Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Cell ; 178(1): 76-90.e22, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31155236

RESUMEN

In ribosome-associated quality control (RQC), Rqc2/NEMF closely supports the E3 ligase Ltn1/listerin in promoting ubiquitylation and degradation of aberrant nascent-chains obstructing large (60S) ribosomal subunits-products of ribosome stalling during translation. However, while Ltn1 is eukaryote-specific, Rqc2 homologs are also found in bacteria and archaea; whether prokaryotic Rqc2 has an RQC-related function has remained unknown. Here, we show that, as in eukaryotes, a bacterial Rqc2 homolog (RqcH) recognizes obstructed 50S subunits and promotes nascent-chain proteolysis. Unexpectedly, RqcH marks nascent-chains for degradation in a direct manner, by appending C-terminal poly-alanine tails that act as degrons recognized by the ClpXP protease. Furthermore, RqcH acts redundantly with tmRNA/ssrA and protects cells against translational and environmental stresses. Our results uncover a proteolytic-tagging mechanism with implications toward the function of related modifications in eukaryotes and suggest that RQC was already active in the last universal common ancestor (LUCA) to help cope with incomplete translation.


Asunto(s)
Alanina/metabolismo , Bacillus subtilis/metabolismo , Células Procariotas/metabolismo , Proteolisis , Subunidades Ribosómicas Grandes Bacterianas/metabolismo , Células Eucariotas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Proteínas de Unión al ARN/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
2.
Cell ; 161(4): 845-57, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25957688

RESUMEN

Macromolecular machines, such as the ribosome, undergo large-scale conformational changes during their functional cycles. Although their mode of action is often compared to that of mechanical machines, a crucial difference is that, at the molecular dimension, thermodynamic effects dominate functional cycles, with proteins fluctuating stochastically between functional states defined by energetic minima on an energy landscape. Here, we have used cryo-electron microscopy to image ex-vivo-derived human polysomes as a source of actively translating ribosomes. Multiparticle refinement and 3D variability analysis allowed us to visualize a variety of native translation intermediates. Significantly populated states include not only elongation cycle intermediates in pre- and post-translocational states, but also eEF1A-containing decoding and termination/recycling complexes. Focusing on the post-translocational state, we extended this assessment to the single-residue level, uncovering striking details of ribosome-ligand interactions and identifying both static and functionally important dynamic elements.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas/química , Ribosomas/ultraestructura , Secuencia de Aminoácidos , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , ARN de Transferencia/química , Alineación de Secuencia , Termodinámica
3.
Cell ; 158(1): 121-31, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995983

RESUMEN

The extent to which bacterial ribosomes and the significantly larger eukaryotic ribosomes share the same mechanisms of ribosomal elongation is unknown. Here, we present subnanometer resolution cryoelectron microscopy maps of the mammalian 80S ribosome in the posttranslocational state and in complex with the eukaryotic eEF1A⋅Val-tRNA⋅GMPPNP ternary complex, revealing significant differences in the elongation mechanism between bacteria and mammals. Surprisingly, and in contrast to bacterial ribosomes, a rotation of the small subunit around its long axis and orthogonal to the well-known intersubunit rotation distinguishes the posttranslocational state from the classical pretranslocational state ribosome. We term this motion "subunit rolling." Correspondingly, a mammalian decoding complex visualized in substates before and after codon recognition reveals structural distinctions from the bacterial system. These findings suggest how codon recognition leads to GTPase activation in the mammalian system and demonstrate that in mammalia subunit rolling occurs during tRNA selection.


Asunto(s)
Mamíferos/metabolismo , Ribosomas/química , Secuencia de Aminoácidos , Animales , Anticodón/metabolismo , Codón/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos , Datos de Secuencia Molecular , Extensión de la Cadena Peptídica de Translación , ARN de Transferencia/metabolismo , Conejos , Saccharomyces cerevisiae/metabolismo , Tetrahymena thermophila/metabolismo
4.
Mol Cell ; 81(6): 1200-1215.e9, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33639093

RESUMEN

Ribosome biogenesis is a fundamental multi-step cellular process that culminates in the formation of ribosomal subunits, whose production and modification are regulated by numerous biogenesis factors. In this study, we analyze physiologic prokaryotic ribosome biogenesis by isolating bona fide pre-50S subunits from an Escherichia coli strain with the biogenesis factor ObgE, affinity tagged at its native gene locus. Our integrative structural approach reveals a network of interacting biogenesis factors consisting of YjgA, RluD, RsfS, and ObgE on the immature pre-50S subunit. In addition, our study provides mechanistic insight into how the GTPase ObgE, in concert with other biogenesis factors, facilitates the maturation of the 50S functional core and reveals both conserved and divergent evolutionary features of ribosome biogenesis between prokaryotes and eukaryotes.


Asunto(s)
Proteínas de Escherichia coli , Evolución Molecular , Sitios Genéticos , Hidroliasas , Proteínas de Unión al GTP Monoméricas , Subunidades Ribosómicas Grandes Bacterianas , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hidroliasas/química , Hidroliasas/genética , Hidroliasas/metabolismo , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Subunidades Ribosómicas Grandes Bacterianas/química , Subunidades Ribosómicas Grandes Bacterianas/genética , Subunidades Ribosómicas Grandes Bacterianas/metabolismo
5.
Mol Cell ; 81(2): 304-322.e16, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33357414

RESUMEN

Protein synthesis must be finely tuned in the developing nervous system as the final essential step of gene expression. This study investigates the architecture of ribosomes from the neocortex during neurogenesis, revealing Ebp1 as a high-occupancy 60S peptide tunnel exit (TE) factor during protein synthesis at near-atomic resolution by cryoelectron microscopy (cryo-EM). Ribosome profiling demonstrated Ebp1-60S binding is highest during start codon initiation and N-terminal peptide elongation, regulating ribosome occupancy of these codons. Membrane-targeting domains emerging from the 60S tunnel, which recruit SRP/Sec61 to the shared binding site, displace Ebp1. Ebp1 is particularly abundant in the early-born neural stem cell (NSC) lineage and regulates neuronal morphology. Ebp1 especially impacts the synthesis of membrane-targeted cell adhesion molecules (CAMs), measured by pulsed stable isotope labeling by amino acids in cell culture (pSILAC)/bioorthogonal noncanonical amino acid tagging (BONCAT) mass spectrometry (MS). Therefore, Ebp1 is a central component of protein synthesis, and the ribosome TE is a focal point of gene expression control in the molecular specification of neuronal morphology during development.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Neocórtex/metabolismo , Neuronas/metabolismo , Biosíntesis de Proteínas , Proteostasis/genética , Proteínas de Unión al ARN/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética , Animales , Animales Recién Nacidos , Sitios de Unión , Moléculas de Adhesión Celular Neuronal/química , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular Tumoral , Microscopía por Crioelectrón , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Masculino , Ratones , Neocórtex/citología , Neocórtex/crecimiento & desarrollo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/citología , Cultivo Primario de Células , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/ultraestructura , Partícula de Reconocimiento de Señal/química , Partícula de Reconocimiento de Señal/genética , Partícula de Reconocimiento de Señal/metabolismo
6.
Nature ; 595(7869): 741-745, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234344

RESUMEN

Peptide-chain elongation during protein synthesis entails sequential aminoacyl-tRNA selection and translocation reactions that proceed rapidly (2-20 per second) and with a low error rate (around 10-3 to 10-5 at each step) over thousands of cycles1. The cadence and fidelity of ribosome transit through mRNA templates in discrete codon increments is a paradigm for movement in biological systems that must hold for diverse mRNA and tRNA substrates across domains of life. Here we use single-molecule fluorescence methods to guide the capture of structures of early translocation events on the bacterial ribosome. Our findings reveal that the bacterial GTPase elongation factor G specifically engages spontaneously achieved ribosome conformations while in an active, GTP-bound conformation to unlock and initiate peptidyl-tRNA translocation. These findings suggest that processes intrinsic to the pre-translocation ribosome complex can regulate the rate of protein synthesis, and that energy expenditure is used later in the translocation mechanism than previously proposed.


Asunto(s)
Factor G de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Aminoacil-ARN de Transferencia/genética , Ribosomas/metabolismo , Codón , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , ARN Mensajero/genética
7.
Mol Cell ; 74(1): 143-157.e5, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30795892

RESUMEN

Bacteriophage λN protein, a model anti-termination factor, binds nascent RNA and host Nus factors, rendering RNA polymerase resistant to all pause and termination signals. A 3.7-Å-resolution cryo-electron microscopy structure and structure-informed functional analyses reveal a multi-pronged strategy by which the intrinsically unstructured λN directly modifies RNA polymerase interactions with the nucleic acids and subverts essential functions of NusA, NusE, and NusG to reprogram the transcriptional apparatus. λN repositions NusA and remodels the ß subunit flap tip, which likely precludes folding of pause or termination RNA hairpins in the exit tunnel and disrupts termination-supporting interactions of the α subunit C-terminal domains. λN invades and traverses the RNA polymerase hybrid cavity, likely stabilizing the hybrid and impeding pause- or termination-related conformational changes of polymerase. λN also lines upstream DNA, seemingly reinforcing anti-backtracking and anti-swiveling by NusG. Moreover, λN-repositioned NusA and NusE sequester the NusG C-terminal domain, counteracting ρ-dependent termination. Other anti-terminators likely utilize similar mechanisms to enable processive transcription.


Asunto(s)
Bacteriófago lambda/metabolismo , Escherichia coli/metabolismo , ARN Bacteriano/biosíntesis , Factores de Transcripción/metabolismo , Terminación de la Transcripción Genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Bacteriófago lambda/genética , Sitios de Unión , Microscopía por Crioelectrón , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/genética , Escherichia coli/virología , Regulación Bacteriana de la Expresión Génica , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica , ARN Bacteriano/química , ARN Bacteriano/genética , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/genética , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/genética
8.
Mol Cell ; 70(5): 881-893.e3, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29883607

RESUMEN

The assembly of ribosomal subunits is an essential prerequisite for protein biosynthesis in all domains of life. Although biochemical and biophysical approaches have advanced our understanding of ribosome assembly, our mechanistic comprehension of this process is still limited. Here, we perform an in vitro reconstitution of the Escherichia coli 50S ribosomal subunit. Late reconstitution products were subjected to high-resolution cryo-electron microscopy and multiparticle refinement analysis to reconstruct five distinct precursors of the 50S subunit with 4.3-3.8 Å resolution. These assembly intermediates define a progressive maturation pathway culminating in a late assembly particle, whose structure is more than 96% identical to a mature 50S subunit. Our structures monitor the formation and stabilization of structural elements in a nascent particle in unprecedented detail and identify the maturation of the rRNA-based peptidyl transferase center as the final critical step along the 50S assembly pathway.


Asunto(s)
Escherichia coli/metabolismo , ARN Bacteriano/metabolismo , ARN Ribosómico 23S/metabolismo , Subunidades Ribosómicas Grandes Bacterianas/metabolismo , Microscopía por Crioelectrón , Escherichia coli/genética , Escherichia coli/ultraestructura , Modelos Moleculares , Conformación de Ácido Nucleico , Conformación Proteica , ARN Bacteriano/genética , ARN Bacteriano/ultraestructura , ARN Ribosómico 23S/genética , ARN Ribosómico 23S/ultraestructura , Subunidades Ribosómicas Grandes Bacterianas/genética , Subunidades Ribosómicas Grandes Bacterianas/ultraestructura , Relación Estructura-Actividad
9.
PLoS Pathog ; 17(8): e1009775, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34339457

RESUMEN

Viruses have evolved means to manipulate the host's ubiquitin-proteasome system, in order to down-regulate antiviral host factors. The Vpx/Vpr family of lentiviral accessory proteins usurp the substrate receptor DCAF1 of host Cullin4-RING ligases (CRL4), a family of modular ubiquitin ligases involved in DNA replication, DNA repair and cell cycle regulation. CRL4DCAF1 specificity modulation by Vpx and Vpr from certain simian immunodeficiency viruses (SIV) leads to recruitment, poly-ubiquitylation and subsequent proteasomal degradation of the host restriction factor SAMHD1, resulting in enhanced virus replication in differentiated cells. To unravel the mechanism of SIV Vpr-induced SAMHD1 ubiquitylation, we conducted integrative biochemical and structural analyses of the Vpr protein from SIVs infecting Cercopithecus cephus (SIVmus). X-ray crystallography reveals commonalities between SIVmus Vpr and other members of the Vpx/Vpr family with regard to DCAF1 interaction, while cryo-electron microscopy and cross-linking mass spectrometry highlight a divergent molecular mechanism of SAMHD1 recruitment. In addition, these studies demonstrate how SIVmus Vpr exploits the dynamic architecture of the multi-subunit CRL4DCAF1 assembly to optimise SAMHD1 ubiquitylation. Together, the present work provides detailed molecular insight into variability and species-specificity of the evolutionary arms race between host SAMHD1 restriction and lentiviral counteraction through Vpx/Vpr proteins.


Asunto(s)
Proteínas Cullin/química , Productos del Gen vpr/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Proteína 1 que Contiene Dominios SAM y HD/química , Ubiquitinación , Replicación Viral , Secuencia de Aminoácidos , Animales , Microscopía por Crioelectrón , Proteínas Cullin/metabolismo , Productos del Gen vpr/genética , Proteína NEDD8/química , Proteína NEDD8/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
10.
Pharmacol Res ; 197: 106971, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38032292

RESUMEN

The class B2 of GPCRs known as adhesion G protein-coupled receptors (aGPCRs) has come under increasing academic and nonacademic research focus over the past decade due to their physiological importance as mechano-sensors in cell-cell and cell-matrix contexts. A major advance in understanding signal transduction of aGPCRs was achieved by the identification of the so-called Stachel sequence, which acts as an intramolecular agonist at the interface between the N terminus (Nt) and the seven-transmembrane helix domain (7TMD). Distinct extracellular signals received by the Nt are integrated at the Stachel into structural changes of the 7TMD towards an active state conformation. Until recently, little information was available on how the activation process of aGPCRs is realized at the molecular level. In the past three years several structures of the 7TMD plus the Stachel in complex with G proteins have been determined, which provide new insights into the architecture and molecular function of this receptor class. Herein, we review this structural information to extract common and distinct aGPCR features with particular focus on the Stachel binding site within the 7TMD. Our analysis extends the current view of aGPCR activation and exposes similarities and differences not only between diverse aGPCR members, but also compared to other GPCR classes.


Asunto(s)
Evolución Biológica , Transducción de Señal , Sitios de Unión , Dominios Proteicos
11.
Mol Cell ; 57(3): 389-90, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25658201

RESUMEN

Utilizing their previously established minimal in vitro ubiquitination system (Shao and Hegde, 2014), Shao et al. (2015) now show how NEMF supports the binding of Listerin to stalled 60S-RNCs, a major substrate of ribosomal quality control.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Aminoacil-ARN de Transferencia/química , Ribosomas/química , Ubiquitina-Proteína Ligasas/metabolismo , Humanos
12.
Mol Cell ; 57(3): 422-32, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25601755

RESUMEN

The cricket paralysis virus (CrPV) uses an internal ribosomal entry site (IRES) to hijack the ribosome. In a remarkable RNA-based mechanism involving neither initiation factor nor initiator tRNA, the CrPV IRES jumpstarts translation in the elongation phase from the ribosomal A site. Here, we present cryoelectron microscopy (cryo-EM) maps of 80S⋅CrPV-STOP ⋅ eRF1 ⋅ eRF3 ⋅ GMPPNP and 80S⋅CrPV-STOP ⋅ eRF1 complexes, revealing a previously unseen binding state of the IRES and directly rationalizing that an eEF2-dependent translocation of the IRES is required to allow the first A-site occupation. During this unusual translocation event, the IRES undergoes a pronounced conformational change to a more stretched conformation. At the same time, our structural analysis provides information about the binding modes of eRF1 ⋅ eRF3 ⋅ GMPPNP and eRF1 in a minimal system. It shows that neither eRF3 nor ABCE1 are required for the active conformation of eRF1 at the intersection between eukaryotic termination and recycling.


Asunto(s)
Dicistroviridae/genética , Factores de Terminación de Péptidos/metabolismo , ARN Mensajero/química , ARN Viral/química , Ribosomas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Microscopía por Crioelectrón/métodos , Dicistroviridae/química , Factores Eucarióticos de Iniciación/química , Factores Eucarióticos de Iniciación/metabolismo , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Factores de Terminación de Péptidos/química , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN de Transferencia/química , ARN de Transferencia/metabolismo , ARN Viral/metabolismo , Ribosomas/química
13.
PLoS Pathog ; 16(2): e1008263, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32092125

RESUMEN

The Type III Secretion Systems (T3SS) needle complex is a conserved syringe-shaped protein translocation nanomachine with a mass of about 3.5 MDa essential for the survival and virulence of many Gram-negative bacterial pathogens. This system is composed of a membrane-embedded basal body and an extracellular needle that deliver effector proteins into host cells. High-resolution structures of the T3SS from different organisms and infection stages are needed to understand the underlying molecular mechanisms of effector translocation. Here, we present the cryo-electron microscopy structure of the isolated Shigella T3SS needle complex. The inner membrane (IM) region of the basal body adopts 24-fold rotational symmetry and forms a channel system that connects the bacterial periplasm with the export apparatus cage. The secretin oligomer adopts a heterogeneous architecture with 16- and 15-fold cyclic symmetry in the periplasmic N-terminal connector and C-terminal outer membrane ring, respectively. Two out of three IM subunits bind the secretin connector via a ß-sheet augmentation. The cryo-EM map also reveals the helical architecture of the export apparatus core, the inner rod, the needle and their intervening interfaces.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Membrana Celular/ultraestructura , Microscopía por Crioelectrón , Shigella/ultraestructura , Sistemas de Secreción Tipo III/ultraestructura , Proteínas Bacterianas/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Conformación Proteica en Lámina beta , Dominios Proteicos , Shigella/genética , Shigella/metabolismo , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo
14.
Trends Biochem Sci ; 42(8): 655-668, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28684008

RESUMEN

Internal initiation is a 5'-end-independent mode of translation initiation engaged by many virus- and putatively some cell-encoded templates. Internal initiation is facilitated by specific RNA tertiary folds, called internal ribosomal entry sites (IRESs), in the 5' untranslated region (UTR) of the respective transcripts. In this review we discuss recent structural insight into how established IRESs first capture and then manipulate the eukaryotic translation machinery through non-canonical interactions and by guiding the intrinsic conformational flexibility of the eukaryotic ribosome. Because IRESs operate with reduced complexity and constitute minimal systems of initiation, comparison with canonical initiation may allow common mechanistic principles of the ribosome to be delineated.


Asunto(s)
Sitios Internos de Entrada al Ribosoma/genética , Ribosomas/metabolismo , Regiones no Traducidas 5'/genética , Conformación de Ácido Nucleico , Ribosomas/química , Ribosomas/genética
15.
EMBO J ; 34(24): 3042-58, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26604301

RESUMEN

Internal ribosomal entry sites (IRESs) are structured cis-acting RNAs that drive an alternative, cap-independent translation initiation pathway. They are used by many viruses to hijack the translational machinery of the host cell. IRESs facilitate translation initiation by recruiting and actively manipulating the eukaryotic ribosome using only a subset of canonical initiation factor and IRES transacting factors. Here we present cryo-EM reconstructions of the ribosome 80S- and 40S-bound Hepatitis C Virus (HCV) IRES. The presence of four subpopulations for the 80S•HCV IRES complex reveals dynamic conformational modes of the complex. At a global resolution of 3.9 Šfor the most stable complex, a derived atomic model reveals a complex fold of the IRES RNA and molecular details of its interaction with the ribosome. The comparison of obtained structures explains how a modular architecture facilitates mRNA loading and tRNA binding to the P-site. This information provides the structural foundation for understanding the mechanism of HCV IRES RNA-driven translation initiation.


Asunto(s)
Sitios Internos de Entrada al Ribosoma , ARN Viral/química , Subunidades Ribosómicas/química , Secuencia de Aminoácidos , Secuencia de Bases , Hepatitis C/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Unión Proteica
16.
Mol Cell ; 44(2): 214-24, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-22017870

RESUMEN

Although the structural core of the ribosome is conserved in all kingdoms of life, eukaryotic ribosomes are significantly larger and more complex than their bacterial counterparts. The extent to which these differences influence the molecular mechanism of translation remains elusive. Multiparticle cryo-electron microscopy and single-molecule FRET investigations of the mammalian pretranslocation complex reveal spontaneous, large-scale conformational changes, including an intersubunit rotation of the ribosomal subunits. Through structurally related processes, tRNA substrates oscillate between classical and at least two distinct hybrid configurations facilitated by localized changes in their L-shaped fold. Hybrid states are favored within the mammalian complex. However, classical tRNA positions can be restored by tRNA binding to the E site or by the eukaryotic-specific antibiotic and translocation inhibitor cycloheximide. These findings reveal critical distinctions in the structural and energetic features of bacterial and mammalian ribosomes, providing a mechanistic basis for divergent translation regulation strategies and species-specific antibiotic action.


Asunto(s)
Aminoacil-ARN de Transferencia/química , Ribosomas/química , Animales , Antibacterianos/química , Sitios de Unión , Microscopía por Crioelectrón , Cicloheximida/química , Transferencia Resonante de Energía de Fluorescencia , Modelos Moleculares , Conformación de Ácido Nucleico , Aminoacil-ARN de Transferencia/metabolismo , Conejos , Ribosomas/metabolismo
17.
Int J Mol Sci ; 20(1)2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30626072

RESUMEN

In all kingdoms of life, proteins are synthesized by ribosomes in a process referred to as translation. The amplitude of translational regulation exceeds the sum of transcription, mRNA degradation and protein degradation. Therefore, it is essential to investigate translation in a global scale. Like the other "omics"-methods, translatomics investigates the totality of the components in the translation process, including but not limited to translating mRNAs, ribosomes, tRNAs, regulatory RNAs and nascent polypeptide chains. Technical advances in recent years have brought breakthroughs in the investigation of these components at global scale, both for their composition and dynamics. These methods have been applied in a rapidly increasing number of studies to reveal multifaceted aspects of translation control. The process of translation is not restricted to the conversion of mRNA coding sequences into polypeptide chains, it also controls the composition of the proteome in a delicate and responsive way. Therefore, translatomics has extended its unique and innovative power to many fields including proteomics, cancer research, bacterial stress response, biological rhythmicity and plant biology. Rational design in translation can enhance recombinant protein production for thousands of times. This brief review summarizes the main state-of-the-art methods of translatomics, highlights recent discoveries made in this field and introduces applications of translatomics on basic biological and biomedical research.


Asunto(s)
Biosíntesis de Proteínas , Proteómica , Animales , Enfermedad , Humanos , Internet , ARN Mensajero/metabolismo , Ribosomas/metabolismo
18.
Nature ; 485(7399): 526-9, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22622583

RESUMEN

Bacterial ribosomes stalled at the 3' end of malfunctioning messenger RNAs can be rescued by transfer-messenger RNA (tmRNA)-mediated trans-translation. The SmpB protein forms a complex with the tmRNA, and the transfer-RNA-like domain (TLD) of the tmRNA then enters the A site of the ribosome. Subsequently, the TLD-SmpB module is translocated to the P site, a process that is facilitated by the elongation factor EF-G, and translation is switched to the mRNA-like domain (MLD) of the tmRNA. Accurate loading of the MLD into the mRNA path is an unusual initiation mechanism. Despite various snapshots of different ribosome-tmRNA complexes at low to intermediate resolution, it is unclear how the large, highly structured tmRNA is translocated and how the MLD is loaded. Here we present a cryo-electron microscopy reconstruction of a fusidic-acid-stalled ribosomal 70S-tmRNA-SmpB-EF-G complex (carrying both of the large ligands, that is, EF-G and tmRNA) at 8.3 Å resolution. This post-translocational intermediate (TI(POST)) presents the TLD-SmpB module in an intrasubunit ap/P hybrid site and a tRNA(fMet) in an intrasubunit pe/E hybrid site. Conformational changes in the ribosome and tmRNA occur in the intersubunit space and on the solvent side. The key underlying event is a unique extra-large swivel movement of the 30S head, which is crucial for both tmRNA-SmpB translocation and MLD loading, thereby coupling translocation to MLD loading. This mechanism exemplifies the versatile, dynamic nature of the ribosome, and it shows that the conformational modes of the ribosome that normally drive canonical translation can also be used in a modified form to facilitate more complex tasks in specialized non-canonical pathways.


Asunto(s)
Escherichia coli/química , Factor G de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , ARN Bacteriano/química , ARN Bacteriano/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Secuencia de Bases , Microscopía por Crioelectrón , Ácido Fusídico/metabolismo , Ligandos , Modelos Moleculares , Conformación de Ácido Nucleico , Factor G de Elongación Peptídica/química , Factor G de Elongación Peptídica/ultraestructura , Unión Proteica , Conformación Proteica , ARN Bacteriano/genética , ARN Bacteriano/ultraestructura , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/ultraestructura , Subunidades Ribosómicas/química , Subunidades Ribosómicas/genética , Subunidades Ribosómicas/metabolismo , Subunidades Ribosómicas/ultraestructura , Ribosomas/química , Ribosomas/genética , Ribosomas/ultraestructura
19.
Anal Biochem ; 532: 72-82, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28576440

RESUMEN

Maltose frequently occurs as intermediate of the central carbon metabolism of prokaryotic and eukaryotic cells. Various mutants possess elevated maltose levels. Maltose exists as two anomers, (α- and ß-form) which are rapidly interconverted without requiring enzyme-mediated catalysis. As maltose is often abundant together with other oligoglucans, selective quantification is essential. In this communication, we present a photometric maltose assay using 4-alpha-glucanotransferase (AtDPE2) from Arabidopsis thaliana. Under in vitro conditions, AtDPE2 utilizes maltose as glucosyl donor and glycogen as acceptor releasing the other hexosyl unit as free glucose which is photometrically quantified following enzymatic phosphorylation and oxidation. Under the conditions used, DPE2 does not noticeably react with other di- or oligosaccharides. Selectivity compares favorably with that of maltase frequently used in maltose assays. Reducing end interconversion of the two maltose anomers is in rapid equilibrium and, therefore, the novel assay measures total maltose contents. Furthermore, an AtDPE2-based continuous photometric assay is presented which allows to quantify ß-amylase activity and was found to be superior to a conventional test. Finally, the AtDPE2-based maltose assay was used to quantify leaf maltose contents of both Arabidopsis wild type and AtDPE2-deficient plants throughout the light-dark cycle. These data are presented together with assimilatory starch levels.


Asunto(s)
Arabidopsis/metabolismo , Sistema de la Enzima Desramificadora del Glucógeno/metabolismo , Maltosa/metabolismo , Fotometría/métodos , Plantas Modificadas Genéticamente/metabolismo , Almidón/metabolismo , Sacarosa/metabolismo , Citosol/metabolismo , Pruebas de Enzimas/métodos , Hojas de la Planta/metabolismo , Especificidad por Sustrato
20.
Nature ; 479(7372): 249-52, 2011 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22002606

RESUMEN

Hydrogenases are abundant enzymes that catalyse the reversible interconversion of H(2) into protons and electrons at high rates. Those hydrogenases maintaining their activity in the presence of O(2) are considered to be central to H(2)-based technologies, such as enzymatic fuel cells and for light-driven H(2) production. Despite comprehensive genetic, biochemical, electrochemical and spectroscopic investigations, the molecular background allowing a structural interpretation of how the catalytic centre is protected from irreversible inactivation by O(2) has remained unclear. Here we present the crystal structure of an O(2)-tolerant [NiFe]-hydrogenase from the aerobic H(2) oxidizer Ralstonia eutropha H16 at 1.5 Å resolution. The heterodimeric enzyme consists of a large subunit harbouring the catalytic centre in the H(2)-reduced state and a small subunit containing an electron relay consisting of three different iron-sulphur clusters. The cluster proximal to the active site displays an unprecedented [4Fe-3S] structure and is coordinated by six cysteines. According to the current model, this cofactor operates as an electronic switch depending on the nature of the gas molecule approaching the active site. It serves as an electron acceptor in the course of H(2) oxidation and as an electron-delivering device upon O(2) attack at the active site. This dual function is supported by the capability of the novel iron-sulphur cluster to adopt three redox states at physiological redox potentials. The second structural feature is a network of extended water cavities that may act as a channel facilitating the removal of water produced at the [NiFe] active site. These discoveries will have an impact on the design of biological and chemical H(2)-converting catalysts that are capable of cycling H(2) in air.


Asunto(s)
Cupriavidus necator/enzimología , Hidrogenasas/química , Proteínas Hierro-Azufre/química , Hierro/química , Oxígeno/metabolismo , Azufre/química , Dominio Catalítico , Membrana Celular/metabolismo , Cristalografía por Rayos X , Cisteína/metabolismo , Hidrogenasas/metabolismo , Hierro/análisis , Proteínas Hierro-Azufre/metabolismo , Modelos Moleculares , Oxidación-Reducción , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Protones , Azufre/análisis , Agua/química , Agua/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA