Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.489
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(13): 3262-3283.e23, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38815580

RESUMEN

In eukaryotes, the Suv39 family of proteins tri-methylate lysine 9 of histone H3 (H3K9me) to form constitutive heterochromatin. However, how Suv39 proteins are nucleated at heterochromatin is not fully described. In the fission yeast, current models posit that Argonaute1-associated small RNAs (sRNAs) nucleate the sole H3K9 methyltransferase, Clr4/SUV39H, to centromeres. Here, we show that in the absence of all sRNAs and H3K9me, the Mtl1 and Red1 core (MTREC)/PAXT complex nucleates Clr4/SUV39H at a heterochromatic long noncoding RNA (lncRNA) at which the two H3K9 deacetylases, Sir2 and Clr3, also accumulate by distinct mechanisms. Iterative cycles of H3K9 deacetylation and methylation spread Clr4/SUV39H from the nucleation center in an sRNA-independent manner, generating a basal H3K9me state. This is acted upon by the RNAi machinery to augment and amplify the Clr4/H3K9me signal at centromeres to establish heterochromatin. Overall, our data reveal that lncRNAs and RNA quality control factors can nucleate heterochromatin and function as epigenetic silencers in eukaryotes.


Asunto(s)
Proteínas de Ciclo Celular , Heterocromatina , N-Metiltransferasa de Histona-Lisina , Histonas , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metilación , Metiltransferasas/metabolismo , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/genética , Schizosaccharomyces/metabolismo , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , ARN de Hongos/genética , ARN Interferente Pequeño/genética
2.
Cell ; 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39094570

RESUMEN

Faithful transfer of parental histones to newly replicated daughter DNA strands is critical for inheritance of epigenetic states. Although replication proteins that facilitate parental histone transfer have been identified, how intact histone H3-H4 tetramers travel from the front to the back of the replication fork remains unknown. Here, we use AlphaFold-Multimer structural predictions combined with biochemical and genetic approaches to identify the Mrc1/CLASPIN subunit of the replisome as a histone chaperone. Mrc1 contains a conserved histone-binding domain that forms a brace around the H3-H4 tetramer mimicking nucleosomal DNA and H2A-H2B histones, is required for heterochromatin inheritance, and promotes parental histone recycling during replication. We further identify binding sites for the FACT histone chaperone in Swi1/TIMELESS and DNA polymerase α that are required for heterochromatin inheritance. We propose that Mrc1, in concert with FACT acting as a mobile co-chaperone, coordinates the distribution of parental histones to newly replicated DNA.

3.
Cell ; 186(22): 4936-4955.e26, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37788668

RESUMEN

Intrinsically disordered regions (IDRs) represent a large percentage of overall nuclear protein content. The prevailing dogma is that IDRs engage in non-specific interactions because they are poorly constrained by evolutionary selection. Here, we demonstrate that condensate formation and heterotypic interactions are distinct and separable features of an IDR within the ARID1A/B subunits of the mSWI/SNF chromatin remodeler, cBAF, and establish distinct "sequence grammars" underlying each contribution. Condensation is driven by uniformly distributed tyrosine residues, and partner interactions are mediated by non-random blocks rich in alanine, glycine, and glutamine residues. These features concentrate a specific cBAF protein-protein interaction network and are essential for chromatin localization and activity. Importantly, human disease-associated perturbations in ARID1B IDR sequence grammars disrupt cBAF function in cells. Together, these data identify IDR contributions to chromatin remodeling and explain how phase separation provides a mechanism through which both genomic localization and functional partner recruitment are achieved.


Asunto(s)
Ensamble y Desensamble de Cromatina , Complejos Multiproteicos , Proteínas Nucleares , Humanos , Cromatina , Proteínas de Unión al ADN/química , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo
4.
Cell ; 185(24): 4654-4673.e28, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36334589

RESUMEN

Brown adipose tissue (BAT) regulates metabolic physiology. However, nearly all mechanistic studies of BAT protein function occur in a single inbred mouse strain, which has limited the understanding of generalizable mechanisms of BAT regulation over physiology. Here, we perform deep quantitative proteomics of BAT across a cohort of 163 genetically defined diversity outbred mice, a model that parallels the genetic and phenotypic variation found in humans. We leverage this diversity to define the functional architecture of the outbred BAT proteome, comprising 10,479 proteins. We assign co-operative functions to 2,578 proteins, enabling systematic discovery of regulators of BAT. We also identify 638 proteins that correlate with protection from, or sensitivity to, at least one parameter of metabolic disease. We use these findings to uncover SFXN5, LETMD1, and ATP1A2 as modulators of BAT thermogenesis or adiposity, and provide OPABAT as a resource for understanding the conserved mechanisms of BAT regulation over metabolic physiology.


Asunto(s)
Tejido Adiposo Pardo , Proteoma , Humanos , Ratones , Animales , Tejido Adiposo Pardo/metabolismo , Proteoma/metabolismo , Termogénesis/fisiología , Adiposidad , Obesidad/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/metabolismo
5.
Cell ; 184(11): 3022-3040.e28, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33961781

RESUMEN

Thousands of interactions assemble proteins into modules that impart spatial and functional organization to the cellular proteome. Through affinity-purification mass spectrometry, we have created two proteome-scale, cell-line-specific interaction networks. The first, BioPlex 3.0, results from affinity purification of 10,128 human proteins-half the proteome-in 293T cells and includes 118,162 interactions among 14,586 proteins. The second results from 5,522 immunoprecipitations in HCT116 cells. These networks model the interactome whose structure encodes protein function, localization, and complex membership. Comparison across cell lines validates thousands of interactions and reveals extensive customization. Whereas shared interactions reside in core complexes and involve essential proteins, cell-specific interactions link these complexes, "rewiring" subnetworks within each cell's interactome. Interactions covary among proteins of shared function as the proteome remodels to produce each cell's phenotype. Viewable interactively online through BioPlexExplorer, these networks define principles of proteome organization and enable unknown protein characterization.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas/genética , Proteoma/genética , Biología Computacional/métodos , Células HCT116/metabolismo , Células HEK293/metabolismo , Humanos , Espectrometría de Masas/métodos , Mapas de Interacción de Proteínas/fisiología , Proteoma/metabolismo , Proteómica/métodos
6.
Cell ; 180(5): 968-983.e24, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109415

RESUMEN

Mammalian tissues engage in specialized physiology that is regulated through reversible modification of protein cysteine residues by reactive oxygen species (ROS). ROS regulate a myriad of biological processes, but the protein targets of ROS modification that drive tissue-specific physiology in vivo are largely unknown. Here, we develop Oximouse, a comprehensive and quantitative mapping of the mouse cysteine redox proteome in vivo. We use Oximouse to establish several paradigms of physiological redox signaling. We define and validate cysteine redox networks within each tissue that are tissue selective and underlie tissue-specific biology. We describe a common mechanism for encoding cysteine redox sensitivity by electrostatic gating. Moreover, we comprehensively identify redox-modified disease networks that remodel in aged mice, establishing a systemic molecular basis for the long-standing proposed links between redox dysregulation and tissue aging. We provide the Oximouse compendium as a framework for understanding mechanisms of redox regulation in physiology and aging.


Asunto(s)
Envejecimiento/genética , Cisteína/genética , Proteínas/genética , Proteoma/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Cisteína/metabolismo , Humanos , Ratones , Especificidad de Órganos/genética , Oxidación-Reducción , Estrés Oxidativo/genética , Proteómica/métodos , Especies Reactivas de Oxígeno , Transducción de Señal/genética
7.
Cell ; 183(7): 1848-1866.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301708

RESUMEN

Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.


Asunto(s)
Inmunidad , Neoplasias/inmunología , Neoplasias/metabolismo , Obesidad/metabolismo , Microambiente Tumoral , Adiposidad , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Células HEK293 , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Cinética , Linfocitos Infiltrantes de Tumor , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Análisis de Componente Principal , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteómica
8.
Cell ; 183(1): 62-75.e17, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32946811

RESUMEN

In response to skeletal muscle contraction during exercise, paracrine factors coordinate tissue remodeling, which underlies this healthy adaptation. Here we describe a pH-sensing metabolite signal that initiates muscle remodeling upon exercise. In mice and humans, exercising skeletal muscle releases the mitochondrial metabolite succinate into the local interstitium and circulation. Selective secretion of succinate is facilitated by its transient protonation, which occurs upon muscle cell acidification. In the protonated monocarboxylic form, succinate is rendered a transport substrate for monocarboxylate transporter 1, which facilitates pH-gated release. Upon secretion, succinate signals via its cognate receptor SUCNR1 in non-myofibrillar cells in muscle tissue to control muscle-remodeling transcriptional programs. This succinate-SUCNR1 signaling is required for paracrine regulation of muscle innervation, muscle matrix remodeling, and muscle strength in response to exercise training. In sum, we define a bioenergetic sensor in muscle that utilizes intracellular pH and succinate to coordinate tissue adaptation to exercise.


Asunto(s)
Músculo Esquelético/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ácido Succínico/metabolismo , Animales , Humanos , Concentración de Iones de Hidrógeno , Inflamación/metabolismo , Ratones , Mitocondrias/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Contracción Muscular , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal , Succinatos/metabolismo , Simportadores/metabolismo
9.
Cell ; 180(2): 387-402.e16, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31978347

RESUMEN

Proteins are essential agents of biological processes. To date, large-scale profiling of cell line collections including the Cancer Cell Line Encyclopedia (CCLE) has focused primarily on genetic information whereas deep interrogation of the proteome has remained out of reach. Here, we expand the CCLE through quantitative profiling of thousands of proteins by mass spectrometry across 375 cell lines from diverse lineages to reveal information undiscovered by DNA and RNA methods. We observe unexpected correlations within and between pathways that are largely absent from RNA. An analysis of microsatellite instable (MSI) cell lines reveals the dysregulation of specific protein complexes associated with surveillance of mutation and translation. These and other protein complexes were associated with sensitivity to knockdown of several different genes. These data in conjunction with the wider CCLE are a broad resource to explore cellular behavior and facilitate cancer research.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Neoplasias/metabolismo , Proteoma/metabolismo , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Humanos , Espectrometría de Masas/métodos , Inestabilidad de Microsatélites , Mutación/genética , Proteómica/métodos
10.
Cell ; 182(2): 429-446.e14, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32526206

RESUMEN

The mode of acquisition and causes for the variable clinical spectrum of coronavirus disease 2019 (COVID-19) remain unknown. We utilized a reverse genetics system to generate a GFP reporter virus to explore severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis and a luciferase reporter virus to demonstrate sera collected from SARS and COVID-19 patients exhibited limited cross-CoV neutralization. High-sensitivity RNA in situ mapping revealed the highest angiotensin-converting enzyme 2 (ACE2) expression in the nose with decreasing expression throughout the lower respiratory tract, paralleled by a striking gradient of SARS-CoV-2 infection in proximal (high) versus distal (low) pulmonary epithelial cultures. COVID-19 autopsied lung studies identified focal disease and, congruent with culture data, SARS-CoV-2-infected ciliated and type 2 pneumocyte cells in airway and alveolar regions, respectively. These findings highlight the nasal susceptibility to SARS-CoV-2 with likely subsequent aspiration-mediated virus seeding to the lung in SARS-CoV-2 pathogenesis. These reagents provide a foundation for investigations into virus-host interactions in protective immunity, host susceptibility, and virus pathogenesis.


Asunto(s)
Betacoronavirus/genética , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Neumonía Viral/patología , Neumonía Viral/virología , Sistema Respiratorio/virología , Genética Inversa/métodos , Anciano , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/terapia , Fibrosis Quística/patología , ADN Recombinante , Femenino , Furina/metabolismo , Humanos , Inmunización Pasiva , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Mucosa Nasal/virología , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/inmunología , Sistema Respiratorio/patología , SARS-CoV-2 , Serina Endopeptidasas/metabolismo , Células Vero , Virulencia , Replicación Viral , Sueroterapia para COVID-19
11.
Cell ; 177(2): 352-360.e13, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30853217

RESUMEN

Bacteria exhibit cell-to-cell variability in their resilience to stress, for example, following antibiotic exposure. Higher resilience is typically ascribed to "dormant" non-growing cellular states. Here, by measuring membrane potential dynamics of Bacillus subtilis cells, we show that actively growing bacteria can cope with ribosome-targeting antibiotics through an alternative mechanism based on ion flux modulation. Specifically, we observed two types of cellular behavior: growth-defective cells exhibited a mathematically predicted transient increase in membrane potential (hyperpolarization), followed by cell death, whereas growing cells lacked hyperpolarization events and showed elevated survival. Using structural perturbations of the ribosome and proteomic analysis, we uncovered that stress resilience arises from magnesium influx, which prevents hyperpolarization. Thus, ion flux modulation provides a distinct mechanism to cope with ribosomal stress. These results suggest new approaches to increase the effectiveness of ribosome-targeting antibiotics and reveal an intriguing connection between ribosomes and the membrane potential, two fundamental properties of cells.


Asunto(s)
Membrana Externa Bacteriana/metabolismo , Magnesio/metabolismo , Ribosomas/metabolismo , Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Proteómica , Proteínas Ribosómicas/metabolismo
12.
Cell ; 176(5): 1014-1025.e12, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30794773

RESUMEN

Bioactive molecules can pass between microbiota and host to influence host cellular functions. However, general principles of interspecies communication have not been discovered. We show here in C. elegans that nitric oxide derived from resident bacteria promotes widespread S-nitrosylation of the host proteome. We further show that microbiota-dependent S-nitrosylation of C. elegans Argonaute protein (ALG-1)-at a site conserved and S-nitrosylated in mammalian Argonaute 2 (AGO2)-alters its function in controlling gene expression via microRNAs. By selectively eliminating nitric oxide generation by the microbiota or S-nitrosylation in ALG-1, we reveal unforeseen effects on host development. Thus, the microbiota can shape the post-translational landscape of the host proteome to regulate microRNA activity, gene expression, and host development. Our findings suggest a general mechanism by which the microbiota may control host cellular functions, as well as a new role for gasotransmitters.


Asunto(s)
Interacciones Microbiota-Huesped/genética , MicroARNs/metabolismo , Óxido Nítrico/metabolismo , Animales , Proteínas Argonautas/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células HEK293 , Células HeLa , Humanos , MicroARNs/fisiología , Microbiota/genética , Óxido Nítrico/fisiología , Procesamiento Proteico-Postraduccional/genética , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Proteínas de Unión al ARN/genética
13.
Cell ; 177(6): 1480-1494.e19, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31056283

RESUMEN

Varying pH of luminal fluid along the female reproductive tract is a physiological cue that modulates sperm motility. CatSper is a sperm-specific, pH-sensitive calcium channel essential for hyperactivated motility and male fertility. Multi-subunit CatSper channel complexes organize linear Ca2+ signaling nanodomains along the sperm tail. Here, we identify EF-hand calcium-binding domain-containing protein 9 (EFCAB9) as a bifunctional, cytoplasmic machine modulating the channel activity and the domain organization of CatSper. Knockout mice studies demonstrate that EFCAB9, in complex with the CatSper subunit, CATSPERζ, is essential for pH-dependent and Ca2+-sensitive activation of the CatSper channel. In the absence of EFCAB9, sperm motility and fertility is compromised, and the linear arrangement of the Ca2+ signaling domains is disrupted. EFCAB9 interacts directly with CATSPERζ in a Ca2+-dependent manner and dissociates at elevated pH. These observations suggest that EFCAB9 is a long-sought, intracellular, pH-dependent Ca2+ sensor that triggers changes in sperm motility.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Motilidad Espermática/fisiología , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Proteínas de Unión al Calcio/fisiología , Línea Celular , Membrana Celular/metabolismo , Fertilidad , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espermatozoides/metabolismo
14.
Cell ; 172(1-2): 106-120.e21, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29249356

RESUMEN

Cell fate transitions involve rapid gene expression changes and global chromatin remodeling, yet the underlying regulatory pathways remain incompletely understood. Here, we identified the RNA-processing factor Nudt21 as a novel regulator of cell fate change using transcription-factor-induced reprogramming as a screening assay. Suppression of Nudt21 enhanced the generation of induced pluripotent stem cells, facilitated transdifferentiation into trophoblast stem cells, and impaired differentiation of myeloid precursors and embryonic stem cells, suggesting a broader role for Nudt21 in cell fate change. We show that Nudt21 directs differential polyadenylation of over 1,500 transcripts in cells acquiring pluripotency, although only a fraction changed protein levels. Remarkably, these proteins were strongly enriched for chromatin regulators, and their suppression neutralized the effect of Nudt21 during reprogramming. Collectively, our data uncover Nudt21 as a novel post-transcriptional regulator of cell fate and establish a direct, previously unappreciated link between alternative polyadenylation and chromatin signaling.


Asunto(s)
Reprogramación Celular , Ensamble y Desensamble de Cromatina , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Poliadenilación , Transducción de Señal , Animales , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células HEK293 , Humanos , Ratones
15.
Cell ; 169(2): 338-349.e11, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388415

RESUMEN

G-protein-coupled receptors (GPCRs) play critical roles in regulating physiological processes ranging from neurotransmission to cardiovascular function. Current methods for tracking GPCR signaling suffer from low throughput, modification or overexpression of effector proteins, and low temporal resolution. Here, we show that peroxidase-catalyzed proximity labeling can be combined with isobaric tagging and mass spectrometry to enable quantitative, time-resolved measurement of GPCR agonist response in living cells. Using this technique, termed "GPCR-APEX," we track activation and internalization of the angiotensin II type 1 receptor and the ß2 adrenoceptor. These receptors co-localize with a variety of G proteins even before receptor activation, and activated receptors are largely sequestered from G proteins upon internalization. Additionally, the two receptors show differing internalization kinetics, and we identify the membrane protein LMBRD2 as a potential regulator of ß2 adrenoceptor signaling, underscoring the value of a dynamic view of receptor function.


Asunto(s)
Ascorbato Peroxidasas/química , Receptor de Angiotensina Tipo 1/análisis , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Coloración y Etiquetado/métodos , Ascorbato Peroxidasas/metabolismo , Biotina/química , Proteínas de Unión al GTP/análisis , Células HEK293 , Humanos , Oligopéptidos/farmacología , Ingeniería de Proteínas , Receptor de Angiotensina Tipo 1/agonistas , beta-Arrestinas/química
16.
Mol Cell ; 84(7): 1290-1303.e7, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38401542

RESUMEN

Most eukaryotic proteins are degraded by the 26S proteasome after modification with a polyubiquitin chain. Substrates lacking unstructured segments cannot be degraded directly and require prior unfolding by the Cdc48 ATPase (p97 or VCP in mammals) in complex with its ubiquitin-binding partner Ufd1-Npl4 (UN). Here, we use purified yeast components to reconstitute Cdc48-dependent degradation of well-folded model substrates by the proteasome. We show that a minimal system consists of the 26S proteasome, the Cdc48-UN ATPase complex, the proteasome cofactor Rad23, and the Cdc48 cofactors Ubx5 and Shp1. Rad23 and Ubx5 stimulate polyubiquitin binding to the 26S proteasome and the Cdc48-UN complex, respectively, allowing these machines to compete for substrates before and after their unfolding. Shp1 stimulates protein unfolding by the Cdc48-UN complex rather than substrate recruitment. Experiments in yeast cells confirm that many proteins undergo bidirectional substrate shuttling between the 26S proteasome and Cdc48 ATPase before being degraded.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Proteínas de Saccharomyces cerevisiae , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/metabolismo , Proteína que Contiene Valosina/genética , Proteína que Contiene Valosina/metabolismo
18.
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
19.
Cell ; 165(1): 153-164, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26972053

RESUMEN

Amino acids signal to the mTOR complex I (mTORC1) growth pathway through the Rag GTPases. Multiple distinct complexes regulate the Rags, including GATOR1, a GTPase activating protein (GAP), and GATOR2, a positive regulator of unknown molecular function. Arginine stimulation of cells activates mTORC1, but how it is sensed is not well understood. Recently, SLC38A9 was identified as a putative lysosomal arginine sensor required for arginine to activate mTORC1 but how arginine deprivation represses mTORC1 is unknown. Here, we show that CASTOR1, a previously uncharacterized protein, interacts with GATOR2 and is required for arginine deprivation to inhibit mTORC1. CASTOR1 homodimerizes and can also heterodimerize with the related protein, CASTOR2. Arginine disrupts the CASTOR1-GATOR2 complex by binding to CASTOR1 with a dissociation constant of ~30 µM, and its arginine-binding capacity is required for arginine to activate mTORC1 in cells. Collectively, these results establish CASTOR1 as an arginine sensor for the mTORC1 pathway.


Asunto(s)
Arginina/metabolismo , Proteínas Portadoras/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Multimerización de Proteína , Serina-Treonina Quinasas TOR/metabolismo
20.
Cell ; 167(7): 1705-1718.e13, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984722

RESUMEN

Metformin has utility in cancer prevention and treatment, though the mechanisms for these effects remain elusive. Through genetic screening in C. elegans, we uncover two metformin response elements: the nuclear pore complex (NPC) and acyl-CoA dehydrogenase family member-10 (ACAD10). We demonstrate that biguanides inhibit growth by inhibiting mitochondrial respiratory capacity, which restrains transit of the RagA-RagC GTPase heterodimer through the NPC. Nuclear exclusion renders RagC incapable of gaining the GDP-bound state necessary to stimulate mTORC1. Biguanide-induced inactivation of mTORC1 subsequently inhibits growth through transcriptional induction of ACAD10. This ancient metformin response pathway is conserved from worms to humans. Both restricted nuclear pore transit and upregulation of ACAD10 are required for biguanides to reduce viability in melanoma and pancreatic cancer cells, and to extend C. elegans lifespan. This pathway provides a unified mechanism by which metformin kills cancer cells and extends lifespan, and illuminates potential cancer targets. PAPERCLIP.


Asunto(s)
Metformina/farmacología , Acil-CoA Deshidrogenasa/genética , Envejecimiento , Animales , Tamaño Corporal , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Humanos , Longevidad , Diana Mecanicista del Complejo 1 de la Rapamicina , Mitocondrias/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos/metabolismo , Neoplasias/tratamiento farmacológico , Poro Nuclear/metabolismo , Fosforilación Oxidativa , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA