Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39253483

RESUMEN

Ribosome stalling during co-translational translocation at the endoplasmic reticulum (ER) causes translocon clogging and impairs ER protein biogenesis. Mammalian cells resolve translocon clogging vial a poorly characterized translocation-associated quality control (TAQC) process. Here, we combine genome-wide CRISPR screen with live cell imaging to dissect the molecular linchpin of TAQC. We show that substrates translated from mRNAs bearing a ribosome stalling poly(A) sequence are degraded by lysosomes and the proteasome, while substrates encoded by non-stop mRNAs are degraded by an unconventional ER-associated degradation (ERAD) mechanism involving ER-to-Golgi trafficking and KDEL-dependent substrate retrieval. The triaging diversity appears to result from the heterogeneity of NEMF-mediated CATylation, because a systematic characterization of representative CAT-tail mimetics establishes an AT-rich tail as a "degron" for ERAD, whereas an AG-rich tail can direct a secretory protein to the lysosome. Our study reveals an unexpected protein sorting function for CAT-tailing that safeguards ER protein biogenesis.

2.
Cell Rep ; 43(3): 113860, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38412092

RESUMEN

The ribosome-associated protein quality control (RQC) pathway acts as a translational surveillance mechanism to maintain proteostasis. In mammalian cells, the cytoplasmic RQC pathway involves nuclear export mediator factor (NEMF)-dependent recruitment of the E3 ligase Listerin to ubiquitinate ribosome-stalled nascent polypeptides on the lysine residue for degradation. However, the quality control of ribosome-stalled nuclear-encoded mitochondrial nascent polypeptides remains elusive, as these peptides can be partially imported into mitochondria through translocons, restricting accessibility to the lysine by Listerin. Here, we identify a Listerin-independent organelle-specific mitochondrial RQC pathway that acts on NEMF-mediated carboxy-terminal poly-alanine modification. In the pathway, mitochondrial proteins carrying C-end poly-Ala tails are recognized by the cytosolic E3 ligase Pirh2 and the ClpXP protease in the mitochondria, which coordinately clear ribosome-stalled mitochondrial nascent polypeptides. Defects in this elimination pathway result in NEMF-mediated aggregates and mitochondrial integrity failure, thus providing a potential molecular mechanism of the RQC pathway in mitochondrial-associated human diseases.


Asunto(s)
Péptido Hidrolasas , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ubiquitina-Proteína Ligasas/metabolismo , Péptido Hidrolasas/metabolismo , Biosíntesis de Proteínas , Lisina/metabolismo , Péptidos/metabolismo , Endopeptidasas/metabolismo , Mitocondrias/metabolismo , Ubiquitinación , Mamíferos/metabolismo
3.
Mol Cell ; 83(4): 607-621.e4, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36804914

RESUMEN

Ribosome-associated quality control (RQC) is a conserved process degrading potentially toxic truncated nascent peptides whose malfunction underlies neurodegeneration and proteostasis decline in aging. During RQC, dissociation of stalled ribosomes is followed by elongation of the nascent peptide with alanine and threonine residues, driven by Rqc2 independently of mRNA, the small ribosomal subunit and guanosine triphosphate (GTP)-hydrolyzing factors. The resulting CAT tails (carboxy-terminal tails) and ubiquitination by Ltn1 mark nascent peptides for proteasomal degradation. Here we present ten cryogenic electron microscopy (cryo-EM) structures, revealing the mechanistic basis of individual steps of the CAT tailing cycle covering initiation, decoding, peptidyl transfer, and tRNA translocation. We discovered eIF5A as a crucial eukaryotic RQC factor enabling peptidyl transfer. Moreover, we observed dynamic behavior of RQC factors and tRNAs allowing for processivity of the CAT tailing cycle without additional energy input. Together, these results elucidate key differences as well as common principles between CAT tailing and canonical translation.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Biosíntesis de Proteínas , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Péptidos/química , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Control de Calidad
4.
Brain ; 146(7): 2897-2912, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-36516294

RESUMEN

Protein quality control pathways have evolved to ensure the fidelity of protein synthesis and efficiently clear potentially toxic protein species. Defects in ribosome-associated quality control and its associated factors have been implicated in the accumulation of aberrant proteins and neurodegeneration. C9orf72 repeat-associated non-AUG translation has been suggested to involve inefficient translation elongation, lead to ribosomal pausing and activation of ribosome-associated quality control pathways. However, the role of the ribosome-associated quality control complex in the processing of proteins generated through this non-canonical translation is not well understood. Here we use reporter constructs containing the C9orf72-associated hexanucleotide repeat, ribosome-associated quality control complex deficient cell models and stain for ribosome-associated quality control markers in C9orf72-expansion carrier human tissue to understand its role in dipeptide-repeat protein pathology. Our studies show that canonical ribosome-associated quality control substrates products are efficiently cleared by the ribosome-associated quality control complex in mammalian cells. Furthermore, using stalling reporter constructs, we show that repeats associated with the C9orf72-expansion induce ribosomal stalling when arginine (R)-rich dipeptide-repeat proteins are synthesized in a length-dependent manner. However, despite triggering this pathway, these arginine-rich dipeptide-repeat proteins are not efficiently processed by the core components of the ribosome-associated quality control complex (listerin, nuclear-export mediator factor and valosin containing protein) partly due to lack of lysine residues, which precludes ubiquitination. Deficient processing by this complex may be implicated in C9orf72-expansion associated disease as dipeptide-repeat protein inclusions were observed to be predominantly devoid of ubiquitin and co-localize with nuclear-export mediator factor in mutation carriers' frontal cortex and cerebellum tissue. These findings suggest that impaired processing of these arginine-rich dipeptide-repeat proteins derived from repeat-associated non-AUG translation by the ribosome-associated quality control complex may contribute to protein homeostasis dysregulation observed in C9orf72-expansion amyotrophic lateral sclerosis and frontotemporal degeneration neuropathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedades Neurodegenerativas , Animales , Humanos , Dipéptidos/genética , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Enfermedades Neurodegenerativas/genética , Ribosomas , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Mamíferos/genética , Mamíferos/metabolismo
5.
Mol Cell ; 82(8): 1451-1466, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35452614

RESUMEN

Ribosome-associated quality-control (RQC) surveys incomplete nascent polypeptides produced by interrupted translation. Central players in RQC are the human ribosome- and tRNA-binding protein, NEMF, and its orthologs, yeast Rqc2 and bacterial RqcH, which sense large ribosomal subunits obstructed with nascent chains and then promote nascent-chain proteolysis. In canonical eukaryotic RQC, NEMF stabilizes the LTN1/Listerin E3 ligase binding to obstructed ribosomal subunits for nascent-chain ubiquitylation. Furthermore, NEMF orthologs across evolution modify nascent chains by mediating C-terminal, untemplated polypeptide elongation. In eukaryotes, this process exposes ribosome-buried nascent-chain lysines, the ubiquitin acceptor sites, to LTN1. Remarkably, in both bacteria and eukaryotes, C-terminal tails also have an extra-ribosomal function as degrons. Here, we discuss recent findings on RQC mechanisms and briefly review how ribosomal stalling is sensed upstream of RQC, including via ribosome collisions, from an evolutionary perspective. Because RQC defects impair cellular fitness and cause neurodegeneration, this knowledge provides a framework for pathway-related biology and disease studies.


Asunto(s)
Ribosomas , Proteínas de Saccharomyces cerevisiae , Bacterias/genética , Bacterias/metabolismo , Humanos , Péptidos/metabolismo , Biosíntesis de Proteínas , Ribosomas/genética , Ribosomas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
6.
Mol Cell ; 81(10): 2112-2122.e7, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33909987

RESUMEN

Incompletely synthesized nascent chains obstructing large ribosomal subunits are targeted for degradation by ribosome-associated quality control (RQC). In bacterial RQC, RqcH marks the nascent chains with C-terminal alanine (Ala) tails that are directly recognized by proteasome-like proteases, whereas in eukaryotes, RqcH orthologs (Rqc2/NEMF [nuclear export mediator factor]) assist the Ltn1/Listerin E3 ligase in nascent chain ubiquitylation. Here, we study RQC-mediated proteolytic targeting of ribosome stalling products in mammalian cells. We show that mammalian NEMF has an additional, Listerin-independent proteolytic role, which, as in bacteria, is mediated by tRNA-Ala binding and Ala tailing. However, in mammalian cells Ala tails signal proteolysis indirectly, through a pathway that recognizes C-terminal degrons; we identify the CRL2KLHDC10 E3 ligase complex and the novel C-end rule E3, Pirh2/Rchy1, as bona fide RQC pathway components that directly bind to Ala-tailed ribosome stalling products and target them for degradation. As Listerin mutation causes neurodegeneration in mice, functionally redundant E3s may likewise be implicated in molecular mechanisms of neurodegeneration.


Asunto(s)
Alanina/metabolismo , Mamíferos/metabolismo , Proteolisis , Ribosomas/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Citocinas/metabolismo , Proteínas Salivales Ricas en Prolina/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
7.
Mol Cell ; 81(1): 115-126.e7, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33259810

RESUMEN

In all branches of life, stalled translation intermediates are recognized and processed by ribosome-associated quality control (RQC) pathways. RQC begins with the splitting of stalled ribosomes, leaving an unfinished polypeptide still attached to the large subunit. Ancient and conserved NEMF family RQC proteins target these incomplete proteins for degradation by the addition of C-terminal "tails." How such tailing can occur without the regular suite of translational components is, however, unclear. Using single-particle cryo-electron microscopy (EM) of native complexes, we show that C-terminal tailing in Bacillus subtilis is mediated by NEMF protein RqcH in concert with RqcP, an Hsp15 family protein. Our structures reveal how these factors mediate tRNA movement across the ribosomal 50S subunit to synthesize polypeptides in the absence of mRNA or the small subunit.


Asunto(s)
Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Subunidades Ribosómicas Grandes Bacterianas/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/ultraestructura , Proteínas Bacterianas/genética , Microscopía por Crioelectrón , Subunidades Ribosómicas Grandes Bacterianas/genética , Subunidades Ribosómicas Grandes Bacterianas/ultraestructura
8.
Infect Disord Drug Targets ; 21(2): 268-273, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32167431

RESUMEN

INTRODUCTION: Tuberculosis (TB) remains one of the most important infectious causes of death throughout the world. A wide range of technologies have been used for the diagnosis of TB. However, current diagnostic tests are inadequate. The aim of this study was to evaluate the expression of four genes, namely ASUN, NEMF, PTPRC and DHX29 as candidate biomarkers for the diagnosis of Latent tuberculosis infection (LTBI) and active TB and discrimination of active TB and LTBI. ; Materials and Methods: The expression of the mentioned four genes as well as ACTB as a housekeeping gene was evaluated by real-time PCR. Receiver operating characteristic (ROC) curve analysis was conducted to assess the specificity and sensitivity of each validated biomarker. ; Results: Our results showed that the expression of theASUN gene could discriminate between active TB cases and healthy BCG vaccinated volunteers with an AUC value of 0.76, combing with a sensitivity of 68% and a specificity of 67%. It should be noted that the PTPRC gene also has the potential for the diagnosis of active TB with an AUC value of 0.67 and a sensitivity of 64.5% and a specificity of 70%. The curve revealed that cases with LTBI could be distinguished from healthy BCG vaccinated volunteers according to their expression of the ASUN gene with an AUC value of 0.81. The cut-off value for diagnosing was 11, with a sensitivity of 73% and a specificity of 79%. Moreover, the expression of the NEMF gene might be considered as a diagnostic tool for the diagnosis of LTBI. The analysis showed an AUC value of 0.75. The highest sensitivity (60%) and specificity (81%) were obtained with a cut off value of 12. ; Conclusion: According to our results, the expression of ASUN and NEMF genes might be considered as a diagnostic tool for the diagnosis of LTBI. Our study showed that the expression of ASUN and PTPRC was obviously higher in active TB patients than those in healthy BCG vaccinated controls. On the other hand, DHX29 and PTPRC genes might be helpful in differentiating active TB and LTBI. However, our findings deserve further validation in larger studies.


Asunto(s)
Tuberculosis Latente , Mycobacterium tuberculosis , Tuberculosis , Biomarcadores , Expresión Génica , Humanos , Antígenos Comunes de Leucocito , ARN Helicasas , Curva ROC
9.
Mol Cell ; 72(2): 286-302.e8, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30244831

RESUMEN

The ribosome-associated quality control (RQC) pathway degrades nascent chains (NCs) arising from interrupted translation. First, recycling factors split stalled ribosomes, yielding NC-tRNA/60S ribosome-nascent chain complexes (60S RNCs). 60S RNCs associate with NEMF, which recruits the E3 ubiquitin ligase Listerin that ubiquitinates NCs. The mechanism of subsequent ribosomal release of Ub-NCs remains obscure. We found that, in non-ubiquitinated 60S RNCs and 80S RNCs formed on non-stop mRNAs, tRNA is not firmly fixed in the P site, which allows peptidyl-tRNA hydrolase Ptrh1 to cleave NC-tRNA, suggesting the existence of a pathway involving release of non-ubiquitinated NCs. Association with NEMF and Listerin and ubiquitination of NCs results in accommodation of NC-tRNA, rendering 60S RNCs resistant to Ptrh1 but susceptible to ANKZF1, which induces specific cleavage in the tRNA acceptor arm, releasing proteasome-degradable Ub-NCs linked to four 3'-terminal tRNA nucleotides. We also found that TCF25, a poorly characterized RQC component, ensures preferential formation of the K48-ubiquitin linkage.


Asunto(s)
Proteínas Portadoras/metabolismo , Mamíferos/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Ribosomas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Biosíntesis de Proteínas/fisiología , ARN de Transferencia/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
10.
Elife ; 5: e11794, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26943317

RESUMEN

Ribosome stalling during translation can potentially be harmful, and is surveyed by a conserved quality control pathway that targets the associated mRNA and nascent polypeptide chain (NC). In this pathway, the ribosome-associated quality control (RQC) complex promotes the ubiquitylation and degradation of NCs remaining stalled in the 60S subunit. NC stalling is recognized by the Rqc2/Tae2 RQC subunit, which also stabilizes binding of the E3 ligase, Listerin/Ltn1. Additionally, Rqc2 modifies stalled NCs with a carboxy-terminal, Ala- and Thr-containing extension-the 'CAT tail'. However, the function of CAT tails and fate of CAT tail-modified ('CATylated') NCs has remained unknown. Here we show that CATylation mediates formation of detergent-insoluble NC aggregates. CATylation and aggregation of NCs could be observed either by inactivating Ltn1 or by analyzing NCs with limited ubiquitylation potential, suggesting that inefficient targeting by Ltn1 favors the Rqc2-mediated reaction. These findings uncover a translational stalling-dependent protein aggregation mechanism, and provide evidence that proteins can become specifically marked for aggregation.


Asunto(s)
Péptidos/metabolismo , Agregación Patológica de Proteínas , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(45): 15981-6, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349383

RESUMEN

All organisms have evolved mechanisms to manage the stalling of ribosomes upon translation of aberrant mRNA. In eukaryotes, the large ribosomal subunit-associated quality control complex (RQC), composed of the listerin/Ltn1 E3 ubiquitin ligase and cofactors, mediates the ubiquitylation and extraction of ribosome-stalled nascent polypeptide chains for proteasomal degradation. How RQC recognizes stalled ribosomes and performs its functions has not been understood. Using single-particle cryoelectron microscopy, we have determined the structure of the RQC complex bound to stalled 60S ribosomal subunits. The structure establishes how Ltn1 associates with the large ribosomal subunit and properly positions its E3-catalytic RING domain to mediate nascent chain ubiquitylation. The structure also reveals that a distinguishing feature of stalled 60S particles is an exposed, nascent chain-conjugated tRNA, and that the Tae2 subunit of RQC, which facilitates Ltn1 binding, is responsible for selective recognition of stalled 60S subunits. RQC components are engaged in interactions across a large span of the 60S subunit surface, connecting the tRNA in the peptidyl transferase center to the distally located nascent chain tunnel exit. This work provides insights into a mechanism linking translation and protein degradation that targets defective proteins immediately after synthesis, while ignoring nascent chains in normally translating ribosomes.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Biosíntesis de Proteínas/fisiología , Proteolisis , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Saccharomyces cerevisiae/metabolismo , Ubiquitinación/fisiología , Estructura Terciaria de Proteína , Aminoacil-ARN de Transferencia/genética , Aminoacil-ARN de Transferencia/metabolismo , Proteínas de Unión al ARN , Subunidades Ribosómicas Grandes de Eucariotas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
12.
RNA Biol ; 11(4): 360-72, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24646681

RESUMEN

A protein family including mammalian NEMF, Drosophila caliban, yeast Tae2, and bacterial FpbA-like proteins was first defined over a decade ago and found to be universally distributed across the three domains/superkingdoms of life. Since its initial characterization, this family of proteins has been tantalizingly linked to a wide range of biochemical functions. Tapping the enormous wealth of genome information that has accumulated since the initial characterization of these proteins, we perform a detailed computational analysis of the family, identifying multiple conserved domains. Domains identified include an enzymatic domain related to the formamidopyrimidine (Fpg), MutM, and Nei/EndoVIII family of DNA glycosylases, a novel, predicted RNA-binding domain, and a domain potentially mediating protein-protein interactions. Through this characterization, we predict that the DNA glycosylase-like domain catalytically operates on double-stranded RNA, as part of a hitherto unknown base modification mechanism that probably targets rRNAs. At least in archaea, and possibly eukaryotes, this pathway might additionally include the AMMECR1 family of proteins. The predicted RNA-binding domain associated with this family is also observed in distinct architectural contexts in other proteins across phylogenetically diverse prokaryotes. Here it is predicted to play a key role in a new pathway for tRNA 4-thiouridylation along with TusA-like sulfur transfer proteins.


Asunto(s)
ADN Glicosilasas/química , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , ARN/metabolismo , Secuencia de Aminoácidos , Animales , Biología Computacional , Secuencia Conservada , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Familia de Multigenes , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas/genética , ARN/química , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA