Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 154(5): 983-995, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23993092

RESUMEN

DNA damage triggers polyubiquitylation and degradation of the largest subunit of RNA polymerase II (RNAPII), a "mechanism of last resort" employed during transcription stress. In yeast, this process is dependent on Def1 through a previously unresolved mechanism. Here, we report that Def1 becomes activated through ubiquitylation- and proteasome-dependent processing. Def1 processing results in the removal of a domain promoting cytoplasmic localization, resulting in nuclear accumulation of the clipped protein. Nuclear Def1 then binds RNAPII, utilizing a ubiquitin-binding domain to recruit the Elongin-Cullin E3 ligase complex via a ubiquitin-homology domain in the Ela1 protein. This facilitates polyubiquitylation of Rpb1, triggering its proteasome-mediated degradation. Together, these results outline the multistep mechanism of Rpb1 polyubiquitylation triggered by transcription stress and uncover the key role played by Def1 as a facilitator of Elongin-Cullin ubiquitin ligase function.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Transcripción Genética , Secuencia de Aminoácidos , Proteínas Cromosómicas no Histona/química , Datos de Secuencia Molecular , Complejo de la Endopetidasa Proteasomal/metabolismo , Estructura Terciaria de Proteína , ARN Polimerasa II/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Alineación de Secuencia , Estrés Fisiológico , Complejos de Ubiquitina-Proteína Ligasa/metabolismo
3.
Cell ; 140(4): 477-90, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178741

RESUMEN

Current models imply that the FERM domain protein Merlin, encoded by the tumor suppressor NF2, inhibits mitogenic signaling at or near the plasma membrane. Here, we show that the closed, growth-inhibitory form of Merlin accumulates in the nucleus, binds to the E3 ubiquitin ligase CRL4(DCAF1), and suppresses its activity. Depletion of DCAF1 blocks the promitogenic effect of inactivation of Merlin. Conversely, enforced expression of a Merlin-insensitive mutant of DCAF1 counteracts the antimitogenic effect of Merlin. Re-expression of Merlin and silencing of DCAF1 implement a similar, tumor-suppressive program of gene expression. Tumor-derived mutations invariably disrupt Merlin's ability to interact with or inhibit CRL4(DCAF1). Finally, depletion of DCAF1 inhibits the hyperproliferation of Schwannoma cells from NF2 patients and suppresses the oncogenic potential of Merlin-deficient tumor cell lines. We propose that Merlin suppresses tumorigenesis by translocating to the nucleus to inhibit CRL4(DCAF1).


Asunto(s)
Proteínas Portadoras/metabolismo , Genes Supresores de Tumor , Mesotelioma/metabolismo , Neurilemoma/metabolismo , Neurofibromina 2/metabolismo , Transporte Activo de Núcleo Celular , Animales , Proteínas Portadoras/química , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Humanos , Modelos Moleculares , Proteínas Serina-Treonina Quinasas , Ubiquitina-Proteína Ligasas
4.
Nucleic Acids Res ; 51(13): 6754-6769, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37309898

RESUMEN

The Sirtuin family of NAD+-dependent enzymes plays an important role in maintaining genome stability upon stress. Several mammalian Sirtuins have been linked directly or indirectly to the regulation of DNA damage during replication through Homologous recombination (HR). The role of one of them, SIRT1, is intriguing as it seems to have a general regulatory role in the DNA damage response (DDR) that has not yet been addressed. SIRT1-deficient cells show impaired DDR reflected in a decrease in repair capacity, increased genome instability and decreased levels of γH2AX. Here we unveil a close functional antagonism between SIRT1 and the PP4 phosphatase multiprotein complex in the regulation of the DDR. Upon DNA damage, SIRT1 interacts specifically with the catalytical subunit PP4c and promotes its inhibition by deacetylating the WH1 domain of the regulatory subunits PP4R3α/ß. This in turn regulates γH2AX and RPA2 phosphorylation, two key events in the signaling of DNA damage and repair by HR. We propose a mechanism whereby during stress, SIRT1 signaling ensures a global control of DNA damage signaling through PP4.


Asunto(s)
Daño del ADN , Sirtuina 1 , Animales , Humanos , Mamíferos/metabolismo , Monoéster Fosfórico Hidrolasas , Fosforilación , Transducción de Señal , Sirtuina 1/metabolismo
5.
Genes Dev ; 27(14): 1581-95, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23824326

RESUMEN

Post-translational histone modifications play important roles in regulating chromatin structure and function. Histone H2B ubiquitination and deubiquitination have been implicated in transcriptional regulation, but the function of H2B deubiquitination is not well defined, particularly in higher eukaryotes. Here we report the purification of ubiquitin-specific peptidase 49 (USP49) as a histone H2B-specific deubiquitinase and demonstrate that H2B deubiquitination by USP49 is required for efficient cotranscriptional splicing of a large set of exons. USP49 forms a complex with RuvB-like1 (RVB1) and SUG1 and specifically deubiquitinates histone H2B in vitro and in vivo. USP49 knockdown results in small changes in gene expression but affects the abundance of >9000 isoforms. Exons down-regulated in USP49 knockdown cells show both elevated levels of alternative splicing and a general decrease in splicing efficiency. Importantly, USP49 is relatively enriched at this set of exons. USP49 knockdown increased H2B ubiquitination (uH2B) levels at these exons as well as upstream 3' and downstream 5' intronic splicing elements. Change in H2B ubiquitination level, as modulated by USP49, regulates U1A and U2B association with chromatin and binding to nascent pre-mRNA. Although H3 levels are relatively stable after USP49 depletion, H2B levels at these exons are dramatically increased, suggesting that uH2B may enhance nucleosome stability. Therefore, this study identifies USP49 as a histone H2B-specific deubiquitinase and uncovers a critical role for H2B deubiquitination in cotranscriptional pre-mRNA processing events.


Asunto(s)
Histonas/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , ADN Helicasas/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas con Dominio LIM/metabolismo , Complejo de la Endopetidasa Proteasomal , Factores de Transcripción/metabolismo , Ubiquitina Tiolesterasa/aislamiento & purificación , Ubiquitinación
6.
Mol Cell ; 42(4): 438-50, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21596310

RESUMEN

We have identified human MBT domain-containing protein L3MBTL2 as an integral component of a protein complex that we termed Polycomb repressive complex 1 (PRC1)-like 4 (PRC1L4), given the copresence of PcG proteins RING1, RING2, and PCGF6/MBLR. PRC1L4 also contained E2F6 and CBX3/HP1γ, known to function in transcriptional repression. PRC1L4-mediated repression necessitated L3MBTL2 that compacted chromatin in a histone modification-independent manner. Genome-wide location analyses identified several hundred genes simultaneously bound by L3MBTL2 and E2F6, preferentially around transcriptional start sites that exhibited little overlap with those targeted by other E2Fs or by L3MBTL1, another MBT domain-containing protein that interacts with RB1. L3MBTL2-specific RNAi resulted in increased expression of target genes that exhibited a significant reduction in H2A lysine 119 monoubiquitination. Our findings highlight a PcG/MBT collaboration that attains repressive chromatin without entailing histone lysine methylation marks.


Asunto(s)
Cromatina/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células HEK293 , Histonas/genética , Humanos , Proteínas Nucleares/genética , Proteínas del Grupo Polycomb , Proteínas Represoras/genética , Factores de Transcripción/genética
7.
J Am Soc Nephrol ; 28(6): 1729-1740, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28028135

RESUMEN

Two metrics, a rise in serum creatinine concentration and a decrease in urine output, are considered tantamount to the injury of the kidney tubule and the epithelial cells thereof (AKI). Yet neither criterion emphasizes the etiology or the pathogenetic heterogeneity of acute decreases in kidney excretory function. In fact, whether decreased excretory function due to contraction of the extracellular fluid volume (vAKI) or due to intrinsic kidney injury (iAKI) actually share pathogenesis and should be aggregated in the same diagnostic group remains an open question. To examine this possibility, we created mouse models of iAKI and vAKI that induced a similar increase in serum creatinine concentration. Using laser microdissection to isolate specific domains of the kidney, followed by RNA sequencing, we found that thousands of genes responded specifically to iAKI or to vAKI, but very few responded to both stimuli. In fact, the activated gene sets comprised different, functionally unrelated signal transduction pathways and were expressed in different regions of the kidney. Moreover, we identified distinctive gene expression patterns in human urine as potential biomarkers of either iAKI or vAKI, but not both. Hence, iAKI and vAKI are biologically unrelated, suggesting that molecular analysis should clarify our current definitions of acute changes in kidney excretory function.


Asunto(s)
Lesión Renal Aguda/clasificación , Lesión Renal Aguda/genética , Transcriptoma , Animales , Femenino , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL
8.
J Proteome Res ; 16(3): 1121-1132, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28102081

RESUMEN

Labeling peptides with isobaric tags is a popular strategy in quantitative bottom-up proteomics. In this study, we labeled six breast tumor cell lysates (1.34 mg proteins per channel) using 10-plex tandem mass tag reagents and analyzed the samples on a Q Exactive HF Quadrupole-Orbitrap mass spectrometer. We identified a total of 8,706 proteins and 28,186 phosphopeptides, including 7,394 proteins and 23,739 phosphosites common to all channels. The majority of technical replicates correlated with a R2 ≥ 0.98, indicating minimum variability was introduced after labeling. Unsupervised hierarchical clustering of phosphopeptide data sets successfully classified the breast tumor samples into Her2 (epidermal growth factor receptor 2) positive and Her2 negative groups, whereas mRNA abundance did not. The tyrosine phosphorylation levels of receptor tyrosine kinases, phosphoinositide-3-kinase, protein kinase C delta, and Src homology 2, among others, were significantly higher in the Her2 positive than the Her2 negative group. Despite ratio compression in MS2-based experiments, we demonstrated the ratios calculated using an MS2 method are highly correlated (R2 > 0.65) with ratios obtained using MS3-based quantitation (using a Thermo Orbitrap Fusion mass spectrometer) with reduced ratio suppression. Given the deep coverage of global and phosphoproteomes, our data show that MS2-based quantitation using TMT can be successfully used for large-scale multiplexed quantitative proteomics.


Asunto(s)
Neoplasias de la Mama/patología , Proteómica/métodos , Coloración y Etiquetado , Línea Celular Tumoral , Análisis por Conglomerados , Femenino , Humanos , Espectrometría de Masas/métodos , Fosfopéptidos/análisis , Fosforilación , Receptor ErbB-2/análisis , Tirosina/metabolismo
9.
Nature ; 472(7344): 476-80, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21525932

RESUMEN

Reactive oxygen species (ROS) are essential components of the innate immune response against intracellular bacteria and it is thought that professional phagocytes generate ROS primarily via the phagosomal NADPH oxidase machinery. However, recent studies have suggested that mitochondrial ROS (mROS) also contribute to mouse macrophage bactericidal activity, although the mechanisms linking innate immune signalling to mitochondria for mROS generation remain unclear. Here we demonstrate that engagement of a subset of Toll-like receptors (TLR1, TLR2 and TLR4) results in the recruitment of mitochondria to macrophage phagosomes and augments mROS production. This response involves translocation of a TLR signalling adaptor, tumour necrosis factor receptor-associated factor 6 (TRAF6), to mitochondria, where it engages the protein ECSIT (evolutionarily conserved signalling intermediate in Toll pathways), which is implicated in mitochondrial respiratory chain assembly. Interaction with TRAF6 leads to ECSIT ubiquitination and enrichment at the mitochondrial periphery, resulting in increased mitochondrial and cellular ROS generation. ECSIT- and TRAF6-depleted macrophages have decreased levels of TLR-induced ROS and are significantly impaired in their ability to kill intracellular bacteria. Additionally, reducing macrophage mROS levels by expressing catalase in mitochondria results in defective bacterial killing, confirming the role of mROS in bactericidal activity. These results reveal a novel pathway linking innate immune signalling to mitochondria, implicate mROS as an important component of antibacterial responses and further establish mitochondria as hubs for innate immune signalling.


Asunto(s)
Macrófagos/inmunología , Macrófagos/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Receptores Toll-Like/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Catalasa/genética , Catalasa/metabolismo , Línea Celular , Inmunidad Innata , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagosomas/metabolismo , Salmonella/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
10.
Nature ; 475(7355): 240-3, 2011 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-21725323

RESUMEN

Mediator is a key regulator of eukaryotic transcription, connecting activators and repressors bound to regulatory DNA elements with RNA polymerase II (Pol II). In the yeast Saccharomyces cerevisiae, Mediator comprises 25 subunits with a total mass of more than one megadalton (refs 5, 6) and is organized into three modules, called head, middle/arm and tail. Our understanding of Mediator assembly and its role in regulating transcription has been impeded so far by limited structural information. Here we report the crystal structure of the essential Mediator head module (seven subunits, with a mass of 223 kilodaltons) at a resolution of 4.3 ångströms. Our structure reveals three distinct domains, with the integrity of the complex centred on a bundle of ten helices from five different head subunits. An intricate pattern of interactions within this helical bundle ensures the stable assembly of the head subunits and provides the binding sites for general transcription factors and Pol II. Our structural and functional data suggest that the head module juxtaposes transcription factor IIH and the carboxy-terminal domain of the largest subunit of Pol II, thereby facilitating phosphorylation of the carboxy-terminal domain of Pol II. Our results reveal architectural principles underlying the role of Mediator in the regulation of gene expression.


Asunto(s)
Complejo Mediador/química , Complejo Mediador/metabolismo , Saccharomyces cerevisiae/química , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Fosforilación , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Saccharomyces cerevisiae/enzimología , Relación Estructura-Actividad , Factor de Transcripción TFIIH/química , Factor de Transcripción TFIIH/metabolismo
11.
Mol Cell ; 36(3): 457-68, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19917253

RESUMEN

TGF-beta induces phosphorylation of the transcription factors Smad2 and Smad3 at the C terminus as well as at an interdomain linker region. TGF-beta-induced linker phosphorylation marks the activated Smad proteins for proteasome-mediated destruction. Here, we identify Nedd4L as the ubiquitin ligase responsible for this step. Through its WW domain, Nedd4L specifically recognizes a TGF-beta-induced phosphoThr-ProTyr motif in the linker region, resulting in Smad2/3 polyubiquitination and degradation. Nedd4L is not interchangeable with Smurf1, a ubiquitin ligase that targets BMP-activated, linker-phosphorylated Smad1. Nedd4L limits the half-life of TGF-beta-activated Smads and restricts the amplitude and duration of TGF-beta gene responses, and in mouse embryonic stem cells, it limits the induction of mesoendodermal fates by Smad2/3-activating factors. Hierarchical regulation is provided by SGK1, which phosphorylates Nedd4L to prevent binding of Smad2/3. Previously identified as a regulator of renal sodium channels, Nedd4L is shown here to play a broader role as a general modulator of Smad turnover during TGF-beta signal transduction.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Immunoblotting , Ratones , Datos de Secuencia Molecular , Ubiquitina-Proteína Ligasas Nedd4 , Fosforilación/efectos de los fármacos , Poliubiquitina/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Homología de Secuencia de Aminoácido , Transducción de Señal , Proteína Smad2/genética , Proteína smad3/genética , Ubiquitina-Proteína Ligasas/genética
12.
Mol Cell Proteomics ; 14(9): 2357-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25693799

RESUMEN

There is an increasing need in biology and clinical medicine to robustly and reliably measure tens to hundreds of peptides and proteins in clinical and biological samples with high sensitivity, specificity, reproducibility, and repeatability. Previously, we demonstrated that LC-MRM-MS with isotope dilution has suitable performance for quantitative measurements of small numbers of relatively abundant proteins in human plasma and that the resulting assays can be transferred across laboratories while maintaining high reproducibility and quantitative precision. Here, we significantly extend that earlier work, demonstrating that 11 laboratories using 14 LC-MS systems can develop, determine analytical figures of merit, and apply highly multiplexed MRM-MS assays targeting 125 peptides derived from 27 cancer-relevant proteins and seven control proteins to precisely and reproducibly measure the analytes in human plasma. To ensure consistent generation of high quality data, we incorporated a system suitability protocol (SSP) into our experimental design. The SSP enabled real-time monitoring of LC-MRM-MS performance during assay development and implementation, facilitating early detection and correction of chromatographic and instrumental problems. Low to subnanogram/ml sensitivity for proteins in plasma was achieved by one-step immunoaffinity depletion of 14 abundant plasma proteins prior to analysis. Median intra- and interlaboratory reproducibility was <20%, sufficient for most biological studies and candidate protein biomarker verification. Digestion recovery of peptides was assessed and quantitative accuracy improved using heavy-isotope-labeled versions of the proteins as internal standards. Using the highly multiplexed assay, participating laboratories were able to precisely and reproducibly determine the levels of a series of analytes in blinded samples used to simulate an interlaboratory clinical study of patient samples. Our study further establishes that LC-MRM-MS using stable isotope dilution, with appropriate attention to analytical validation and appropriate quality control measures, enables sensitive, specific, reproducible, and quantitative measurements of proteins and peptides in complex biological matrices such as plasma.


Asunto(s)
Proteínas de Neoplasias/sangre , Neoplasias/metabolismo , Péptidos/análisis , Proteómica/métodos , Cromatografía Liquida/métodos , Humanos , Marcaje Isotópico , Espectrometría de Masas/métodos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/aislamiento & purificación , Neoplasias/sangre , Péptidos/química , Reproducibilidad de los Resultados
13.
EMBO J ; 31(2): 443-56, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22045337

RESUMEN

Regulation of mtDNA expression is critical for maintaining cellular energy homeostasis and may, in principle, occur at many different levels. The leucine-rich pentatricopeptide repeat containing (LRPPRC) protein regulates mitochondrial mRNA stability and an amino-acid substitution of this protein causes the French-Canadian type of Leigh syndrome (LSFC), a neurodegenerative disorder characterized by complex IV deficiency. We have generated conditional Lrpprc knockout mice and show here that the gene is essential for embryonic development. Tissue-specific disruption of Lrpprc in heart causes mitochondrial cardiomyopathy with drastic reduction in steady-state levels of most mitochondrial mRNAs. LRPPRC forms an RNA-dependent protein complex that is necessary for maintaining a pool of non-translated mRNAs in mammalian mitochondria. Loss of LRPPRC does not only decrease mRNA stability, but also leads to loss of mRNA polyadenylation and the appearance of aberrant mitochondrial translation. The translation pattern without the presence of LRPPRC is misregulated with excessive translation of some transcripts and no translation of others. Our findings point to the existence of an elaborate machinery that regulates mammalian mtDNA expression at the post-transcriptional level.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/genética , Enfermedad de Leigh/genética , Mitocondrias Cardíacas/fisiología , Proteínas de Neoplasias/fisiología , Poliadenilación/fisiología , Biosíntesis de Proteínas/fisiología , Animales , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/análisis , Células HeLa , Humanos , Sustancias Macromoleculares , Ratones , Ratones Noqueados , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Especificidad de Órganos , Polinucleotido Adenililtransferasa , Estabilidad del ARN , ARN Mensajero , Proteínas de Unión al ARN/metabolismo
14.
Mol Cell ; 30(4): 498-506, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18498751

RESUMEN

The final outcome of protein polyubiquitylation is often proteasome-mediated proteolysis, meaning that "proofreading" of ubiquitylation by ubiquitin proteases (UBPs) is crucial. Transcriptional arrest can trigger ubiquitin-mediated proteolysis of RNA polymerase II (RNAPII) so a UBP reversing RNAPII ubiquitylation might be expected. Here, we show that Ubp3 deubiquitylates RNAPII in yeast. Genetic characterization of ubp3 cells is consistent with a role in elongation, and Ubp3 can be purified with RNAPII, Def1, and the elongation factors Spt5 and TFIIF. This Ubp3 complex deubiquitylates both mono- and polyubiquitylated RNAPII in vitro, and ubp3 cells have elevated levels of ubiquitylated RNAPII in vivo. Moreover, RNAPII is degraded faster in a ubp3 mutant after UV irradiation. Problems posed by damage-arrested RNAPII are thought to be resolved either by removing the damage or degrading the polymerase. In agreement with this, cells with compromised DNA repair are better equipped to survive UV damage when UPB3 is deleted.


Asunto(s)
Endopeptidasas/metabolismo , ARN Polimerasa II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Antimetabolitos/metabolismo , Supervivencia Celular , Reparación del ADN , Endopeptidasas/genética , Humanos , ARN Polimerasa II/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo , Ubiquitinación , Rayos Ultravioleta , Uracilo/análogos & derivados , Uracilo/metabolismo
15.
Mol Cell Proteomics ; 13(11): 3082-96, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25056937

RESUMEN

Cancer is responsible for many deaths and is a major source of healthcare expenditures. The identification of new, non-invasive biomarkers might allow improvement of the direct diagnostic or prognostic ability of already available tools. Here, we took the innovative approach of interrogating the activity of exopeptidases in the serum of cancer patients with the aim of establishing a distinction based on enzymatic function, instead of simple protein levels, as a means to biomarker discovery. We first analyzed two well-characterized mouse models of prostate cancer, each with a distinct genetic lesion, and established that broad exopeptidase and targeted aminopeptidase activity tests reveal proteolytic changes associated with tumor development. We also describe new peptide-based freeze-frame reagents uniquely suited to probe the altered balance of selected aminopeptidases, as opposed to the full array of exopeptidases, and/or their modulators in patient serum or plasma. One particular proteolytic activity was impaired in animals with aggressive disease relative to cancer-free littermates. We identified the protease in question as dipeptidyl peptidase 4 (DPP4) by analyzing selected knockout mice and evaluating the effect of specific inhibitors. DPP4 activity was also reduced in the sera of patients with metastatic prostate cancer relative to patients with localized disease or healthy controls. However, no significant differences in DPP4 serum levels were observed, which established the loss of activity as the result of impaired enzymatic function. Biochemical analysis indicated that reduced activity was the result not of post-translational modifications or allosteric changes, but instead of a low-molecular-weight inhibitor. After we adjusted for age and total prostate-specific antigen, reduced DPP4 activity remained a significant predictor of cancer status. The results of this proof-of-principle study suggest that DPP4 activity might be a potential blood-based indicator of the presence of metastatic cancer of prostatic origin, either by itself or, more likely, as a means to improve the sensitivity and specificity of existing markers.


Asunto(s)
Biomarcadores de Tumor/sangre , Dipeptidil Peptidasa 4/sangre , Dipeptidil Peptidasa 4/metabolismo , Neoplasias de la Próstata/sangre , Aminopeptidasas/sangre , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Dipeptidil Peptidasa 4/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Neoplasias Experimentales/sangre , Neoplasias Experimentales/diagnóstico , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteína p53 Supresora de Tumor/genética
16.
Nature ; 457(7225): 57-62, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19092802

RESUMEN

DNA double-stranded breaks present a serious challenge for eukaryotic cells. The inability to repair breaks leads to genomic instability, carcinogenesis and cell death. During the double-strand break response, mammalian chromatin undergoes reorganization demarcated by H2A.X Ser 139 phosphorylation (gamma-H2A.X). However, the regulation of gamma-H2A.X phosphorylation and its precise role in chromatin remodelling during the repair process remain unclear. Here we report a new regulatory mechanism mediated by WSTF (Williams-Beuren syndrome transcription factor, also known as BAZ1B)-a component of the WICH complex (WSTF-ISWI ATP-dependent chromatin-remodelling complex). We show that WSTF has intrinsic tyrosine kinase activity by means of a domain that shares no sequence homology to any known kinase fold. We show that WSTF phosphorylates Tyr 142 of H2A.X, and that WSTF activity has an important role in regulating several events that are critical for the DNA damage response. Our work demonstrates a new mechanism that regulates the DNA damage response and expands our knowledge of domains that contain intrinsic tyrosine kinase activity.


Asunto(s)
Daño del ADN , Histonas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Histonas/genética , Humanos , Ratones , Células 3T3 NIH , Nucleosomas/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Estructura Terciaria de Proteína , Factores de Transcripción/química , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
17.
Mol Cell Proteomics ; 12(9): 2623-39, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23689285

RESUMEN

Multiple reaction monitoring (MRM) mass spectrometry coupled with stable isotope dilution (SID) and liquid chromatography (LC) is increasingly used in biological and clinical studies for precise and reproducible quantification of peptides and proteins in complex sample matrices. Robust LC-SID-MRM-MS-based assays that can be replicated across laboratories and ultimately in clinical laboratory settings require standardized protocols to demonstrate that the analysis platforms are performing adequately. We developed a system suitability protocol (SSP), which employs a predigested mixture of six proteins, to facilitate performance evaluation of LC-SID-MRM-MS instrument platforms, configured with nanoflow-LC systems interfaced to triple quadrupole mass spectrometers. The SSP was designed for use with low multiplex analyses as well as high multiplex approaches when software-driven scheduling of data acquisition is required. Performance was assessed by monitoring of a range of chromatographic and mass spectrometric metrics including peak width, chromatographic resolution, peak capacity, and the variability in peak area and analyte retention time (RT) stability. The SSP, which was evaluated in 11 laboratories on a total of 15 different instruments, enabled early diagnoses of LC and MS anomalies that indicated suboptimal LC-MRM-MS performance. The observed range in variation of each of the metrics scrutinized serves to define the criteria for optimized LC-SID-MRM-MS platforms for routine use, with pass/fail criteria for system suitability performance measures defined as peak area coefficient of variation <0.15, peak width coefficient of variation <0.15, standard deviation of RT <0.15 min (9 s), and the RT drift <0.5min (30 s). The deleterious effect of a marginally performing LC-SID-MRM-MS system on the limit of quantification (LOQ) in targeted quantitative assays illustrates the use and need for a SSP to establish robust and reliable system performance. Use of a SSP helps to ensure that analyte quantification measurements can be replicated with good precision within and across multiple laboratories and should facilitate more widespread use of MRM-MS technology by the basic biomedical and clinical laboratory research communities.


Asunto(s)
Cromatografía Liquida/instrumentación , Cromatografía Liquida/métodos , Espectrometría de Masas/instrumentación , Espectrometría de Masas/métodos , Secuencia de Aminoácidos , Animales , Bovinos , Límite de Detección , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Estándares de Referencia , Programas Informáticos , Factores de Tiempo
18.
Nature ; 454(7200): 126-30, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18509338

RESUMEN

With the recent recognition of non-coding RNAs (ncRNAs) flanking many genes, a central issue is to obtain a full understanding of their potential roles in regulated gene transcription programmes, possibly through different mechanisms. Here we show that an RNA-binding protein, TLS (for translocated in liposarcoma), serves as a key transcriptional regulatory sensor of DNA damage signals that, on the basis of its allosteric modulation by RNA, specifically binds to and inhibits CREB-binding protein (CBP) and p300 histone acetyltransferase activities on a repressed gene target, cyclin D1 (CCND1) in human cell lines. Recruitment of TLS to the CCND1 promoter to cause gene-specific repression is directed by single-stranded, low-copy-number ncRNA transcripts tethered to the 5' regulatory regions of CCND1 that are induced in response to DNA damage signals. Our data suggest that signal-induced ncRNAs localized to regulatory regions of transcription units can act cooperatively as selective ligands, recruiting and modulating the activities of distinct classes of RNA-binding co-regulators in response to specific signals, providing an unexpected ncRNA/RNA-binding protein-based strategy to integrate transcriptional programmes.


Asunto(s)
Regulación hacia Abajo , ARN no Traducido/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Transcripción Genética , Regulación Alostérica , Proteína de Unión a CREB/antagonistas & inhibidores , Proteína de Unión a CREB/metabolismo , Línea Celular , Secuencia de Consenso , Ciclina D1/genética , Daño del ADN , Células HeLa , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Humanos , Oligonucleótidos/genética , Regiones Promotoras Genéticas/genética , ARN no Traducido/genética , Proteína FUS de Unión a ARN/genética
19.
Nature ; 454(7207): 961-7, 2008 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-18719582

RESUMEN

Brown fat can increase energy expenditure and protect against obesity through a specialized program of uncoupled respiration. Here we show by in vivo fate mapping that brown, but not white, fat cells arise from precursors that express Myf5, a gene previously thought to be expressed only in the myogenic lineage. We also demonstrate that the transcriptional regulator PRDM16 (PRD1-BF1-RIZ1 homologous domain containing 16) controls a bidirectional cell fate switch between skeletal myoblasts and brown fat cells. Loss of PRDM16 from brown fat precursors causes a loss of brown fat characteristics and promotes muscle differentiation. Conversely, ectopic expression of PRDM16 in myoblasts induces their differentiation into brown fat cells. PRDM16 stimulates brown adipogenesis by binding to PPAR-gamma (peroxisome-proliferator-activated receptor-gamma) and activating its transcriptional function. Finally, Prdm16-deficient brown fat displays an abnormal morphology, reduced thermogenic gene expression and elevated expression of muscle-specific genes. Taken together, these data indicate that PRDM16 specifies the brown fat lineage from a progenitor that expresses myoblast markers and is not involved in white adipogenesis.


Asunto(s)
Adipocitos Marrones/metabolismo , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Músculo Esquelético/metabolismo , Factores de Transcripción/metabolismo , Adipocitos Marrones/citología , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/citología , Animales , Células COS , Diferenciación Celular/genética , Línea Celular , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Masculino , Ratones , Desarrollo de Músculos/genética , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Factor 5 Regulador Miogénico/genética , PPAR gamma/genética , Factores de Transcripción/genética
20.
Proc Natl Acad Sci U S A ; 108(39): 16375-80, 2011 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-21930909

RESUMEN

We previously described four small molecules that reduced the growth of lung adenocarcinoma cell lines with either epidermal growth factor receptor (EGFR) or KRAS mutations in a high-throughout chemical screen. By combining affinity proteomics and gene expression analysis, we now propose superoxide dismutase 1 (SOD1) as the most likely target of one of these small molecules, referred to as lung cancer screen 1 (LCS-1). siRNAs against SOD1 slowed the growth of LCS-1 sensitive cell lines; conversely, expression of a SOD1 cDNA increased proliferation of H358 cells and reduced sensitivity of these cells to LCS-1. In addition, SOD1 enzymatic activity was inhibited in vitro by LCS-1 and two closely related analogs. These results suggest that SOD1 is an LCS-1-binding protein that may act in concert with mutant proteins, such as EGFR and KRAS, to promote cell growth, providing a therapeutic target for compounds like LCS-1.


Asunto(s)
Adenocarcinoma/patología , División Celular/efectos de los fármacos , Neoplasias Pulmonares/patología , Superóxido Dismutasa/metabolismo , Adenocarcinoma/enzimología , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , ARN Interferente Pequeño/genética , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA