Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
J Biol Chem ; 293(47): 18031-18039, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30305391

RESUMEN

Germ cell proliferation is epigenetically controlled, mainly through DNA methylation and histone modifications. However, the pivotal epigenetic regulators of germ cell self-renewal and differentiation in postnatal testis are still poorly defined. The histone methyltransferase enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of Polycomb repressive complex 2, represses target genes through trimethylation of histone H3 at Lys-27 (H3K27me3), and interacts (in)directly with both protein phosphatase 1 (PP1) and nuclear inhibitor of PP1 (NIPP1). Here, we report that postnatal, testis-specific ablation of NIPP1 in mice results in loss of EZH2 and reduces H3K27me3 levels. Mechanistically, the NIPP1 deletion abrogated PP1-mediated EZH2 dephosphorylation at two cyclin-dependent kinase sites (Thr-345/487), thereby generating hyperphosphorylated EZH2, which is a substrate for proteolytic degradation. Accordingly, alanine mutation of these residues prolonged the half-life of EZH2 in male germ cells. Our study discloses a key role for the PP1:NIPP1 holoenzyme in stabilizing EZH2 and maintaining the H3K27me3 mark on genes that are important for germ cell development and spermatogenesis.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Eliminación de Gen , Péptidos y Proteínas de Señalización Intracelular/genética , Testículo/metabolismo , Animales , Proteína Potenciadora del Homólogo Zeste 2/genética , Histonas/genética , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Metilación , Ratones , Ratones Noqueados , Fosforilación , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteolisis , Espermatogénesis , Testículo/crecimiento & desarrollo
3.
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
4.
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
5.
Stem Cells ; 34(8): 2256-62, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27068806

RESUMEN

The Ppp1r8 gene encodes NIPP1, a nuclear interactor of protein phosphatase PP1. The deletion of NIPP1 is embryonic lethal at the gastrulation stage, which has hampered its functional characterization in adult tissues. Here, we describe the effects of a conditional deletion of NIPP1 in mouse liver epithelial cells. Ppp1r8(-/-) livers developed a ductular reaction, that is, bile-duct hyperplasia with associated fibrosis. The increased proliferation of biliary epithelial cells was at least partially due to an expansion of the progenitor cell compartment that was independent of liver injury. Gene-expression analysis confirmed an upregulation of progenitor cell markers in the liver knockout livers but showed no effect on the expression of liver-injury associated regulators of cholangiocyte differentiation markers. Consistent with an inhibitory effect of NIPP1 on progenitor cell proliferation, Ppp1r8(-/-) livers displayed an increased sensitivity to diet-supplemented 3,5-diethoxycarbonyl-1,4-dihydrocollidine, which also causes bile-duct hyperplasia through progenitor cell expansion. In contrast, the liver knockouts responded normally to injuries (partial hepatectomy, single CCl4 administration) that are restored through proliferation of differentiated parenchymal cells. Our data indicate that NIPP1 does not regulate the proliferation of hepatocytes but is a suppressor of biliary epithelial cell proliferation, including progenitor cells, in the adult liver. Stem Cells 2016;34:2256-2262.


Asunto(s)
Eliminación de Gen , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/citología , Células Madre/citología , Animales , Conductos Biliares/patología , Biomarcadores/metabolismo , Proliferación Celular , Hiperplasia , Hígado/metabolismo , Ratones Noqueados , Especificidad de Órganos , Células Madre/metabolismo , Regulación hacia Arriba
6.
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
7.
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
8.
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
9.
FEBS J ; 291(12): 2615-2635, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38303113

RESUMEN

Protein phosphatase-1 (PP1) complexed to nuclear inhibitor of PP1 (NIPP1) limits DNA repair through dephosphorylation of NIPP1-recruited substrates. However, the PP1:NIPP1 holoenzyme is completely inactive under basal conditions, hinting at a DNA damage-regulated activation mechanism. Here, we report that DNA damage caused the activation of PP1:NIPP1 after a time delay of several hours through phosphorylation of NIPP1 at the C-terminal tyrosine 335 (Y335) by a Src-family kinase. PP1:NIPP1 activation partially resulted from the dissociation of the C terminus of NIPP1 from the active site of PP1. In addition, the released Y335-phosphorylated C terminus interacted with the N terminus of NIPP1 to enhance substrate recruitment by the flanking forkhead-associated (FHA) domain. Constitutive activation of PP1:NIPP1 by knock-in of a phospho-mimicking (Y335E) NIPP1 mutant led to the hypo-phosphorylation of FHA ligands and an accumulation of DNA double-strand breaks. Our data indicate that PP1:NIPP1 activation through circularization of NIPP1 is a late response to DNA damage that contributes to the timely recovery from damage repair.


Asunto(s)
Daño del ADN , Proteína Fosfatasa 1 , Familia-src Quinasas , Fosforilación , Humanos , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/química , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/química , Reparación del ADN , Regulación Alostérica , Roturas del ADN de Doble Cadena , Células HEK293 , Unión Proteica , Péptidos y Proteínas de Señalización Intracelular
10.
J Invest Dermatol ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38431220

RESUMEN

NIPP1 is a ubiquitously expressed regulatory subunit of PP1. Its embryonic deletion in keratinocytes causes chronic sterile skin inflammation, epidermal hyperproliferation, and resistance to mutagens in adult mice. To explore the primary effects of NIPP1 deletion, we first examined hair cycle progression of NIPP1 skin knockouts (SKOs). The entry of the first hair cycle in the SKOs was delayed owing to prolonged quiescence of hair follicle stem cells. In contrast, the entry of the second hair cycle in the SKOs was advanced as a result of precocious activation of hair follicle stem cells. The epidermis of SKOs progressively accumulated senescent cells, and this cell-fate switch was accelerated by DNA damage. Primary keratinocytes from SKO neonates and human NIPP1-depleted HaCaT keratinocytes failed to proliferate and showed an increase in the expression of cell cycle inhibitors (p21, p16/Ink4a, and/or p19/Arf) and senescence-associated-secretory-phenotype factors as well as in DNA damage (γH2AX and 53BP1). Our data demonstrate that the primary effect of NIPP1 deletion in keratinocytes is a cell cycle arrest and premature senescence that gradually progresse to chronic senescence and likely contribute to the decreased sensitivity of SKOs to mutagens.

11.
Nat Commun ; 15(1): 5359, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918402

RESUMEN

SDS22 forms an inactive complex with nascent protein phosphatase PP1 and Inhibitor-3. SDS22:PP1:Inhibitor-3 is a substrate for the ATPase p97/VCP, which liberates PP1 for binding to canonical regulatory subunits. The exact role of SDS22 in PP1-holoenzyme assembly remains elusive. Here, we show that SDS22 stabilizes nascent PP1. In the absence of SDS22, PP1 is gradually lost, resulting in substrate hyperphosphorylation and a proliferation arrest. Similarly, we identify a female individual with a severe neurodevelopmental disorder bearing an unstable SDS22 mutant, associated with decreased PP1 levels. We furthermore find that SDS22 directly binds to Inhibitor-3 and that this is essential for the stable assembly of SDS22:PP1: Inhibitor-3, the recruitment of p97/VCP, and the extraction of SDS22 during holoenzyme assembly. SDS22 with a disabled Inhibitor-3 binding site co-transfers with PP1 to canonical regulatory subunits, thereby forming non-functional holoenzymes. Our data show that SDS22, through simultaneous interaction with PP1 and Inhibitor-3, integrates the major steps of PP1 holoenzyme assembly.


Asunto(s)
Proteína Fosfatasa 1 , Femenino , Humanos , Células HEK293 , Holoenzimas/metabolismo , Fosforilación , Unión Proteica , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/genética , Proteína que Contiene Valosina/metabolismo , Proteína que Contiene Valosina/genética
13.
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
14.
Nature ; 439(7078): 871-4, 2006 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-16357870

RESUMEN

The establishment and maintenance of epigenetic gene silencing is fundamental to cell determination and function. The essential epigenetic systems involved in heritable repression of gene activity are the Polycomb group (PcG) proteins and the DNA methylation systems. Here we show that the corresponding silencing pathways are mechanistically linked. We find that the PcG protein EZH2 (Enhancer of Zeste homolog 2) interacts-within the context of the Polycomb repressive complexes 2 and 3 (PRC2/3)-with DNA methyltransferases (DNMTs) and associates with DNMT activity in vivo. Chromatin immunoprecipitations indicate that binding of DNMTs to several EZH2-repressed genes depends on the presence of EZH2. Furthermore, we show by bisulphite genomic sequencing that EZH2 is required for DNA methylation of EZH2-target promoters. Our results suggest that EZH2 serves as a recruitment platform for DNA methyltransferases, thus highlighting a previously unrecognized direct connection between two key epigenetic repression systems.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/clasificación , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Proteínas Represoras/clasificación , Proteínas Represoras/metabolismo , Factores de Transcripción/clasificación , Factores de Transcripción/metabolismo , Inmunoprecipitación de Cromatina , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Células HeLa , Humanos , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas/genética , Unión Proteica , Especificidad por Sustrato
15.
Nucleic Acids Res ; 38(21): 7500-12, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20671031

RESUMEN

Polycomb group (PcG) proteins are key regulators of stem-cell and cancer biology. They mainly act as repressors of differentiation and tumor-suppressor genes. One key silencing step involves the trimethylation of histone H3 on Lys27 (H3K27) by EZH2, a core component of the Polycomb Repressive Complex 2 (PRC2). The mechanism underlying the initial recruitment of mammalian PRC2 complexes is not well understood. Here, we show that NIPP1, a regulator of protein Ser/Thr phosphatase-1 (PP1), forms a complex with PP1 and PRC2 components on chromatin. The knockdown of NIPP1 or PP1 reduced the association of EZH2 with a subset of its target genes, whereas the overexpression of NIPP1 resulted in a retargeting of EZH2 from fully repressed to partially active PcG targets. However, the expression of a PP1-binding mutant of NIPP1 (NIPP1m) did not cause a redistribution of EZH2. Moreover, mapping of the chromatin binding sites with the DamID technique revealed that NIPP1 was associated with multiple PcG target genes, including the Homeobox A cluster, whereas NIPP1m showed a deficient binding at these loci. We propose that NIPP1 associates with a subset of PcG targets in a PP1-dependent manner and thereby contributes to the recruitment of the PRC2 complex.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/análisis , Endorribonucleasas/metabolismo , N-Metiltransferasa de Histona-Lisina/análisis , Fosfoproteínas Fosfatasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/análisis , Sitios de Unión , Línea Celular , Cromatina/química , Cromatina/enzimología , Endorribonucleasas/análisis , Endorribonucleasas/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2 , Histona Metiltransferasas , Humanos , Fosfoproteínas Fosfatasas/análisis , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/fisiología , Interferencia de ARN , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/antagonistas & inhibidores
16.
Nat Commun ; 12(1): 6116, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675204

RESUMEN

Critical cancer pathways often cannot be targeted because of limited efficiency crossing cell membranes. Here we report the development of a Salmonella-based intracellular delivery system to address this challenge. We engineer genetic circuits that (1) activate the regulator flhDC to drive invasion and (2) induce lysis to release proteins into tumor cells. Released protein drugs diffuse from Salmonella containing vacuoles into the cellular cytoplasm where they interact with their therapeutic targets. Control of invasion with flhDC increases delivery over 500 times. The autonomous triggering of lysis after invasion makes the platform self-limiting and prevents drug release in healthy organs. Bacterial delivery of constitutively active caspase-3 blocks the growth of hepatocellular carcinoma and lung metastases, and increases survival in mice. This success in targeted killing of cancer cells provides critical evidence that this approach will be applicable to a wide range of protein drugs for the treatment of solid tumors.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Caspasa 3/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Hepáticas/prevención & control , Neoplasias Pulmonares/tratamiento farmacológico , Salmonella/genética , Animales , Bacteriólisis , Carcinoma Hepatocelular/fisiopatología , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Sistemas de Liberación de Medicamentos/instrumentación , Femenino , Humanos , Neoplasias Hepáticas/secundario , Masculino , Ratones , Salmonella/fisiología , Salmonella typhimurium
17.
Oncogenesis ; 9(3): 30, 2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32123159

RESUMEN

Nuclear Inhibitor of PP1 (NIPP1) is a conserved regulatory subunit of protein phosphatase PP1. The selective deletion of NIPP1 in mouse liver parenchymal cells or skin epidermal cells culminates in a late-onset hyperproliferation of a subset of resident progenitor cells. Although a hyperplastic phenotype is usually tumor promoting, we show here that the absence of NIPP1 conferred a strong resistance to chemically induced hepatocellular or skin carcinoma. The ablation of NIPP1 did not affect the metabolism of the administered mutagens (diethylnitrosamine or 7,12-dimethylbenz[a]anthracene), but reduced the conversion of mutagen-induced covalent DNA modifications into cancer-initiating mutations. This reduced sensitivity to mutagens correlated with an enhanced DNA-damage response and an augmented expression of rate-limiting DNA-repair proteins (MGMT in liver, XPD and XPG in skin), hinting at an increased DNA-repair capacity. Our data identify NIPP1 as a repressor of DNA repair and as a promising target for novel cancer prevention and treatment therapies.

18.
J Invest Dermatol ; 140(8): 1576-1588, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31972250

RESUMEN

Nuclear inhibitor of protein phosphatase 1 (NIPP1) is a ubiquitously expressed nuclear protein that regulates functions of protein serine/threonine phosphatase-1 in cell proliferation and lineage specification. The role of NIPP1 in tissue homeostasis is not fully understood. This study shows that the selective deletion of NIPP1 in mouse epidermis resulted in epidermal hyperproliferation, a reduced adherence of basal keratinocytes, and a gradual decrease in the stemness of hair follicle stem cells, culminating in hair loss. This complex phenotype was associated with chronic sterile skin inflammation and could be partially rescued by dexamethasone treatment. NIPP1-deficient keratinocytes massively expressed proinflammatory chemokines and immunomodulatory proteins in a cell-autonomous manner. Chemokines subsequently induced the recruitment and activation of immune cells, in particular conventional dendritic cells and Langerhans cells, accounting for the chronic inflammation phenotype. The data identifies NIPP1 as a key regulator of epidermal homeostasis and as a potential target for the treatment of inflammatory skin diseases.


Asunto(s)
Alopecia/inmunología , Quimiocinas/metabolismo , Dermatitis/inmunología , Epidermis/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Alopecia/genética , Alopecia/patología , Animales , Adhesión Celular/inmunología , Proliferación Celular/genética , Quimiocinas/inmunología , Dermatitis/genética , Dermatitis/patología , Modelos Animales de Enfermedad , Epidermis/inmunología , Folículo Piloso/inmunología , Folículo Piloso/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Queratinocitos/inmunología , Queratinocitos/patología , Ratones , Ratones Noqueados
19.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 16-30, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30056088

RESUMEN

Protein phosphatase 1 (PP1) catalyzes more than half of all phosphoserine/threonine dephosphorylation reactions in mammalian cells. In vivo PP1 does not exist as a free catalytic subunit but is always associated with at least one regulatory PP1-interacting protein (PIP) to generate a large set of distinct holoenzymes. Each PP1 complex controls the dephosphorylation of only a small subset of PP1 substrates. We screened the literature for genetically engineered mouse models and identified models for all PP1 isoforms and 104 PIPs. PP1 itself and at least 49 PIPs were connected to human disease-associated phenotypes. Additionally, phenotypes related to 17 PIPs were clearly linked to altered PP1 function, while such information was lacking for 32 other PIPs. We propose structural reverse genetics, which combines structural characterization of proteins with mouse genetics, to identify new PP1-related therapeutic targets. The available mouse models confirm the pleiotropic action of PP1 in health and diseases.


Asunto(s)
Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/fisiología , Empalme Alternativo , Animales , Enfermedad , Genotipo , Holoenzimas/metabolismo , Holoenzimas/fisiología , Humanos , Ratones , Modelos Animales , Fenotipo , Fosforilación , Isoformas de Proteínas , Genética Inversa/métodos , Especificidad por Sustrato/fisiología
20.
Biochim Biophys Acta ; 1769(9-10): 541-5, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17804093

RESUMEN

NIPP1 is a ubiquitously expressed nuclear protein that represses the transcription of targeted genes. Here we show that the transcriptional repression by NIPP1 is alleviated by the RNAi-mediated knockdown of EED and EZH2, two core components of the Polycomb Repressive Complex 2 (PRC2), and by the overexpression of a catalytically dead mutant of the histone methyltransferase EZH2. NIPP1 is present in a complex with EED and EZH2 in vivo and has distinct binding sites for these proteins. These data disclose an essential role for the PRC2 complex in the transcriptional repression by NIPP1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endorribonucleasas/metabolismo , Complejos Multiproteicos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Sitios de Unión/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Endorribonucleasas/genética , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Complejos Multiproteicos/genética , Fosfoproteínas Fosfatasas/genética , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Unión Proteica/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA