Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 79(1): 155-166.e9, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32454028

RESUMEN

To understand gene function, the encoding DNA or mRNA transcript can be manipulated and the consequences observed. However, these approaches do not have a direct effect on the protein product of the gene, which is either permanently abrogated or depleted at a rate defined by the half-life of the protein. We therefore developed a single-component system that could induce the rapid degradation of the specific endogenous protein itself. A construct combining the RING domain of ubiquitin E3 ligase RNF4 with a protein-specific camelid nanobody mediates target destruction by the ubiquitin proteasome system, a process we describe as antibody RING-mediated destruction (ARMeD). The technique is highly specific because we observed no off-target protein destruction. Furthermore, bacterially produced nanobody-RING fusion proteins electroporated into cells induce degradation of target within minutes. With increasing availability of protein-specific nanobodies, this method will allow rapid and specific degradation of a wide range of endogenous proteins.


Asunto(s)
Endopeptidasas/metabolismo , Proteína NEDD8/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Anticuerpos de Dominio Único/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Endopeptidasas/inmunología , Células HeLa , Humanos , Proteína NEDD8/inmunología , Proteínas Nucleares/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Proteolisis , Anticuerpos de Dominio Único/inmunología , Factores de Transcripción/inmunología , Ubiquitinación
2.
Mol Cell ; 75(3): 483-497.e9, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31253574

RESUMEN

In mammals, ∼100 deubiquitinases act on ∼20,000 intracellular ubiquitination sites. Deubiquitinases are commonly regarded as constitutively active, with limited regulatory and targeting capacity. The BRCA1-A and BRISC complexes serve in DNA double-strand break repair and immune signaling and contain the lysine-63 linkage-specific BRCC36 subunit that is functionalized by scaffold subunits ABRAXAS and ABRO1, respectively. The molecular basis underlying BRCA1-A and BRISC function is currently unknown. Here we show that in the BRCA1-A complex structure, ABRAXAS integrates the DNA repair protein RAP80 and provides a high-affinity binding site that sequesters the tumor suppressor BRCA1 away from the break site. In the BRISC structure, ABRO1 binds SHMT2α, a metabolic enzyme enabling cancer growth in hypoxic environments, which we find prevents BRCC36 from binding and cleaving ubiquitin chains. Our work explains modularity in the BRCC36 DUB family, with different adaptor subunits conferring diversified targeting and regulatory functions.


Asunto(s)
Proteína BRCA1/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Enzimas Desubicuitinizantes/genética , Chaperonas de Histonas/genética , Neoplasias/genética , Sitios de Unión/genética , Proteínas Portadoras/genética , Núcleo Celular/genética , Núcleo Celular/inmunología , Citoplasma/genética , Citoplasma/inmunología , Roturas del ADN de Doble Cadena , Reparación del ADN/inmunología , Enzimas Desubicuitinizantes/inmunología , Células HeLa , Humanos , Inmunidad Celular/genética , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Neoplasias/inmunología , Proteínas Asociadas a Matriz Nuclear/genética , Unión Proteica/genética , Ubiquitina/genética , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/genética
3.
EMBO J ; 40(18): e107413, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34346517

RESUMEN

DNA-protein crosslinks (DPCs) obstruct essential DNA transactions, posing a serious threat to genome stability and functionality. DPCs are proteolytically processed in a ubiquitin- and DNA replication-dependent manner by SPRTN and the proteasome but can also be resolved via targeted SUMOylation. However, the mechanistic basis of SUMO-mediated DPC resolution and its interplay with replication-coupled DPC repair remain unclear. Here, we show that the SUMO-targeted ubiquitin ligase RNF4 defines a major pathway for ubiquitylation and proteasomal clearance of SUMOylated DPCs in the absence of DNA replication. Importantly, SUMO modifications of DPCs neither stimulate nor inhibit their rapid DNA replication-coupled proteolysis. Instead, DPC SUMOylation provides a critical salvage mechanism to remove DPCs formed after DNA replication, as DPCs on duplex DNA do not activate interphase DNA damage checkpoints. Consequently, in the absence of the SUMO-RNF4 pathway cells are able to enter mitosis with a high load of unresolved DPCs, leading to defective chromosome segregation and cell death. Collectively, these findings provide mechanistic insights into SUMO-driven pathways underlying replication-independent DPC resolution and highlight their critical importance in maintaining chromosome stability and cellular fitness.


Asunto(s)
Reparación del ADN , Replicación del ADN , Proteínas Nucleares/metabolismo , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/metabolismo , Inestabilidad Genómica , Humanos , Unión Proteica , Procesamiento Proteico-Postraduccional , Sumoilación , Ubiquitina/metabolismo , Ubiquitinación
4.
PLoS Pathog ; 19(7): e1011477, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37410772

RESUMEN

SUMO modifications regulate the function of many proteins and are important in controlling herpesvirus infections. We performed a site-specific proteomic analysis of SUMO1- and SUMO2-modified proteins in Epstein-Barr virus (EBV) latent and lytic infection to identify proteins that change in SUMO modification status in response to EBV reactivation. Major changes were identified in all three components of the TRIM24/TRIM28/TRIM33 complex, with TRIM24 being rapidly degraded and TRIM33 being phosphorylated and SUMOylated in response to EBV lytic infection. Further experiments revealed TRIM24 and TRIM33 repress expression of the EBV BZLF1 lytic switch gene, suppressing EBV reactivation. However, BZLF1 was shown to interact with TRIM24 and TRIM33, resulting in disruption of TRIM24/TRIM28/TRIM33 complexes, degradation of TRIM24 and modification followed by degradation of TRIM33. Therefore, we have identified TRIM24 and TRIM33 as cellular antiviral defence factors against EBV lytic infection and established the mechanism by which BZLF1 disables this defence.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Humanos , Herpesvirus Humano 4/genética , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteómica , Activación Viral , Latencia del Virus , Factores de Transcripción/metabolismo , Proteínas Portadoras
5.
Mol Cell ; 65(1): 66-77, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27939944

RESUMEN

During Caenorhabditis elegans oocyte meiosis, a multi-protein ring complex (RC) localized between homologous chromosomes, promotes chromosome congression through the action of the chromokinesin KLP-19. While some RC components are known, the mechanism of RC assembly has remained obscure. We show that SUMO E3 ligase GEI-17/PIAS is required for KLP-19 recruitment to the RC, and proteomic analysis identified KLP-19 as a SUMO substrate in vivo. In vitro analysis revealed that KLP-19 is efficiently sumoylated in a GEI-17-dependent manner, while GEI-17 undergoes extensive auto-sumoylation. GEI-17 and another RC component, the kinase BUB-1, contain functional SUMO interaction motifs (SIMs), allowing them to recruit SUMO modified proteins, including KLP-19, into the RC. Thus, dynamic SUMO modification and the presence of SIMs in RC components generate a SUMO-SIM network that facilitates assembly of the RC. Our results highlight the importance of SUMO-SIM networks in regulating the assembly of dynamic protein complexes.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Posicionamiento de Cromosoma , Segregación Cromosómica , Cinesinas/metabolismo , Ligasas/metabolismo , Meiosis , Oocitos/metabolismo , Sumoilación , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Femenino , Genotipo , Cinesinas/genética , Ligasas/genética , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Tiempo , Ubiquitina-Proteína Ligasas/genética
6.
J Virol ; 97(11): e0079123, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37916833

RESUMEN

IMPORTANCE: Human adenoviruses (HAdVs) generally cause mild and self-limiting diseases of the upper respiratory and gastrointestinal tracts but pose a serious risk to immunocompromised patients and children. Moreover, they are widely used as vectors for vaccines and vector-based gene therapy approaches. It is therefore vital to thoroughly characterize HAdV gene products and especially HAdV virulence factors. Early region 1B 55 kDa protein (E1B-55K) is a multifunctional HAdV-encoded oncoprotein involved in various viral and cellular pathways that promote viral replication and cell transformation. We analyzed the E1B-55K dependency of SUMOylation, a post-translational protein modification, in infected cells using quantitative proteomics. We found that HAdV increases overall cellular SUMOylation and that this increased SUMOylation can target antiviral cellular pathways that impact HAdV replication. Moreover, we showed that E1B-55K orchestrates the SUMO-dependent degradation of certain cellular antiviral factors. These results once more emphasize the key role of E1B-55K in the regulation of viral and cellular proteins in productive HAdV infections.


Asunto(s)
Infecciones por Adenoviridae , Adenovirus Humanos , Factores de Restricción Antivirales , Humanos , Adenoviridae/genética , Infecciones por Adenoviridae/metabolismo , Adenovirus Humanos/fisiología , Factores de Restricción Antivirales/metabolismo , Sumoilación
7.
J Biol Chem ; 298(12): 102690, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36372232

RESUMEN

RNA Polymerase I (Pol I) synthesizes rRNA, which is the first and rate-limiting step in ribosome biogenesis. Factors governing the stability of the polymerase complex are not known. Previous studies characterizing Pol I inhibitor BMH-21 revealed a transcriptional stress-dependent pathway for degradation of the largest subunit of Pol I, RPA194. To identify the E3 ligase(s) involved, we conducted a cell-based RNAi screen for ubiquitin pathway genes. We establish Skp-Cullin-F-box protein complex F-box protein FBXL14 as an E3 ligase for RPA194. We show that FBXL14 binds to RPA194 and mediates RPA194 ubiquitination and degradation in cancer cells treated with BMH-21. Mutation analysis in yeast identified lysines 1150, 1153, and 1156 on Rpa190 relevant for the protein degradation. These results reveal the regulated turnover of Pol I, showing that the stability of the catalytic subunit is controlled by the F-box protein FBXL14 in response to transcription stress.


Asunto(s)
Proteínas F-Box , Proteínas Ligasas SKP Cullina F-box , Transcripción Genética , Dominio Catalítico , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ubiquitinación , Humanos , Transcripción Genética/genética
8.
Mol Cell Proteomics ; 20: 100164, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34673284

RESUMEN

To investigate the role of SUMO modification in the maintenance of pluripotent stem cells, we used ML792, a potent and selective inhibitor of SUMO Activating Enzyme. Treatment of human induced pluripotent stem cells with ML792 resulted in the loss of key pluripotency markers. To identify putative effector proteins and establish sites of SUMO modification, cells were engineered to stably express either SUMO1 or SUMO2 with C-terminal TGG to KGG mutations that facilitate GlyGly-K peptide immunoprecipitation and identification. A total of 976 SUMO sites were identified in 427 proteins. STRING enrichment created three networks of proteins with functions in regulation of gene expression, ribosome biogenesis, and RNA splicing, although the latter two categories represented only 5% of the total GGK peptide intensity. The rest have roles in transcription and the regulation of chromatin structure. Many of the most heavily SUMOylated proteins form a network of zinc-finger transcription factors centered on TRIM28 and associated with silencing of retroviral elements. At the level of whole proteins, there was only limited evidence for SUMO paralogue-specific modification, although at the site level there appears to be a preference for SUMO2 modification over SUMO1 in acidic domains. We show that SUMO influences the pluripotent state in hiPSCs and identify many chromatin-associated proteins as bona fide SUMO substrates in human induced pluripotent stem cells.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Línea Celular , Humanos , Proteómica , Sumoilación
9.
Mol Cell ; 53(6): 880-92, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24656128

RESUMEN

Dimeric RING E3 ligases interact with protein substrates and conformationally restrain the ubiquitin-E2-conjugating enzyme thioester complex such that it is primed for catalysis. RNF4 is an E3 ligase containing an N-terminal domain that binds its polySUMO substrates and a C-terminal RING domain responsible for dimerization. To investigate how RNF4 activity is controlled, we increased polySUMO substrate concentration by ablating expression of SUMO protease SENP6. Accumulation of SUMO chains in vivo leads to ubiquitin-mediated proteolysis of RNF4. In vitro we demonstrate that at concentrations equivalent to those found in vivo RNF4 is predominantly monomeric and inactive as an ubiquitin E3 ligase. However, in the presence of SUMO chains, RNF4 is activated by dimerization, leading to both substrate ubiquitylation and autoubiquitylation, responsible for degradation of RNF4. Thus the ubiquitin E3 ligase activity of RNF4 is directly linked to the availability of its polySUMO substrates.


Asunto(s)
Cisteína Endopeptidasas/genética , Regulación de la Expresión Génica , Proteínas Nucleares/genética , Multimerización de Proteína , Proteína SUMO-1/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Factores de Transcripción/genética , Sitios de Unión , Línea Celular Tumoral , Cisteína Endopeptidasas/metabolismo , Humanos , Microscopía Fluorescente , Proteínas Nucleares/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína SUMO-1/metabolismo , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación
10.
Mol Cell ; 56(3): 453-461, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25306921

RESUMEN

The ubiquitin-proteasome system is the major pathway for protein degradation in eukaryotic cells. Proteins to be degraded are conjugated to ubiquitin chains that act as recognition signals for the 26S proteasome. The proteasome subunits Rpn10 and Rpn13 are known to bind ubiquitin, but genetic and biochemical data suggest the existence of at least one other substrate receptor. Here, we show that the phylogenetically conserved proteasome subunit Dss1 (Sem1) binds ubiquitin chains linked by K63 and K48. Atomic resolution data show that Dss1 is disordered and binds ubiquitin by binding sites characterized by acidic and hydrophobic residues. The complementary binding region in ubiquitin is composed of a hydrophobic patch formed by I13, I44, and L69 flanked by two basic regions. Mutations in the ubiquitin-binding site of Dss1 cause growth defects and accumulation of ubiquitylated proteins.


Asunto(s)
Proteínas Portadoras/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Ubiquitina/metabolismo , Sitios de Unión , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN , Proteínas de Schizosaccharomyces pombe/química , Ubiquitina/química
11.
Biochem J ; 478(13): 2399-2403, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34198321

RESUMEN

The coronavirus pandemic has had a huge impact on public health with over 165 million people infected, 3.4 million deaths and a hugely deleterious effect on most economies. While vaccination effectively protects against the disease it is likely that viruses will evolve that can replicate in hosts immunised with the present vaccines. Thus, there is a great unmet need for effective antivirals that can block the development of serious disease in infected patients. The seven papers published in this issue of the Biochemical Journal address this need by expressing and purifying components required for viral replication, developing biochemical assays for these components and using the assays to screen a library of pre-existing pharmaceuticals for drugs that inhibited the target in vitro and inhibited viral replication in cell culture. The candidate drugs obtained are potential antivirals that may protect against SARS-CoV-2 infection. While not all the antiviral candidates will make it through to the clinic, they will be useful tool compounds and can act as the starting point for further drug discovery programmes.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , COVID-19/virología , Evaluación Preclínica de Medicamentos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , Replicación Viral/efectos de los fármacos , Humanos , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo
12.
PLoS Genet ; 15(11): e1008427, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31765407

RESUMEN

Replication fork stalling and accumulation of single-stranded DNA trigger the S phase checkpoint, a signalling cascade that, in budding yeast, leads to the activation of the Rad53 kinase. Rad53 is essential in maintaining cell viability, but its targets of regulation are still partially unknown. Here we show that Rad53 drives the hyper-SUMOylation of Pol2, the catalytic subunit of DNA polymerase ε, principally following replication forks stalling induced by nucleotide depletion. Pol2 is the main target of SUMOylation within the replisome and its modification requires the SUMO-ligase Mms21, a subunit of the Smc5/6 complex. Moreover, the Smc5/6 complex co-purifies with Pol ε, independently of other replisome components. Finally, we map Pol2 SUMOylation to a single site within the N-terminal catalytic domain and identify a SUMO-interacting motif at the C-terminus of Pol2. These data suggest that the S phase checkpoint regulate Pol ε during replication stress through Pol2 SUMOylation and SUMO-binding ability.


Asunto(s)
Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , ADN Polimerasa II/genética , ADN/biosíntesis , Proteína SUMO-1/genética , Proteínas de Saccharomyces cerevisiae/genética , Sumoilación/genética , Dominio Catalítico/genética , ADN/genética , Replicación del ADN/genética , Complejos Multiproteicos/genética , Unión Proteica , Fase S/genética , Saccharomyces cerevisiae/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética
13.
Proc Natl Acad Sci U S A ; 116(35): 17399-17408, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31391303

RESUMEN

Dynamic small ubiquitin-like modifier (SUMO) linkages to diverse cellular protein groups are critical to orchestrate resolution of stresses such as genome damage, hypoxia, or proteotoxicity. Defense against pathogen insult (often reliant upon host recognition of "non-self" nucleic acids) is also modulated by SUMO, but the underlying mechanisms are incompletely understood. Here, we used quantitative SILAC-based proteomics to survey pan-viral host SUMOylation responses, creating a resource of almost 600 common and unique SUMO remodeling events that are mounted during influenza A and B virus infections, as well as during viral innate immune stimulation. Subsequent mechanistic profiling focused on a common infection-induced loss of the SUMO-modified form of TRIM28/KAP1, a host transcriptional repressor. By integrating knockout and reconstitution models with system-wide transcriptomics, we provide evidence that influenza virus-triggered loss of SUMO-modified TRIM28 leads to derepression of endogenous retroviral (ERV) elements, unmasking this cellular source of "self" double-stranded (ds)RNA. Consequently, loss of SUMO-modified TRIM28 potentiates canonical cytosolic dsRNA-activated IFN-mediated defenses that rely on RIG-I, MAVS, TBK1, and JAK1. Intriguingly, although wild-type influenza A virus robustly triggers this SUMO switch in TRIM28, the induction of IFN-stimulated genes is limited unless expression of the viral dsRNA-binding protein NS1 is abrogated. This may imply a viral strategy to antagonize such a host response by sequestration of induced immunostimulatory ERV dsRNAs. Overall, our data reveal that a key nuclear mechanism that normally prevents aberrant expression of ERV elements (ERVs) has been functionally co-opted via a stress-induced SUMO switch to augment antiviral immunity.


Asunto(s)
Retrovirus Endógenos/inmunología , Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/virología , Interacciones Microbianas , Proteína SUMO-1/metabolismo , Animales , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Modelos Biológicos , ARN Bicatenario/metabolismo , Sumoilación , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Replicación Viral
14.
J Cell Sci ; 132(14)2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31243051

RESUMEN

Oocyte meiotic spindles in most species lack centrosomes and the mechanisms that underlie faithful chromosome segregation in acentrosomal meiotic spindles are not well understood. In C. elegans oocytes, spindle microtubules exert a poleward force on chromosomes that is dependent on the microtubule-stabilising protein CLS-2, the orthologue of the mammalian CLASP proteins. The checkpoint kinase BUB-1 and CLS-2 localise in the central spindle and display a dynamic localisation pattern throughout anaphase, but the signals regulating their anaphase-specific localisation remains unknown. We have shown previously that SUMO regulates BUB-1 localisation during metaphase I. Here, we found that SUMO modification of BUB-1 is regulated by the SUMO E3 ligase GEI-17 and the SUMO protease ULP-1. SUMO and GEI-17 are required for BUB-1 localisation between segregating chromosomes during early anaphase I. We also show that CLS-2 is subject to SUMO-mediated regulation; CLS-2 precociously localises in the midbivalent when either SUMO or GEI-17 are depleted. Overall, we provide evidence for a novel, SUMO-mediated control of protein dynamics during early anaphase I in oocytes.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Segregación Cromosómica , Meiosis , Oocitos/citología , Oocitos/metabolismo , Sumoilación , Anafase , Animales , Modelos Biológicos , Transporte de Proteínas , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Huso Acromático/metabolismo
15.
Nat Rev Mol Cell Biol ; 10(8): 564-8, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19474794

RESUMEN

Although the post-translational modification of proteins with small ubiquitin-like modifier (SUMO) has a role in many biological processes, it was thought that SUMO, unlike ubiquitin, does not target proteins for degradation. However, these views need to be revised, as recent findings in yeast and human cells indicate that SUMO can act as a signal for the recruitment of E3 ubiquitin ligases, which leads to the ubiquitylation and degradation of the modified protein.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Datos de Secuencia Molecular , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
16.
Genes Dev ; 26(11): 1196-208, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661230

RESUMEN

Here we demonstrate that RNF4, a highly conserved small ubiquitin-like modifier (SUMO)-targeted ubiquitin E3 ligase, plays a critical role in the response of mammalian cells to DNA damage. Human cells in which RNF4 expression was ablated by siRNA or chicken DT40 cells with a homozygous deletion of the RNF4 gene displayed increased sensitivity to DNA-damaging agents. Recruitment of RNF4 to double-strand breaks required its RING and SUMO interaction motif (SIM) domains and DNA damage factors such as NBS1, mediator of DNA damage checkpoint 1 (MDC1), RNF8, 53BP1, and BRCA1. In the absence of RNF4, these factors were still recruited to sites of DNA damage, but 53BP1, RNF8, and RNF168 displayed delayed clearance from such foci. SILAC-based proteomics of SUMO substrates revealed that MDC1 was SUMO-modified in response to ionizing radiation. As a consequence of SUMO modification, MDC1 recruited RNF4, which mediated ubiquitylation at the DNA damage site. Failure to recruit RNF4 resulted in defective loading of replication protein A (RPA) and Rad51 onto ssDNA. This appeared to be a consequence of reduced recruitment of the CtIP nuclease, resulting in inefficient end resection. Thus, RNF4 is a novel DNA damage-responsive protein that plays a role in homologous recombination and integrates SUMO modification and ubiquitin signaling in the cellular response to genotoxic stress.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Células HeLa , Recombinación Homóloga , Humanos , Proteínas Nucleares/genética , Ratas , Transactivadores/metabolismo , Factores de Transcripción/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
17.
Mol Cell Proteomics ; 16(2): 310-326, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27913581

RESUMEN

Aspirin, or acetylsalicylic acid is widely used to control pain, inflammation and fever. Important to this function is its ability to irreversibly acetylate cyclooxygenases at active site serines. Aspirin has the potential to acetylate other amino acid side-chains, leading to the possibility that aspirin-mediated lysine acetylation could explain some of its as-yet unexplained drug actions or side-effects. Using isotopically labeled aspirin-d3, in combination with acetylated lysine purification and LC-MS/MS, we identified over 12000 sites of lysine acetylation from cultured human cells. Although aspirin amplifies endogenous acetylation signals at the majority of detectable endogenous sites, cells tolerate aspirin mediated acetylation very well unless cellular deacetylases are inhibited. Although most endogenous acetylations are amplified by orders of magnitude, lysine acetylation site occupancies remain very low even after high doses of aspirin. This work shows that while aspirin has enormous potential to alter protein function, in the majority of cases aspirin-mediated acetylations do not accumulate to levels likely to elicit biological effects. These findings are consistent with an emerging model for cellular acetylation whereby stoichiometry correlates with biological relevance, and deacetylases act to minimize the biological consequences of nonspecific chemical acetylations.


Asunto(s)
Aspirina/farmacología , Lisina/análisis , Proteoma/química , Proteómica/métodos , Acetilación , Sitios de Unión , Cromatografía Liquida , Células HeLa , Histona Desacetilasas/metabolismo , Humanos , Marcaje Isotópico , Lisina/química , Lisina/efectos de los fármacos , Espectrometría de Masas en Tándem
18.
J Cell Sci ; 129(3): 580-91, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26675234

RESUMEN

Promyelocytic leukemia (PML) protein forms the basis of PML nuclear bodies (PML NBs), which control many important processes. We have screened an shRNA library targeting ubiquitin pathway proteins for effects on PML NBs, and identified RNF8 and RNF168 DNA-damage response proteins as negative regulators of PML NBs. Additional studies confirmed that depletion of either RNF8 or RNF168 increased the levels of PML NBs and proteins, whereas overexpression induced loss of PML NBs. RNF168 partially localized to PML NBs through its UMI/MIU1 ubiquitin-interacting region and associated with NBs formed by any PML isoform. The association of RNF168 with PML NBs resulted in increased ubiquitylation and SUMO2 modification of PML. In addition, RNF168 was found to associate with proteins modified by SUMO2 and/or SUMO3 in a manner dependent on its ubiquitin-binding sequences, suggesting that hybrid SUMO-ubiquitin chains can be bound. In vitro assays confirmed that RNF168, preferentially, binds hybrid SUMO2-K63 ubiquitin chains compared with K63-ubiquitin chains or individual SUMO2. Our study identified previously unrecognized roles for RNF8 and RNF168 in the regulation of PML, and a so far unknown preference of RNF168 for hybrid SUMO-ubiquitin chains.


Asunto(s)
Cuerpos de Inclusión Intranucleares/metabolismo , Leucemia Mieloide/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Humanos , Proteínas de Neoplasias/metabolismo , Proteína de la Leucemia Promielocítica , Unión Proteica/fisiología , Isoformas de Proteínas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología
19.
FASEB J ; 31(6): 2446-2459, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28254759

RESUMEN

Brain regions affected by Alzheimer disease (AD) display well-recognized early neuropathologic features in the endolysosomal and autophagy systems of neurons, including enlargement of endosomal compartments, progressive accumulation of autophagic vacuoles, and lysosomal dysfunction. Although the primary causes of these disturbances are still under investigation, a growing body of evidence suggests that the amyloid precursor protein (APP) intracellular C-terminal fragment ß (C99), generated by cleavage of APP by ß-site APP cleaving enzyme 1 (BACE-1), is the primary cause of the endosome enlargement in AD and the earliest initiator of synaptic plasticity and long-term memory impairment. The aim of the present study was to evaluate the possible relationship between the endolysosomal degradation pathway and autophagy on the proteolytic processing and turnover of C99. We found that pharmacologic treatments that either inhibit autophagosome formation or block the fusion of autophagosomes to endolysosomal compartments caused an increase in C99 levels. We also found that inhibition of autophagosome formation by depletion of Atg5 led to higher levels of C99 and to its massive accumulation in the lumen of enlarged perinuclear, lysosomal-associated membrane protein 1 (LAMP1)-positive organelles. In contrast, activation of autophagosome formation, either by starvation or by inhibition of the mammalian target of rapamycin, enhanced lysosomal clearance of C99. Altogether, our results indicate that autophagosomes are key organelles to help avoid C99 accumulation preventing its deleterious effects.-González, A. E., Muñoz, V. C., Cavieres, V. A., Bustamante, H. A., Cornejo, V.-H., Januário, Y. C., González, I., Hetz, C., daSilva, L. L., Rojas-Fernández, A., Hay, R. T., Mardones, G. A., Burgos, P. V. Autophagosomes cooperate in the degradation of intracellular C-terminal fragments of the amyloid precursor protein via the MVB/lysosomal pathway.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Autofagosomas/fisiología , Lisosomas/fisiología , Cuerpos Multivesiculares/fisiología , Precursor de Proteína beta-Amiloide/genética , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Regulación de la Expresión Génica/fisiología , Silenciador del Gen , Humanos , Naftiridinas/farmacología , Neuroglía , ARN Interferente Pequeño , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Nature ; 489(7414): 115-20, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22842904

RESUMEN

Ubiquitin modification is mediated by a large family of specificity determining ubiquitin E3 ligases. To facilitate ubiquitin transfer, RING E3 ligases bind both substrate and a ubiquitin E2 conjugating enzyme linked to ubiquitin via a thioester bond, but the mechanism of transfer has remained elusive. Here we report the crystal structure of the dimeric RING domain of rat RNF4 in complex with E2 (UbcH5A) linked by an isopeptide bond to ubiquitin. While the E2 contacts a single protomer of the RING, ubiquitin is folded back onto the E2 by contacts from both RING protomers. The carboxy-terminal tail of ubiquitin is locked into an active site groove on the E2 by an intricate network of interactions, resulting in changes at the E2 active site. This arrangement is primed for catalysis as it can deprotonate the incoming substrate lysine residue and stabilize the consequent tetrahedral transition-state intermediate.


Asunto(s)
Biocatálisis , Proteínas Nucleares/química , Factores de Transcripción/química , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina/metabolismo , Dedos de Zinc , Animales , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Hidrólisis , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina/química , Ubiquitina/genética , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA