Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(25): 4770-4787.e20, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36493755

RESUMEN

The ATP-dependent ring-shaped chaperonin TRiC/CCT is essential for cellular proteostasis. To uncover why some eukaryotic proteins can only fold with TRiC assistance, we reconstituted the folding of ß-tubulin using human prefoldin and TRiC. We find unstructured ß-tubulin is delivered by prefoldin to the open TRiC chamber followed by ATP-dependent chamber closure. Cryo-EM resolves four near-atomic-resolution structures containing progressively folded ß-tubulin intermediates within the closed TRiC chamber, culminating in native tubulin. This substrate folding pathway appears closely guided by site-specific interactions with conserved regions in the TRiC chamber. Initial electrostatic interactions between the TRiC interior wall and both the folded tubulin N domain and its C-terminal E-hook tail establish the native substrate topology, thus enabling C-domain folding. Intrinsically disordered CCT C termini within the chamber promote subsequent folding of tubulin's core and middle domains and GTP-binding. Thus, TRiC's chamber provides chemical and topological directives that shape the folding landscape of its obligate substrates.


Asunto(s)
Chaperonina con TCP-1 , Tubulina (Proteína) , Humanos , Chaperonina con TCP-1/química , Tubulina (Proteína)/metabolismo , Pliegue de Proteína , Proteostasis , Adenosina Trifosfato/metabolismo
2.
Cell ; 184(25): 6037-6051.e14, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34852237

RESUMEN

RNA viruses generate defective viral genomes (DVGs) that can interfere with replication of the parental wild-type virus. To examine their therapeutic potential, we created a DVG by deleting the capsid-coding region of poliovirus. Strikingly, intraperitoneal or intranasal administration of this genome, which we termed eTIP1, elicits an antiviral response, inhibits replication, and protects mice from several RNA viruses, including enteroviruses, influenza, and SARS-CoV-2. While eTIP1 replication following intranasal administration is limited to the nasal cavity, its antiviral action extends non-cell-autonomously to the lungs. eTIP1 broad-spectrum antiviral effects are mediated by both local and distal type I interferon responses. Importantly, while a single eTIP1 dose protects animals from SARS-CoV-2 infection, it also stimulates production of SARS-CoV-2 neutralizing antibodies that afford long-lasting protection from SARS-CoV-2 reinfection. Thus, eTIP1 is a safe and effective broad-spectrum antiviral generating short- and long-term protection against SARS-CoV-2 and other respiratory infections in animal models.


Asunto(s)
Proteínas de la Cápside/genética , Virus Interferentes Defectuosos/metabolismo , Replicación Viral/efectos de los fármacos , Administración Intranasal , Animales , Antivirales/farmacología , Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos ampliamente neutralizantes/farmacología , COVID-19 , Proteínas de la Cápside/metabolismo , Línea Celular , Virus Interferentes Defectuosos/patogenicidad , Modelos Animales de Enfermedad , Genoma Viral/genética , Humanos , Gripe Humana , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Poliovirus/genética , Poliovirus/metabolismo , Infecciones del Sistema Respiratorio/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad
3.
Cell ; 177(3): 751-765.e15, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955883

RESUMEN

Maintaining proteostasis in eukaryotic protein folding involves cooperation of distinct chaperone systems. To understand how the essential ring-shaped chaperonin TRiC/CCT cooperates with the chaperone prefoldin/GIMc (PFD), we integrate cryoelectron microscopy (cryo-EM), crosslinking-mass-spectrometry and biochemical and cellular approaches to elucidate the structural and functional interplay between TRiC/CCT and PFD. We find these hetero-oligomeric chaperones associate in a defined architecture, through a conserved interface of electrostatic contacts that serves as a pivot point for a TRiC-PFD conformational cycle. PFD alternates between an open "latched" conformation and a closed "engaged" conformation that aligns the PFD-TRiC substrate binding chambers. PFD can act after TRiC bound its substrates to enhance the rate and yield of the folding reaction, suppressing non-productive reaction cycles. Disrupting the TRiC-PFD interaction in vivo is strongly deleterious, leading to accumulation of amyloid aggregates. The supra-chaperone assembly formed by PFD and TRiC is essential to prevent toxic conformations and ensure effective cellular proteostasis.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Chaperonas Moleculares/metabolismo , Proteostasis/fisiología , Actinas/química , Actinas/metabolismo , Chaperonina con TCP-1/química , Chaperonina con TCP-1/genética , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Pliegue de Proteína , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Saccharomyces cerevisiae/metabolismo , Electricidad Estática
5.
Annu Rev Biochem ; 86: 97-122, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28489421

RESUMEN

A healthy proteome is essential for cell survival. Protein misfolding is linked to a rapidly expanding list of human diseases, ranging from neurodegenerative diseases to aging and cancer. Many of these diseases are characterized by the accumulation of misfolded proteins in intra- and extracellular inclusions, such as amyloid plaques. The clear link between protein misfolding and disease highlights the need to better understand the elaborate machinery that manages proteome homeostasis, or proteostasis, in the cell. Proteostasis depends on a network of molecular chaperones and clearance pathways involved in the recognition, refolding, and/or clearance of aberrant proteins. Recent studies reveal that an integral part of the cellular management of misfolded proteins is their spatial sequestration into several defined compartments. Here, we review the properties, function, and formation of these compartments. Spatial sequestration plays a central role in protein quality control and cellular fitness and represents a critical link to the pathogenesis of protein aggregation-linked diseases.


Asunto(s)
Envejecimiento/metabolismo , Chaperonas Moleculares/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Agregación Patológica de Proteínas/metabolismo , Deficiencias en la Proteostasis/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/genética , Proteínas Amiloidogénicas/metabolismo , Compartimento Celular , Regulación de la Expresión Génica , Humanos , Chaperonas Moleculares/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Proteínas Priónicas/química , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Biosíntesis de Proteínas , Conformación Proteica , Pliegue de Proteína , Replegamiento Proteico , Proteolisis , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/patología
6.
Mol Cell ; 83(17): 3123-3139.e8, 2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37625406

RESUMEN

How the essential eukaryotic chaperonin TRiC/CCT assembles from eight distinct subunits into a unique double-ring architecture remains undefined. We show TRiC assembly involves a hierarchical pathway that segregates subunits with distinct functional properties until holocomplex (HC) completion. A stable, likely early intermediate arises from small oligomers containing CCT2, CCT4, CCT5, and CCT7, contiguous subunits that constitute the negatively charged hemisphere of the TRiC chamber, which has weak affinity for unfolded actin. The remaining subunits CCT8, CCT1, CCT3, and CCT6, which comprise the positively charged chamber hemisphere that binds unfolded actin more strongly, join the ring individually. Unincorporated late-assembling subunits are highly labile in cells, which prevents their accumulation and premature substrate binding. Recapitulation of assembly in a recombinant system demonstrates that the subunits in each hemisphere readily form stable, noncanonical TRiC-like HCs with aberrant functional properties. Thus, regulation of TRiC assembly along a biochemical axis disfavors the formation of stable alternative chaperonin complexes.


Asunto(s)
Chaperonina con TCP-1 , Actinas , Chaperonina con TCP-1/química , Chaperonina con TCP-1/metabolismo , Humanos , Animales
7.
Cell ; 163(5): 1108-1123, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26582131

RESUMEN

Viral protein homeostasis depends entirely on the machinery of the infected cell. Accordingly, viruses can illuminate the interplay between cellular proteostasis components and their distinct substrates. Here, we define how the Hsp70 chaperone network mediates the dengue virus life cycle. Cytosolic Hsp70 isoforms are required at distinct steps of the viral cycle, including entry, RNA replication, and virion biogenesis. Hsp70 function at each step is specified by nine distinct DNAJ cofactors. Of these, DnaJB11 relocalizes to virus-induced replication complexes to promote RNA synthesis, while DnaJB6 associates with capsid protein and facilitates virion biogenesis. Importantly, an allosteric Hsp70 inhibitor, JG40, potently blocks infection of different dengue serotypes in human primary blood cells without eliciting viral resistance or exerting toxicity to the host cells. JG40 also blocks replication of other medically-important flaviviruses including yellow fever, West Nile and Japanese encephalitis viruses. Thus, targeting host Hsp70 subnetworks provides a path for broad-spectrum antivirals.


Asunto(s)
Dengue/virología , Proteínas HSP70 de Choque Térmico/metabolismo , Replicación Viral , Animales , Proteínas de la Cápside/metabolismo , Culicidae/virología , Dengue/metabolismo , Virus del Dengue , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Humanos , Replicación Viral/efectos de los fármacos
8.
Nature ; 626(7997): 119-127, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38200310

RESUMEN

The evolution of reproductive barriers is the first step in the formation of new species and can help us understand the diversification of life on Earth. These reproductive barriers often take the form of hybrid incompatibilities, in which alleles derived from two different species no longer interact properly in hybrids1-3. Theory predicts that hybrid incompatibilities may be more likely to arise at rapidly evolving genes4-6 and that incompatibilities involving multiple genes should be common7,8, but there has been sparse empirical data to evaluate these predictions. Here we describe a mitonuclear incompatibility involving three genes whose protein products are in physical contact within respiratory complex I of naturally hybridizing swordtail fish species. Individuals homozygous for mismatched protein combinations do not complete embryonic development or die as juveniles, whereas those heterozygous for the incompatibility have reduced complex I function and unbalanced representation of parental alleles in the mitochondrial proteome. We find that the effects of different genetic interactions on survival are non-additive, highlighting subtle complexity in the genetic architecture of hybrid incompatibilities. Finally, we document the evolutionary history of the genes involved, showing signals of accelerated evolution and evidence that an incompatibility has been transferred between species via hybridization.


Asunto(s)
Núcleo Celular , Complejo I de Transporte de Electrón , Peces , Genes Letales , Especiación Genética , Hibridación Genética , Proteínas Mitocondriales , Animales , Alelos , Complejo I de Transporte de Electrón/genética , Peces/clasificación , Peces/embriología , Peces/genética , Peces/crecimiento & desarrollo , Homocigoto , Genes Letales/genética , Especificidad de la Especie , Desarrollo Embrionario/genética , Proteínas Mitocondriales/genética , Núcleo Celular/genética , Heterocigoto , Evolución Molecular
9.
Cell ; 159(5): 1042-1055, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416944

RESUMEN

The eukaryotic chaperonin TRiC (also called CCT) is the obligate chaperone for many essential proteins. TRiC is hetero-oligomeric, comprising two stacked rings of eight different subunits each. Subunit diversification from simpler archaeal chaperonins appears linked to proteome expansion. Here, we integrate structural, biophysical, and modeling approaches to identify the hitherto unknown substrate-binding site in TRiC and uncover the basis of substrate recognition. NMR and modeling provided a structural model of a chaperonin-substrate complex. Mutagenesis and crosslinking-mass spectrometry validated the identified substrate-binding interface and demonstrate that TRiC contacts full-length substrates combinatorially in a subunit-specific manner. The binding site of each subunit has a distinct, evolutionarily conserved pattern of polar and hydrophobic residues specifying recognition of discrete substrate motifs. The combinatorial recognition of polypeptides broadens the specificity of TRiC and may direct the topology of bound polypeptides along a productive folding trajectory, contributing to TRiC's unique ability to fold obligate substrates.


Asunto(s)
Chaperonina con TCP-1/química , Chaperonina con TCP-1/metabolismo , Eucariontes/química , Pliegue de Proteína , Animales , Archaea/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Bovinos , Chaperonina con TCP-1/genética , Eucariontes/citología , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato
10.
Cell ; 159(6): 1389-403, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25467444

RESUMEN

Telomere maintenance by telomerase is impaired in the stem cell disease dyskeratosis congenita and during human aging. Telomerase depends upon a complex pathway for enzyme assembly, localization in Cajal bodies, and association with telomeres. Here, we identify the chaperonin CCT/TRiC as a critical regulator of telomerase trafficking using a high-content genome-wide siRNA screen in human cells for factors required for Cajal body localization. We find that TRiC is required for folding the telomerase cofactor TCAB1, which controls trafficking of telomerase and small Cajal body RNAs (scaRNAs). Depletion of TRiC causes loss of TCAB1 protein, mislocalization of telomerase and scaRNAs to nucleoli, and failure of telomere elongation. DC patient-derived mutations in TCAB1 impair folding by TRiC, disrupting telomerase function and leading to severe disease. Our findings establish a critical role for TRiC-mediated protein folding in the telomerase pathway and link proteostasis, telomere maintenance, and human disease.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Disqueratosis Congénita/genética , Disqueratosis Congénita/patología , Humanos , Hibridación Fluorescente in Situ , Chaperonas Moleculares , Pliegue de Proteína , Telomerasa/química
11.
Cell ; 152(1-2): 196-209, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23332755

RESUMEN

In eukaryotic cells a molecular chaperone network associates with translating ribosomes, assisting the maturation of emerging nascent polypeptides. Hsp70 is perhaps the major eukaryotic ribosome-associated chaperone and the first reported to bind cotranslationally to nascent chains. However, little is known about the underlying principles and function of this interaction. Here, we use a sensitive and global approach to define the cotranslational substrate specificity of the yeast Hsp70 SSB. We find that SSB binds to a subset of nascent polypeptides whose intrinsic properties and slow translation rates hinder efficient cotranslational folding. The SSB-ribosome cycle and substrate recognition is modulated by its ribosome-bound cochaperone, RAC. Deletion of SSB leads to widespread aggregation of newly synthesized polypeptides. Thus, cotranslationally acting Hsp70 meets the challenge of folding the eukaryotic proteome by stabilizing its longer, more slowly translated, and aggregation-prone nascent polypeptides.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Biosíntesis de Proteínas , Pliegue de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Ribosomas/metabolismo
12.
Nature ; 601(7894): 637-642, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35046576

RESUMEN

Ageing is accompanied by a decline in cellular proteostasis, which underlies many age-related protein misfolding diseases1,2. Yet, how ageing impairs proteostasis remains unclear. As nascent polypeptides represent a substantial burden on the proteostasis network3, we hypothesized that altered translational efficiency during ageing could help to drive the collapse of proteostasis. Here we show that ageing alters the kinetics of translation elongation in both Caenorhabditis elegans and Saccharomyces cerevisiae. Ribosome pausing was exacerbated at specific positions in aged yeast and worms, including polybasic stretches, leading to increased ribosome collisions known to trigger ribosome-associated quality control (RQC)4-6. Notably, aged yeast cells exhibited impaired clearance and increased aggregation of RQC substrates, indicating that ageing overwhelms this pathway. Indeed, long-lived yeast mutants reduced age-dependent ribosome pausing, and extended lifespan correlated with greater flux through the RQC pathway. Further linking altered translation to proteostasis collapse, we found that nascent polypeptides exhibiting age-dependent ribosome pausing in C. elegans were strongly enriched among age-dependent protein aggregates. Notably, ageing increased the pausing and aggregation of many components of proteostasis, which could initiate a cycle of proteostasis collapse. We propose that increased ribosome pausing, leading to RQC overload and nascent polypeptide aggregation, critically contributes to proteostasis impairment and systemic decline during ageing.


Asunto(s)
Proteostasis , Proteínas de Saccharomyces cerevisiae , Envejecimiento , Animales , Caenorhabditis elegans/metabolismo , Péptidos/metabolismo , Biosíntesis de Proteínas , Ribosomas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
13.
Mol Cell ; 78(2): 329-345.e9, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32268122

RESUMEN

Neural stem and progenitor cells (NSPCs) are critical for continued cellular replacement in the adult brain. Lifelong maintenance of a functional NSPC pool necessitates stringent mechanisms to preserve a pristine proteome. We find that the NSPC chaperone network robustly maintains misfolded protein solubility and stress resilience through high levels of the ATP-dependent chaperonin TRiC/CCT. Strikingly, NSPC differentiation rewires the cellular chaperone network, reducing TRiC/CCT levels and inducing those of the ATP-independent small heat shock proteins (sHSPs). This switches the proteostasis strategy in neural progeny cells to promote sequestration of misfolded proteins into protective inclusions. The chaperone network of NSPCs is more effective than that of differentiated cells, leading to improved management of proteotoxic stress and amyloidogenic proteins. However, NSPC proteostasis is impaired by brain aging. The less efficient chaperone network of differentiated neural progeny may contribute to their enhanced susceptibility to neurodegenerative diseases characterized by aberrant protein misfolding and aggregation.


Asunto(s)
Envejecimiento/genética , Chaperonas Moleculares/genética , Células-Madre Neurales/metabolismo , Agregación Patológica de Proteínas/genética , Adenosina Trifosfato/genética , Envejecimiento/patología , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Chaperoninas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Ratones , Chaperonas Moleculares/metabolismo , Células-Madre Neurales/patología , Pliegue de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Células Madre/metabolismo , Células Madre/patología
14.
Cell ; 150(2): 413-25, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22817900

RESUMEN

Protein function is often regulated by posttranslational modifications (PTMs), and recent advances in mass spectrometry have resulted in an exponential increase in PTM identification. However, the functional significance of the vast majority of these modifications remains unknown. To address this problem, we compiled nearly 200,000 phosphorylation, acetylation, and ubiquitination sites from 11 eukaryotic species, including 2,500 newly identified ubiquitylation sites for Saccharomyces cerevisiae. We developed methods to prioritize the functional relevance of these PTMs by predicting those that likely participate in cross-regulatory events, regulate domain activity, or mediate protein-protein interactions. PTM conservation within domain families identifies regulatory "hot spots" that overlap with functionally important regions, a concept that we experimentally validated on the HSP70 domain family. Finally, our analysis of the evolution of PTM regulation highlights potential routes for neutral drift in regulatory interactions and suggests that only a fraction of modification sites are likely to have a significant biological role.


Asunto(s)
Eucariontes/metabolismo , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Procesamiento Proteico-Postraduccional , Acetilación , Secuencia de Aminoácidos , Animales , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Ubiquitinación
15.
Nature ; 596(7873): 558-564, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34408324

RESUMEN

Viral pathogens are an ongoing threat to public health worldwide. Analysing their dependence on host biosynthetic pathways could lead to effective antiviral therapies1. Here we integrate proteomic analyses of polysomes with functional genomics and pharmacological interventions to define how enteroviruses and flaviviruses remodel host polysomes to synthesize viral proteins and disable host protein production. We find that infection with polio, dengue or Zika virus markedly modifies polysome composition, without major changes to core ribosome stoichiometry. These viruses use different strategies to evict a common set of translation initiation and RNA surveillance factors from polysomes while recruiting host machineries that are specifically required for viral biogenesis. Targeting these specialized viral polysomes could provide a new approach for antiviral interventions. For example, we find that both Zika and dengue use the collagen proline hydroxylation machinery to mediate cotranslational modification of conserved proline residues in the viral polyprotein. Genetic or pharmacological inhibition of proline hydroxylation impairs nascent viral polyprotein folding and induces its aggregation and degradation. Notably, such interventions prevent viral polysome remodelling and lower virus production. Our findings delineate the modular nature of polysome specialization at the virus-host interface and establish a powerful strategy to identify targets for selective antiviral interventions.


Asunto(s)
Flavivirus/crecimiento & desarrollo , Flavivirus/metabolismo , Interacciones Huésped-Patógeno , Hidroxilación , Procolágeno-Prolina Dioxigenasa/metabolismo , Prolina/metabolismo , Biosíntesis de Proteínas , Línea Celular , Colágeno/química , Colágeno/metabolismo , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Flavivirus/química , Regulación Viral de la Expresión Génica , Genómica , Factores Celulares Derivados del Huésped/antagonistas & inhibidores , Factores Celulares Derivados del Huésped/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Sitios Internos de Entrada al Ribosoma , Chaperonas Moleculares/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Poliovirus/genética , Poliovirus/crecimiento & desarrollo , Polirribosomas/química , Polirribosomas/metabolismo , Agregado de Proteínas , Pliegue de Proteína , Mapas de Interacción de Proteínas , Proteolisis , Proteómica , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
16.
Mol Cell ; 75(6): 1117-1130.e5, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31400849

RESUMEN

Cotranslational protein folding requires assistance from elaborate ribosome-associated chaperone networks. It remains unclear how the changing information in a growing nascent polypeptide dictates the recruitment of functionally distinct chaperones. Here, we used ribosome profiling to define the principles governing the cotranslational action of the chaperones TRiC/CCT and Hsp70/Ssb. We show that these chaperones are sequentially recruited to specific sites within domain-encoding regions of select nascent polypeptides. Hsp70 associates first, binding select sites throughout domains, whereas TRiC associates later, upon the emergence of nearly complete domains that expose an unprotected hydrophobic surface. This suggests that transient topological properties of nascent folding intermediates drive sequential chaperone association. Moreover, cotranslational recruitment of both TRiC and Hsp70 correlated with translation elongation slowdowns. We propose that the temporal modulation of the nascent chain structural landscape is coordinated with local elongation rates to regulate the hierarchical action of Hsp70 and TRiC for cotranslational folding.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Extensión de la Cadena Peptídica de Translación/fisiología , Pliegue de Proteína , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Ribosomas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
17.
Mol Cell ; 74(4): 729-741.e7, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30982745

RESUMEN

The nascent polypeptide-associated complex (NAC) is a conserved ribosome-associated protein biogenesis factor. Whether NAC exerts chaperone activity and whether this function is restricted to de novo protein synthesis is unknown. Here, we demonstrate that NAC directly exerts chaperone activity toward structurally diverse model substrates including polyglutamine (PolyQ) proteins, firefly luciferase, and Aß40. Strikingly, we identified the positively charged ribosome-binding domain in the N terminus of the ßNAC subunit (N-ßNAC) as a major chaperone entity of NAC. N-ßNAC by itself suppressed aggregation of PolyQ-expanded proteins in vitro, and the positive charge of this domain was critical for this activity. Moreover, we found that NAC also exerts a ribosome-independent chaperone function in vivo. Consistently, we found that a substantial fraction of NAC is non-ribosomal bound in higher eukaryotes. In sum, NAC is a potent suppressor of aggregation and proteotoxicity of mutant PolyQ-expanded proteins associated with human diseases like Huntington's disease and spinocerebellar ataxias.


Asunto(s)
Péptidos beta-Amiloides/genética , Chaperonas Moleculares/genética , Agregación Patológica de Proteínas/genética , Péptidos beta-Amiloides/química , Sitios de Unión/genética , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Luciferasas/química , Luciferasas/genética , Chaperonas Moleculares/química , Péptidos/química , Péptidos/genética , Unión Proteica/genética , Biosíntesis de Proteínas/genética , Dominios Proteicos/genética , Pliegue de Proteína , Ribosomas/genética , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
18.
Genes Dev ; 33(15-16): 1027-1030, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371460

RESUMEN

Gene dosage alterations caused by aneuploidy are a common feature of most cancers yet pose severe proteotoxic challenges. Therefore, cells have evolved various dosage compensation mechanisms to limit the damage caused by the ensuing protein level imbalances. For instance, for heteromeric protein complexes, excess nonstoichiometric subunits are rapidly recognized and degraded. In this issue of Genes & Development, Brennan et al. (pp. 1031-1047) reveal that sequestration of nonstoichiometric subunits into aggregates is an alternative mechanism for dosage compensation in aneuploid budding yeast and human cell lines. Using a combination of proteomic and genetic techniques, they found that excess proteins undergo either degradation or aggregation but not both. Which route is preferred depends on the half-life of the protein in question. Given the multitude of diseases linked to either aneuploidy or protein aggregation, this study could serve as a springboard for future studies with broad-spanning implications.


Asunto(s)
Agregado de Proteínas , Proteómica , Aneuploidia , Compensación de Dosificación (Genética) , Humanos
19.
PLoS Pathog ; 20(2): e1011535, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38335237

RESUMEN

A better mechanistic understanding of virus-host dependencies can help reveal vulnerabilities and identify opportunities for therapeutic intervention. Of particular interest are essential interactions that enable production of viral proteins, as those could target an early step in the virus lifecycle. Here, we use subcellular proteomics, ribosome profiling analyses and reporter assays to detect changes in protein synthesis dynamics during SARS-CoV-2 (CoV2) infection. We identify specific translation factors and molecular chaperones that are used by CoV2 to promote the synthesis and maturation of its own proteins. These can be targeted to inhibit infection, without major toxicity to the host. We also find that CoV2 non-structural protein 1 (Nsp1) cooperates with initiation factors EIF1 and 1A to selectively enhance translation of viral RNA. When EIF1/1A are depleted, more ribosomes initiate translation from a conserved upstream CUG start codon found in all genomic and subgenomic viral RNAs. This results in higher translation of an upstream open reading frame (uORF1) and lower translation of the main ORF, altering the stoichiometry of viral proteins and attenuating infection. Replacing the upstream CUG with AUG strongly inhibits translation of the main ORF independently of Nsp1, EIF1, or EIF1A. Taken together, our work describes multiple dependencies of CoV2 on host biosynthetic networks and proposes a model for dosage control of viral proteins through Nsp1-mediated control of translation start site selection.


Asunto(s)
COVID-19 , ARN Viral , Humanos , ARN Viral/genética , SARS-CoV-2/genética , COVID-19/genética , Factores de Iniciación de Péptidos , Proteínas Virales
20.
Cell ; 144(2): 240-52, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21241893

RESUMEN

Group II chaperonins are ATP-dependent ring-shaped complexes that bind nonnative polypeptides and facilitate protein folding in archaea and eukaryotes. A built-in lid encapsulates substrate proteins within the central chaperonin chamber. Here, we describe the fate of the substrate during the nucleotide cycle of group II chaperonins. The chaperonin substrate-binding sites are exposed, and the lid is open in both the ATP-free and ATP-bound prehydrolysis states. ATP hydrolysis has a dual function in the folding cycle, triggering both lid closure and substrate release into the central chamber. Notably, substrate release can occur in the absence of a lid, and lid closure can occur without substrate release. However, productive folding requires both events, so that the polypeptide is released into the confined space of the closed chamber where it folds. Our results show that ATP hydrolysis coordinates the structural and functional determinants that trigger productive folding.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas Arqueales/metabolismo , Chaperoninas del Grupo II/metabolismo , Methanococcus/metabolismo , Pliegue de Proteína , Adenosina Trifosfatasas/metabolismo , Regulación Alostérica , Proteínas Arqueales/química , Sitios de Unión , Chaperoninas del Grupo II/química , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA