Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nucleic Acids Res ; 51(9): 4602-4612, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36999600

RESUMEN

Kinetoplastid parasites, such as trypanosomes or leishmania, rely on RNA-templated RNA editing to mature mitochondrial cryptic pre-mRNAs into functional protein-coding transcripts. Processive pan-editing of multiple editing blocks within a single transcript is dependent on the 20-subunit RNA editing substrate binding complex (RESC) that serves as a platform to orchestrate the interactions between pre-mRNA, guide RNAs (gRNAs), the catalytic RNA editing complex (RECC), and a set of RNA helicases. Due to the lack of molecular structures and biochemical studies with purified components, neither the spacio-temporal interplay of these factors nor the selection mechanism for the different RNA components is understood. Here we report the cryo-EM structure of Trypanosoma brucei RESC1-RESC2, a central hub module of the RESC complex. The structure reveals that RESC1 and RESC2 form an obligatory domain-swapped dimer. Although the tertiary structures of both subunits closely resemble each other, only RESC2 selectively binds 5'-triphosphate-nucleosides, a defining characteristic of gRNAs. We therefore propose RESC2 as the protective 5'-end binding site for gRNAs within the RESC complex. Overall, our structure provides a starting point for the study of the assembly and function of larger RNA-bound kinetoplast RNA editing modules and might aid in the design of anti-parasite drugs.


Asunto(s)
Complejos Multiproteicos , Proteínas Protozoarias , Edición de ARN , ARN Guía de Kinetoplastida , ARN , Trypanosoma brucei brucei , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/ultraestructura , ARN/química , ARN/genética , ARN/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Protozoario/química , ARN Protozoario/genética , ARN Protozoario/metabolismo , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Microscopía por Crioelectrón , Multimerización de Proteína , Estructura Terciaria de Proteína , Especificidad por Sustrato , Sitios de Unión , Unión Proteica
2.
EMBO Rep ; 20(11): e48451, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31535454

RESUMEN

Stable capsid structures of viruses protect viral RNA while they also require controlled disassembly for releasing the viral genome in the host cell. A detailed understanding of viral disassembly processes and the involved structural switches is still lacking. This process has been extensively studied using tobacco mosaic virus (TMV), and carboxylate interactions are assumed to play a critical part in this process. Here, we present two cryo-EM structures of the helical TMV assembly at 2.0 and 1.9 Å resolution in conditions of high Ca2+ concentration at low pH and in water. Based on our atomic models, we identify the conformational details of the disassembly switch mechanism: In high Ca2+ /acidic pH environment, the virion is stabilized between neighboring subunits through carboxyl groups E95 and E97 in close proximity to a Ca2+ binding site that is shared between two subunits. Upon increase in pH and lower Ca2+ levels, mutual repulsion of the E95/E97 pair and Ca2+ removal destabilize the network of interactions between adjacent subunits at lower radius and release the switch for viral disassembly.


Asunto(s)
Modelos Moleculares , Virus del Mosaico del Tabaco/fisiología , Ensamble de Virus , Calcio/química , Cápside/química , Concentración de Iones de Hidrógeno , Fenotipo , Reproducibilidad de los Resultados , Virión
3.
PLoS Genet ; 11(12): e1005677, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26624285

RESUMEN

Ribosome biogenesis is a ubiquitous and essential process in cells. Defects in ribosome biogenesis and function result in a group of human disorders, collectively known as ribosomopathies. In this study, we describe a zebrafish mutant with a loss-of-function mutation in nol9, a gene that encodes a non-ribosomal protein involved in rRNA processing. nol9sa1022/sa1022 mutants have a defect in 28S rRNA processing. The nol9sa1022/sa1022 larvae display hypoplastic pancreas, liver and intestine and have decreased numbers of hematopoietic stem and progenitor cells (HSPCs), as well as definitive erythrocytes and lymphocytes. In addition, ultrastructural analysis revealed signs of pathological processes occurring in endothelial cells of the caudal vein, emphasizing the complexity of the phenotype observed in nol9sa1022/sa1022 larvae. We further show that both the pancreatic and hematopoietic deficiencies in nol9sa1022/sa1022 embryos were due to impaired cell proliferation of respective progenitor cells. Interestingly, genetic loss of Tp53 rescued the HSPCs but not the pancreatic defects. In contrast, activation of mRNA translation via the mTOR pathway by L-Leucine treatment did not revert the erythroid or pancreatic defects. Together, we present the nol9sa1022/sa1022 mutant, a novel zebrafish ribosomopathy model, which recapitulates key human disease characteristics. The use of this genetically tractable model will enhance our understanding of the tissue-specific mechanisms following impaired ribosome biogenesis in the context of an intact vertebrate.


Asunto(s)
Morfogénesis/genética , Polinucleótido 5'-Hidroxil-Quinasa/biosíntesis , Ribosomas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Modelos Animales de Enfermedad , Hematopoyesis/genética , Células Madre Hematopoyéticas/patología , Humanos , Páncreas/metabolismo , Páncreas/patología , Polinucleótido 5'-Hidroxil-Quinasa/genética , ARN Ribosómico 28S/genética , Ribosomas/patología , Pez Cebra
4.
EMBO J ; 29(22): 3810-8, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-20953161

RESUMEN

Ribosomes mediate protein synthesis by decoding the information carried by messenger RNAs (mRNAs) and catalysing peptide bond formation between amino acids. When bacterial ribosomes stall on incomplete messages, the trans-translation quality control mechanism is activated by the transfer-messenger RNA bound to small protein B (tmRNA-SmpB ribonucleoprotein complex). Trans-translation liberates the stalled ribosomes and triggers degradation of the incomplete proteins. Here, we present the cryo-electron microscopy structures of tmRNA-SmpB accommodated or translocated into stalled ribosomes. Two atomic models for each state are proposed. This study reveals how tmRNA-SmpB crosses the ribosome and how, as the problematic mRNA is ejected, the tmRNA resume codon is placed onto the ribosomal decoding site by new contacts between SmpB and the nucleotides upstream of the tag-encoding sequence. This provides a structural basis for the transit of the large tmRNA-SmpB complex through the ribosome and for the means by which the tmRNA internal frame is set for translation to resume.


Asunto(s)
Proteínas Bacterianas/metabolismo , ARN Bacteriano/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Thermus thermophilus/metabolismo , Proteínas Bacterianas/química , Secuencia de Bases , Microscopía por Crioelectrón , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Biosíntesis de Proteínas , Conformación Proteica , ARN Bacteriano/química , Proteínas de Unión al ARN/química , Ribosomas/química , Thermus thermophilus/química
5.
Science ; 381(6655): 313-319, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37384673

RESUMEN

Loss of H2A-H2B histone dimers is a hallmark of actively transcribed genes, but how the cellular machinery functions in the context of noncanonical nucleosomal particles remains largely elusive. In this work, we report the structural mechanism for adenosine 5'-triphosphate-dependent chromatin remodeling of hexasomes by the INO80 complex. We show how INO80 recognizes noncanonical DNA and histone features of hexasomes that emerge from the loss of H2A-H2B. A large structural rearrangement switches the catalytic core of INO80 into a distinct, spin-rotated mode of remodeling while its nuclear actin module remains tethered to long stretches of unwrapped linker DNA. Direct sensing of an exposed H3-H4 histone interface activates INO80, independently of the H2A-H2B acidic patch. Our findings reveal how the loss of H2A-H2B grants remodelers access to a different, yet unexplored layer of energy-driven chromatin regulation.


Asunto(s)
Chaetomium , Ensamble y Desensamble de Cromatina , Cromatina , Histonas , Nucleosomas , Cromatina/química , ADN/química , Histonas/química , Nucleosomas/química , Microscopía por Crioelectrón , Chaetomium/química , Chaetomium/ultraestructura
6.
Nat Commun ; 14(1): 7068, 2023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37923743

RESUMEN

Legionella pneumophila (LP) secretes more than 300 effectors into the host cytosol to facilitate intracellular replication. One of these effectors, SidH, 253 kDa in size with no sequence similarity to proteins of known function is toxic when overexpressed in host cells. SidH is regulated by the LP metaeffector LubX which targets SidH for degradation in a temporal manner during LP infection. The mechanism underlying the toxicity of SidH and its role in LP infection are unknown. Here, we determined the cryo-EM structure of SidH at 2.7 Å revealing a unique alpha helical arrangement with no overall similarity to known protein structures. Surprisingly, purified SidH came bound to a E. coli EF-Tu/t-RNA/GTP ternary complex which could be modeled into the cryo-EM density. Mutation of residues disrupting the SidH-tRNA interface and SidH-EF-Tu interface abolish the toxicity of overexpressed SidH in human cells, a phenotype confirmed in infection of Acanthamoeba castellani. We also present the cryo-EM structure of SidH in complex with a U-box domain containing ubiquitin ligase LubX delineating the mechanism of regulation of SidH. Our data provide the basis for the toxicity of SidH and into its regulation by the metaeffector LubX.


Asunto(s)
Legionella pneumophila , Enfermedad de los Legionarios , Humanos , Legionella pneumophila/metabolismo , Escherichia coli/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Bacterianas/metabolismo
7.
RNA ; 16(2): 299-306, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20038631

RESUMEN

In eubacteria, translation of defective messenger RNAs (mRNAs) produces truncated polypeptides that stall on the ribosome. A quality control mechanism referred to as trans-translation is performed by transfer-messenger RNA (tmRNA), a specialized RNA acting as both a tRNA and an mRNA, associated with small protein B (SmpB). So far, a clear view of the structural movements of both the protein and RNA necessary to perform accommodation is still lacking. By using a construct containing the tRNA-like domain as well as the extended helix H2 of tmRNA, we present a cryo-electron microscopy study of the process of accommodation. The structure suggests how tmRNA and SmpB move into the ribosome decoding site after the release of EF-Tu.GDP. While two SmpB molecules are bound per ribosome in a preaccommodated state, our results show that during accommodation the SmpB protein interacting with the small subunit decoding site stays in place while the one interacting with the large subunit moves away. Relative to canonical translation, an additional movement is observed due to the rotation of H2. This suggests that the larger movement required to resume translation on a tmRNA internal open reading frame starts during accommodation.


Asunto(s)
Proteínas Bacterianas/metabolismo , ARN Bacteriano/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Ribosomas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Microscopía por Crioelectrón , Imagenología Tridimensional , Sustancias Macromoleculares , Modelos Moleculares , Conformación de Ácido Nucleico , Biosíntesis de Proteínas , Conformación Proteica , ARN Bacteriano/química , ARN Bacteriano/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Thermus thermophilus/genética , Thermus thermophilus/metabolismo , Thermus thermophilus/ultraestructura
8.
Science ; 375(6576): 50-57, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34822310

RESUMEN

Recognition of the intron branch site (BS) by the U2 small nuclear ribonucleoprotein (snRNP) is a critical event during spliceosome assembly. In mammals, BS sequences are poorly conserved, and unambiguous intron recognition cannot be achieved solely through a base-pairing mechanism. We isolated human 17S U2 snRNP and reconstituted in vitro its adenosine 5´-triphosphate (ATP)­dependent remodeling and binding to the pre­messenger RNA substrate. We determined a series of high-resolution (2.0 to 2.2 angstrom) structures providing snapshots of the BS selection process. The substrate-bound U2 snRNP shows that SF3B6 stabilizes the BS:U2 snRNA duplex, which could aid binding of introns with poor sequence complementarity. ATP-dependent remodeling uncoupled from substrate binding captures U2 snRNA in a conformation that competes with BS recognition, providing a selection mechanism based on branch helix stability.


Asunto(s)
Intrones , Precursores del ARN/química , Ribonucleoproteína Nuclear Pequeña U2/química , Empalmosomas/química , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Unión Proteica , Conformación Proteica , Precursores del ARN/metabolismo , Empalme del ARN , Factores de Empalme de ARN/química , Factores de Empalme de ARN/metabolismo , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Empalmosomas/metabolismo , Transactivadores/química , Transactivadores/metabolismo
9.
Elife ; 112022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35060902

RESUMEN

Cryogenic electron microscopy has become an essential tool for structure determination of biological macromolecules. In practice, the difficulty to reliably prepare samples with uniform ice thickness still represents a barrier for routine high-resolution imaging and limits the current throughput of the technique. We show that a nanofluidic sample support with well-defined geometry can be used to prepare cryo-EM specimens with reproducible ice thickness from picoliter sample volumes. The sample solution is contained in electron-transparent nanochannels that provide uniform thickness gradients without further optimisation and eliminate the potentially destructive air-water interface. We demonstrate the possibility to perform high-resolution structure determination with three standard protein specimens. Nanofabricated sample supports bear potential to automate the cryo-EM workflow, and to explore new frontiers for cryo-EM applications such as time-resolved imaging and high-throughput screening.


Asunto(s)
Microscopía por Crioelectrón/métodos , Microfluídica/instrumentación , Microfluídica/métodos , Análisis por Matrices de Proteínas/métodos , Manejo de Especímenes/instrumentación , Manejo de Especímenes/métodos , Microscopía por Crioelectrón/instrumentación , Complejo de la Endopetidasa Proteasomal , Agua/química
10.
Nat Commun ; 13(1): 6737, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36347890

RESUMEN

The essential deamination of adenosine A34 to inosine at the wobble base is the individual tRNA modification with the greatest effects on mRNA decoding, empowering a single tRNA to translate three different codons. To date, many aspects of how eukaryotic deaminases specifically select their multiple substrates remain unclear. Here, using cryo-EM, we present the structure of a eukaryotic ADAT2/3 deaminase bound to a full-length tRNA, revealing that the enzyme distorts the anticodon loop, but in contrast to the bacterial enzymes, selects its substrate via sequence-independent contacts of eukaryote-acquired flexible or intrinsically unfolded motifs distal from the conserved catalytic core. A gating mechanism for substrate entry to the active site is identified. Our multi-step tRNA recognition model yields insights into how RNA editing by A34 deamination evolved, shaped the genetic code, and directly impacts the eukaryotic proteome.


Asunto(s)
Adenosina Desaminasa , Eucariontes , Adenosina Desaminasa/metabolismo , Eucariontes/genética , Eucariontes/metabolismo , Inosina/metabolismo , ARN de Transferencia/metabolismo , Anticodón/genética
11.
J Biol Chem ; 285(13): 9525-9534, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20110359

RESUMEN

Hsp90 and tubulin are among the most abundant proteins in the cytosol of eukaryotic cells. Although Hsp90 plays key roles in maintaining its client proteins in their active state, tubulin is essential for fundamental processes such as cell morphogenesis and division. Several studies have suggested a possible connection between Hsp90 and the microtubule cytoskeleton. Because tubulin is a labile protein in its soluble form, we investigated whether Hsp90 protects it against thermal denaturation. Both proteins were purified from porcine brain, and their interaction was characterized in vitro by using spectrophotometry, sedimentation assays, video-enhanced differential interference contrast light microscopy, and native polyacrylamide gel electrophoresis. Our results show that Hsp90 protects tubulin against thermal denaturation and keeps it in a state compatible with microtubule polymerization. We demonstrate that Hsp90 cannot resolve tubulin aggregates but that it likely binds early unfolding intermediates, preventing their aggregation. Protection was maximal at a stoichiometry of two molecules of Hsp90 for one of tubulin. This protection does not require ATP binding and hydrolysis by Hsp90, but it is counteracted by geldanamycin, a specific inhibitor of Hsp90.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Tubulina (Proteína)/química , Adenosina Trifosfato/química , Animales , Benzoquinonas/farmacología , Encéfalo/metabolismo , Citosol/metabolismo , Electroforesis en Gel de Poliacrilamida/métodos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Calor , Lactamas Macrocíclicas/farmacología , Luz , Microscopía de Interferencia/métodos , Microtúbulos/metabolismo , Desnaturalización Proteica , Espectrofotometría/métodos , Porcinos
12.
J Vis Exp ; (169)2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33818563

RESUMEN

Cryogenic electron tomography (cryoET) is a powerful method to study the 3D structure of biological samples in a close-to-native state. Current state-of-the-art cryoET combined with subtomogram averaging analysis enables the high-resolution structural determination of macromolecular complexes that are present in multiple copies within tomographic reconstructions. Tomographic experiments usually require a vast amount of tilt series to be acquired by means of high-end transmission electron microscopes with important operational running-costs. Although the throughput and reliability of automated data acquisition routines have constantly improved over the recent years, the process of selecting regions of interest at which a tilt series will be acquired cannot be easily automated and it still relies on the user's manual input. Therefore, the set-up of a large-scale data collection session is a time-consuming procedure that can considerably reduce the remaining microscope time available for tilt series acquisition. Here, the protocol describes the recently developed implementations based on the SerialEM package and the PyEM software that significantly improve the time-efficiency of grid screening and large-scale tilt series data collection. The presented protocol illustrates how to use SerialEM scripting functionalities to fully automate grid mapping, grid square mapping, and tilt series acquisition. Furthermore, the protocol describes how to use PyEM to select additional acquisition targets in off-line mode after automated data collection is initiated. To illustrate this protocol, its application in the context of high-end data collection of Sars-Cov-2 tilt series is described. The presented pipeline is particularly suited to maximizing the time-efficiency of tomography experiments that require a careful selection of acquisition targets and at the same time a large amount of tilt series to be collected.


Asunto(s)
Microscopía por Crioelectrón/métodos , Tomografía con Microscopio Electrónico/métodos , SARS-CoV-2 , Procesamiento de Imagen Asistido por Computador/métodos , Sustancias Macromoleculares , Reproducibilidad de los Resultados , Programas Informáticos
13.
Nat Commun ; 12(1): 6174, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702826

RESUMEN

Legionella pneumophila (LP) avoids phagocytosis by secreting nearly 300 effector proteins into the host cytosol. SidE family of effectors (SdeA, SdeB, SdeC and SidE) employ phosphoribosyl ubiquitination to target multiple host Rab GTPases and innate immune factors. To suppress the deleterious toxicity of SidE enzymes in a timely manner, LP employs a metaeffector named SidJ. Upon activation by host Calmodulin (CaM), SidJ executes an ATP-dependent glutamylation to modify the catalytic residue Glu860 in the mono-ADP-ribosyl transferase (mART) domain of SdeA. SidJ is a unique glutamylase that adopts a kinase-like fold but contains two nucleotide-binding pockets. There is a lack of consensus about the substrate recognition and catalytic mechanism of SidJ. Here, we determined the cryo-EM structure of SidJ in complex with its substrate SdeA in two different states of catalysis. Our structures reveal that both phosphodiesterase (PDE) and mART domains of SdeA make extensive contacts with SidJ. In the pre-glutamylation state structure of the SidJ-SdeA complex, adenylylated E860 of SdeA is inserted into the non-canonical (migrated) nucleotide-binding pocket of SidJ. Structure-based mutational analysis indicates that SidJ employs its migrated pocket for the glutamylation of SdeA. Finally, using mass spectrometry, we identified several transient autoAMPylation sites close to both the catalytic pockets of SidJ. Our data provide unique insights into the substrate recognition and the mechanism of protein glutamylation by the pseudokinase SidJ.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Sitios de Unión , Calmodulina/química , Calmodulina/metabolismo , Catálisis , Microscopía por Crioelectrón , Legionella pneumophila , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Factores de Virulencia/genética
14.
Acta Crystallogr D Struct Biol ; 76(Pt 8): 724-728, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32744254

RESUMEN

Cryo-electron microscopy (cryo-EM) can be used to elucidate the 3D structure of macromolecular complexes. Driven by technological breakthroughs in electron-microscope and electron-detector development, coupled with improved image-processing procedures, it is now possible to reach high resolution both in single-particle analysis and in cryo-electron tomography and subtomogram-averaging approaches. As a consequence, the way in which cryo-EM data are collected has changed and new challenges have arisen in terms of microscope alignment, aberration correction and imaging parameters. This review describes how high-end data collection is performed at the EMBL Heidelberg cryo-EM platform, presenting recent microscope implementations that allow an increase in throughput while maintaining aberration-free imaging and the optimization of acquisition parameters to collect high-resolution data.


Asunto(s)
Microscopía por Crioelectrón/métodos , Recolección de Datos , Tomografía con Microscopio Electrónico/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Imagenología Tridimensional/métodos , Sustancias Macromoleculares/química , Ensayos Analíticos de Alto Rendimiento , Programas Informáticos
15.
Nat Struct Mol Biol ; 27(3): 233-239, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066964

RESUMEN

Genome regulation requires control of chromosome organization by SMC-kleisin complexes. The cohesin complex contains the Smc1 and Smc3 subunits that associate with the kleisin Scc1 to form a ring-shaped complex that can topologically engage chromatin to regulate chromatin structure. Release from chromatin involves opening of the ring at the Smc3-Scc1 interface in a reaction that is controlled by acetylation and engagement of the Smc ATPase head domains. To understand the underlying molecular mechanisms, we have determined the 3.2-Šresolution cryo-electron microscopy structure of the ATPγS-bound, heterotrimeric cohesin ATPase head module and the 2.1-Šresolution crystal structure of a nucleotide-free Smc1-Scc1 subcomplex from Saccharomyces cerevisiae and Chaetomium thermophilium. We found that ATP-binding and Smc1-Smc3 heterodimerization promote conformational changes within the ATPase that are transmitted to the Smc coiled-coil domains. Remodeling of the coiled-coil domain of Smc3 abrogates the binding surface for Scc1, thus leading to ring opening at the Smc3-Scc1 interface.


Asunto(s)
Adenosina Trifosfatasas/química , Proteínas de Ciclo Celular/química , Proteínas Cromosómicas no Histona/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Chaetomium/química , Chaetomium/genética , Chaetomium/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Clonación Molecular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Cohesinas
16.
Elife ; 82019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30913026

RESUMEN

RNA polymerase (Pol) I is a 14-subunit enzyme that solely transcribes pre-ribosomal RNA. Cryo-electron microscopy (EM) structures of Pol I initiation and elongation complexes have given first insights into the molecular mechanisms of Pol I transcription. Here, we present cryo-EM structures of yeast Pol I elongation complexes (ECs) bound to the nucleotide analog GMPCPP at 3.2 to 3.4 Å resolution that provide additional insight into the functional interplay between the Pol I-specific transcription-like factors A49-A34.5 and A12.2. Strikingly, most of the nucleotide-bound ECs lack the A49-A34.5 heterodimer and adopt a Pol II-like conformation, in which the A12.2 C-terminal domain is bound in a previously unobserved position at the A135 surface. Our structural and biochemical data suggest a mechanism where reversible binding of the A49-A34.5 heterodimer could contribute to the regulation of Pol I transcription initiation and elongation.


Asunto(s)
Microscopía por Crioelectrón , ARN Polimerasa I/ultraestructura , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/enzimología
17.
Nat Commun ; 10(1): 5543, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804486

RESUMEN

RNA polymerase I (Pol I) assembles with core factor (CF) and Rrn3 on the rDNA core promoter for transcription initiation. Here, we report cryo-EM structures of closed, intermediate and open Pol I initiation complexes from 2.7 to 3.7 Å resolution to visualize Pol I promoter melting and to structurally and biochemically characterize the recognition mechanism of Pol I promoter DNA. In the closed complex, double-stranded DNA runs outside the DNA-binding cleft. Rotation of CF and upstream DNA with respect to Pol I and Rrn3 results in the spontaneous loading and opening of the promoter followed by cleft closure and positioning of the Pol I A49 tandem winged helix domain (tWH) onto DNA. Conformational rearrangement of A49 tWH leads to a clash with Rrn3 to initiate complex disassembly and promoter escape. Comprehensive insight into the Pol I transcription initiation cycle allows comparisons with promoter opening by Pol II and Pol III.


Asunto(s)
ADN de Hongos/genética , Regiones Promotoras Genéticas/genética , ARN Polimerasa I/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Transcripción Genética , Sitios de Unión/genética , Microscopía por Crioelectrón , ADN de Hongos/química , ADN de Hongos/metabolismo , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , ARN Polimerasa I/química , ARN Polimerasa I/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Iniciación de la Transcripción Genética
18.
Sci Adv ; 5(7): eaaw2326, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31309145

RESUMEN

The highly conserved Elongator complex modifies transfer RNAs (tRNAs) in their wobble base position, thereby regulating protein synthesis and ensuring proteome stability. The precise mechanisms of tRNA recognition and its modification reaction remain elusive. Here, we show cryo-electron microscopy structures of the catalytic subcomplex of Elongator and its tRNA-bound state at resolutions of 3.3 and 4.4 Å. The structures resolve details of the catalytic site, including the substrate tRNA, the iron-sulfur cluster, and a SAM molecule, which are all validated by mutational analyses in vitro and in vivo. tRNA binding induces conformational rearrangements, which precisely position the targeted anticodon base in the active site. Our results provide the molecular basis for substrate recognition of Elongator, essential to understand its cellular function and role in neurodegenerative diseases and cancer.


Asunto(s)
Complejos Multiproteicos/metabolismo , Factores de Elongación de Péptidos/metabolismo , ARN de Transferencia/genética , Anticodón/química , Sitios de Unión , Dominio Catalítico , Histona Acetiltransferasas/química , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Modelos Moleculares , Conformación Molecular , Complejos Multiproteicos/química , Factores de Elongación de Péptidos/química , Factores de Elongación de Péptidos/genética , Unión Proteica , ARN de Transferencia/química , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
19.
Nat Commun ; 9(1): 4420, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30356040

RESUMEN

Understanding the structural biology of the insulin receptor and how it signals is of key importance in the development of insulin analogs to treat diabetes. We report here a cryo-electron microscopy structure of a single insulin bound to a physiologically relevant, high-affinity version of the receptor ectodomain, the latter generated through attachment of C-terminal leucine zipper elements to overcome the conformational flexibility associated with ectodomain truncation. The resolution of the cryo-electron microscopy maps is 3.2 Å in the insulin-binding region and 4.2 Å in the membrane-proximal region. The structure reveals how the membrane proximal domains of the receptor come together to effect signalling and how insulin's negative cooperativity of binding likely arises. Our structure further provides insight into the high affinity of certain super-mitogenic insulins. Together, these findings provide a new platform for insulin analog investigation and design.


Asunto(s)
Receptor de Insulina/química , Receptor de Insulina/ultraestructura , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología
20.
Science ; 359(6383): 1533-1536, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29599245

RESUMEN

The integrated stress response (ISR) is a conserved translational and transcriptional program affecting metabolism, memory, and immunity. The ISR is mediated by stress-induced phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) that attenuates the guanine nucleotide exchange factor eIF2B. A chemical inhibitor of the ISR, ISRIB, reverses the attenuation of eIF2B by phosphorylated eIF2α, protecting mice from neurodegeneration and traumatic brain injury. We describe a 4.1-angstrom-resolution cryo-electron microscopy structure of human eIF2B with an ISRIB molecule bound at the interface between the ß and δ regulatory subunits. Mutagenesis of residues lining this pocket altered the hierarchical cellular response to ISRIB analogs in vivo and ISRIB binding in vitro. Our findings point to a site in eIF2B that can be exploited by ISRIB to regulate translation.


Asunto(s)
Acetamidas/química , Ciclohexilaminas/química , Factor 2B Eucariótico de Iniciación/química , Acetamidas/farmacología , Animales , Microscopía por Crioelectrón , Ciclohexilaminas/farmacología , Factor 2B Eucariótico de Iniciación/genética , Células HeLa , Humanos , Ratones , Mutagénesis , Fosforilación , Unión Proteica , Biosíntesis de Proteínas/efectos de los fármacos , Conformación Proteica , Estrés Fisiológico/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA