Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(7): 3357-3374, 2023 04 24.
Article in English | MEDLINE | ID: mdl-36869663

ABSTRACT

The conserved H/ACA RNPs consist of one H/ACA RNA and 4 core proteins: dyskerin, NHP2, NOP10, and GAR1. Its assembly requires several assembly factors. A pre-particle containing the nascent RNAs, dyskerin, NOP10, NHP2 and NAF1 is assembled co-transcriptionally. NAF1 is later replaced by GAR1 to form mature RNPs. In this study, we explore the mechanism leading to the assembly of H/ACA RNPs. We performed the analysis of GAR1, NHP2, SHQ1 and NAF1 proteomes by quantitative SILAC proteomic, and analyzed purified complexes containing these proteins by sedimentation on glycerol gradient. We propose the formation of several distinct intermediate complexes during H/ACA RNP assembly, notably the formation of early protein-only complexes containing at least the core proteins dyskerin, NOP10, and NHP2, and the assembly factors SHQ1 and NAF1. We also identified new proteins associated with GAR1, NHP2, SHQ1 and NAF1, which can be important for box H/ACA assembly or function. Moreover, even though GAR1 is regulated by methylations, the nature, localization, and functions of these methylations are not well known. Our MS analysis of purified GAR1 revealed new sites of arginine methylations. Additionally, we showed that unmethylated GAR1 is correctly incorporated in H/ACA RNPs, even though with less efficiency than methylated ones.


Subject(s)
Proteomics , Ribonucleoproteins , Ribonucleoproteins/genetics , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , RNA-Binding Proteins , RNA/genetics
3.
J Mol Biol ; 434(19): 167760, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35901867

ABSTRACT

DPCD is a protein that may play a role in cilia formation and whose absence leads to primary ciliary dyskinesia (PCD), a rare disease caused by impairment of ciliated cells. Except for high-throughput studies that identified DPCD as a possible RUVBL1 (R1) and RUVBL2 (R2) partner, no in-depth cellular, biochemical, and structural investigation involving DPCD have been reported so far. R1 and R2 proteins are ubiquitous highly conserved AAA + family ATPases that assemble and mature a plethora of macromolecular complexes and are pivotal in numerous cellular processes, especially by guaranteeing a co-chaperoning function within R2TP or R2TP-like machineries. In the present study, we identified DPCD as a new R1R2 partner in vivo. We show that DPCD interacts directly with R1 and R2 in vitro and in cells. We characterized the physico-chemical properties of DPCD in solution and built a 3D model of DPCD. In addition, we used a variety of orthogonal biophysical techniques including small-angle X-ray scattering, structural mass spectrometry and electron microscopy to assess the molecular determinants of DPCD interaction with R1R2. Interestingly, DPCD disrupts the dodecameric state of R1R2 complex upon binding and this interaction occurs mainly via the DII domains of R1R2.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Carrier Proteins , DNA Helicases , Multiprotein Complexes , Proteins , ATPases Associated with Diverse Cellular Activities/chemistry , Carrier Proteins/chemistry , DNA Helicases/chemistry , Humans , Multiprotein Complexes/chemistry , Proteins/chemistry
4.
Nucleic Acids Res ; 50(4): 2172-2189, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35150569

ABSTRACT

MicroRNAs silence mRNAs by guiding the RISC complex. RISC assembly occurs following cleavage of pre-miRNAs by Dicer, assisted by TRBP or PACT, and the transfer of miRNAs to AGO proteins. The R2TP complex is an HSP90 co-chaperone involved in the assembly of ribonucleoprotein particles. Here, we show that the R2TP component RPAP3 binds TRBP but not PACT. The RPAP3-TPR1 domain interacts with the TRBP-dsRBD3, and the 1.5 Å resolution crystal structure of this complex identifies key residues involved in the interaction. Remarkably, binding of TRBP to RPAP3 or Dicer is mutually exclusive. Additionally, we found that AGO(1/2), TRBP and Dicer are all sensitive to HSP90 inhibition, and that TRBP sensitivity is increased in the absence of RPAP3. Finally, RPAP3 seems to impede miRNA activity, raising the possibility that the R2TP chaperone might sequester TRBP to regulate the miRNA pathway.


Subject(s)
MicroRNAs , RNA-Induced Silencing Complex , Gene Silencing , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Receptor Coactivators/chemistry , Ribonuclease III/genetics , Ribonuclease III/metabolism
5.
Nat Commun ; 12(1): 4810, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376666

ABSTRACT

The R2TP chaperone cooperates with HSP90 to integrate newly synthesized proteins into multi-subunit complexes, yet its role in tissue homeostasis is unknown. Here, we generated conditional, inducible knock-out mice for Rpap3 to inactivate this core component of R2TP in the intestinal epithelium. In adult mice, Rpap3 invalidation caused destruction of the small intestinal epithelium and death within 10 days. Levels of R2TP substrates decreased, with strong effects on mTOR, ATM and ATR. Proliferative stem cells and progenitors deficient for Rpap3 failed to import RNA polymerase II into the nucleus and they induced p53, cell cycle arrest and apoptosis. Post-mitotic, differentiated cells did not display these alterations, suggesting that R2TP clients are preferentially built in actively proliferating cells. In addition, high RPAP3 levels in colorectal tumors from patients correlate with bad prognosis. Here, we show that, in the intestine, the R2TP chaperone plays essential roles in normal and tumoral proliferation.


Subject(s)
Cell Proliferation , Epithelial Cells/metabolism , HSP90 Heat-Shock Proteins/metabolism , Intestinal Mucosa/metabolism , Molecular Chaperones/metabolism , Animals , Cells, Cultured , Epithelial Cells/cytology , Humans , Intestinal Mucosa/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Protein Binding , Stem Cells/cytology , Stem Cells/metabolism
6.
Nat Commun ; 12(1): 3646, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131137

ABSTRACT

U5 snRNP is a complex particle essential for RNA splicing. U5 snRNPs undergo intricate biogenesis that ensures that only a fully mature particle assembles into a splicing competent U4/U6•U5 tri-snRNP and enters the splicing reaction. During splicing, U5 snRNP is substantially rearranged and leaves as a U5/PRPF19 post-splicing particle, which requires re-generation before the next round of splicing. Here, we show that a previously uncharacterized protein TSSC4 is a component of U5 snRNP that promotes tri-snRNP formation. We provide evidence that TSSC4 associates with U5 snRNP chaperones, U5 snRNP and the U5/PRPF19 particle. Specifically, TSSC4 interacts with U5-specific proteins PRPF8, EFTUD2 and SNRNP200. We also identified TSSC4 domains critical for the interaction with U5 snRNP and the PRPF19 complex, as well as for TSSC4 function in tri-snRNP assembly. TSSC4 emerges as a specific chaperone that acts in U5 snRNP de novo biogenesis as well as post-splicing recycling.


Subject(s)
Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/metabolism , Down-Regulation , HeLa Cells , Humans , Nuclear Proteins/metabolism , Peptide Elongation Factors , Protein Domains , Protein Interaction Domains and Motifs , RNA Splicing , RNA Splicing Factors/metabolism , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins , Ribonucleoproteins, Small Nuclear/chemistry , Transcription Factors , Tumor Suppressor Proteins/genetics
7.
Nucleic Acids Res ; 49(2): 1094-1113, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33367824

ABSTRACT

The PAQosome is a large complex composed of the HSP90/R2TP chaperone and a prefoldin-like module. It promotes the biogenesis of cellular machineries but it is unclear how it discriminates closely related client proteins. Among the main PAQosome clients are C/D snoRNPs and in particular their core protein NOP58. Using NOP58 mutants and proteomic experiments, we identify different assembly intermediates and show that C12ORF45, which we rename NOPCHAP1, acts as a bridge between NOP58 and PAQosome. NOPCHAP1 makes direct physical interactions with the CC-NOP domain of NOP58 and domain II of RUVBL1/2 AAA+ ATPases. Interestingly, NOPCHAP1 interaction with RUVBL1/2 is disrupted upon ATP binding. Moreover, while it robustly binds both yeast and human NOP58, it makes little interactions with NOP56 and PRPF31, two other closely related CC-NOP proteins. Expression of NOP58, but not NOP56 or PRPF31, is decreased in NOPCHAP1 KO cells. We propose that NOPCHAP1 is a client-loading PAQosome cofactor that selects NOP58 to promote box C/D snoRNP assembly.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Carrier Proteins/metabolism , DNA Helicases/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Ribonucleoproteins, Small Nucleolar/biosynthesis , Adenosine Triphosphate/metabolism , Eye Proteins/metabolism , Gene Knockout Techniques , Genes, Reporter , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Multiprotein Complexes , Protein Domains , Protein Interaction Mapping , Proteomics/methods , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Saccharomyces cerevisiae Proteins/metabolism
8.
Structure ; 26(9): 1196-1209.e8, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30033218

ABSTRACT

RPAP3 and PIH1D1 are part of the HSP90 co-chaperone R2TP complex involved in the assembly process of many molecular machines. In this study, we performed a deep structural investigation of the HSP binding abilities of the two TPR domains of RPAP3. We combined 3D NMR, non-denaturing MS, and ITC techniques with Y2H, IP-LUMIER, FRET, and ATPase activity assays and explain the fundamental role played by the second TPR domain of RPAP3 in the specific recruitment of HSP90. We also established the 3D structure of an RPAP3:PIH1D1 sub-complex demonstrating the need for a 34-residue insertion, specific of RPAP3 isoform 1, for the tight binding of PIH1D1. We also confirm the existence of a complex lacking PIH1D1 in human cells (R2T), which shows differential binding to certain clients. These results highlight similarities and differences between the yeast and human R2TP complexes, and document the diversification of this family of co-chaperone complexes in human.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Binding Sites , Cell Line , HSP72 Heat-Shock Proteins/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Multimerization
9.
Nat Commun ; 9(1): 2093, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844425

ABSTRACT

R2TP is an HSP90 co-chaperone that assembles important macro-molecular machineries. It is composed of an RPAP3-PIH1D1 heterodimer, which binds the two essential AAA+ATPases RUVBL1/RUVBL2. Here, we resolve the structure of the conserved C-terminal domain of RPAP3, and we show that it directly binds RUVBL1/RUVBL2 hexamers. The human genome encodes two other proteins bearing RPAP3-C-terminal-like domains and three containing PIH-like domains. Systematic interaction analyses show that one RPAP3-like protein, SPAG1, binds PIH1D2 and RUVBL1/2 to form an R2TP-like complex termed R2SP. This co-chaperone is enriched in testis and among 68 of the potential clients identified, some are expressed in testis and others are ubiquitous. One substrate is liprin-α2, which organizes large signaling complexes. Remarkably, R2SP is required for liprin-α2 expression and for the assembly of liprin-α2 complexes, indicating that R2SP functions in quaternary protein folding. Effects are stronger at 32 °C, suggesting that R2SP could help compensating the lower temperate of testis.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , DNA Helicases/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Testis/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Cell Line , GTP-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Male , Membrane Proteins/metabolism , Protein Binding , Protein Folding , Protein Structure, Secondary , Signal Transduction
10.
J Cell Biol ; 216(6): 1579-1596, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28515276

ABSTRACT

Splicing is catalyzed by the spliceosome, a complex of five major small nuclear ribonucleoprotein particles (snRNPs). The pre-mRNA splicing factor PRPF8 is a crucial component of the U5 snRNP, and together with EFTUD2 and SNRNP200, it forms a central module of the spliceosome. Using quantitative proteomics, we identified assembly intermediates containing PRPF8, EFTUD2, and SNRNP200 in association with the HSP90/R2TP complex, its ZNHIT2 cofactor, and additional proteins. HSP90 and R2TP bind unassembled U5 proteins in the cytoplasm, stabilize them, and promote the formation of the U5 snRNP. We further found that PRPF8 mutants causing Retinitis pigmentosa assemble less efficiently with the U5 snRNP and bind more strongly to R2TP, with one mutant retained in the cytoplasm in an R2TP-dependent manner. We propose that the HSP90/R2TP chaperone system promotes the assembly of a key module of U5 snRNP while assuring the quality control of PRPF8. The proteomics data further reveal new interactions between R2TP and the tuberous sclerosis complex (TSC), pointing to a potential link between growth signals and the assembly of key cellular machines.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U5 Small Nuclear/metabolism , Calcium-Binding Proteins/metabolism , HeLa Cells , Humans , Multiprotein Complexes , Mutation , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Proteomics/methods , RNA Interference , RNA Precursors/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Transfection
11.
Cell Rep ; 18(11): 2635-2650, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297668

ABSTRACT

The nuclear cap-binding complex (CBC) stimulates processing reactions of capped RNAs, including their splicing, 3'-end formation, degradation, and transport. CBC effects are particular for individual RNA families, but how such selectivity is achieved remains elusive. Here, we analyze three main CBC partners known to impact different RNA species. ARS2 stimulates 3'-end formation/transcription termination of several transcript types, ZC3H18 stimulates degradation of a diverse set of RNAs, and PHAX functions in pre-small nuclear RNA/small nucleolar RNA (pre-snRNA/snoRNA) transport. Surprisingly, these proteins all bind capped RNAs without strong preferences for given transcripts, and their steady-state binding correlates poorly with their function. Despite this, PHAX and ZC3H18 compete for CBC binding and we demonstrate that this competitive binding is functionally relevant. We further show that CBC-containing complexes are short lived in vivo, and we therefore suggest that RNA fate involves the transient formation of mutually exclusive CBC complexes, which may only be consequential at particular checkpoints during RNA biogenesis.


Subject(s)
Nuclear Cap-Binding Protein Complex/metabolism , RNA/metabolism , HEK293 Cells , HeLa Cells , Humans , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
RNA Biol ; 14(6): 680-692, 2017 06 03.
Article in English | MEDLINE | ID: mdl-27715451

ABSTRACT

Box C/D and box H/ACA snoRNAs are abundant non-coding RNAs that localize in the nucleolus and mostly function as guides for nucleotide modifications. While a large pool of snoRNAs modifies rRNAs, an increasing number of snoRNAs could also potentially target mRNAs. ScaRNAs belong to a family of specific RNAs that localize in Cajal bodies and that are structurally similar to snoRNAs. Most scaRNAs are involved in snRNA modification, while telomerase RNA, which contains H/ACA motifs, functions in telomeric DNA synthesis. In this review, we describe how box C/D and H/ACA snoRNAs are processed and assembled with core proteins to form functional RNP particles. Their biogenesis involve several transport factors that first direct pre-snoRNPs to Cajal bodies, where some processing steps are believed to take place, and then to nucleoli. Assembly of core proteins involves the HSP90/R2TP chaperone-cochaperone system for both box C/D and H/ACA RNAs, but also several factors specific for each family. These assembly factors chaperone unassembled core proteins, regulate the formation and disassembly of pre-snoRNP intermediates, and control the activity of immature particles. The AAA+ ATPase RUVBL1 and RUVBL2 belong to the R2TP co-chaperones and play essential roles in snoRNP biogenesis, as well as in the formation of other macro-molecular complexes. Despite intensive research, their mechanisms of action are still incompletely understood.


Subject(s)
RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Animals , Carrier Proteins , Coiled Bodies/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , RNA Processing, Post-Transcriptional , RNA, Small Nucleolar/chemistry , Signal Transduction , Transcription, Genetic
14.
Nucleic Acids Res ; 43(18): 8973-89, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26275778

ABSTRACT

The Sm proteins are loaded on snRNAs by the SMN complex, but how snRNP-specific proteins are assembled remains poorly characterized. U4 snRNP and box C/D snoRNPs have structural similarities. They both contain the 15.5K and proteins with NOP domains (PRP31 for U4, NOP56/58 for snoRNPs). Biogenesis of box C/D snoRNPs involves NUFIP and the HSP90/R2TP chaperone system and here, we explore the function of this machinery in U4 RNP assembly. We show that yeast Prp31 interacts with several components of the NUFIP/R2TP machinery, and that these interactions are separable from each other. In human cells, PRP31 mutants that fail to stably associate with U4 snRNA still interact with components of the NUFIP/R2TP system, indicating that these interactions precede binding of PRP31 to U4 snRNA. Knock-down of NUFIP leads to mislocalization of PRP31 and decreased association with U4. Moreover, NUFIP is associated with the SMN complex through direct interactions with Gemin3 and Gemin6. Altogether, our data suggest a model in which the NUFIP/R2TP system is connected with the SMN complex and facilitates assembly of U4 snRNP-specific proteins.


Subject(s)
Eye Proteins/metabolism , Molecular Chaperones/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribosomal Proteins/metabolism , SMN Complex Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism , Cell Line , Coiled Bodies/metabolism , Cytoplasm/metabolism , Eye Proteins/chemistry , Eye Proteins/genetics , HeLa Cells , Humans , Mutagenesis, Insertional , RNA, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes/genetics
15.
J Cell Biol ; 207(4): 463-80, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25404746

ABSTRACT

In vitro, assembly of box C/D small nucleolar ribonucleoproteins (snoRNPs) involves the sequential recruitment of core proteins to snoRNAs. In vivo, however, assembly factors are required (NUFIP, BCD1, and the HSP90-R2TP complex), and it is unknown whether a similar sequential scheme applies. In this paper, we describe systematic quantitative stable isotope labeling by amino acids in cell culture proteomic experiments and the crystal structure of the core protein Snu13p/15.5K bound to a fragment of the assembly factor Rsa1p/NUFIP. This revealed several unexpected features: (a) the existence of a protein-only pre-snoRNP complex containing five assembly factors and two core proteins, 15.5K and Nop58; (b) the characterization of ZNHIT3, which is present in the protein-only complex but gets released upon binding to C/D snoRNAs; (c) the dynamics of the R2TP complex, which appears to load/unload RuvBL AAA(+) adenosine triphosphatase from pre-snoRNPs; and (d) a potential mechanism for preventing premature activation of snoRNP catalytic activity. These data provide a framework for understanding the assembly of box C/D snoRNPs.


Subject(s)
Nuclear Proteins/chemistry , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nucleolar/metabolism , ATPases Associated with Diverse Cellular Activities , Amino Acid Sequence , Binding Sites , Carrier Proteins/metabolism , Cell Line, Tumor , Crystallography, X-Ray , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding , Proteomics/methods , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribonucleases/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Saccharomyces cerevisiae/genetics , Sequence Alignment , Transcription Factors
16.
Nucleic Acids Res ; 42(13): 8663-77, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25013170

ABSTRACT

Mammalian mRNAs are generated by complex and coordinated biogenesis pathways and acquire 5'-end m(7)G caps that play fundamental roles in processing and translation. Here we show that several selenoprotein mRNAs are not recognized efficiently by translation initiation factor eIF4E because they bear a hypermethylated cap. This cap modification is acquired via a 5'-end maturation pathway similar to that of the small nucle(ol)ar RNAs (sn- and snoRNAs). Our findings also establish that the trimethylguanosine synthase 1 (Tgs1) interacts with selenoprotein mRNAs for cap hypermethylation and that assembly chaperones and core proteins devoted to sn- and snoRNP maturation contribute to recruiting Tgs1 to selenoprotein mRNPs. We further demonstrate that the hypermethylated-capped selenoprotein mRNAs localize to the cytoplasm, are associated with polysomes and thus translated. Moreover, we found that the activity of Tgs1, but not of eIF4E, is required for the synthesis of the GPx1 selenoprotein in vivo.


Subject(s)
RNA Caps/metabolism , RNA, Messenger/metabolism , Selenoproteins/genetics , Cell Line , Eukaryotic Initiation Factor-4E/metabolism , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/genetics , Humans , Methylation , Methyltransferases/metabolism , Nuclear Proteins/metabolism , Polyribosomes/chemistry , Protein Biosynthesis , RNA, Messenger/analysis , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , SMN Complex Proteins/metabolism , Selenoproteins/biosynthesis , Selenoproteins/metabolism , Glutathione Peroxidase GPX1
17.
Nat Struct Mol Biol ; 20(12): 1358-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24270878

ABSTRACT

The nuclear cap-binding complex (CBC) stimulates multiple steps in several RNA maturation pathways, but how it functions in humans is incompletely understood. For small, capped RNAs such as pre-snRNAs, the CBC recruits PHAX. Here, we identify the CBCAP complex, composed of CBC, ARS2 and PHAX, and show that both CBCAP and CBC-ARS2 complexes can be reconstituted from recombinant proteins. ARS2 stimulates PHAX binding to the CBC and snRNA 3'-end processing, thereby coupling maturation with export. In vivo, CBC and ARS2 bind similar capped noncoding and coding RNAs and stimulate their 3'-end processing. The strongest effects are for cap-proximal polyadenylation sites, and this favors premature transcription termination. ARS2 functions partly through the mRNA 3'-end cleavage factor CLP1, which binds RNA Polymerase II through PCF11. ARS2 is thus a major CBC effector that stimulates functional and cryptic 3'-end processing sites.


Subject(s)
Models, Genetic , Nuclear Cap-Binding Protein Complex/physiology , Nuclear Proteins/physiology , Nucleocytoplasmic Transport Proteins/physiology , Phosphoproteins/physiology , RNA 3' End Processing , HeLa Cells , Humans , Nuclear Cap-Binding Protein Complex/chemistry , Nuclear Cap-Binding Protein Complex/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Poly A/chemistry , Poly A/metabolism
18.
Nat Struct Mol Biol ; 20(12): 1367-76, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24270879

ABSTRACT

Nuclear processing and quality control of eukaryotic RNA is mediated by the RNA exosome, which is regulated by accessory factors. However, the mechanism of exosome recruitment to its ribonucleoprotein (RNP) targets remains poorly understood. Here we report a physical link between the human exosome and the cap-binding complex (CBC). The CBC associates with the ARS2 protein to form CBC-ARS2 (CBCA) and then further connects, together with the ZC3H18 protein, to the nuclear exosome targeting (NEXT) complex, thus forming CBC-NEXT (CBCN). RNA immunoprecipitation using CBCN factors as well as the analysis of combinatorial depletion of CBCN and exosome components underscore the functional relevance of CBC-exosome bridging at the level of target RNA. Specifically, CBCA suppresses read-through products of several RNA families by promoting their transcriptional termination. We suggest that the RNP 5' cap links transcription termination to exosomal RNA degradation through CBCN.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/physiology , Nuclear Cap-Binding Protein Complex/physiology , Exosome Multienzyme Ribonuclease Complex/chemistry , Exosome Multienzyme Ribonuclease Complex/metabolism , Humans , Immunoprecipitation , Nuclear Cap-Binding Protein Complex/chemistry , Nuclear Cap-Binding Protein Complex/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , RNA Stability , Transcription Termination, Genetic
19.
Nucleus ; 3(2): 132-7, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22555597

ABSTRACT

Here, we review the sn- and sno-RNA transport pathways in S. cerevisiae and humans, aiming at understanding how they evolved and how common factors can have distinct functions depending on the RNA they bind. We give a particular emphasis on Tgs1, the cap hypermethylase that is conserved from yeast to humans and appears to play a central role in both sn- and sno-RNA biogenesis. In yeast, Tgs1 hypermethylates sn- and sno-RNAs in the nucleolus. In humans, Tgs1 occurs in two forms: a long isoform (Tgs1 LF), which locates in the cytoplasm and Cajal bodies, which is predominantly associated with snRNAs and a short isoform (Tgs1 SF), which is nuclear and mainly associates with snoRNAs. We show that Tgs1 LF is exported by CRM1 and that interaction with CRM1 competes for binding with the C-terminal domain of the core protein Nop58, which contains the Nucleolar localization signal of Box C/D snoRNPs (NoLS). Our data suggest a model where CRM1 removes Tgs1 LF from snoRNPs, thereby promoting nucleolar targeting via activation of their NoLS. In this review, we argue that CRM1, while first described as an export receptor, can also control the composition of nucleoplasmic complexes. Thus, it could coordinate the fate of these complexes with the general nucleo-cytoplasmic trafficking.


Subject(s)
Cell Nucleolus/metabolism , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Humans , Karyopherins/chemistry , Methyltransferases/chemistry , Methyltransferases/metabolism , Molecular Sequence Data , Receptors, Cytoplasmic and Nuclear/chemistry , Ribonucleoproteins, Small Nucleolar/biosynthesis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Exportin 1 Protein
20.
EMBO J ; 30(11): 2205-18, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21522132

ABSTRACT

Transport of C/D snoRNPs to nucleoli involves nuclear export factors. In particular, CRM1 binds nascent snoRNPs, but its precise role remains unknown. We show here that both CRM1 and nucleocytoplasmic trafficking are required to transport snoRNPs to nucleoli, but the snoRNPs do not transit through the cytoplasm. Instead, CRM1 controls the composition of nucleoplasmic pre-snoRNP complexes. We observed that Tgs1 long form (Tgs1 LF), the long isoform of the cap hypermethylase, contains a leucine-rich nuclear export signal, shuttles in a CRM1-dependent manner, and binds to the nucleolar localization signal (NoLS) of the core snoRNP protein Nop58. In vitro data indicate that CRM1 binds Tgs1 LF and promotes its dissociation from Nop58 NoLS, and immunoprecipitation experiments from cells indicate that the association of Tgs1 LF with snoRNPs increases upon CRM1 inhibition. Thus, CRM1 appears to promote nucleolar transport of snoRNPs by removing Tgs1 LF from the Nop58 NoLS. Microarray/IP data show that this occurs on most snoRNPs, from both C/D and H/ACA families, and on the telomerase RNA. Hence, CRM1 provides a general molecular link between nuclear events and nucleocytoplasmic trafficking.


Subject(s)
Cell Nucleus/metabolism , Karyopherins/metabolism , RNA, Small Nucleolar/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Active Transport, Cell Nucleus , Cell Line , Humans , Methyltransferases/metabolism , Nuclear Proteins/metabolism , Protein Binding , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...