Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598269

ABSTRACT

Heat stress can cause cell death by triggering the aggregation of essential proteins. In bacteria, aggregated proteins are rescued by the canonical Hsp70/AAA+ (ClpB) bi-chaperone disaggregase. Man-made, severe stress conditions applied during, e.g., food processing represent a novel threat for bacteria by exceeding the capacity of the Hsp70/ClpB system. Here, we report on the potent autonomous AAA+ disaggregase ClpL from Listeria monocytogenes that provides enhanced heat resistance to the food-borne pathogen enabling persistence in adverse environments. ClpL shows increased thermal stability and enhanced disaggregation power compared to Hsp70/ClpB, enabling it to withstand severe heat stress and to solubilize tight aggregates. ClpL binds to protein aggregates via aromatic residues present in its N-terminal domain (NTD) that adopts a partially folded and dynamic conformation. Target specificity is achieved by simultaneous interactions of multiple NTDs with the aggregate surface. ClpL shows remarkable structural plasticity by forming diverse higher assembly states through interacting ClpL rings. NTDs become largely sequestered upon ClpL ring interactions. Stabilizing ring assemblies by engineered disulfide bonds strongly reduces disaggregation activity, suggesting that they represent storage states.


Subject(s)
Listeria monocytogenes , Neural Tube Defects , Humans , Animals , Cell Death , Estrus , Food , HSP70 Heat-Shock Proteins
2.
EMBO Rep ; 24(12): e57984, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37921038

ABSTRACT

The rixosome defined in Schizosaccharomyces pombe and humans performs diverse roles in pre-ribosomal RNA processing and gene silencing. Here, we isolate and describe the conserved rixosome from Chaetomium thermophilum, which consists of two sub-modules, the sphere-like Rix1-Ipi3-Ipi1 and the butterfly-like Las1-Grc3 complex, connected by a flexible linker. The Rix1 complex of the rixosome utilizes Sda1 as landing platform on nucleoplasmic pre-60S particles to wedge between the 5S rRNA tip and L1-stalk, thereby facilitating the 180° rotation of the immature 5S RNP towards its mature conformation. Upon rixosome positioning, the other sub-module with Las1 endonuclease and Grc3 polynucleotide-kinase can reach a strategic position at the pre-60S foot to cleave and 5' phosphorylate the nearby ITS2 pre-rRNA. Finally, inward movement of the L1 stalk permits the flexible Nop53 N-terminus with its AIM motif to become positioned at the base of the L1-stalk to facilitate Mtr4 helicase-exosome participation for completing ITS2 removal. Thus, the rixosome structure elucidates the coordination of two central ribosome biogenesis events, but its role in gene silencing may adapt similar strategies.


Subject(s)
Saccharomyces cerevisiae Proteins , Schizosaccharomyces , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Nuclear Proteins/metabolism , Rotation , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , RNA Processing, Post-Transcriptional , Ribosomal Proteins/genetics
3.
Nat Struct Mol Biol ; 30(8): 1119-1131, 2023 08.
Article in English | MEDLINE | ID: mdl-37291423

ABSTRACT

The 5S ribonucleoprotein (RNP) is assembled from its three components (5S rRNA, Rpl5/uL18 and Rpl11/uL5) before being incorporated into the pre-60S subunit. However, when ribosome synthesis is disturbed, a free 5S RNP can enter the MDM2-p53 pathway to regulate cell cycle and apoptotic signaling. Here we reconstitute and determine the cryo-electron microscopy structure of the conserved hexameric 5S RNP with fungal or human factors. This reveals how the nascent 5S rRNA associates with the initial nuclear import complex Syo1-uL18-uL5 and, upon further recruitment of the nucleolar factors Rpf2 and Rrs1, develops into the 5S RNP precursor that can assemble into the pre-ribosome. In addition, we elucidate the structure of another 5S RNP intermediate, carrying the human ubiquitin ligase Mdm2, which unravels how this enzyme can be sequestered from its target substrate p53. Our data provide molecular insight into how the 5S RNP can mediate between ribosome biogenesis and cell proliferation.


Subject(s)
RNA, Ribosomal, 5S , Tumor Suppressor Protein p53 , Humans , RNA, Ribosomal, 5S/chemistry , Tumor Suppressor Protein p53/metabolism , Cryoelectron Microscopy , Ribosomal Proteins/metabolism , Ribonucleoproteins/metabolism , Ribosomes/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism
4.
Cell Rep ; 39(1): 110640, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385737

ABSTRACT

Synthesis of ribosomes begins in the nucleolus with formation of the 90S pre-ribosome, during which the pre-40S and pre-60S pathways diverge by pre-rRNA cleavage. However, it remains unclear how, after this uncoupling, the earliest pre-60S subunit continues to develop. Here, we reveal a large-subunit intermediate at the beginning of its construction when still linked to the 90S, the precursor to the 40S subunit. This primordial pre-60S is characterized by the SPOUT domain methyltransferase Upa1-Upa2, large α-solenoid scaffolds, Mak5, one of several RNA helicases, and two small nucleolar RNA (snoRNAs), C/D box snR190 and H/ACA box snR37. The emerging pre-60S does not efficiently disconnect from the 90S pre-ribosome in a dominant mak5 helicase mutant, allowing a 70-nm 90S-pre-60S bipartite particle to be visualized by electron microscopy. Our study provides insight into the assembly pathway when the still-connected nascent 40S and 60S subunits are beginning to separate.


Subject(s)
Ribosome Subunits, Large, Eukaryotic , Ribosomes , Saccharomyces cerevisiae Proteins , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
5.
Commun Biol ; 4(1): 600, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34017052

ABSTRACT

The eukaryotic signal recognition particle (SRP) contains an Alu domain, which docks into the factor binding site of translating ribosomes and confers translation retardation. The canonical Alu domain consists of the SRP9/14 protein heterodimer and a tRNA-like folded Alu RNA that adopts a strictly 'closed' conformation involving a loop-loop pseudoknot. Here, we study the structure of the Alu domain from Plasmodium falciparum (PfAlu), a divergent apicomplexan protozoan that causes human malaria. Using NMR, SAXS and cryo-EM analyses, we show that, in contrast to its prokaryotic and eukaryotic counterparts, the PfAlu domain adopts an 'open' Y-shaped conformation. We show that cytoplasmic P. falciparum ribosomes are non-discriminative and recognize both the open PfAlu and closed human Alu domains with nanomolar affinity. In contrast, human ribosomes do not provide high affinity binding sites for either of the Alu domains. Our analyses extend the structural database of Alu domains to the protozoan species and reveal species-specific differences in the recognition of SRP Alu domains by ribosomes.


Subject(s)
Alu Elements , Plasmodium falciparum/metabolism , Ribosomes/metabolism , Signal Recognition Particle/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Nucleic Acid Conformation , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Ribosomes/genetics , Scattering, Small Angle
6.
Mol Cell ; 81(2): 293-303.e4, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33326748

ABSTRACT

Ribosome assembly is catalyzed by numerous trans-acting factors and coupled with irreversible pre-rRNA processing, driving the pathway toward mature ribosomal subunits. One decisive step early in this progression is removal of the 5' external transcribed spacer (5'-ETS), an RNA extension at the 18S rRNA that is integrated into the huge 90S pre-ribosome structure. Upon endo-nucleolytic cleavage at an internal site, A1, the 5'-ETS is separated from the 18S rRNA and degraded. Here we present biochemical and cryo-electron microscopy analyses that depict the RNA exosome, a major 3'-5' exoribonuclease complex, in a super-complex with the 90S pre-ribosome. The exosome is docked to the 90S through its co-factor Mtr4 helicase, a processive RNA duplex-dismantling helicase, which strategically positions the exosome at the base of 5'-ETS helices H9-H9', which are dislodged in our 90S-exosome structures. These findings suggest a direct role of the exosome in structural remodeling of the 90S pre-ribosome to drive eukaryotic ribosome synthesis.


Subject(s)
DEAD-box RNA Helicases/chemistry , Endoribonucleases/chemistry , Exonucleases/chemistry , Exosome Multienzyme Ribonuclease Complex/ultrastructure , RNA, Ribosomal, 18S/chemistry , Ribosomes/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Binding Sites , Cryoelectron Microscopy , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Exonucleases/genetics , Exonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Models, Molecular , Protein Binding , Protein Biosynthesis , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA Stability , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
Mol Cell ; 80(1): 72-86.e7, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32910895

ABSTRACT

Membrane protein biogenesis faces the challenge of chaperoning hydrophobic transmembrane helices for faithful membrane insertion. The guided entry of tail-anchored proteins (GET) pathway targets and inserts tail-anchored (TA) proteins into the endoplasmic reticulum (ER) membrane with an insertase (yeast Get1/Get2 or mammalian WRB/CAML) that captures the TA from a cytoplasmic chaperone (Get3 or TRC40, respectively). Here, we present cryo-electron microscopy reconstructions, native mass spectrometry, and structure-based mutagenesis of human WRB/CAML/TRC40 and yeast Get1/Get2/Get3 complexes. Get3 binding to the membrane insertase supports heterotetramer formation, and phosphatidylinositol binding at the heterotetramer interface stabilizes the insertase for efficient TA insertion in vivo. We identify a Get2/CAML cytoplasmic helix that forms a "gating" interaction with Get3/TRC40 important for TA insertion. Structural homology with YidC and the ER membrane protein complex (EMC) implicates an evolutionarily conserved insertion mechanism for divergent substrates utilizing a hydrophilic groove. Thus, we provide a detailed structural and mechanistic framework to understand TA membrane insertion.


Subject(s)
Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Multiprotein Complexes/metabolism , Cell Line , Conserved Sequence , Evolution, Molecular , Humans , Membrane Proteins/metabolism , Models, Molecular , Phosphatidylinositols/metabolism , Protein Binding , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
8.
Nat Commun ; 11(1): 776, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034140

ABSTRACT

Human Ebp1 is a member of the proliferation-associated 2G4 (PA2G4) family and plays an important role in cancer regulation. Ebp1 shares the methionine aminopeptidase (MetAP) fold and binds to mature 80S ribosomes for translational control. Here, we present a cryo-EM single particle analysis reconstruction of Ebp1 bound to non-translating human 80S ribosomes at a resolution range from 3.3 to ~8 Å. Ebp1 blocks the tunnel exit with major interactions to the general uL23/uL29 docking site for nascent chain-associated factors complemented by eukaryote-specific eL19 and rRNA helix H59. H59 is defined as dynamic adaptor undergoing significant remodeling upon Ebp1 binding. Ebp1 recruits rRNA expansion segment ES27L to the tunnel exit via specific interactions with rRNA consensus sequences. The Ebp1-ribosome complex serves as a template for MetAP binding and provides insights into the structural principles for spatial coordination of co-translational events and molecular triage at the ribosomal tunnel exit.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribosome Subunits/metabolism , Cryoelectron Microscopy , Humans , Models, Molecular , Molecular Docking Simulation , Protein Conformation , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Ribosome Subunits/chemistry
9.
Nature ; 578(7795): 467-471, 2020 02.
Article in English | MEDLINE | ID: mdl-31856152

ABSTRACT

Microtubules are dynamic polymers of α- and ß-tubulin and have crucial roles in cell signalling, cell migration, intracellular transport and chromosome segregation1. They assemble de novo from αß-tubulin dimers in an essential process termed microtubule nucleation. Complexes that contain the protein γ-tubulin serve as structural templates for the microtubule nucleation reaction2. In vertebrates, microtubules are nucleated by the 2.2-megadalton γ-tubulin ring complex (γ-TuRC), which comprises γ-tubulin, five related γ-tubulin complex proteins (GCP2-GCP6) and additional factors3. GCP6 is unique among the GCP proteins because it carries an extended insertion domain of unknown function. Our understanding of microtubule formation in cells and tissues is limited by a lack of high-resolution structural information on the γ-TuRC. Here we present the cryo-electron microscopy structure of γ-TuRC from Xenopus laevis at 4.8 Å global resolution, and identify a 14-spoked arrangement of GCP proteins and γ-tubulins in a partially flexible open left-handed spiral with a uniform sequence of GCP variants. By forming specific interactions with other GCP proteins, the GCP6-specific insertion domain acts as a scaffold for the assembly of the γ-TuRC. Unexpectedly, we identify actin as a bona fide structural component of the γ-TuRC with functional relevance in microtubule nucleation. The spiral geometry of γ-TuRC is suboptimal for microtubule nucleation and a controlled conformational rearrangement of the γ-TuRC is required for its activation. Collectively, our cryo-electron microscopy reconstructions provide detailed insights into the molecular organization, assembly and activation mechanism of vertebrate γ-TuRC, and will serve as a framework for the mechanistic understanding of fundamental biological processes associated with microtubule nucleation, such as meiotic and mitotic spindle formation and centriole biogenesis4.


Subject(s)
Cryoelectron Microscopy , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/ultrastructure , Microtubules/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Xenopus , Actins/chemistry , Actins/metabolism , Actins/ultrastructure , Animals , Microtubule-Associated Proteins/metabolism , Microtubules/chemistry , Models, Molecular , Tubulin/chemistry , Tubulin/metabolism , Tubulin/ultrastructure
10.
Nat Commun ; 8(1): 1787, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29176610

ABSTRACT

Removal of internal transcribed spacer 2 (ITS2) from pre-ribosomal RNA is essential to make functional ribosomes. This complicated processing reaction begins with a single endonucleolytic cleavage followed by exonucleolytic trimming at both new cleavage sites to generate mature 5.8S and 25S rRNA. We reconstituted the 7S→5.8S processing branch within ITS2 using purified exosome and its nuclear cofactors. We find that both Rrp44's ribonuclease activities are required for initial RNA shortening followed by hand over to the exonuclease Rrp6. During the in vitro reaction, ITS2-associated factors dissociate and the underlying 'foot' structure of the pre-60S particle is dismantled. 7S pre-rRNA processing is independent of 5S RNP rotation, but 26S→25S trimming is a precondition for subsequent 7S→5.8S processing. To complete the in vitro assay, we reconstituted the entire cycle of ITS2 removal with a total of 18 purified factors, catalysed by the integrated activities of the two participating RNA-processing machines, the Las1 complex and nuclear exosome.


Subject(s)
RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/physiology , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/physiology , Cell Nucleus/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Nuclear Proteins/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal, 5.8S/metabolism , Saccharomyces cerevisiae Proteins/metabolism
11.
Nat Struct Mol Biol ; 24(12): 1107-1115, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29083413

ABSTRACT

Assembly of fully functional ribosomes is a prerequisite for failsafe translation. This explains why maturing preribosomal subunits have to pass through an array of quality-control checkpoints, including nuclear export, to ensure that only properly assembled ribosomes engage in translation. Despite these safeguards, we found that nuclear pre-60S particles unable to remove a transient structure composed of ITS2 pre-rRNA and associated assembly factors, termed the 'foot', escape to the cytoplasm, where they can join with mature 40S subunits to catalyze protein synthesis. However, cells harboring these abnormal ribosomes show translation defects indicated by the formation of 80S ribosomes poised with pre-60S subunits carrying tRNAs in trapped hybrid states. To overcome this translational stress, the cytoplasmic surveillance machineries RQC and Ski-exosome target these malfunctioning ribosomes. Thus, pre-60S subunits that escape nuclear quality control can enter translation, but are caught by cytoplasmic surveillance mechanisms.


Subject(s)
Cell Nucleus/metabolism , DNA, Ribosomal Spacer/genetics , Protein Biosynthesis/genetics , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Protein Biosynthesis/physiology , RNA Precursors/genetics , Saccharomyces cerevisiae/metabolism
12.
Protein Sci ; 26(2): 327-342, 2017 02.
Article in English | MEDLINE | ID: mdl-27863450

ABSTRACT

Ribosome biogenesis in eukaryotic cells is a highly dynamic and complex process innately linked to cell proliferation. The assembly of ribosomes is driven by a myriad of biogenesis factors that shape pre-ribosomal particles by processing and folding the ribosomal RNA and incorporating ribosomal proteins. Biochemical approaches allowed the isolation and characterization of pre-ribosomal particles from Saccharomyces cerevisiae, which lead to a spatiotemporal map of biogenesis intermediates along the path from the nucleolus to the cytoplasm. Here, we cloned almost the entire set (∼180) of ribosome biogenesis factors from the thermophilic fungus Chaetomium thermophilum in order to perform an in-depth analysis of their protein-protein interaction network as well as exploring the suitability of these thermostable proteins for structural studies. First, we performed a systematic screen, testing about 80 factors for crystallization and structure determination. Next, we performed a yeast 2-hybrid analysis and tested about 32,000 binary combinations, which identified more than 1000 protein-protein contacts between the thermophilic ribosome assembly factors. To exemplary verify several of these interactions, we performed biochemical reconstitution with the focus on the interaction network between 90S pre-ribosome factors forming the ctUTP-A and ctUTP-B modules, and the Brix-domain containing assembly factors of the pre-60S subunit. Our work provides a rich resource for biochemical reconstitution and structural analyses of the conserved ribosome assembly machinery from a eukaryotic thermophile.


Subject(s)
Chaetomium/chemistry , Fungal Proteins/chemistry , Ribosomal Proteins/chemistry , Ribosomes/chemistry , Chaetomium/metabolism , Fungal Proteins/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism
13.
J Cell Biol ; 215(6): 823-840, 2016 Dec 19.
Article in English | MEDLINE | ID: mdl-27920216

ABSTRACT

MOZART1/Mzt1 is required for the localization of γ-tubulin complexes to microtubule (MT)-organizing centers from yeast to human cells. Nevertheless, the molecular function of MOZART1/Mzt1 is largely unknown. Taking advantage of the minimal MT nucleation system of Candida albicans, we reconstituted the interactions of Mzt1, γ-tubulin small complex (γ-TuSC), and γ-tubulin complex receptors (γ-TuCRs) Spc72 and Spc110 in vitro. With affinity measurements, domain deletion, and swapping, we show that Spc110 and Mzt1 bind to distinct regions of the γ-TuSC. In contrast, both Mzt1 and γ-TuSC interact with the conserved CM1 motif of Spc110/Spc72. Spc110/Spc72 and Mzt1 constitute "oligomerization chaperones," cooperatively promoting and directing γ-TuSC oligomerization into MT nucleation-competent rings. Consistent with the functions of Mzt1, human MOZART1 directly interacts with the CM1-containing region of the γ-TuCR CEP215. MOZART1 depletion in human cells destabilizes the large γ-tubulin ring complex and abolishes CEP215CM1-induced ectopic MT nucleation. Together, we reveal conserved functions of MOZART1/Mzt1 through interactions with γ-tubulin complex subunits and γ-TuCRs.


Subject(s)
Candida albicans/metabolism , Fungal Proteins/metabolism , Microtubules/metabolism , Tubulin/metabolism , Fungal Proteins/chemistry , Humans , Microtubule-Associated Proteins/metabolism , Models, Biological , Protein Binding , Protein Domains , Protein Multimerization , Protein Stability , Saccharomyces cerevisiae/metabolism , Spindle Apparatus/metabolism , Spindle Pole Bodies/metabolism
14.
Cell ; 166(2): 380-393, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27419870

ABSTRACT

The 90S pre-ribosome is an early biogenesis intermediate formed during co-transcriptional ribosome formation, composed of ∼70 assembly factors and several small nucleolar RNAs (snoRNAs) that associate with nascent pre-rRNA. We report the cryo-EM structure of the Chaetomium thermophilum 90S pre-ribosome, revealing how a network of biogenesis factors including 19 ß-propellers and large α-solenoid proteins engulfs the pre-rRNA. Within the 90S pre-ribosome, we identify the UTP-A, UTP-B, Mpp10-Imp3-Imp4, Bms1-Rcl1, and U3 snoRNP modules, which are organized around 5'-ETS and partially folded 18S rRNA. The U3 snoRNP is strategically positioned at the center of the 90S particle to perform its multiple tasks during pre-rRNA folding and processing. The architecture of the elusive 90S pre-ribosome gives unprecedented structural insight into the early steps of pre-rRNA maturation. Nascent rRNA that is co-transcriptionally folded and given a particular shape by encapsulation within a dedicated mold-like structure is reminiscent of how polypeptides use chaperone chambers for their protein folding.


Subject(s)
Chaetomium/chemistry , Organelle Biogenesis , Ribosomes/chemistry , Saccharomyces cerevisiae/chemistry , Chaetomium/classification , Cryoelectron Microscopy , Models, Molecular , RNA, Ribosomal, 18S/chemistry , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosomes/ultrastructure
15.
Nat Struct Mol Biol ; 23(1): 37-44, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26619264

ABSTRACT

Ribosome synthesis is catalyzed by ∼200 assembly factors, which facilitate efficient production of mature ribosomes. Here, we determined the cryo-EM structure of a Saccharomyces cerevisiae nucleoplasmic pre-60S particle containing the dynein-related 550-kDa Rea1 AAA(+) ATPase and the Rix1 subcomplex. This particle differs from its preceding state, the early Arx1 particle, by two massive structural rearrangements: an ∼180° rotation of the 5S ribonucleoprotein complex and the central protuberance (CP) rRNA helices, and the removal of the 'foot' structure from the 3' end of the 5.8S rRNA. Progression from the Arx1 to the Rix1 particle was blocked by mutational perturbation of the Rix1-Rea1 interaction but not by a dominant-lethal Rea1 AAA(+) ATPase-ring mutant. After remodeling, the Rix1 subcomplex and Rea1 become suitably positioned to sense correct structural maturation of the CP, which allows unidirectional progression toward mature ribosomes.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Organelle Biogenesis , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosome Subunits, Large, Eukaryotic/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Cryoelectron Microscopy , Models, Molecular , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism
16.
Mol Cell ; 60(5): 808-815, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638174

ABSTRACT

The rapidly evolving internal transcribed spacer 2 (ITS2) in the pre-ribosomal RNA is one of the most commonly applied phylogenetic markers at species and genus level. Yet, during ribosome biogenesis ITS2 is removed in all eukaryotes by a common, but still unknown, mechanism. Here we describe the existence of an RNA processome, assembled from four conserved subunits, Las1-Grc3-Rat1-Rai1, that carries all the necessary RNA processing enzymes to mediate coordinated ITS2 rRNA removal. Las1 is the long-sought-after endonuclease cleaving 27SB pre-rRNA at site C2 to yield a 5'-OH end at the 26S pre-rRNA and 2',3' cyclic phosphate at the 3' end of 7S pre-rRNA. Subsequently, polynucleotide kinase Grc3 catalyzes ATP-dependent 5'-OH phosphorylation of 26S pre-rRNA, which in turn enables Rat1-Rai1 exonuclease to generate 25S' pre-rRNA. ITS2 processing is reminiscent of tRNA splicing, but instead of subsequent tRNA ligation, the Las1 complex carries along an exonuclease tool to degrade the ITS2 rRNA.


Subject(s)
DNA, Ribosomal Spacer/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Exoribonucleases/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , RNA, Fungal/metabolism , RNA, Ribosomal/metabolism , RNA-Binding Proteins , Saccharomyces cerevisiae/enzymology
17.
Structure ; 23(7): 1246-57, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26051714

ABSTRACT

The TREX-2 complex integrates mRNA nuclear export into the gene expression pathway and is based on a Sac3 scaffold to which Thp1, Sem1, Sus1, and Cdc31 bind. TREX-2 also binds the mRNA nuclear export factor, Mex67:Mtr2, through the Sac3 N-terminal region (Sac3N). Here, we characterize Chaetomium thermophilum TREX-2, show that the in vitro reconstituted complex has an annular structure, and define the structural basis for interactions between Sac3, Sus1, Cdc31, and Mex67:Mtr2. Crystal structures show that the binding of C. thermophilum Sac3N to the Mex67 NTF2-like domain (Mex67(NTF2L)) is mediated primarily through phenylalanine residues present in a series of repeating sequence motifs that resemble those seen in many nucleoporins, and Mlp1 also binds Mex67:Mtr2 using a similar motif. Deletion of Sac3N generated growth and mRNA export defects in Saccharomyces cerevisiae, and we propose TREX-2 and Mlp1 function to facilitate export by concentrating mature messenger ribonucleoparticles at the nuclear pore entrance.


Subject(s)
Chaetomium , Fungal Proteins/chemistry , Nucleocytoplasmic Transport Proteins/chemistry , Active Transport, Cell Nucleus , Amino Acid Sequence , Crystallography, X-Ray , Fungal Proteins/ultrastructure , Models, Molecular , Molecular Sequence Data , Nucleocytoplasmic Transport Proteins/ultrastructure , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Secondary , RNA Transport , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/ultrastructure , Sequence Homology, Amino Acid
18.
Mol Cell ; 58(5): 854-62, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25936803

ABSTRACT

Eukaryotic ribosome biogenesis requires nuclear import and hierarchical incorporation of ∼80 ribosomal proteins (RPs) into the ribosomal RNA core. In contrast to prokaryotes, many eukaryotic RPs possess long extensions that interdigitate in the mature ribosome. RpL4 is a prime example, with an âˆ¼80-residue-long surface extension of unknown function. Here, we identify assembly chaperone Acl4 that initially binds the universally conserved internal loop of newly synthesized RpL4 via its superhelical TPR domain, thereby restricting RpL4 loop insertion at its cognate nascent rRNA site. RpL4 release from Acl4 is orchestrated with pre-ribosome assembly, during which the eukaryote-specific RpL4 extension makes several distinct interactions with the 60S surface, including a co-evolved site on neighboring RpL18. Consequently, mutational inactivation of this contact site, on either RpL4 or RpL18, impairs RpL4-Acl4 disassembly and RpL4 pre-ribosome incorporation. We propose that hierarchical ribosome assembly can be achieved by eukaryotic RP extensions and dedicated assembly chaperones.


Subject(s)
Fungal Proteins/chemistry , Ribosomal Proteins/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Fungal Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Ribosomal Proteins/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Saccharomyces cerevisiae
19.
J Cell Biol ; 208(3): 283-97, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25646085

ABSTRACT

Nuclear pore complexes (NPCs) are huge assemblies formed from ∼30 different nucleoporins, typically organized in subcomplexes. One module, the conserved Nup82 complex at the cytoplasmic face of NPCs, is crucial to terminate mRNA export. To gain insight into the structure, assembly, and function of the cytoplasmic pore filaments, we reconstituted in yeast the Nup82-Nup159-Nsp1-Dyn2 complex, which was suitable for biochemical, biophysical, and electron microscopy analyses. Our integrative approach revealed that the yeast Nup82 complex forms an unusual asymmetric structure with a dimeric array of subunits. Based on all these data, we developed a three-dimensional structural model of the Nup82 complex that depicts how this module might be anchored to the NPC scaffold and concomitantly can interact with the soluble nucleocytoplasmic transport machinery.


Subject(s)
Nuclear Pore Complex Proteins/ultrastructure , Nuclear Pore/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Amino Acid Sequence , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Saccharomyces cerevisiae/ultrastructure
20.
Methods Cell Biol ; 122: 99-115, 2014.
Article in English | MEDLINE | ID: mdl-24857727

ABSTRACT

The discovery of dynein light chain 2 (Dyn2) as a member of the nucleoporins in yeast led to a series of applications to study NPC structure and function. Its intriguing ability to act as a hub for the parallel dimerization of two short amino acid sequence motifs (DID) prompted us to utilize it as a tool for probing nucleocytoplasmic transport in vivo. Further, the distinct structure of the Dyn2-DID rod, which is easily visible in the electron microscope, allowed us to develop a precise structural label on proteins or protein complexes. This label was used to identify the position of subunits in NPC subcomplexes or to derive at pseudo-atomic models of single large Nups. The versatility for various applications of the DID-Dyn2 system makes it an attractive molecular tool beyond the nuclear pore and transport field.


Subject(s)
Dyneins/metabolism , Nuclear Pore Complex Proteins/ultrastructure , Nuclear Pore/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...