Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Biol Rep ; 49(5): 4079-4087, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35397714

RESUMEN

BACKGROUND: The cell division cycle is a process that is exquisitely controlled by a complex interplay between E3 ubiquitin ligases and deubiquitinating enzymes (DUBs). We have previously reported that the DUB USP13 regulates Aurora B levels along the cell cycle. That observation prompted us to explore any possible connection between USP13 and the APC/CCDH1, the major E3 controlling Aurora B levels in cells. METHODS: We performed immunoprecipitation assays followed by western-blotting to assess the interaction between USP13 and CDH1. The cellular effects of USP13 gain or loss of function were analyzed by transfection of FLAG-tagged USP13 plasmid or small interfering RNAs and short hairpin RNAs directed against USP13. The levels of CDH1 and other proteins were quantified in cell extracts by western-blotting. RESULTS: We found that USP13 binds to the APC/C adaptor CDH1. In addition, we report for the first time that USP13 controls CDH1 protein levels in cells: overexpression of USP13 increased CDH1 levels, whereas depletion of USP13 decreased CDH1 levels. CONCLUSIONS: We unveil the existing interplay between USP13 and CDH1: USP13 is capable of stabilizing CDH1 levels. We previously reported that USP13 stabilizes Aurora B in cells, a known substrate of the APC/CCDH1 E3 ubiquitin ligase, before their entry into mitosis. Altogether, our data identify and establish the USP13-CDH1-Aurora B axis as a new regulatory module required for flawless cell cycle progression in mammalian cells, whose misfunction may be involved in the rewiring of cell cycle pathways linked to cancer development.


Asunto(s)
Mitosis , Proteínas Serina-Treonina Quinasas , Ciclosoma-Complejo Promotor de la Anafase/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Mamíferos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
2.
Int J Cancer ; 148(10): 2579-2593, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33210294

RESUMEN

In non-small cell lung cancer (NSCLC), activating mutations in the epidermal growth factor receptor (EGFR) induce sensitivity to EGFR tyrosine kinase inhibitors. Despite impressive clinical responses, patients ultimately relapse as a reservoir of drug-tolerant cells persist, which ultimately leads to acquired resistance mechanisms. We performed an unbiased high-throughput siRNA screen to identify proteins that abrogate the response of EGFR-mutant NSCLC to EGFR-targeted therapy. The deubiquitinase USP13 was a top hit resulting from this screen. Targeting USP13 increases the sensitivity to EGFR inhibition with small molecules in vitro and in vivo. USP13 selectively stabilizes mutant EGFR in a peptidase-independent manner by counteracting the action of members of the Cbl family of E3 ubiquitin ligases. We conclude that USP13 is a strong mutant EGFR-specific cotarget that could improve the treatment efficacy of EGFR-targeted therapies.

3.
Int J Mol Sci ; 22(2)2021 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-33435251

RESUMEN

Lung cancer is one of the most frequently diagnosed cancers accounting for the highest number of cancer-related deaths in the world. Despite significant progress including targeted therapies and immunotherapy, the treatment of advanced lung cancer remains challenging. Targeted therapies are highly efficacious at prolonging life, but not curative. In prior work we have identified Ubiquitin Specific Protease 13 (USP13) as a potential target to significantly enhance the efficacy of mutant EGFR inhibition. The current study aimed to develop lead molecules for the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) by developing potent USP13 inhibitors initially starting from Spautin-1, the only available USP13 inhibitor. A SAR study was performed which revealed that increasing the chain length between the secondary amine and phenyl group and introducing a halogen capable of inducing a halogen bond at position 4' of the phenyl group, dramatically increased the activity. However, we could not confirm the binding between Spautin-1 (or its analogues) and USP13 using isothermal titration calorimetry (ITC) or thermal shift assay (TSA) but do not exclude binding under physiological conditions. Nevertheless, we found that the anti-proliferative activity displayed by Spautin-1 towards EGFR-mutant NSCLC cells in vitro was at least partially associated with kinase inhibition. In this work, we present N-[2-(substituted-phenyl)ethyl]-6-fluoro-4-quinazolinamines as promising lead compounds for the treatment of NSCLC. These analogues are significantly more effective towards EGFR-mutant NSCLC cells than Spautin-1 and act as potent never in mitosis A related kinase 4 (NEK4) inhibitors (IC50~1 µM) with moderate selectivity over other kinases.


Asunto(s)
Bencilaminas/farmacología , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Neoplasias Pulmonares/enzimología , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Quinazolinas/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bencilaminas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Relación Estructura-Actividad
4.
Dev Growth Differ ; 62(4): 232-242, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32130724

RESUMEN

Polycomb group (PcG) RING finger protein 5 (PCGF5) is a core component of the so-called Polycomb repressive complex 1.5 (PRC1.5), which is involved in epigenetic transcriptional repression. To explore the developmental function of Pcgf5, we generated Pcgf5 knockout (Pcgf5-/- ) mouse embryonic stem cell (mESC) lines with the help of CRISPR/Cas9 technology. We subjected the Pcgf5-/- and wild-type (WT) mESCs to a differentiation protocol toward mesodermal-cardiac cell types as aggregated embryoid bodies (EBs) and we found that knockout of Pcgf5 delayed the generation of the three germ layers, especially the ectoderm. Further, disruption of Pcgf5 impacted the epithelial-mesenchymal transition during EB morphogenesis and differentially affected the gene expression of essential developmental signaling pathways such as Nodal and Wnt. Finally, we also unveiled that loss of Pcgf5 induced the repression of genes involved in the Notch pathway, which may explain the enhancement of cardiomyocyte maturation and the dampening of ectodermal-neural differentiation observed in the Pcgf5-/- EBs.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Transducción de Señal , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Ratones , Ratones Noqueados , Proteínas del Grupo Polycomb/deficiencia , Proteínas del Grupo Polycomb/genética , Transducción de Señal/genética
5.
Proc Natl Acad Sci U S A ; 114(22): E4462-E4471, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28500272

RESUMEN

The molecular pathogenesis of bipolar disorder (BPD) is poorly understood. Using human-induced pluripotent stem cells (hiPSCs) to unravel such mechanisms in polygenic diseases is generally challenging. However, hiPSCs from BPD patients responsive to lithium offered unique opportunities to discern lithium's target and hence gain molecular insight into BPD. By profiling the proteomics of BDP-hiPSC-derived neurons, we found that lithium alters the phosphorylation state of collapsin response mediator protein-2 (CRMP2). Active nonphosphorylated CRMP2, which binds cytoskeleton, is present throughout the neuron; inactive phosphorylated CRMP2, which dissociates from cytoskeleton, exits dendritic spines. CRMP2 elimination yields aberrant dendritogenesis with diminished spine density and lost lithium responsiveness (LiR). The "set-point" for the ratio of pCRMP2:CRMP2 is elevated uniquely in hiPSC-derived neurons from LiR BPD patients, but not with other psychiatric (including lithium-nonresponsive BPD) and neurological disorders. Lithium (and other pathway modulators) lowers pCRMP2, increasing spine area and density. Human BPD brains show similarly elevated ratios and diminished spine densities; lithium therapy normalizes the ratios and spines. Consistent with such "spine-opathies," human LiR BPD neurons with abnormal ratios evince abnormally steep slopes for calcium flux; lithium normalizes both. Behaviorally, transgenic mice that reproduce lithium's postulated site-of-action in dephosphorylating CRMP2 emulate LiR in BPD. These data suggest that the "lithium response pathway" in BPD governs CRMP2's phosphorylation, which regulates cytoskeletal organization, particularly in spines, modulating neural networks. Aberrations in the posttranslational regulation of this developmentally critical molecule may underlie LiR BPD pathogenesis. Instructively, examining the proteomic profile in hiPSCs of a functional agent-even one whose mechanism-of-action is unknown-might reveal otherwise inscrutable intracellular pathogenic pathways.


Asunto(s)
Trastorno Bipolar , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Litio/farmacología , Modelos Biológicos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Animales , Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo , Trastorno Bipolar/fisiopatología , Química Encefálica , Calcio/metabolismo , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteómica
6.
Proc Natl Acad Sci U S A ; 108(22): 9119-24, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21571647

RESUMEN

The ubiquitin-recognition protein Ufd1 facilitates clearance of misfolded proteins through the endoplasmic reticulum (ER)-associated degradation (ERAD) pathway. Here we report that prolonged ER stress represses Ufd1 expression to trigger cell cycle delay, which contributes to ERAD. Remarkably, down-regulation of Ufd1 enhances ubiquitination and destabilization of Skp2 mediated by the anaphase-promoting complex or cyclosome bound to Cdh1 (APC/C(Cdh1)), resulting in accumulation of the cyclin-dependent kinase inhibitor p27 and a concomitant cell cycle delay during the G1 phase that enables more efficient clearance of misfolded proteins. Mechanistically, nuclear Ufd1 recruits the deubiquitinating enzyme USP13 to counteract APC/C(Cdh1)-mediated ubiquitination of Skp2. Our data identify a coordinated cell cycle response to prolonged ER stress through regulation of the Cdh1-Skp2-p27 axis by Ufd1 and USP13.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas/fisiología , Saccharomyces cerevisiae/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Sitios de Unión , Ciclo Celular , Separación Celular , Regulación hacia Abajo , Citometría de Flujo , Regulación Fúngica de la Expresión Génica , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mutación , Estructura Terciaria de Proteína , Proteínas/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Tunicamicina/farmacología
7.
Nat Cell Biol ; 8(10): 1084-94, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16964245

RESUMEN

Xenopus RINGO/Speedy (XRINGO) is a potent inducer of oocyte meiotic maturation that can directly activate Cdk1 and Cdk2. Here, we show that endogenous XRINGO protein accumulates transiently during meiosis I entry and then is downregulated. This tight regulation of XRINGO expression is the consequence of two interconnected mechanisms: processing and degradation. XRINGO processing involves recognition of at least three distinct phosphorylated recognition motifs by the SCF(betaTrCP) ubiquitin ligase, followed by proteasome-mediated limited degradation, resulting in an amino-terminal XRINGO fragment. XRINGO processing is directly stimulated by several kinases, including protein kinase A and glycogen synthase kinase-3beta, and may contribute to the maintenance of G2 arrest. On the other hand, XRINGO degradation after meiosis I is mediated by the ubiquitin ligase Siah-2, which probably requires phosphorylation of XRINGO on Ser 243 and may be important for the omission of S phase at the meiosis-I-meiosis-II transition in Xenopus oocytes.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Fase G2 , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Meiosis , Oocitos/citología , Oocitos/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Ligasas SKP Cullina F-box/metabolismo , Xenopus laevis
8.
Cancers (Basel) ; 15(9)2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37174055

RESUMEN

Tyrosine kinase inhibitors (TKI) targeting the epidermal growth factor receptor (EGFR) have significantly prolonged survival in EGFR-mutant non-small cell lung cancer patients. However, the development of resistance mechanisms prohibits the curative potential of EGFR TKIs. Combination therapies emerge as a valuable approach to preventing or delaying disease progression. Here, we investigated the combined inhibition of polo-like kinase 1 (PLK1) and EGFR in TKI-sensitive EGFR-mutant NSCLC cells. The pharmacological inhibition of PLK1 destabilized EGFR levels and sensitized NSCLC cells to Osimertinib through induction of apoptosis. In addition, we found that c-Cbl, a ubiquitin ligase of EGFR, is a direct phosphorylation target of PLK1 and PLK1 impacts the stability of c-Cbl in a kinase-dependent manner. In conclusion, we describe a novel interaction between mutant EGFR and PLK1 that may be exploited in the clinic. Co-targeting PLK1 and EGFR may improve and prolong the clinical response to EGFR TKI in patients with an EGFR-mutated NSCLC.

9.
Proc Natl Acad Sci U S A ; 106(31): 12676-81, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19651615

RESUMEN

The p53 tumor suppressor protein is a key regulator of cellular proliferation and survival whose function is tightly regulated at the levels of transcription and protein stability. Here, we unveil the fine control of p53 on translationally active polysomes. We have previously reported that Ubc13, an E2 ubiquitin-conjugating enzyme, directly regulates p53 localization and transcriptional activity. We now demonstrate that the association of p53 and Ubc13 on polysomes requires ongoing translation and results in p53 ubiquitination that interferes with its tetramerization. JNK phosphorylation of p53 at Threonine 81 occurring on polysomes is required for the dissociation of Ubc13 from p53, leading to p53 multimerization and transcriptional activation. Inhibition of JNK activity or expression of a nonphosphorylatable mutant of p53 maintains an Ubc13-p53 complex that inhibits p53 multimerization. Our findings reveal a layer in the regulation of p53 multimerization that requires the concerted action of JNK and Ubc13 on polysome-bound p53.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Proteína p53 Supresora de Tumor/química , Enzimas Ubiquitina-Conjugadoras/fisiología , Sistema de Señalización de MAP Quinasas , Polirribosomas/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/química
10.
Cell Death Dis ; 13(7): 611, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840561

RESUMEN

Non-small cell lung cancer (NSCLC) patients harboring activating mutations in epidermal growth factor receptor (EGFR) are sensitive to therapy with EGFR tyrosine kinase inhibitors (TKI). Despite remarkable clinical responses using EGFR TKI, surviving drug tolerant cells serve as a reservoir from which drug resistant tumors may emerge. This study addresses the need for improved efficacy of EGFR TKI by identifying targets involved in functional drug tolerance against them. To this aim, a high-throughput siRNA kinome screen was performed using two EGFR TKI-sensitive EGFR-mutant NSCLC cell lines in the presence/absence of the second-generation EGFR TKI afatinib. From the screen, Serine/Threonine/Tyrosine Kinase 1 (STYK1) was identified as a target that when downregulated potentiates the effects of EGFR inhibition in vitro. We found that chemical inhibition of EGFR combined with the siRNA-mediated knockdown of STYK1 led to a significant decrease in cancer cell viability and anchorage-independent cell growth. Further, we show that STYK1 selectively interacts with mutant EGFR and that the interaction is disrupted upon EGFR inhibition. Finally, we identified fibroblast growth factor 1 (FGF1) as a downstream effector of STYK1 in NSCLC cells. Accordingly, downregulation of STYK1 counteracted the afatinib-induced upregulation of FGF1. Altogether, we unveil STYK1 as a valuable target to repress the pool of surviving drug tolerant cells arising upon EGFR inhibition. Co-targeting of EGFR and STYK1 could lead to a better overall outcome for NSCLC patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Tolerancia a Medicamentos , Neoplasias Pulmonares , Inhibidores de Proteínas Quinasas , Afatinib/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Resistencia a Antineoplásicos/genética , Tolerancia a Medicamentos/genética , Tolerancia a Medicamentos/fisiología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo
11.
J Biol Chem ; 285(19): 14217-28, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20220133

RESUMEN

c-Jun NH(2)-terminal Kinases (JNKs) play a central role in the cellular response to a wide variety of stress signals. After their activation, JNKs induce phosphorylation of substrates, which control proliferation, migration, survival, and differentiation. Recent studies suggest that JNKs may also play a role in cell cycle control, although the underlying mechanisms are largely unexplored. Here we show that JNK directly phosphorylates Cdc25C at serine 168 during G(2) phase of the cell cycle. Cdc25C phosphorylation by JNK negatively regulates its phosphatase activity and thereby Cdk1 activation, enabling a timely control of mitosis onset. Unrestrained phosphorylation by JNK, as obtained by a cell cycle-stabilized form of JNK or as seen in some human tumors, results in aberrant cell cycle progression. Additionally, UV irradiation-induced G(2)/M checkpoint requires inactivation of Cdc25C by JNK phosphorylation. JNK phosphorylation of Cdc25C as well as Cdc25A establishes a novel link between stress signaling and unperturbed cell cycle and checkpoint pathways.


Asunto(s)
División Celular/fisiología , Daño del ADN , Fase G2/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Mitosis/fisiología , Fosfatasas cdc25/metabolismo , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Fosforilación
12.
Nat Cell Biol ; 4(5): 337-42, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11951092

RESUMEN

Par-1 kinase is critical for polarization of the Drosophila melanogaster oocyte and the one-cell Caenorhabditis elegans embryo. Although Par-1 localizes specifically to the posterior pole in both cells, neither its targets nor its function at the posterior pole have been elucidated. Here we show that Drosophila Par-1 phosphorylates the posterior determinant Oskar (Osk) and demonstrate genetically that Par-1 is required for accumulation of Osk protein. We show in cell-free extracts that Osk protein is intrinsically unstable and that it is stabilized after phosphorylation by Par-1. Our data indicate that posteriorly localized Par-1 regulates posterior patterning by stabilizing Osk.


Asunto(s)
Tipificación del Cuerpo , Proteínas de Caenorhabditis elegans , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Regiones no Traducidas 3'/metabolismo , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Embrión no Mamífero/anatomía & histología , Femenino , Modelos Biológicos , Complejos Multienzimáticos/metabolismo , Ovario/química , Ovario/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes de Fusión/metabolismo , Extractos de Tejidos/química , Extractos de Tejidos/metabolismo , Ubiquitina/metabolismo , Xenopus laevis/metabolismo
13.
Trends Biochem Sci ; 31(6): 324-32, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16647857

RESUMEN

Proteolysis mediated by the ubiquitin-proteasome system is a crucial regulatory mechanism in signal transduction cascades of temporal cellular processes such as cell division. Two principal subtypes of modular ubiquitin ligase, the anaphase-promoting complex or cyclosome (APC/C) and the Skp1/Cullin-1/F-box protein complex, have emerged as essential regulators of key events in the cell cycle. The importance of these ligases is best illustrated by their roles in the checkpoint and repair pathways or in response to multiple stresses, where they affect activation of the M-phase-promoting factor or proper formation and/or maintenance of the mitotic spindle. Recent studies have considerably improved our understanding of the function of the concerted action of the phosphorylation and ubiquitin or SUMO systems in the regulation of the stability and activity of key components of the mitotic checkpoint.


Asunto(s)
Anafase/fisiología , División Celular/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína SUMO-1/metabolismo , Transducción de Señal/fisiología , Ubiquitina/metabolismo , Animales , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional/fisiología
14.
Biochimie ; 175: 120-124, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32454068

RESUMEN

The archaeal model organism Sulfolobus acidocaldarius possesses a TetR-like transcription factor that represses a 30-kb gene cluster encoding fatty acid metabolism enzymes. Interaction of this regulator, FadRSa, with acyl-CoA molecules causes a DNA dissociation, which may lead to a derepression of the gene cluster. Previously, a phosphoproteome analysis revealed the phosphorylation of three consecutive amino acids in the acyl-CoA ligand binding pocket. Here, we study this phosphorylation event and show that ArnC, a Hanks-type protein kinase, targets a threonine within the phosphoacceptor motif in vitro. Electrophoretic mobility shift assays using a phosphomimetic mutant of FadRSa demonstrate that the presence of negatively charged groups on the phosphoacceptor motif causes an inhibition of the ligand binding that desensitizes the responsiveness of the regulator to acyl-CoA molecules. Based on these observations, we propose a model in which phosphorylation of FadRSa in its ligand-binding pocket acts as an additional regulatory layer silencing acyl-CoA responsive derepression of fatty acid and lipid degradation. Moreover, given the recently discovered interplay between FadRSa and the chromosome structuring protein coalescin, FadRSa phosphorylation could also influence local chromosome conformation under specific cellular conditions.


Asunto(s)
Proteínas Arqueales/química , Modelos Moleculares , Proteínas Represoras/química , Sulfolobus acidocaldarius/química , Proteínas Arqueales/metabolismo , Sitios de Unión , Fosforilación , Proteínas Represoras/metabolismo , Sulfolobus acidocaldarius/metabolismo
15.
Oncogene ; 39(37): 6009-6023, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32772043

RESUMEN

Aurora B kinase plays essential roles in mitosis. Its protein levels increase before the onset of mitosis and sharply decrease during mitosis exit. The latter decrease is due to a balance between the actions of the E3 ubiquitin ligase anaphase-promoting complex or cyclosome (activated by the Cdh1 adapter), and the deubiquitinating enzyme USP35. Aurora B also executes important functions in interphase. Abnormal modulation of Aurora B in interphase leads to cell cycle defects often linked to aberrant chromosomal condensation and segregation. Very little is however known about how Aurora B levels are regulated in interphase. Here we found that USP13-associates with and stabilizes Aurora B in cells, especially before their entry into mitosis. In order for USP13 to exert its stabilizing effect on Aurora B, their association is promoted by the Aurora B-mediated phosphorylation of USP13 at Serine 114. We also present evidence that USP13 instigates Aurora B deubiquitination and/or protect it from degradation in a non-catalytic manner. In addition, we report that genetic or chemical modulation of the cellular levels/activity of USP13 affects unperturbed cell-cycle progression. Overall our study unveils the molecular and cellular connections of the USP13-Aurora B axis, which potentially participates in the rewiring of the cell cycle happening in cancer cells.


Asunto(s)
Aurora Quinasa B/metabolismo , Ciclo Celular/genética , Endopeptidasas/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Endopeptidasas/metabolismo , Estabilidad de Enzimas , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Fosforilación , Unión Proteica , Serina/metabolismo , Proteasas Ubiquitina-Específicas
16.
Methods Mol Biol ; 1449: 251-65, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27613041

RESUMEN

The anaphase-promoting complex or cyclosome (APC/C) is one of the major orchestrators of the cell division cycle in mammalian cells. The APC/C acts as a ubiquitin ligase that triggers sequential ubiquitylation of a significant number of substrates which will be eventually degraded by proteasomes during major transitions of the cell cycle. In this chapter, we present accessible methodologies to assess both in in vitro conditions and in cellular systems ubiquitylation reactions mediated by the APC/C. In addition, we also describe techniques to evidence the changes in protein stability provoked by modulation of the activity of the APC/C. Finally, specific methods to analyze interactors or posttranslational modifications of particular APC/C subunits are also discussed. Given the crucial role played by the APC/C in the regulation of the cell cycle, this review only focuses on its action and effects in actively proliferating cells.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclo Celular/fisiología , Ubiquitinación/fisiología , Ciclosoma-Complejo Promotor de la Anafase/genética , Animales , Ciclo Celular/genética , Humanos , Fosforilación/genética , Fosforilación/fisiología , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Ubiquitina/metabolismo , Ubiquitinación/genética
17.
Methods Mol Biol ; 1449: 267-78, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27613042

RESUMEN

SUMOylation is a widely used protein posttranslational mechanism capable of regulating substrates localization, stability, and/or activity. Identification and characterization of bona fide SUMO substrates is a laborious task but its discovery can shed light to exquisite and crucial regulatory signaling events occurring within the cell. Experiments performed in the SUMOylation field often demand a good understanding of the putative substrate's function and necessitate a solid knowledge regarding both in vitro and in vivo approaches. This contribution offers a simplified view into some of the most common experiments performed in biochemical and cell biological research of the SUMO pathway in mammalian systems. It also summarizes and updates well established protocols and tricks in order to improve the likelihood to obtain reliable and reproducible results.


Asunto(s)
Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación/fisiología , Animales , Humanos , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación/genética
18.
Stem Cell Reports ; 7(3): 527-542, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27569059

RESUMEN

Controlled differentiation of human embryonic stem cells (hESCs) can be utilized for precise analysis of cell type identities during early development. We established a highly efficient neural induction strategy and an improved analytical platform, and determined proteomic and phosphoproteomic profiles of hESCs and their specified multipotent neural stem cell derivatives (hNSCs). This quantitative dataset (nearly 13,000 proteins and 60,000 phosphorylation sites) provides unique molecular insights into pluripotency and neural lineage entry. Systems-level comparative analysis of proteins (e.g., transcription factors, epigenetic regulators, kinase families), phosphorylation sites, and numerous biological pathways allowed the identification of distinct signatures in pluripotent and multipotent cells. Furthermore, as predicted by the dataset, we functionally validated an autocrine/paracrine mechanism by demonstrating that the secreted protein midkine is a regulator of neural specification. This resource is freely available to the scientific community, including a searchable website, PluriProt.


Asunto(s)
Diferenciación Celular , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteoma , Proteómica , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Biología Computacional/métodos , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neuronas/citología , Proteómica/métodos , Transducción de Señal , Transcriptoma
19.
BMC Biochem ; 6: 27, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16321160

RESUMEN

BACKGROUND: The two myogenic regulatory factors Myf5 and MyoD are basic helix-loop-helix muscle transcription factors undergoing differential cell cycle dependent proteolysis in proliferating myoblasts. This regulated degradation results in the striking expression of these two factors at distinct phases of the cell cycle, and suggests that their precise and alternated disappearance is an important feature of myoblasts, maybe connected to the maintenance of the proliferative status and/or commitment to the myogenic lineage of these cells. One way to understand the biological function(s) of the cyclic expression of these proteins is to specifically alter their degradation, and to analyze the effects of their stabilization on cells. To this aim, we undertook the biochemical analysis of the mechanisms governing Myf5 mitotic degradation, using heterologous systems. RESULTS: We show here that mitotic degradation of Myf5 is conserved in non-myogenic cells, and is thus strictly under the control of the cell cycle apparatus. Using Xenopus egg extracts as an in vitro system to dissect the main steps of Myf5 mitotic proteolysis, we show that (1) Myf5 stability is regulated by a complex interplay of phosphorylation/dephosphorylation, probably involving various kinases and phosphatases, (2) Myf5 is ubiquitylated in mitotic extracts, and this is a prerequisite to its degradation by the proteasome and (3) at least in the Xenopus system, the E3 responsible for its mitotic degradation is not the APC/C (the major E3 during mitosis). CONCLUSION: Altogether, our data strongly suggest that the mitotic degradation of Myf5 by the ubiquitin-proteasome system is precisely controlled by multiple phosphorylation of the protein, and that the APC/C is not involved in this process.


Asunto(s)
Mitosis/fisiología , Factor 5 Regulador Miogénico/metabolismo , Animales , Femenino , Células HeLa , Humanos , Ratones , Mitosis/genética , Factor 5 Regulador Miogénico/genética , Fosforilación , Conejos , Xenopus
20.
FEBS Lett ; 518(1-3): 23-8, 2002 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-11997011

RESUMEN

Here we show that during the meiotic maturation of Xenopus oocytes, histone H3 becomes phosphorylated on serine-10 at about the time of maturation promoting factor activation and meiosis I entry. However, overexpression of cAMP-dependent protein kinase that blocks entry into M phase, also leads to massive serine-10 phosphorylation of histone H3 in intact Xenopus oocytes but does not cause chromosome condensation. We also show that the phosphorylation of histone H3 during oocyte maturation requires the activation of the mitogen-activated protein kinase/p90Rsk pathway. Our results indicate that in G2-arrested oocytes, which are about to enter M phase, histone H3 phosphorylation is not sufficient for chromosome condensation.


Asunto(s)
Histonas/metabolismo , Sistema de Señalización de MAP Quinasas , Oocitos/crecimiento & desarrollo , Proteínas Quinasas S6 Ribosómicas/fisiología , Xenopus laevis/embriología , Animales , Células Cultivadas , Cromosomas/ultraestructura , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , ADN/ultraestructura , Histonas/química , Meiosis , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oocitos/metabolismo , Oocitos/ultraestructura , Fosforilación , Fosfoserina/metabolismo , Transfección , Xenopus laevis/genética , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA