Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.045
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(16): 4284-4298.e27, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34233164

RESUMEN

Many organisms evolved strategies to survive desiccation. Plant seeds protect dehydrated embryos from various stressors and can lay dormant for millennia. Hydration is the key trigger to initiate germination, but the mechanism by which seeds sense water remains unresolved. We identified an uncharacterized Arabidopsis thaliana prion-like protein we named FLOE1, which phase separates upon hydration and allows the embryo to sense water stress. We demonstrate that biophysical states of FLOE1 condensates modulate its biological function in vivo in suppressing seed germination under unfavorable environments. We find intragenic, intraspecific, and interspecific natural variation in FLOE1 expression and phase separation and show that intragenic variation is associated with adaptive germination strategies in natural populations. This combination of molecular, organismal, and ecological studies uncovers FLOE1 as a tunable environmental sensor with direct implications for the design of drought-resistant crops, in the face of climate change.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/crecimiento & desarrollo , Germinación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Priones/metabolismo , Semillas/crecimiento & desarrollo , Agua/metabolismo , Arabidopsis/genética , Arabidopsis/ultraestructura , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/ultraestructura , Deshidratación , Imagenología Tridimensional , Péptidos y Proteínas de Señalización Intercelular/química , Mutación/genética , Latencia en las Plantas , Plantas Modificadas Genéticamente , Dominios Proteicos , Isoformas de Proteínas/metabolismo , Semillas/ultraestructura
2.
Cell ; 180(5): 928-940.e14, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109413

RESUMEN

Covalent modifications to histones are essential for development, establishing distinct and functional chromatin domains from a common genetic sequence. Whereas repressed chromatin is robustly inherited, no mechanism that facilitates inheritance of an activated domain has been described. Here, we report that the Set3C histone deacetylase scaffold Snt1 can act as a prion that drives the emergence and transgenerational inheritance of an activated chromatin state. This prion, which we term [ESI+] for expressed sub-telomeric information, is triggered by transient Snt1 phosphorylation upon cell cycle arrest. Once engaged, the prion reshapes the activity of Snt1 and the Set3C complex, recruiting RNA pol II and interfering with Rap1 binding to activate genes in otherwise repressed sub-telomeric domains. This transcriptional state confers broad resistance to environmental stress, including antifungal drugs. Altogether, our results establish a robust means by which a prion can facilitate inheritance of an activated chromatin state to provide adaptive benefit.


Asunto(s)
Cromatina/genética , Histona Desacetilasas/genética , Proteínas Nucleares/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Unión a Telómeros/genética , Factores de Transcripción/genética , Puntos de Control del Ciclo Celular/genética , Código de Histonas/genética , Histonas/genética , Fosforilación/genética , Priones/genética , ARN Polimerasa II/genética , Saccharomyces cerevisiae , Complejo Shelterina , Telómero/genética , Transcripción Genética
3.
Cell ; 178(6): 1403-1420.e21, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491385

RESUMEN

Prion-like proteins can assume distinct conformational and physical states in the same cell. Sequence analysis suggests that prion-like proteins are prevalent in various species; however, it remains unclear what functional space they occupy in multicellular organisms. Here, we report the identification of a prion-like protein, Herzog (CG5830), through a multimodal screen in Drosophila melanogaster. Herzog functions as a membrane-associated phosphatase and controls embryonic patterning, likely being involved in TGF-ß/BMP and FGF/EGF signaling pathways. Remarkably, monomeric Herzog is enzymatically inactive and becomes active upon amyloid-like assembly. The prion-like domain of Herzog is necessary for both its assembly and membrane targeting. Removal of the prion-like domain impairs activity, while restoring assembly on the membrane using a heterologous prion-like domain and membrane-targeting motif can restore phosphatase activity. This study provides an example of a prion-like domain that allows an enzyme to gain essential functionality via amyloid-like assembly to control animal development.


Asunto(s)
Proteínas Amiloidogénicas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Desarrollo Embrionario , Fosfoproteínas Fosfatasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/genética , Animales , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/genética , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/genética , Priones/química , Dominios Proteicos
4.
Cell ; 174(3): 688-699.e16, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-29961577

RESUMEN

Proteins such as FUS phase separate to form liquid-like condensates that can harden into less dynamic structures. However, how these properties emerge from the collective interactions of many amino acids remains largely unknown. Here, we use extensive mutagenesis to identify a sequence-encoded molecular grammar underlying the driving forces of phase separation of proteins in the FUS family and test aspects of this grammar in cells. Phase separation is primarily governed by multivalent interactions among tyrosine residues from prion-like domains and arginine residues from RNA-binding domains, which are modulated by negatively charged residues. Glycine residues enhance the fluidity, whereas glutamine and serine residues promote hardening. We develop a model to show that the measured saturation concentrations of phase separation are inversely proportional to the product of the numbers of arginine and tyrosine residues. These results suggest it is possible to predict phase-separation properties based on amino acid sequences.


Asunto(s)
Proteína FUS de Unión a ARN/genética , Proteínas de Unión al ARN/fisiología , Secuencia de Aminoácidos , Aminoácidos/química , Animales , Arginina/química , Simulación por Computador , Células HeLa , Humanos , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/fisiología , Transición de Fase , Proteínas Priónicas/química , Proteínas Priónicas/genética , Priones/genética , Priones/fisiología , Dominios Proteicos , Proteína FUS de Unión a ARN/fisiología , Proteínas de Unión al ARN/aislamiento & purificación , Células Sf9 , Tirosina/química
5.
Cell ; 173(3): 677-692.e20, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29677512

RESUMEN

RNA-binding proteins (RBPs) with prion-like domains (PrLDs) phase transition to functional liquids, which can mature into aberrant hydrogels composed of pathological fibrils that underpin fatal neurodegenerative disorders. Several nuclear RBPs with PrLDs, including TDP-43, FUS, hnRNPA1, and hnRNPA2, mislocalize to cytoplasmic inclusions in neurodegenerative disorders, and mutations in their PrLDs can accelerate fibrillization and cause disease. Here, we establish that nuclear-import receptors (NIRs) specifically chaperone and potently disaggregate wild-type and disease-linked RBPs bearing a NLS. Karyopherin-ß2 (also called Transportin-1) engages PY-NLSs to inhibit and reverse FUS, TAF15, EWSR1, hnRNPA1, and hnRNPA2 fibrillization, whereas Importin-α plus Karyopherin-ß1 prevent and reverse TDP-43 fibrillization. Remarkably, Karyopherin-ß2 dissolves phase-separated liquids and aberrant fibrillar hydrogels formed by FUS and hnRNPA1. In vivo, Karyopherin-ß2 prevents RBPs with PY-NLSs accumulating in stress granules, restores nuclear RBP localization and function, and rescues degeneration caused by disease-linked FUS and hnRNPA2. Thus, NIRs therapeutically restore RBP homeostasis and mitigate neurodegeneration.


Asunto(s)
Transporte Activo de Núcleo Celular , Priones/química , Proteínas de Unión al ARN/química , Receptores Citoplasmáticos y Nucleares/química , Adulto , Anciano , Animales , Citoplasma/química , Proteínas de Unión al ADN/química , Drosophila melanogaster , Femenino , Proteínas Fluorescentes Verdes/química , Células HEK293 , Células HeLa , Homeostasis , Humanos , Carioferinas/química , Masculino , Persona de Mediana Edad , Chaperonas Moleculares/química , Mutación , Enfermedades Neurodegenerativas/patología , Dominios Proteicos , Proteína EWS de Unión a ARN/química , Factores Asociados con la Proteína de Unión a TATA/química , beta Carioferinas/química
6.
Cell ; 171(1): 30-31, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28938120

RESUMEN

Prion-like domains have emerged as important drivers of neurodegenerative disease. Now, Boulay et al. establish that the translocated prion-like domain of the oncogenic EWS-FLI1 fusion protein enables phase-separation events, which inappropriately recruit chromatin-remodeling factors to elicit the aberrant transcriptional programs underlying Ewing's sarcoma.


Asunto(s)
Proteína EWS de Unión a ARN , Sarcoma de Ewing , Humanos , Proteínas de Fusión Oncogénica , Priones , Proteína Proto-Oncogénica c-fli-1 , Factores de Transcripción
7.
Cell ; 171(5): 1001-1014, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29149602

RESUMEN

Protein conformational states-from intrinsically disordered ensembles to amyloids that underlie the self-templating, infectious properties of prion-like proteins-have attracted much attention. Here, we highlight the diversity, including differences in biophysical properties, that drive distinct biological functions and pathologies among self-templating proteins. Advances in chemical genomics, gene editing, and model systems now permit deconstruction of the complex interplay between these protein states and the host factors that react to them. These methods reveal that conformational switches modulate normal and abnormal information transfer and that intimate relationships exist between the intrinsic function of proteins and the deleterious consequences of their misfolding.


Asunto(s)
Priones/química , Conformación Proteica , Amiloide/química , Amiloide/metabolismo , Animales , Humanos , Modelos Animales , Enfermedades Neurodegenerativas/metabolismo , Priones/metabolismo , Saccharomyces cerevisiae/metabolismo
8.
Cell ; 171(4): 966-979.e18, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29056345

RESUMEN

Protein aggregation is a hallmark of many diseases but also underlies a wide range of positive cellular functions. This phenomenon has been difficult to study because of a lack of quantitative and high-throughput cellular tools. Here, we develop a synthetic genetic tool to sense and control protein aggregation. We apply the technology to yeast prions, developing sensors to track their aggregation states and employing prion fusions to encode synthetic memories in yeast cells. Utilizing high-throughput screens, we identify prion-curing mutants and engineer "anti-prion drives" that reverse the non-Mendelian inheritance pattern of prions and eliminate them from yeast populations. We extend our technology to yeast RNA-binding proteins (RBPs) by tracking their propensity to aggregate, searching for co-occurring aggregates, and uncovering a group of coalescing RBPs through screens enabled by our platform. Our work establishes a quantitative, high-throughput, and generalizable technology to study and control diverse protein aggregation processes in cells.


Asunto(s)
Técnicas Genéticas , Priones/genética , Ingeniería Genética , Técnicas Genéticas/economía , Proteínas de Unión al ARN/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Biología Sintética/métodos , Factores de Escisión y Poliadenilación de ARNm/metabolismo
9.
Cell ; 165(5): 1055-1066, 2016 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-27203110

RESUMEN

We here attempt to achieve an integrated understanding of the structure and dynamics of a number of higher-order assemblies, including amyloids, various kinds of signalosomes, and cellular granules. We propose that the synergy between folded domains, linear motifs, and intrinsically disordered regions regulates the formation and intrinsic fuzziness of all higher-order assemblies, creating a structural and dynamic continuum. We describe how such regulatory mechanisms could be influenced under pathological conditions.


Asunto(s)
Orgánulos/química , Orgánulos/metabolismo , Amiloide/química , Amiloide/metabolismo , Humanos , Complejos Multiproteicos/química , Priones/química , Priones/metabolismo , Estructura Cuaternaria de Proteína , Transducción de Señal
11.
Cell ; 167(2): 369-381.e12, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27693355

RESUMEN

Prions are a paradigm-shifting mechanism of inheritance in which phenotypes are encoded by self-templating protein conformations rather than nucleic acids. Here, we examine the breadth of protein-based inheritance across the yeast proteome by assessing the ability of nearly every open reading frame (ORF; ∼5,300 ORFs) to induce heritable traits. Transient overexpression of nearly 50 proteins created traits that remained heritable long after their expression returned to normal. These traits were beneficial, had prion-like patterns of inheritance, were common in wild yeasts, and could be transmitted to naive cells with protein alone. Most inducing proteins were not known prions and did not form amyloid. Instead, they are highly enriched in nucleic acid binding proteins with large intrinsically disordered domains that have been widely conserved across evolution. Thus, our data establish a common type of protein-based inheritance through which intrinsically disordered proteins can drive the emergence of new traits and adaptive opportunities.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/metabolismo , Carácter Cuantitativo Heredable , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Amiloide/metabolismo , Evolución Molecular , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Sistemas de Lectura Abierta , Priones/química , Priones/metabolismo , Proteoma , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
12.
Cell ; 166(3): 637-650, 2016 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-27471966

RESUMEN

Most vertebrate oocytes contain a Balbiani body, a large, non-membrane-bound compartment packed with RNA, mitochondria, and other organelles. Little is known about this compartment, though it specifies germline identity in many non-mammalian vertebrates. We show Xvelo, a disordered protein with an N-terminal prion-like domain, is an abundant constituent of Xenopus Balbiani bodies. Disruption of the prion-like domain of Xvelo, or substitution with a prion-like domain from an unrelated protein, interferes with its incorporation into Balbiani bodies in vivo. Recombinant Xvelo forms amyloid-like networks in vitro. Amyloid-like assemblies of Xvelo recruit both RNA and mitochondria in binding assays. We propose that Xenopus Balbiani bodies form by amyloid-like assembly of Xvelo, accompanied by co-recruitment of mitochondria and RNA. Prion-like domains are found in germ plasm organizing proteins in other species, suggesting that Balbiani body formation by amyloid-like assembly could be a conserved mechanism that helps oocytes function as long-lived germ cells.


Asunto(s)
Amiloide/metabolismo , Biogénesis de Organelos , Proteínas de Dominio T Box/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Benzotiazoles , Femenino , Colorantes Fluorescentes , Mitocondrias/metabolismo , Oocitos/citología , Orgánulos/metabolismo , Priones/química , Dominios Proteicos , Transporte de Proteínas , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Sf9 , Proteínas de Dominio T Box/química , Proteínas de Dominio T Box/genética , Tiazoles , Proteínas de Xenopus/química , Proteínas de Xenopus/genética , Xenopus laevis , Pez Cebra
13.
Cell ; 163(3): 620-8, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26456111

RESUMEN

Biological processes occur in complex environments containing a myriad of potential interactors. Unfortunately, limitations on the sensitivity of biophysical techniques normally restrict structural investigations to purified systems, at concentrations that are orders of magnitude above endogenous levels. Dynamic nuclear polarization (DNP) can dramatically enhance the sensitivity of nuclear magnetic resonance (NMR) spectroscopy and enable structural studies in biologically complex environments. Here, we applied DNP NMR to investigate the structure of a protein containing both an environmentally sensitive folding pathway and an intrinsically disordered region, the yeast prion protein Sup35. We added an exogenously prepared isotopically labeled protein to deuterated lysates, rendering the biological environment "invisible" and enabling highly efficient polarization transfer for DNP. In this environment, structural changes occurred in a region known to influence biological activity but intrinsically disordered in purified samples. Thus, DNP makes structural studies of proteins at endogenous levels in biological contexts possible, and such contexts can influence protein structure.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular/métodos , Factores de Terminación de Péptidos/química , Priones/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Factores de Terminación de Péptidos/metabolismo , Priones/metabolismo , Pliegue de Proteína , Estructura Secundaria de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo
14.
Cell ; 162(5): 1066-77, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26317470

RESUMEN

Many proteins contain disordered regions of low-sequence complexity, which cause aging-associated diseases because they are prone to aggregate. Here, we study FUS, a prion-like protein containing intrinsically disordered domains associated with the neurodegenerative disease ALS. We show that, in cells, FUS forms liquid compartments at sites of DNA damage and in the cytoplasm upon stress. We confirm this by reconstituting liquid FUS compartments in vitro. Using an in vitro "aging" experiment, we demonstrate that liquid droplets of FUS protein convert with time from a liquid to an aggregated state, and this conversion is accelerated by patient-derived mutations. We conclude that the physiological role of FUS requires forming dynamic liquid-like compartments. We propose that liquid-like compartments carry the trade-off between functionality and risk of aggregation and that aberrant phase transitions within liquid-like compartments lie at the heart of ALS and, presumably, other age-related diseases.


Asunto(s)
Envejecimiento/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Mutación , Proteína FUS de Unión a ARN/química , Proteína FUS de Unión a ARN/genética , Envejecimiento/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Núcleo Celular/química , Citoplasma/química , Humanos , Priones/química , Agregado de Proteínas , Estructura Terciaria de Proteína , Proteína FUS de Unión a ARN/metabolismo
15.
Mol Cell ; 82(22): 4290-4306.e11, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36272412

RESUMEN

Manifestation of aggregate pathology in Huntington's disease is thought to be facilitated by a preferential vulnerability of affected brain cells to age-dependent proteostatic decline. To understand how specific cellular backgrounds may facilitate pathologic aggregation, we utilized the yeast model in which polyQ-expanded Huntingtin forms aggregates only when the endogenous prion-forming protein Rnq1 is in its amyloid-like prion [PIN+] conformation. We employed optogenetic clustering of polyQ protein as an orthogonal method to induce polyQ aggregation in prion-free [pin-] cells. Optogenetic aggregation circumvented the prion requirement for the formation of detergent-resistant polyQ inclusions but bypassed the formation of toxic polyQ oligomers, which accumulated specifically in [PIN+] cells. Reconstitution of aggregation in vitro suggested that these polyQ oligomers formed through direct templating on Rnq1 prions. These findings shed light on the mechanism of prion-mediated formation of oligomers, which may play a role in triggering polyQ pathology in the patient brain.


Asunto(s)
Priones , Proteínas de Saccharomyces cerevisiae , Humanos , Priones/genética , Priones/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Péptidos/genética , Péptidos/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
16.
Nat Immunol ; 18(2): 214-224, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27992402

RESUMEN

The signaling adaptor MAVS forms prion-like aggregates to activate an innate antiviral immune response after viral infection. However, the molecular mechanisms that regulate MAVS aggregation are poorly understood. Here we identified TRIM31, an E3 ubiquitin ligase of the TRIM family of proteins, as a regulator of MAVS aggregation. TRIM31 was recruited to mitochondria after viral infection and specifically regulated antiviral signaling mediated by RLR pattern-recognition receptors. TRIM31-deficient mice were more susceptible to infection with RNA virus than were wild-type mice. TRIM31 interacted with MAVS and catalyzed the Lys63 (K63)-linked polyubiquitination of Lys10, Lys311 and Lys461 on MAVS. This modification promoted the formation of prion-like aggregates of MAVS after viral infection. Our findings reveal new insights in the molecular regulation of MAVS aggregation and the cellular antiviral response through TRIM31-mediated K63-linked polyubiquitination of MAVS.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Macrófagos/fisiología , Proteínas Nucleares/metabolismo , Priones/inmunología , Virosis/inmunología , Animales , Proteínas Portadoras/genética , Células Cultivadas , Inmunidad Innata/genética , Lisina/genética , Lisina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Agregación de Receptores/genética , Transducción de Señal/genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Ubiquitinación/genética
17.
Immunity ; 53(6): 1123-1125, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326760

RESUMEN

EBF1 is a pioneer transcription factor involved in B lymphocyte specification. In this issue of Immunity, Wang et al. localize EBF1's pioneering activity to a prion-like domain that mediates recruitment of the nucleosome remodeler Brg1 and FUS-assisted liquid-liquid phase separation.


Asunto(s)
Cromatina , Priones , Linfocitos B/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Immunity ; 53(6): 1151-1167.e6, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33159853

RESUMEN

Establishment of B-lineage-specific gene expression requires the binding of transcription factors to inaccessible chromatin of progenitors. The transcription factor EBF1 can bind genomic regions prior to the detection of chromatin accessibility in a manner dependent on EBF1's C-terminal domain (CTD) and independent of cooperating transcription factors. Here, we studied the mechanism whereby the CTD enables this pioneering function. The CTD of EBF1 was dispensable for initial chromatin targeting but stabilized occupancy via recruitment of the chromatin remodeler Brg1. We found that the CTD harbors a prion-like domain (PLD) with an ability of liquid-liquid phase separation, which was enhanced by interaction of EBF1 with the RNA-binding protein FUS. Brg1 also partitioned into phase-separated FUS condensates and coincided with EBF1 and FUS foci in pro-B cells. Heterologous PLDs conferred pioneering function on EBF1ΔCTD. Thus, the phase separation ability of EBF1 facilitates Brg1-mediated chromatin opening and the transition of naive progenitor chromatin to B-lineage-committed chromatin.


Asunto(s)
Linfocitos B/metabolismo , Cromatina/metabolismo , Priones/química , Transactivadores/metabolismo , Secuencia de Aminoácidos , Linfocitos B/citología , ADN Helicasas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Transición de Fase , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Dominios Proteicos , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/química , Factores de Transcripción/metabolismo
19.
Cell ; 158(3): 522-33, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083867

RESUMEN

Proteins destined for the cell surface are first assessed in the endoplasmic reticulum (ER) for proper folding before release into the secretory pathway. This ensures that defective proteins are normally prevented from entering the extracellular environment, where they could be disruptive. Here, we report that, when ER folding capacity is saturated during stress, misfolded glycosylphosphatidylinositol-anchored proteins dissociate from resident ER chaperones, engage export receptors, and quantitatively leave the ER via vesicular transport to the Golgi. Clearance from the ER commences within minutes of acute ER stress, before the transcriptional component of the unfolded protein response is activated. These aberrant proteins then access the cell surface transiently before destruction in lysosomes. Inhibiting this stress-induced pathway by depleting the ER-export receptors leads to aggregation of the ER-retained misfolded protein. Thus, this rapid response alleviates the elevated burden of misfolded proteins in the ER at the onset of ER stress, promoting protein homeostasis in the ER.


Asunto(s)
Estrés del Retículo Endoplásmico , Lisosomas/metabolismo , Vías Secretoras , Animales , Línea Celular , Humanos , Ratones , Priones/metabolismo , Pliegue de Proteína , Ratas , Respuesta de Proteína Desplegada
20.
Cell ; 156(6): 1127-1129, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24630715

RESUMEN

Microbial and danger signals result in inflammasome activation and release of inflammatory cytokines through mechanisms that remain elusive. Cai et al. and Lu et al. show that triggering of inflammasome sensors induces prion-like polymerization of the adaptor ASC into filaments. These structures function as platforms for inflammatory cytokine production and represent a unified mechanism for inflammasome assembly.


Asunto(s)
Evolución Biológica , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Inmunidad Innata , Inflamasomas/química , Inflamasomas/inmunología , Priones/metabolismo , Transducción de Señal , Levaduras/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA