Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Cell ; 164(3): 460-75, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824657

RESUMEN

Neurogenesis is initiated by the transient expression of the highly conserved proneural proteins, bHLH transcriptional regulators. Here, we discover a conserved post-translational switch governing the duration of proneural protein activity that is required for proper neuronal development. Phosphorylation of a single Serine at the same position in Scute and Atonal proneural proteins governs the transition from active to inactive forms by regulating DNA binding. The equivalent Neurogenin2 Threonine also regulates DNA binding and proneural activity in the developing mammalian neocortex. Using genome editing in Drosophila, we show that Atonal outlives its mRNA but is inactivated by phosphorylation. Inhibiting the phosphorylation of the conserved proneural Serine causes quantitative changes in expression dynamics and target gene expression resulting in neuronal number and fate defects. Strikingly, even a subtle change from Serine to Threonine appears to shift the duration of Atonal activity in vivo, resulting in neuronal fate defects.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neurogénesis , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Drosophila , Proteínas de Drosophila , Ojo/crecimiento & desarrollo , Ojo/ultraestructura , Discos Imaginales/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Retina/crecimiento & desarrollo , Alineación de Secuencia
2.
Mol Cell ; 68(4): 715-730.e5, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29129638

RESUMEN

The spindle assembly checkpoint (SAC) generates a diffusible protein complex that prevents anaphase until all chromosomes are properly attached to spindle microtubules. A key step in SAC initiation is the recruitment of MAD1 to kinetochores, which is generally thought to be governed by the microtubule-kinetochore (MT-KT) attachment status. However, we demonstrate that the recruitment of MAD1 via BUB1, a conserved kinetochore receptor, is not affected by MT-KT interactions in human cells. Instead, BUB1:MAD1 interaction depends on BUB1 phosphorylation, which is controlled by a biochemical timer that integrates counteracting kinase and phosphatase effects on BUB1 into a pulse-generating incoherent feedforward loop. We propose that this attachment-independent timer serves to rapidly activate the SAC at mitotic entry, before the attachment-sensing MAD1 receptors have become fully operational. The BUB1-centered timer is largely impervious to conventional anti-mitotic drugs, and it is, therefore, a promising therapeutic target to induce cell death through permanent SAC activation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/metabolismo , Proteínas de Ciclo Celular/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Huso Acromático/genética
3.
Chembiochem ; 22(5): 834-838, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33085143

RESUMEN

Phosphoprotein phosphatase-1 (PP1) is a key player in the regulation of phospho-serine (pSer) and phospho-threonine (pThr) dephosphorylation and is involved in a large fraction of cellular signaling pathways. Aberrant activity of PP1 has been linked to many diseases, including cancer and heart failure. Besides a well-established activity control by regulatory proteins, an inhibitory function for phosphorylation (p) of a Thr residue in the C-terminal intrinsically disordered tail of PP1 has been demonstrated. The associated phenotype of cell-cycle arrest was repeatedly proposed to be due to autoinhibition of PP1 through either conformational changes or substrate competition. Here, we use PP1 variants created by mutations and protein semisynthesis to differentiate between these hypotheses. Our data support the hypothesis that pThr exerts its inhibitory function by mediating protein complex formation rather than by a direct mechanism of structural changes or substrate competition.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/farmacología , Proteína Fosfatasa 1/antagonistas & inhibidores , Serina/química , Treonina/química , Humanos , Fosforilación , Unión Proteica , Conformación Proteica , Dominios Proteicos , Proteína Fosfatasa 1/genética
4.
J Cell Sci ; 131(13)2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29898919

RESUMEN

The ubiquitously expressed nuclear protein NIPP1 (also known as PPP1R8) recruits phosphoproteins for regulated dephosphorylation by the associated protein phosphatase PP1. To bypass the PP1 titration artifacts seen upon NIPP1 overexpression, we have engineered covalently linked fusions of PP1 and NIPP1, and demonstrate their potential to selectively explore the function of the PP1:NIPP1 holoenzyme. By using inducible stable cell lines, we show that PP1-NIPP1 fusions cause replication stress in a manner that requires both PP1 activity and substrate recruitment via the ForkHead Associated domain of NIPP1. More specifically, PP1-NIPP1 expression resulted in the build up of RNA-DNA hybrids (R-loops), enhanced chromatin compaction and a diminished repair of DNA double-strand breaks (DSBs), culminating in the accumulation of DSBs. These effects were associated with a reduced expression of DNA damage signaling and repair proteins. Our data disclose a key role for dephosphorylation of PP1:NIPP1 substrates in setting the threshold for DNA repair, and indicate that activators of this phosphatase hold therapeutic potential as sensitizers for DNA-damaging agents.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Endorribonucleasas/genética , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 1/genética , Proteínas de Unión al ARN/genética , Cromatina/genética , Cromatina/metabolismo , Dimerización , Endorribonucleasas/química , Endorribonucleasas/metabolismo , Expresión Génica , Células HEK293 , Humanos , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/metabolismo , ARN/genética , ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo
5.
J Biol Chem ; 293(39): 15152-15162, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30115685

RESUMEN

The protein Ser/Thr phosphatase PP1 catalyzes an important fraction of protein dephosphorylation events and forms highly specific holoenzymes through an association with regulatory interactors of protein phosphatase one (RIPPOs). The functional characterization of individual PP1 holoenzymes is hampered by the lack of straightforward strategies for substrate mapping. Because efficient substrate recruitment often involves binding to both PP1 and its associated RIPPO, here we examined whether PP1-RIPPO fusions can be used to trap substrates for further analysis. Fusions of an hypoactive point mutant of PP1 and either of four tested RIPPOs accumulated in HEK293T cells with their associated substrates and were co-immunoprecipitated for subsequent identification of the substrates by immunoblotting or MS analysis. Hypoactive fusions were also used to study RIPPOs themselves as substrates for associated PP1. In contrast, substrate trapping was barely detected with active PP1-RIPPO fusions or with nonfused PP1 or RIPPO subunits. Our results suggest that hypoactive fusions of PP1 subunits represent an easy-to-use tool for substrate identification of individual holoenzymes.


Asunto(s)
Núcleo Celular/química , Holoenzimas/química , Proteína Fosfatasa 1/química , Receptores de Neuropéptido Y/química , Animales , Sitios de Unión , Células COS , Núcleo Celular/genética , Chlorocebus aethiops/genética , Células HEK293 , Holoenzimas/genética , Humanos , Inmunoprecipitación , Fosforilación , Unión Proteica , Proteína Fosfatasa 1/genética , Receptores de Neuropéptido Y/genética , Especificidad por Sustrato
6.
EMBO J ; 33(22): 2704-20, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25298395

RESUMEN

Faithful chromosome segregation during mitosis is tightly regulated by opposing activities of Aurora B kinase and protein phosphatase-1 (PP1). PP1 function at kinetochores has been linked to SDS22, but the exact localization of SDS22 and how it affects PP1 are controversial. Here, we confirm that SDS22 is required for PP1 activity, but show that SDS22 does not normally localize to kinetochores. Instead, SDS22 is kept in solution by formation of a ternary complex with PP1 and inhibitor-3 (I3). Depletion of I3 does not affect the amount of PP1 at kinetochores but causes quantitative association of SDS22 with PP1 on KNL1 at the kinetochore. Such accumulation of SDS22 at kinetochores interferes with PP1 activity and inhibits Aurora B threonine-232 dephosphorylation, which leads to increased Aurora B activity in metaphase and persistence in anaphase accompanied with segregation defects. We propose a model in which I3 regulates an SDS22-mediated PP1 activation step in solution that precedes SDS22 dissociation and transfer of PP1 to kinetochores, and which is required for PP1 to efficiently antagonize Aurora B.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinetocoros/metabolismo , Modelos Biológicos , Proteína Fosfatasa 1/metabolismo , Huso Acromático/metabolismo , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Fosforilación/fisiología , Proteína Fosfatasa 1/genética , Huso Acromático/genética , Ubiquitina-Proteína Ligasas
7.
J Cell Sci ; 128(24): 4526-37, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26542020

RESUMEN

The serine/threonine protein phosphatase-1 (PP1) complex is a key regulator of the cell cycle. However, the redundancy of PP1 isoforms and the lack of specific inhibitors have hampered studies on the global role of PP1 in cell cycle progression in vertebrates. Here, we show that the overexpression of nuclear inhibitor of PP1 (NIPP1; also known as PPP1R8) in HeLa cells culminated in a prometaphase arrest, associated with severe spindle-formation and chromosome-congression defects. In addition, the spindle assembly checkpoint was activated and checkpoint silencing was hampered. Eventually, most cells either died by apoptosis or formed binucleated cells. The NIPP1-induced mitotic arrest could be explained by the inhibition of PP1 that was titrated away from other mitotic PP1 interactors. Consistent with this notion, the mitotic-arrest phenotype could be rescued by the overexpression of PP1 or the inhibition of the Aurora B kinase, which acts antagonistically to PP1. Finally, we demonstrate that the overexpression of NIPP1 also hampered colony formation and tumor growth in xenograft assays in a PP1-dependent manner. Our data show that the selective inhibition of PP1 can be used to induce cancer cell death through mitotic catastrophe.


Asunto(s)
Endorribonucleasas/metabolismo , Mitosis , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Muerte Celular , Endorribonucleasas/genética , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteínas de Unión al ARN/genética
8.
Nucleic Acids Res ; 43(12): 5771-84, 2015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-25990731

RESUMEN

Protein phosphatase-1 (PP1) is a key regulator of transcription and is targeted to promoter regions via associated proteins. However, the chromatin binding sites of PP1 have never been studied in a systematic and genome-wide manner. Methylation-based DamID profiling in HeLa cells has enabled us to map hundreds of promoter binding sites of PP1 and three of its major nuclear interactors, i.e. RepoMan, NIPP1 and PNUTS. Our data reveal that the α, ß and γ isoforms of PP1 largely bind to distinct subsets of promoters and can also be differentiated by their promoter binding pattern. PP1ß emerged as the major promoter-associated isoform and shows an overlapping binding profile with PNUTS at dozens of active promoters. Surprisingly, most promoter binding sites of PP1 are not shared with RepoMan, NIPP1 or PNUTS, hinting at the existence of additional, largely unidentified chromatin-targeting subunits. We also found that PP1 is not required for the global chromatin targeting of RepoMan, NIPP1 and PNUTS, but alters the promoter binding specificity of NIPP1. Our data disclose an unexpected specificity and complexity in the promoter binding of PP1 isoforms and their chromatin-targeting subunits.


Asunto(s)
Regiones Promotoras Genéticas , Proteína Fosfatasa 1/metabolismo , Animales , Sitios de Unión , Bovinos , Núcleo Celular/enzimología , Núcleo Celular/genética , Proteínas de Unión al ADN/metabolismo , Genoma , Células HeLa , Holoenzimas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Subunidades de Proteína/metabolismo , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Transcripción Genética
9.
Nucleic Acids Res ; 41(2): 842-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23241245

RESUMEN

The histone methyltransferase EZH2 regulates cell proliferation and differentiation by silencing Polycomb group target genes. NIPP1, a nuclear regulator of serine/threonine protein phosphatase 1 (PP1), has been implicated in the regulation of EZH2 occupancy at target loci, but the underlying mechanism is not understood. Here, we demonstrate that the phosphorylation of EZH2 by cyclin-dependent kinases at Thr416 creates a docking site for the ForkHead-associated domain of NIPP1. Recruited NIPP1 enables the net phosphorylation of EZH2 by inhibiting its dephosphorylation by PP1. Accordingly, a NIPP1-binding mutant of EZH2 is hypophosphorylated, and the knockdown of NIPP1 results in a reduced phosphorylation of endogenous EZH2. Conversely, the loss of PP1 is associated with a hyperphosphorylation of EZH2. A genome-wide promoter-binding profiling in HeLa cells revealed that the NIPP1-binding mutant shows a deficient association with about a third of the Polycomb target genes, and these are enriched for functions in proliferation. Our data identify PP1 as an EZH2 phosphatase and demonstrate that the phosphorylation-regulated association of EZH2 with proliferation-related targets depends on associated NIPP1.


Asunto(s)
Endorribonucleasas/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Proteínas de Unión al ARN/metabolismo , Animales , Proliferación Celular , Endorribonucleasas/química , Proteína Potenciadora del Homólogo Zeste 2 , Células HEK293 , Células HeLa , Humanos , Ratones , Modelos Moleculares , Fosfoproteínas Fosfatasas/química , Fosforilación , Complejo Represivo Polycomb 2/química , Dominios y Motivos de Interacción de Proteínas , Proteína Fosfatasa 1/metabolismo , Proteínas de Unión al ARN/química , Treonina/metabolismo
10.
Trends Biochem Sci ; 35(8): 450-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20399103

RESUMEN

Protein Ser/Thr phosphatase-1 (PP1) catalyzes the majority of eukaryotic protein dephosphorylation reactions in a highly regulated and selective manner. Recent studies have identified an unusually diversified PP1 interactome with the properties of a regulatory toolkit. PP1-interacting proteins (PIPs) function as targeting subunits, substrates and/or inhibitors. As targeting subunits, PIPs contribute to substrate selection by bringing PP1 into the vicinity of specific substrates and by modulating substrate specificity via additional substrate docking sites or blocking substrate-binding channels. Many of the nearly 200 established mammalian PIPs are predicted to be intrinsically disordered, a property that facilitates their binding to a large surface area of PP1 via multiple docking motifs. These novel insights offer perspectives for the therapeutic targeting of PP1 by interfering with the binding of PIPs or substrates.


Asunto(s)
Dominio Catalítico , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/metabolismo , Secuencias de Aminoácidos , Animales , Modelos Moleculares , Unión Proteica , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/genética , Especificidad por Sustrato
11.
J Biol Chem ; 288(33): 24200-12, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23836907

RESUMEN

Maternal embryonic leucine zipper kinase (MELK) belongs to the subfamily of AMP-activated Ser/Thr protein kinases. The expression of MELK is very high in glioblastoma-type brain tumors, but it is not clear how this contributes to tumor growth. Here we show that the siRNA-mediated loss of MELK in U87 MG glioblastoma cells causes a G1/S phase cell cycle arrest accompanied by cell death or a senescence-like phenotype that can be rescued by the expression of siRNA-resistant MELK. This cell cycle arrest is mediated by an increased expression of p21(WAF1/CIP1), an inhibitor of cyclin-dependent kinases, and is associated with the hypophosphorylation of the retinoblastoma protein and the down-regulation of E2F target genes. The increased expression of p21 can be explained by the consecutive activation of ATM (ataxia telangiectasia mutated), Chk2, and p53. Intriguingly, the activation of p53 in MELK-deficient cells is not due to an increased stability of p53 but stems from the loss of MDMX (mouse double minute-X), an inhibitor of p53 transactivation. The activation of the ATM-Chk2 pathway in MELK-deficient cells is associated with the accumulation of DNA double-strand breaks during replication, as demonstrated by the appearance of γH2AX foci. Replication stress in these cells is also illustrated by an increased number of stalled replication forks and a reduced fork progression speed. Our data indicate that glioblastoma cells have elevated MELK protein levels to better cope with replication stress during unperturbed S phase. Hence, MELK inhibitors hold great potential for the treatment of glioblastomas as such or in combination with DNA-damaging therapies.


Asunto(s)
Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Replicación del ADN , Glioblastoma/enzimología , Glioblastoma/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Fisiológico , Animales , Línea Celular Tumoral , Senescencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Roturas del ADN de Doble Cadena , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Ratones , Modelos Biológicos , Fenotipo , Proteína de Retinoblastoma/metabolismo , Fase S , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
12.
Stem Cells ; 31(6): 1051-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23404835

RESUMEN

Glioblastoma multiforme (GBM) is a life-threatening brain tumor. Accumulating evidence suggests that eradication of glioma stem-like cells (GSCs) in GBM is essential to achieve cure. The transcription factor FOXM1 has recently gained attention as a master regulator of mitotic progression of cancer cells in various organs. Here, we demonstrate that FOXM1 forms a protein complex with the mitotic kinase MELK in GSCs, leading to phosphorylation and activation of FOXM1 in a MELK kinase-dependent manner. This MELK-dependent activation of FOXM1 results in a subsequent increase in mitotic regulatory genes in GSCs. MELK-driven FOXM1 activation is regulated by the binding and subsequent trans-phosphorylation of FOXM1 by another kinase PLK1. Using mouse neural progenitor cells (NPCs), we found that transgenic expression of FOXM1 enhances, while siRNA-mediated gene silencing diminishes neurosphere formation, suggesting that FOXM1 is required for NPC growth. During tumorigenesis, FOXM1 expression sequentially increases as cells progress from NPCs, to pretumorigenic progenitors and GSCs. The antibiotic Siomycin A disrupts MELK-mediated FOXM1 signaling with a greater sensitivity in GSC compared to neural stem cell. Treatment with the first-line chemotherapy agent for GBM, Temozolomide, paradoxically enriches for both FOXM1 (+) and MELK (+) cells in GBM cells, and addition of Siomycin A to Temozolomide treatment in mice harboring GSC-derived intracranial tumors enhances the effects of the latter. Collectively, our data indicate that FOXM1 signaling through its direct interaction with MELK regulates key mitotic genes in GSCs in a PLK1-dependent manner and thus, this protein complex is a potential therapeutic target for GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Factores de Transcripción Forkhead/metabolismo , Glioblastoma/patología , Células Madre Neoplásicas/patología , Células-Madre Neurales/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Factores de Transcripción Forkhead/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Células HEK293 , Humanos , Ratones , Mitosis/efectos de los fármacos , Mitosis/genética , Mitosis/fisiología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Péptidos/farmacología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Temozolomida , Regulación hacia Arriba/efectos de los fármacos , Quinasa Tipo Polo 1
13.
Biochem J ; 456(1): 119-28, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23937259

RESUMEN

A cluster of phosphorylation sites in LRRK2 (leucine-rich repeat kinase 2), including Ser910, Ser935, Ser955 and Ser973, is important for PD (Parkinson's disease) pathogenesis as several PD-linked LRRK2 mutants are dephosphorylated at these sites. LRRK2 is also dephosphorylated in cells after pharmacological inhibition of its kinase activity, which is currently proposed as a strategy for disease-modifying PD therapy. Despite this importance of LRRK2 dephosphorylation in mutant LRRK2 pathological mechanism(s) and in LRRK2's response to inhibition, the mechanism by which this occurs is unknown. Therefore we aimed to identify the phosphatase for LRRK2. Using a panel of recombinant phosphatases, we found that PP1 (protein phosphatase 1) efficiently dephosphorylates LRRK2 in vitro. PP1 activity on LRRK2 dephosphorylation was confirmed in cells using PP1 inhibition to reverse LRRK2 dephosphorylation induced by the potent LRRK2 kinase inhibitor LRRK2-IN1 as well as in R1441G mutant LRRK2. We also found that PP1 and LRRK2 can form a complex in cells. Furthermore, we observed that PP1 inhibition modulates LRRK2's cellular phenotype by reducing skein-like LRRK2-positive structures associated with dephosphorylation. In conclusion, the present study reveals PP1 as the physiological LRRK2 phosphatase, responsible for LRRK2 dephosphorylation observed in PD mutant LRRK2 and after LRRK2 kinase inhibition.


Asunto(s)
Proteína Fosfatasa 1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Benzodiazepinonas/farmacología , Línea Celular , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Mutación , Enfermedad de Parkinson/enzimología , Fosforilación , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Pirimidinas/farmacología
14.
Nucleic Acids Res ; 40(9): 4025-39, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22210893

RESUMEN

Alternative pre-mRNA processing is a central element of eukaryotic gene regulation. The cell frequently alters the use of alternative exons in response to physiological stimuli. Ceramides are lipid-signaling molecules composed of sphingosine and a fatty acid. Previously, water-insoluble ceramides were shown to change alternative splicing and decrease SR-protein phosphorylation by activating protein phosphatase-1 (PP1). To gain further mechanistical insight into ceramide-mediated alternative splicing, we analyzed the effect of C6 pyridinium ceramide (PyrCer) on alternative splice site selection. PyrCer is a water-soluble ceramide analog that is under investigation as a cancer drug. We found that PyrCer binds to the PP1 catalytic subunit and inhibits the dephosphorylation of several splicing regulatory proteins containing the evolutionarily conserved RVxF PP1-binding motif (including PSF/SFPQ, Tra2-beta1 and SF2/ASF). In contrast to natural ceramides, PyrCer promotes phosphorylation of splicing factors. Exons that are regulated by PyrCer have in common suboptimal splice sites, are unusually short and share two 4-nt motifs, GAAR and CAAG. They are dependent on PSF/SFPQ, whose phosphorylation is regulated by PyrCer. Our results indicate that lipids can influence pre-mRNA processing by regulating the phosphorylation status of specific regulatory factors, which is mediated by protein phosphatase activity.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Ceramidas/farmacología , Inhibidores Enzimáticos/farmacología , Proteína Fosfatasa 1/antagonistas & inhibidores , Compuestos de Piridinio/farmacología , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Sitios de Unión , Exones , Células HEK293 , Células HeLa , Humanos , Fosforilación/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo
16.
Biochem J ; 443(1): 173-83, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22242915

RESUMEN

P-Rex1 is a GEF (guanine-nucleotide-exchange factor) for the small G-protein Rac that is activated by PIP3 (phosphatidylinositol 3,4,5-trisphosphate) and Gßγ subunits and inhibited by PKA (protein kinase A). In the present study we show that PP1α (protein phosphatase 1α) binds P-Rex1 through an RVxF-type docking motif. PP1α activates P-Rex1 directly in vitro, both independently of and additively to PIP3 and Gßγ. PP1α also substantially activates P-Rex1 in vivo, both in basal and PDGF (platelet-derived growth factor)- or LPA (lysophosphatidic acid)-stimulated cells. The phosphatase activity of PP1α is required for P-Rex1 activation. PP1ß, a close homologue of PP1α, is also able to activate P-Rex1, but less effectively. PP1α stimulates P-Rex1-mediated Rac-dependent changes in endothelial cell morphology. MS analysis of wild-type P-Rex1 and a PP1α-binding-deficient mutant revealed that endogenous PP1α dephosphorylates P-Rex1 on at least three residues, Ser834, Ser1001 and Ser1165. Site-directed mutagenesis of Ser1165 to alanine caused activation of P-Rex1 to a similar degree as did PP1α, confirming Ser1165 as a dephosphorylation site important in regulating P-Rex1 Rac-GEF activity. In summary, we have identified a novel mechanism for direct activation of P-Rex1 through PP1α-dependent dephosphorylation.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Proteína Fosfatasa 1/química , Secuencias de Aminoácidos , Animales , Aorta/citología , Forma de la Célula , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Unión Proteica , Proteína Fosfatasa 1/metabolismo , Estructura Terciaria de Proteína , Conejos , Porcinos , Proteína de Unión al GTP rac1/metabolismo
17.
Biochem J ; 443(1): 193-203, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22233421

RESUMEN

Recombinant muscle GYS1 (glycogen synthase 1) and recombinant liver GYS2 were phosphorylated by recombinant AMPK (AMP-activated protein kinase) in a time-dependent manner and to a similar stoichiometry. The phosphorylation site in GYS2 was identified as Ser7, which lies in a favourable consensus for phosphorylation by AMPK. Phosphorylation of GYS1 or GYS2 by AMPK led to enzyme inactivation by decreasing the affinity for both UDP-Glc (UDP-glucose) [assayed in the absence of Glc-6-P (glucose-6-phosphate)] and Glc-6-P (assayed at low UDP-Glc concentrations). Incubation of freshly isolated rat hepatocytes with the pharmacological AMPK activators AICA riboside (5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside) or A769662 led to persistent GYS inactivation and Ser7 phosphorylation, whereas inactivation by glucagon treatment was transient. In hepatocytes from mice harbouring a liver-specific deletion of the AMPK catalytic α1/α2 subunits, GYS2 inactivation by AICA riboside and A769662 was blunted, whereas inactivation by glucagon was unaffected. The results suggest that GYS inactivation by AMPK activators in hepatocytes is due to GYS2 Ser7 phosphorylation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucógeno Sintasa/metabolismo , Hepatocitos/enzimología , Hígado/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Quinasas Activadas por AMP/química , Secuencia de Aminoácidos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Apraxia Ideomotora , Compuestos de Bifenilo , Células Cultivadas , Secuencia de Consenso , Proteínas Quinasas Dependientes de AMP Cíclico/química , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/farmacología , Glucógeno Sintasa/química , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Pironas/farmacología , Ratas , Ratas Wistar , Ribonucleótidos/farmacología , Tiofenos/farmacología
18.
J Biol Chem ; 286(5): 3798-804, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21098020

RESUMEN

CDK9/cyclin T1, a key enzyme in HIV-1 transcription, is negatively regulated by 7SK RNA and the HEXIM1 protein. Dephosphorylation of CDK9 on Thr(186) by protein phosphatase 1 (PP1) in stress-induced cells or by protein phosphatase M1A in normally growing cells activates CDK9. Our previous studies showed that HIV-1 Tat protein binds to PP1 through the Tat Q(35)VCF(38) sequence, which is similar to the PP1-binding RVXF motif and that this interaction facilitates HIV-1 transcription. In the present study, we analyzed the effect of expression of the central domain of nuclear inhibitor of PP1 (cdNIPP1) in an engineered cell line and also when cdNIPP1 was expressed as part of HIV-1 pNL4-3 in place of nef. Stable expression of cdNIPP1 increased CDK9 phosphorylation on Thr(186) and the association of CDK9 with 7SK RNA. The stable expression of cdNIPP1 disrupted the interaction of Tat and PP1 and inhibited HIV-1 transcription. Expression of cdNIPP1 as a part of the HIV-1 genome inhibited HIV-1 replication. Our study provides a proof-of-concept for the future development of PP1-targeting compounds as inhibitors of HIV-1 replication.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Endorribonucleasas/fisiología , VIH-1/genética , Fosfoproteínas Fosfatasas/fisiología , Proteínas de Unión al ARN/fisiología , Transcripción Genética , Animales , Fármacos Anti-VIH , Línea Celular , Endorribonucleasas/genética , Productos del Gen tat/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Fosfoproteínas Fosfatasas/genética , Fosforilación , Proteína Fosfatasa 1/metabolismo , Proteínas de Unión al ARN/genética , Conejos , Treonina/metabolismo , Replicación Viral
19.
J Biol Chem ; 286(12): 10126-36, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21220421

RESUMEN

Alternative pre-mRNA splicing is a central element of eukaryotic gene expression. Its deregulation can lead to disease, and methods to change splice site selection are developed as potential therapies. Spinal muscular atrophy is caused by the loss of the SMN1 (survival of motoneuron 1) gene. A therapeutic avenue for spinal muscular atrophy treatment is to promote exon 7 inclusion of the almost identical SMN2 (survival of motoneuron 2) gene. The splicing factor tra2-beta1 promotes inclusion of this exon and is antagonized by protein phosphatase (PP) 1. To identify new compounds that promote exon 7 inclusion, we synthesized analogs of cantharidin, an inhibitor of PP1, and PP2A. Three classes of compounds emerged from these studies. The first class blocks PP1 and PP2A activity, blocks constitutive splicing in vitro, and promotes exon 7 inclusion in vivo. The second class has no measurable effect on PP1 activity but activates PP2A. This class represents the first compounds described with these properties. These compounds cause a dephosphorylation of Thr-33 of tra2-beta1, which promotes exon 7 inclusion. The third class had no detectable effect on phosphatase activity and could promote exon 7 via allosteric effects. Our data show that subtle changes in similar compounds can turn a phosphatase inhibitor into an activator. These chemically related compounds influence alternative splicing by distinct mechanisms.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Exones , Fibroblastos/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN/efectos de los fármacos , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Células Cultivadas , Niño , Humanos , Masculino , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Precursores del ARN/genética , Empalme del ARN/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
20.
Retrovirology ; 9: 94, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23140174

RESUMEN

BACKGROUND: HIV-1 transcription is activated by the viral Tat protein that recruits host positive transcription elongation factor-b (P-TEFb) containing CDK9/cyclin T1 to the HIV-1 promoter. P-TEFb in the cells exists as a lower molecular weight CDK9/cyclin T1 dimer and a high molecular weight complex of 7SK RNA, CDK9/cyclin T1, HEXIM1 dimer and several additional proteins. Our previous studies implicated CDK2 in HIV-1 transcription regulation. We also found that inhibition of CDK2 by iron chelators leads to the inhibition of CDK9 activity, suggesting a functional link between CDK2 and CDK9. Here, we investigate whether CDK2 phosphorylates CDK9 and regulates its activity. RESULTS: The siRNA-mediated knockdown of CDK2 inhibited CDK9 kinase activity and reduced CDK9 phosphorylation. Stable shRNA-mediated CDK2 knockdown inhibited HIV-1 transcription, but also increased the overall level of 7SK RNA. CDK9 contains a motif (90SPYNR94) that is consensus CDK2 phosphorylation site. CDK9 was phosphorylated on Ser90 by CDK2 in vitro. In cultured cells, CDK9 phosphorylation was reduced when Ser90 was mutated to an Ala. Phosphorylation of CDK9 on Ser90 was also detected with phospho-specific antibodies and it was reduced after the knockdown of CDK2. CDK9 expression decreased in the large complex for the CDK9-S90A mutant and was correlated with a reduced activity and an inhibition of HIV-1 transcription. In contrast, the CDK9-S90D mutant showed a slight decrease in CDK9 expression in both the large and small complexes but induced Tat-dependent HIV-1 transcription. Molecular modeling showed that Ser 90 of CDK9 is located on a flexible loop exposed to solvent, suggesting its availability for phosphorylation. CONCLUSION: Our data indicate that CDK2 phosphorylates CDK9 on Ser 90 and thereby contributes to HIV-1 transcription. The phosphorylation of Ser90 by CDK2 represents a novel mechanism of HIV-1 regulated transcription and provides a new strategy for activation of latent HIV-1 provirus.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 9 Dependiente de la Ciclina/metabolismo , Regulación Viral de la Expresión Génica , VIH-1/genética , Transcripción Genética , Línea Celular , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/química , Activación Enzimática/genética , Silenciador del Gen , Humanos , Modelos Moleculares , Mutación , Fosforilación , Factor B de Elongación Transcripcional Positiva/metabolismo , Unión Proteica , Conformación Proteica , Interferencia de ARN , ARN Viral/genética , ARN Viral/metabolismo , Serina/química
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda