A residues in mRNAs.


Subject(s)
RNA-Binding Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Homologous Recombination , Meiosis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , RNA-Binding Proteins/metabolism , Methylation
4.
RNA Biol ; 18(12): 2450-2465, 2021 12.
Article in English | MEDLINE | ID: mdl-34060423

ABSTRACT

Antiproliferative BTG/Tob proteins interact directly with the CAF1 deadenylase subunit of the CCR4-NOT complex. This binding requires the presence of two conserved motifs, boxA and boxB, characteristic of the BTG/Tob APRO domain. Consistently, these proteins were shown to stimulate mRNA deadenylation and decay in several instances. Two members of the family, BTG1 and BTG2, were reported further to associate with the protein arginine methyltransferase PRMT1 through a motif, boxC, conserved only in this subset of proteins. We recently demonstrated that BTG1 and BTG2 also contact the first RRM domain of the cytoplasmic poly(A) binding protein PABPC1. To decipher the mode of interaction of BTG1 and BTG2 with partners, we performed nuclear magnetic resonance experiments as well as mutational and biochemical analyses. Our data demonstrate that, in the context of an APRO domain, the boxC motif is necessary and sufficient to allow interaction with PABPC1 but, unexpectedly, that it is not required for BTG2 association with PRMT1. We show further that the presence of a boxC motif in an APRO domain endows it with the ability to stimulate deadenylation in cellulo and in vitro. Overall, our results identify the molecular interface allowing BTG1 and BTG2 to activate deadenylation, a process recently shown to be necessary for maintaining T-cell quiescence.


Subject(s)
Immediate-Early Proteins/metabolism , Neoplasm Proteins/metabolism , Poly A/metabolism , Polyadenylation , Protein-Arginine N-Methyltransferases/metabolism , RNA, Messenger/chemistry , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Neoplasm Proteins/genetics , Poly A/genetics , Protein Binding , Protein-Arginine N-Methyltransferases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
5.
Nucleic Acids Res ; 48(11): 6353-6366, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32396195

ABSTRACT

Most eukaryotic mRNAs harbor a characteristic 5' m7GpppN cap that promotes pre-mRNA splicing, mRNA nucleocytoplasmic transport and translation while also protecting mRNAs from exonucleolytic attacks. mRNA caps are eliminated by Dcp2 during mRNA decay, allowing 5'-3' exonucleases to degrade mRNA bodies. However, the Dcp2 decapping enzyme is poorly active on its own and requires binding to stable or transient protein partners to sever the cap of target mRNAs. Here, we analyse the role of one of these partners, the yeast Pby1 factor, which is known to co-localize into P-bodies together with decapping factors. We report that Pby1 uses its C-terminal domain to directly bind to the decapping enzyme. We solved the structure of this Pby1 domain alone and bound to the Dcp1-Dcp2-Edc3 decapping complex. Structure-based mutant analyses reveal that Pby1 binding to the decapping enzyme is required for its recruitment into P-bodies. Moreover, Pby1 binding to the decapping enzyme stimulates growth in conditions in which decapping activation is compromised. Our results point towards a direct connection of Pby1 with decapping and P-body formation, both stemming from its interaction with the Dcp1-Dcp2 holoenzyme.


Subject(s)
DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Catalytic Domain , DNA-Binding Proteins/chemistry , Endopeptidases/chemistry , Endopeptidases/metabolism , Endoribonucleases/chemistry , Holoenzymes/chemistry , Holoenzymes/metabolism , Ligases/metabolism , Models, Molecular , Organelles/enzymology , Organelles/metabolism , Protein Binding , Protein Domains , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/chemistry
6.
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
7.
Proc Natl Acad Sci U S A ; 114(45): E9493-E9501, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078363

ABSTRACT

The Pat1 protein is a central player of eukaryotic mRNA decay that has also been implicated in translational control. It is commonly considered a central platform responsible for the recruitment of several RNA decay factors. We demonstrate here that a yeast-specific C-terminal region from Pat1 interacts with several short motifs, named helical leucine-rich motifs (HLMs), spread in the long C-terminal region of yeast Dcp2 decapping enzyme. Structures of Pat1-HLM complexes reveal the basis for HLM recognition by Pat1. We also identify a HLM present in yeast Xrn1, the main 5'-3' exonuclease involved in mRNA decay. We show further that the ability of yeast Pat1 to bind HLMs is required for efficient growth and normal mRNA decay. Overall, our analyses indicate that yeast Pat1 uses a single binding surface to successively recruit several mRNA decay factors and show that interaction between those factors is highly polymorphic between species.


Subject(s)
Endoribonucleases/metabolism , Exoribonucleases/metabolism , Fungal Proteins/metabolism , RNA, Messenger/metabolism , Yeasts/metabolism , Protein Binding/physiology , Protein Domains/physiology , RNA Stability/physiology , RNA-Binding Proteins/metabolism
8.
Enzymes ; 41: 117-149, 2017.
Article in English | MEDLINE | ID: mdl-28601220

ABSTRACT

Elongator is a highly conserved eukaryotic protein complex consisting of two sets of six Elp proteins, while homologues of its catalytic subunit Elp3 are found in all the kingdoms of life. Although it was originally described as a transcription elongation factor, cumulating evidence suggests that its primary function is catalyzing tRNA modifications. In humans, defects in Elongator subunits are associated with neurological disorders and cancer. Although further studies are still required, a clearer picture of the molecular mechanism of action of Elongator and its cofactors has started to emerge within recent years that have witnessed significant development in the field. In this review we summarize recent Elongator-related findings provided largely by crystal structures of several subunits of the complex, the electron microscopy structure of the entire yeast holoenzyme, as well as the structure of the Elongator cofactor complex Kti11/Kti13.


Subject(s)
Histone Acetyltransferases/chemistry , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Peptide Elongation Factors/chemistry , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Repressor Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/ultrastructure , Holoenzymes/chemistry , Holoenzymes/metabolism , Holoenzymes/ultrastructure , Humans , Multiprotein Complexes/ultrastructure , Peptide Elongation Factors/metabolism , Peptide Elongation Factors/ultrastructure , RNA, Transfer/genetics , Repressor Proteins/metabolism , Repressor Proteins/ultrastructure , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure
9.
RNA ; 23(6): 968-981, 2017 06.
Article in English | MEDLINE | ID: mdl-28348170

ABSTRACT

Spliceosomal proteins Hsh49p and Cus1p are components of SF3b, which together with SF3a, Msl1p/Lea1p, Sm proteins, and U2 snRNA, form U2 snRNP, which plays a crucial role in pre-mRNA splicing. Hsh49p, comprising two RRMs, forms a heterodimer with Cus1p. We determined the crystal structures of Saccharomyces cerevisiae full-length Hsh49p as well as its RRM1 in complex with a minimal binding region of Cus1p (residues 290-368). The structures show that the Cus1 fragment binds to the α-helical surface of Hsh49p RRM1, opposite the four-stranded ß-sheet, leaving the canonical RNA-binding surface available to bind RNA. Hsh49p binds the 5' end region of U2 snRNA via RRM1. Its affinity is increased in complex with Cus1(290-368)p, partly because an extended RNA-binding surface forms across the protein-protein interface. The Hsh49p RRM1-Cus1(290-368)p structure fits well into cryo-EM density of the Bact spliceosome, corroborating the biological relevance of our crystal structure.


Subject(s)
Models, Molecular , Protein Conformation , Ribonucleoprotein, U2 Small Nuclear/chemistry , Amino Acid Sequence , Binding Sites , Conserved Sequence , Multiprotein Complexes/metabolism , Proline-Rich Protein Domains , Protein Binding , Protein Interaction Domains and Motifs , RNA/chemistry , RNA/genetics , RNA/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism
10.
EMBO Rep ; 18(2): 264-279, 2017 02.
Article in English | MEDLINE | ID: mdl-27974378

ABSTRACT

The highly conserved eukaryotic Elongator complex performs specific chemical modifications on wobble base uridines of tRNAs, which are essential for proteome stability and homeostasis. The complex is formed by six individual subunits (Elp1-6) that are all equally important for its tRNA modification activity. However, its overall architecture and the detailed reaction mechanism remain elusive. Here, we report the structures of the fully assembled yeast Elongator and the Elp123 sub-complex solved by an integrative structure determination approach showing that two copies of the Elp1, Elp2, and Elp3 subunits form a two-lobed scaffold, which binds Elp456 asymmetrically. Our topological models are consistent with previous studies on individual subunits and further validated by complementary biochemical analyses. Our study provides a structural framework on how the tRNA modification activity is carried out by Elongator.


Subject(s)
Fungal Proteins/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Mutation , Protein Binding , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Protein Transport , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
11.
Nat Struct Mol Biol ; 23(11): 982-986, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27694841

ABSTRACT

Elimination of the 5' cap of eukaryotic mRNAs, known as decapping, is considered to be a crucial, irreversible and highly regulated step required for the rapid degradation of mRNA by Xrn1, the major cytoplasmic 5'-3' exonuclease. Decapping is accomplished by the recruitment of a protein complex formed by the Dcp2 catalytic subunit and its Dcp1 cofactor. However, this complex has a low intrinsic enzymatic activity and requires several accessory proteins such as the Lsm1-7 complex, Pat1, Edc1-Edc2 and/or Edc3 to be fully active. Here we present the crystal structure of the active form of the yeast Kluyveromyces lactis Dcp1-Dcp2 enzyme bound to its product (m7GDP) and its potent activator Edc3. This structure of the Dcp1-Dcp2 complex bound to a cap analog further explains previously published data on substrate binding and provides hints as to the mechanism of Edc3-mediated Dcp2 activation.


Subject(s)
Endoribonucleases/metabolism , Fungal Proteins/metabolism , Guanosine Diphosphate/analogs & derivatives , Saccharomycetales/metabolism , Crystallography, X-Ray , Endoribonucleases/chemistry , Enzyme Activation , Fungal Proteins/chemistry , Guanosine Diphosphate/metabolism , Models, Molecular , Protein Binding , Protein Conformation , RNA Stability , Saccharomycetales/chemistry
12.
Mol Cell ; 63(6): 927-38, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27635759

ABSTRACT

Acetylation of histones and transcription-related factors is known to exert epigenetic and transcriptional control of gene expression. Here we report that histone acetyltransferases (HATs) and histone deacetylases (HDACs) also regulate gene expression at the posttranscriptional level by controlling poly(A) RNA stability. Inhibition of HDAC1 and HDAC2 induces massive and widespread degradation of normally stable poly(A) RNA in mammalian and Drosophila cells. Acetylation-induced RNA decay depends on the HATs p300 and CBP, which acetylate the exoribonuclease CAF1a, a catalytic subunit of the CCR4-CAF1-NOT deadenlyase complex and thereby contribute to accelerating poly(A) RNA degradation. Taking adipocyte differentiation as a model, we observe global stabilization of poly(A) RNA during differentiation, concomitant with loss of CBP/p300 expression. Our study uncovers reversible acetylation as a fundamental switch by which HATs and HDACs control the overall turnover of poly(A) RNA.


Subject(s)
Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Poly A/genetics , RNA, Messenger/genetics , p300-CBP Transcription Factors/genetics , 3T3-L1 Cells , Acetylation , Amino Acid Sequence , Animals , Cell Differentiation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , HEK293 Cells , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Poly A/metabolism , RNA Stability , RNA, Messenger/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
13.
Nat Struct Mol Biol ; 23(9): 794-802, 2016 09.
Article in English | MEDLINE | ID: mdl-27455459

ABSTRACT

During translation elongation, decoding is based on the recognition of codons by corresponding tRNA anticodon triplets. Molecular mechanisms that regulate global protein synthesis via specific base modifications in tRNA anticodons are receiving increasing attention. The conserved eukaryotic Elongator complex specifically modifies uridines located in the wobble base position of tRNAs. Mutations in Elongator subunits are associated with certain neurodegenerative diseases and cancer. Here we present the crystal structure of D. mccartyi Elp3 (DmcElp3) at 2.15-Å resolution. Our results reveal an unexpected arrangement of Elp3 lysine acetyltransferase (KAT) and radical S-adenosyl methionine (SAM) domains, which share a large interface and form a composite active site and tRNA-binding pocket, with an iron-sulfur cluster located in the dimerization interface of two DmcElp3 molecules. Structure-guided mutagenesis studies of yeast Elp3 confirmed the relevance of our findings for eukaryotic Elp3s and should aid in understanding the cellular functions and pathophysiological roles of Elongator.


Subject(s)
Bacterial Proteins/chemistry , Histone Acetyltransferases/chemistry , RNA, Transfer/chemistry , Catalytic Domain , Chloroflexi/enzymology , Crystallography, X-Ray , Protein Binding , Protein Conformation, alpha-Helical , Protein Multimerization , RNA, Bacterial/chemistry , Substrate Specificity
14.
Nat Commun ; 7: 10811, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26912148

ABSTRACT

While BTG2 plays an important role in cellular differentiation and cancer, its precise molecular function remains unclear. BTG2 interacts with CAF1 deadenylase through its APRO domain, a defining feature of BTG/Tob factors. Our previous experiments revealed that expression of BTG2 promoted mRNA poly(A) tail shortening through an undefined mechanism. Here we report that the APRO domain of BTG2 interacts directly with the first RRM domain of the poly(A)-binding protein PABPC1. Moreover, PABPC1 RRM and BTG2 APRO domains are sufficient to stimulate CAF1 deadenylase activity in vitro in the absence of other CCR4-NOT complex subunits. Our results unravel thus the mechanism by which BTG2 stimulates mRNA deadenylation, demonstrating its direct role in poly(A) tail length control. Importantly, we also show that the interaction of BTG2 with the first RRM domain of PABPC1 is required for BTG2 to control cell proliferation.


Subject(s)
Cell Proliferation , Immediate-Early Proteins/metabolism , Poly(A)-Binding Protein I/metabolism , RNA, Messenger/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Blotting, Western , Cell Line, Tumor , HEK293 Cells , Humans , Immunoprecipitation , In Vitro Techniques , Protein Structure, Tertiary
15.
Nucleic Acids Res ; 44(1): 437-48, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26602689

ABSTRACT

Metazoan SR and SR-like proteins are important regulatory factors in RNA splicing, export, translation and RNA decay. We determined the NMR structures and nucleic acid interaction modes of Gbp2 and Hrb1, two paralogous budding yeast proteins with similarities to mammalian SR proteins. Gbp2 RRM1 and RRM2 recognise preferentially RNAs containing the core motif GGUG. Sequence selectivity resides in a non-canonical interface in RRM2 that is highly related to the SRSF1 pseudoRRM. The atypical Gbp2/Hrb1 C-terminal RRM domains (RRM3) do not interact with RNA/DNA, likely because of their novel N-terminal extensions that block the canonical RNA binding interface. Instead, we discovered that RRM3 is crucial for interaction with the THO/TREX complex and identified key residues essential for this interaction. Moreover, Gbp2 interacts genetically with Tho2 as the double deletion shows a synthetic phenotype and preventing Gbp2 interaction with the THO/TREX complex partly supresses gene expression defect associated with inactivation of the latter complex. These findings provide structural and functional insights into the contribution of SR-like proteins in the post-transcriptional control of gene expression.


Subject(s)
Multiprotein Complexes/metabolism , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Carbon-13 Magnetic Resonance Spectroscopy , DNA/metabolism , Models, Molecular , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/metabolism , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/metabolism , Protein Binding , Protein Conformation , Proton Magnetic Resonance Spectroscopy , RNA/metabolism , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Telomere/genetics , Telomere/metabolism
16.
Hum Mol Genet ; 24(11): 3163-71, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25712129

ABSTRACT

mRNA decay is an essential and active process that allows cells to continuously adapt gene expression to internal and environmental cues. There are two mRNA degradation pathways: 3' to 5' and 5' to 3'. The DCPS protein is the scavenger mRNA decapping enzyme which functions in the last step of the 3' end mRNA decay pathway. We have identified a DCPS pathogenic mutation in a large family with three affected individuals presenting with a novel recessive syndrome consisting of craniofacial anomalies, intellectual disability and neuromuscular defects. Using patient's primary cells, we show that this homozygous splice mutation results in a DCPS loss-of-function allele. Diagnostic biochemical analyses using various m7G cap derivatives as substrates reveal no DCPS enzymatic activity in patient's cells. Our results implicate DCPS and more generally RNA catabolism, as a critical cellular process for neurological development, normal cognition and organismal homeostasis in humans.


Subject(s)
Abnormalities, Multiple/genetics , Endoribonucleases/genetics , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Cells, Cultured , Child , Child, Preschool , Consanguinity , DNA Mutational Analysis , Endoribonucleases/deficiency , Genetic Association Studies , Humans , Male , Pedigree , RNA Splice Sites , Syndrome
17.
FEBS J ; 282(5): 819-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25604895

ABSTRACT

UNLABELLED: Modification of wobble uridines of many eukaryotic tRNAs requires the Elongator complex, a highly conserved six-subunit eukaryotic protein assembly, as well as the Killer toxin-insensitive (Kti) proteins 11-14. Kti11 was additionally shown to be implicated in the biosynthesis of diphthamide, a post-translationally modified histidine of translation elongation factor 2. Recent data indicate that iron-bearing Kti11 functions as an electron donor to the [4Fe-4S] cluster of radical S-Adenosylmethionine enzymes, triggering the subsequent radical reaction. We show here that recombinant yeast Kti11 forms a stable 1 : 1 complex with Kti13. To obtain insights into the function of this heterodimer, the Kti11/Kti13 complex was purified to homogeneity, crystallized, and its structure determined at 1.45 Å resolution. The importance of several residues mediating complex formation was confirmed by mutagenesis. Kti13 adopts a fold characteristic of RCC1-like proteins. The seven-bladed ß-propeller consists of a unique mixture of four- and three-stranded blades. In the complex, Kti13 orients Kti11 and restricts access to its electron-carrying iron atom, constraining the electron transfer capacity of Kti11. Based on these findings, we propose a role for Kti13, and discuss the possible functional implications of complex formation. DATABASE: Structural data have been submitted to the Protein Data Bank under accession number 4X33.


Subject(s)
RNA, Transfer/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Electron Transport , Iron/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , RNA, Transfer/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Static Electricity
18.
Nucleic Acids Res ; 43(1): 482-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25432955

ABSTRACT

Eukaryotic 5' mRNA cap structures participate to the post-transcriptional control of gene expression before being released by the two main mRNA decay pathways. In the 3'-5' pathway, the exosome generates free cap dinucleotides (m7GpppN) or capped oligoribonucleotides that are hydrolyzed by the Scavenger Decapping Enzyme (DcpS) forming m7GMP. In the 5'-3' pathway, the decapping enzyme Dcp2 generates m7GDP. We investigated the fate of m7GDP and m7GpppN produced by RNA decay in extracts and cells. This defined a pathway involving DcpS, NTPs and the nucleoside diphosphate kinase for m7GDP elimination. Interestingly, we identified and characterized in vitro and in vivo a new scavenger decapping enzyme involved in m7GpppN degradation. We show that activities mediating cap elimination identified in yeast are essentially conserved in human. Their alteration may contribute to pathologies, possibly through the interference of cap (di)nucleotide with cellular function.


Subject(s)
Acid Anhydride Hydrolases/metabolism , Endoribonucleases/metabolism , Neoplasm Proteins/metabolism , RNA Caps/metabolism , RNA Stability , RNA, Messenger/metabolism , Adenosine Triphosphate/metabolism , Dinucleoside Phosphates/metabolism , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/metabolism , HEK293 Cells , Humans , N-Glycosyl Hydrolases/metabolism , Nucleoside-Diphosphate Kinase/metabolism , RNA Cap Analogs/metabolism , Saccharomyces cerevisiae Proteins/metabolism
19.
Nucleic Acids Res ; 42(20): 12847-60, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25352554

ABSTRACT

Splicing reactions generally combine high speed with accuracy. However, some of the pre-mRNAs escape the nucleus with a retained intron. Intron retention can control gene expression and increase proteome diversity. We calculated the escape rate for the yeast PTC7 intron and pre-mRNA. This prediction was facilitated by the observation that splicing is a linear process and by deriving simple algebraic expressions from a model of co- and post-transcriptional splicing and RNA surveillance that determines the rate of the nonsense-mediated decay (NMD) of the pre-mRNAs with the retained intron. The escape rate was consistent with the observed threshold of splicing rate below which the mature mRNA level declined. When an mRNA contains multiple introns, the outcome of splicing becomes more difficult to predict since not only the escape rate of the pre-mRNA has to be considered, but also the possibility that the splicing of each intron is influenced by the others. We showed that the two adjacent introns in the SUS1 mRNA are spliced cooperatively, but this does not counteract the escape of the partially spliced mRNA. These findings will help to infer promoter activity and to predict the behavior of and to control splicing regulatory networks.


Subject(s)
RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , Introns , Models, Genetic , Nuclear Proteins/genetics , Protein Phosphatase 2/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics
20.
J Biol Chem ; 289(41): 28640-50, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25160624

ABSTRACT

The retention and splicing (RES) complex is a conserved spliceosome-associated module that was shown to enhance splicing of a subset of transcripts and promote the nuclear retention of unspliced pre-mRNAs in yeast. The heterotrimeric RES complex is organized around the Snu17p protein that binds to both the Bud13p and Pml1p subunits. Snu17p exhibits an RRM domain that resembles a U2AF homology motif (UHM) and Bud13p harbors a Trp residue reminiscent of an UHM-ligand motif (ULM). It has therefore been proposed that the interaction between Snu17p and Bud13p resembles canonical UHM-ULM complexes. Here, we have used biochemical and NMR structural analysis to characterize the structure of the yeast Snu17p-Bud13p complex. Unlike known UHMs that sequester the Trp residue of the ULM ligand in a hydrophobic pocket, Snu17p and Bud13p utilize a large interaction surface formed around the two helices of the Snu17p domain. In total 18 residues of the Bud13p ligand wrap around the Snu17p helical surface in an U-turn-like arrangement. The invariant Trp(232) in Bud13p is located in the center of the turn, and contacts surface residues of Snu17p. The structural data are supported by mutational analysis and indicate that Snu17p provides an extended binding surface with Bud13p that is notably distinct from canonical UHM-ULM interactions. Our data highlight structural diversity in RRM-protein interactions, analogous to the one seen for nucleic acid interactions.


Subject(s)
Carrier Proteins/chemistry , RNA Precursors/biosynthesis , RNA, Fungal/biosynthesis , Ribonucleoprotein, U2 Small Nuclear/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Spliceosomes/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , RNA Precursors/genetics , RNA Splicing , RNA, Fungal/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Spliceosomes/metabolism , Tryptophan/chemistry , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...