Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Commun ; 15(1): 5359, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918402

ABSTRACT

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.


Subject(s)
Protein Phosphatase 1 , Female , Humans , HEK293 Cells , Holoenzymes/metabolism , Phosphorylation , Protein Binding , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Valosin Containing Protein/metabolism , Valosin Containing Protein/genetics
2.
J Invest Dermatol ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38431220

ABSTRACT

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.

3.
FEBS J ; 291(12): 2615-2635, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38303113

ABSTRACT

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.


Subject(s)
DNA Damage , Protein Phosphatase 1 , src-Family Kinases , Phosphorylation , Humans , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Protein Phosphatase 1/chemistry , src-Family Kinases/metabolism , src-Family Kinases/genetics , src-Family Kinases/chemistry , DNA Repair , Allosteric Regulation , DNA Breaks, Double-Stranded , HEK293 Cells , Protein Binding , Intracellular Signaling Peptides and Proteins
4.
Nat Commun ; 12(1): 6116, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675204

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Caspase 3/administration & dosage , Drug Delivery Systems/methods , Liver Neoplasms/prevention & control , Lung Neoplasms/drug therapy , Salmonella/genetics , Animals , Bacteriolysis , Carcinoma, Hepatocellular/physiopathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Delivery Systems/instrumentation , Female , Humans , Liver Neoplasms/secondary , Male , Mice , Salmonella/physiology , Salmonella typhimurium
5.
Oncogenesis ; 9(3): 30, 2020 Mar 02.
Article in English | MEDLINE | ID: mdl-32123159

ABSTRACT

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.

6.
J Invest Dermatol ; 140(8): 1576-1588, 2020 08.
Article in English | MEDLINE | ID: mdl-31972250

ABSTRACT

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.


Subject(s)
Alopecia/immunology , Chemokines/metabolism , Dermatitis/immunology , Epidermis/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Alopecia/genetics , Alopecia/pathology , Animals , Cell Adhesion/immunology , Cell Proliferation/genetics , Chemokines/immunology , Dermatitis/genetics , Dermatitis/pathology , Disease Models, Animal , Epidermis/immunology , Hair Follicle/immunology , Hair Follicle/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Keratinocytes/immunology , Keratinocytes/pathology , Mice , Mice, Knockout
7.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 16-30, 2019 01.
Article in English | MEDLINE | ID: mdl-30056088

ABSTRACT

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.


Subject(s)
Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/physiology , Alternative Splicing , Animals , Disease , Genotype , Holoenzymes/metabolism , Holoenzymes/physiology , Humans , Mice , Models, Animal , Phenotype , Phosphorylation , Protein Isoforms , Reverse Genetics/methods , Substrate Specificity/physiology
9.
J Biol Chem ; 293(47): 18031-18039, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30305391

ABSTRACT

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.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Deletion , Intracellular Signaling Peptides and Proteins/genetics , Testis/metabolism , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Histones/genetics , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Methylation , Mice , Mice, Knockout , Phosphorylation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proteolysis , Spermatogenesis , Testis/growth & development
10.
J Biol Chem ; 293(39): 15152-15162, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30115685

ABSTRACT

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.


Subject(s)
Cell Nucleus/chemistry , Holoenzymes/chemistry , Protein Phosphatase 1/chemistry , Receptors, Neuropeptide Y/chemistry , Animals , Binding Sites , COS Cells , Cell Nucleus/genetics , Chlorocebus aethiops/genetics , HEK293 Cells , Holoenzymes/genetics , Humans , Immunoprecipitation , Phosphorylation , Protein Binding , Protein Phosphatase 1/genetics , Receptors, Neuropeptide Y/genetics , Substrate Specificity
11.
J Cell Sci ; 131(13)2018 07 09.
Article in English | MEDLINE | ID: mdl-29898919

ABSTRACT

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.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Endoribonucleases/genetics , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 1/genetics , RNA-Binding Proteins/genetics , Chromatin/genetics , Chromatin/metabolism , Dimerization , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Gene Expression , HEK293 Cells , Humans , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/metabolism , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
12.
Sci Rep ; 7(1): 13364, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042623

ABSTRACT

NIPP1 is one of the major nuclear interactors of protein phosphatase PP1. The deletion of NIPP1 in mice is early embryonic lethal, which has precluded functional studies in adult tissues. Hence, we have generated an inducible NIPP1 knockout model using a tamoxifen-inducible Cre recombinase transgene. The inactivation of the NIPP1 encoding alleles (Ppp1r8) in adult mice occurred very efficiently in testis and resulted in a gradual loss of germ cells, culminating in a Sertoli-cell only phenotype. Before the overt development of this phenotype Ppp1r8 -/- testis showed a decreased proliferation and survival capacity of cells of the spermatogenic lineage. A reduced proliferation was also detected after the tamoxifen-induced removal of NIPP1 from cultured testis slices and isolated germ cells enriched for undifferentiated spermatogonia, hinting at a testis-intrinsic defect. Consistent with the observed phenotype, RNA sequencing identified changes in the transcript levels of cell-cycle and apoptosis regulating genes in NIPP1-depleted testis. We conclude that NIPP1 is essential for mammalian spermatogenesis because it is indispensable for the proliferation and survival of progenitor germ cells, including (un)differentiated spermatogonia.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Protein Phosphatase 1/metabolism , Spermatogenesis , Animals , Biomarkers , Gene Deletion , Germ Cells/drug effects , Germ Cells/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Male , Mammals , Mice , Mice, Knockout , Mice, Transgenic , Spermatogenesis/drug effects , Spermatogenesis/genetics
14.
FEBS Lett ; 591(2): 415-424, 2017 01.
Article in English | MEDLINE | ID: mdl-28032891

ABSTRACT

The biotin identification (BioID) protocol uses a mutant of the biotin ligase BirA (BirA*) fused to a protein-of-interest to biotinylate proximate proteins in intact cells. Here, we show that two inactive halves of BirA* separately fused to a catalytic and regulatory subunit of protein phosphatase PP1 reconstitute a functional BirA* enzyme upon heterodimerization of the phosphatase subunits. We also demonstrate that this BirA* fragment complementation approach, termed split-BioID, can be used to screen for substrates and other protein interactors of PP1 holoenzymes. Split-BioID is a novel and versatile tool for the identification of (transient) interactors of protein dimers.


Subject(s)
Biological Assay/methods , Dimerization , Protein Interaction Mapping , Biotinylation , Genetic Complementation Test , HEK293 Cells , Humans , Reproducibility of Results
15.
Stem Cells ; 34(8): 2256-62, 2016 08.
Article in English | MEDLINE | ID: mdl-27068806

ABSTRACT

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.


Subject(s)
Gene Deletion , Intracellular Signaling Peptides and Proteins/metabolism , Liver/cytology , Stem Cells/cytology , Animals , Bile Ducts/pathology , Biomarkers/metabolism , Cell Proliferation , Hyperplasia , Liver/metabolism , Mice, Knockout , Organ Specificity , Stem Cells/metabolism , Up-Regulation
16.
J Cell Sci ; 128(24): 4526-37, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26542020

ABSTRACT

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.


Subject(s)
Endoribonucleases/metabolism , Mitosis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 1/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Cell Death , Endoribonucleases/genetics , HeLa Cells , Humans , Neoplasm Proteins/genetics , Neoplasms/genetics , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , RNA-Binding Proteins/genetics
17.
Biosci Rep ; 35(6)2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26431963

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK), a serine/threonine protein kinase, has oncogenic properties and is overexpressed in many cancer cells. The oncogenic function of MELK is attributed to its capacity to disable critical cell-cycle checkpoints and reduce replication stress. Most functional studies have relied on the use of siRNA/shRNA-mediated gene silencing. In the present study, we have explored the biological function of MELK using MELK-T1, a novel and selective small-molecule inhibitor. Strikingly, MELK-T1 triggered a rapid and proteasome-dependent degradation of the MELK protein. Treatment of MCF-7 (Michigan Cancer Foundation-7) breast adenocarcinoma cells with MELK-T1 induced the accumulation of stalled replication forks and double-strand breaks that culminated in a replicative senescence phenotype. This phenotype correlated with a rapid and long-lasting ataxia telangiectasia-mutated (ATM) activation and phosphorylation of checkpoint kinase 2 (CHK2). Furthermore, MELK-T1 induced a strong phosphorylation of p53 (cellular tumour antigen p53), a prolonged up-regulation of p21 (cyclin-dependent kinase inhibitor 1) and a down-regulation of FOXM1 (Forkhead Box M1) target genes. Our data indicate that MELK is a key stimulator of proliferation by its ability to increase the threshold for DNA-damage tolerance (DDT). Thus, targeting MELK by the inhibition of both its catalytic activity and its protein stability might sensitize tumours to DNA-damaging agents or radiation therapy by lowering the DNA-damage threshold.


Subject(s)
Azepines/administration & dosage , Benzamides/administration & dosage , Breast Neoplasms/genetics , DNA Damage/drug effects , Enzyme Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/biosynthesis , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics
18.
PLoS One ; 10(6): e0130651, 2015.
Article in English | MEDLINE | ID: mdl-26086362

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is a very heterogeneous disease with respect to clinical outcome. This study explored differential DNA methylation in a priori selected genes to diagnose PCa and predict clinical failure (CF) in high-risk patients. METHODS: A quantitative multiplex, methylation-specific PCR assay was developed to assess promoter methylation of the APC, CCND2, GSTP1, PTGS2 and RARB genes in formalin-fixed, paraffin-embedded tissue samples from 42 patients with benign prostatic hyperplasia and radical prostatectomy specimens of patients with high-risk PCa, encompassing training and validation cohorts of 147 and 71 patients, respectively. Log-rank tests, univariate and multivariate Cox models were used to investigate the prognostic value of the DNA methylation. RESULTS: Hypermethylation of APC, CCND2, GSTP1, PTGS2 and RARB was highly cancer-specific. However, only GSTP1 methylation was significantly associated with CF in both independent high-risk PCa cohorts. Importantly, trichotomization into low, moderate and high GSTP1 methylation level subgroups was highly predictive for CF. Patients with either a low or high GSTP1 methylation level, as compared to the moderate methylation groups, were at a higher risk for CF in both the training (Hazard ratio [HR], 3.65; 95% CI, 1.65 to 8.07) and validation sets (HR, 4.27; 95% CI, 1.03 to 17.72) as well as in the combined cohort (HR, 2.74; 95% CI, 1.42 to 5.27) in multivariate analysis. CONCLUSIONS: Classification of primary high-risk tumors into three subtypes based on DNA methylation can be combined with clinico-pathological parameters for a more informative risk-stratification of these PCa patients.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Prostatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Cell Line, Tumor , Glutathione S-Transferase pi/genetics , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Promoter Regions, Genetic , Proportional Hazards Models , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Risk , Treatment Failure
19.
Nucleic Acids Res ; 43(12): 5771-84, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25990731

ABSTRACT

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.


Subject(s)
Promoter Regions, Genetic , Protein Phosphatase 1/metabolism , Animals , Binding Sites , Cattle , Cell Nucleus/enzymology , Cell Nucleus/genetics , DNA-Binding Proteins/metabolism , Genome , HeLa Cells , Holoenzymes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Subunits/metabolism , RNA Polymerase II/metabolism , RNA-Binding Proteins/metabolism , Rats , Transcription, Genetic
20.
FEBS Lett ; 589(12): 1314-21, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25907536

ABSTRACT

The deletion of the protein phosphatase-1 (PP1) regulator known as Nuclear Inhibitor of PP1 (NIPP1) is embryonic lethal during gastrulation, hinting at a key role of PP1-NIPP1 in lineage specification. Consistent with this notion we show here that a mild, stable overexpression of NIPP1 in HeLa cells caused a massive induction of genes of the mesenchymal lineage, in particular smooth/cardiac-muscle and matrix markers. This reprogramming was associated with the formation of actin-based stress fibers and retracting filopodia, and a reduced proliferation potential. The NIPP1-induced mesenchymal transition required functional substrate and PP1-binding domains, suggesting that it involves the selective dephosphorylation of substrates of PP1-NIPP1.


Subject(s)
Endoribonucleases/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Neoplasm Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Processing, Post-Translational , RNA-Binding Proteins/metabolism , Transcriptional Activation , Binding Sites , Biomarkers/metabolism , Cell Proliferation , Cell Transdifferentiation , Endoribonucleases/chemistry , Endoribonucleases/genetics , HeLa Cells , Humans , Ligands , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Interaction Domains and Motifs , Protein Stability , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...