Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Chembiochem ; 22(5): 834-838, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33085143

ABSTRACT

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.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , Protein Phosphatase 1/antagonists & inhibitors , Serine/chemistry , Threonine/chemistry , Humans , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Protein Phosphatase 1/genetics
2.
Structure ; 27(3): 507-518.e5, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30661852

ABSTRACT

SDS22 is an ancient regulator of protein phosphatase-1 (PP1). Our crystal structure of SDS22 shows that its twelve leucine-rich repeats adopt a banana-shaped fold that is shielded from solvent by capping domains at its extremities. Subsequent modeling and biochemical studies revealed that the concave side of SDS22 likely interacts with PP1 helices α5 and α6, which are distal from the binding sites of many previously described PP1 interactors. Accordingly, we found that SDS22 acts as a "third" subunit of multiple PP1 holoenzymes. The crystal structure of SDS22 also revealed a large basic surface patch that enables binding of a phosphorylated form of splicing factor BCLAF1. Taken together, our data provide insights into the formation of PP1:SDS22 and the recruitment of additional interaction proteins, such as BCLAF1.


Subject(s)
Protein Phosphatase 1/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Humans , Models, Molecular , Phosphorylation , Protein Binding , Protein Phosphatase 1/chemistry , Protein Structure, Secondary
3.
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
5.
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
6.
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
7.
Mol Cell ; 68(4): 715-730.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29129638

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/metabolism , Kinetochores/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Cell Cycle Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/genetics
9.
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
10.
Nat Struct Mol Biol ; 23(7): 663-72, 2016 07.
Article in English | MEDLINE | ID: mdl-27273514

ABSTRACT

Control of mRNA levels, a fundamental aspect in the regulation of gene expression, is achieved through a balance between mRNA synthesis and decay. E26-related gene (Erg) proteins are canonical transcription factors whose previously described functions are confined to the control of mRNA synthesis. Here, we report that ERG also regulates gene expression by affecting mRNA stability and identify the molecular mechanisms underlying this function in human cells. ERG is recruited to mRNAs via interaction with the RNA-binding protein RBPMS, and it promotes mRNA decay by binding CNOT2, a component of the CCR4-NOT deadenylation complex. Transcriptome-wide mRNA stability analysis revealed that ERG controls the degradation of a subset of mRNAs highly connected to Aurora signaling, whose decay during S phase is necessary for mitotic progression. Our data indicate that control of gene expression by mammalian transcription factors may follow a more complex scheme than previously anticipated, integrating mRNA synthesis and degradation.


Subject(s)
Mitosis , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Aurora Kinases/genetics , Aurora Kinases/metabolism , Cell Line, Tumor , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
11.
Oncoimmunology ; 5(2): e1083669, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27057467

ABSTRACT

Dendritic cell (DC)-based immunotherapy has yielded promising results against high-grade glioma (HGG). However, the efficacy of DC vaccines is abated by HGG-induced immunosuppression and lack of attention toward the immunogenicity of the tumor lysate/cells used for pulsing DCs. A literature analysis of DC vaccination clinical trials in HGG patients delineated the following two most predominantly applied methods for tumor lysate preparation: freeze-thaw (FT)-induced necrosis or FT-necrosis followed by X-ray irradiation. However, from the available clinical evidence, it is unclear which of both methodologies has superior immunogenic potential. Using an orthotopic HGG murine model (GL261-C57BL/6), we observed that prophylactic vaccination with DCs pulsed with irradiated FT-necrotic cells (compared to FT-necrotic cells only) prolonged overall survival by increasing tumor rejection in glioma-challenged mice. This was associated, both in prophylactic and curative vaccination setups, with an increase in brain-infiltrating Th1 cells and cytotoxic T lymphocytes (CTL), paralleled by a reduced accumulation of regulatory T cells, tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC). Further analysis showed that irradiation treatment of FT-necrotic cells considerably increased the levels of carbonylated proteins - a surrogate-marker of oxidation-associated molecular patterns (OAMPs). Through further application of antioxidants and hydrogen peroxide, we found a striking correlation between the amount of lysate-associated protein carbonylation/OAMPs and DC vaccine-mediated tumor rejection capacity thereby suggesting for the first time a role for protein carbonylation/OAMPs in at least partially mediating antitumor immunity. Together, these data strongly advocate the use of protein oxidation-inducing modalities like irradiation for increasing the immunogenicity of tumor lysate/cells used for pulsing DC vaccines.

12.
Cell ; 164(3): 460-75, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824657

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Neurogenesis , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila , Drosophila Proteins , Eye/growth & development , Eye/ultrastructure , Imaginal Discs/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Phosphorylation , Retina/growth & development , Sequence Alignment
13.
Nat Commun ; 6: 10215, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26674376

ABSTRACT

RepoMan is a scaffold for signalling by mitotic phosphatases at the chromosomes. During (pro)metaphase, RepoMan-associated protein phosphatases PP1 and PP2A-B56 regulate the chromosome targeting of Aurora-B kinase and RepoMan, respectively. Here we show that this task division is critically dependent on the phosphorylation of RepoMan by protein kinase Cyclin-dependent kinase 1 (Cdk1), which reduces the binding of PP1 but facilitates the recruitment of PP2A-B56. The inactivation of Cdk1 in early anaphase reverses this phosphatase switch, resulting in the accumulation of PP1-RepoMan to a level that is sufficient to catalyse its own chromosome targeting in a PP2A-independent and irreversible manner. Bulk-targeted PP1-RepoMan also inactivates Aurora B and initiates nuclear-envelope reassembly through dephosphorylation-mediated recruitment of Importin ß. Bypassing the Cdk1 regulation of PP1-RepoMan causes the premature dephosphorylation of its mitotic-exit substrates in prometaphase. Hence, the regulation of RepoMan-associated phosphatases by Cdk1 is essential for the timely dephosphorylation of their mitotic substrates.


Subject(s)
Aurora Kinase B/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/genetics , Mitosis/genetics , Nuclear Proteins/metabolism , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , beta Karyopherins/metabolism , Anaphase , CDC2 Protein Kinase , Cell Line, Tumor , Chromosomes/metabolism , Cyclin-Dependent Kinases/metabolism , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Humans , Microscopy, Confocal , Nuclear Envelope/metabolism , Phosphoric Monoester Hydrolases , Prometaphase
14.
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
15.
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
16.
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
17.
J Inorg Biochem ; 149: 1-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25890482

ABSTRACT

Protein phosphatase-1 (PP1) is a major protein Ser/Thr phosphatase in eukaryotic cells. Its activity depends on two metal ions in the catalytic site, which were identified as manganese in the bacterially expressed phosphatase. However, the identity of the metal ions in native PP1 is unknown. In this study, total reflection X-ray fluorescence (TXRF) was used to detect iron and zinc in PP1 that was purified from rabbit skeletal muscle. Metal exchange experiments confirmed that the distinct substrate specificity of recombinant and native PP1 is determined by the nature of their associated metals. We also found that the iron level associated with native PP1 is decreased by incubation with inhibitor-2, consistent with a function of inhibitor-2 as a PP1 chaperone.


Subject(s)
Catalytic Domain , Iron/chemistry , Protein Phosphatase 1/chemistry , Zinc/chemistry , Animals , Muscle, Skeletal/enzymology , Protein Phosphatase 1/antagonists & inhibitors , Rabbits
18.
Clin Transl Med ; 4: 11, 2015.
Article in English | MEDLINE | ID: mdl-25852826

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK) is a highly conserved serine/threonine kinase initially found to be expressed in a wide range of early embryonic cellular stages, and as a result has been implicated in embryogenesis and cell cycle control. Recent evidence has identified a broader spectrum of tissue expression pattern for this kinase than previously appreciated. MELK is expressed in several human cancers and stem cell populations. Unique spatial and temporal patterns of expression within these tissues suggest that MELK plays a prominent role in cell cycle control, cell proliferation, apoptosis, cell migration, cell renewal, embryogenesis, oncogenesis, and cancer treatment resistance and recurrence. These findings have important implications for our understanding of development, disease, and cancer therapeutics. Furthermore understanding MELK signaling may elucidate an added dimension of stem cell control.

19.
EMBO J ; 33(22): 2704-20, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25298395

ABSTRACT

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.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Kinetochores/metabolism , Models, Biological , Protein Phosphatase 1/metabolism , Spindle Apparatus/metabolism , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation/physiology , Protein Phosphatase 1/genetics , Spindle Apparatus/genetics , Ubiquitin-Protein Ligases
20.
J Cancer Res Clin Oncol ; 140(11): 1849-61, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24938434

ABSTRACT

PURPOSE: To explore differential methylation of HAAO, HOXD3, LGALS3, PITX2, RASSF1 and TDRD1 as a molecular tool to predict biochemical recurrence (BCR) in patients with high-risk prostate cancer (PCa). METHODS: A multiplexed nested methylation-specific PCR was applied to quantify promoter methylation of the selected markers in five cell lines, 42 benign prostatic hyperplasia (BPH) and 71 high-risk PCa tumor samples. Uni- and multivariate Cox regression models were used to assess the importance of the methylation level in predicting BCR. RESULTS: A PCa-specific methylation marker HAAO in combination with HOXD3 and a hypomethylation marker TDRD1 distinguished PCa samples (>90 % of tumor cells each) from BPH with a sensitivity of 0.99 and a specificity of 0.95. High methylation of PITX2, HOXD3 and RASSF1, as well as low methylation of TDRD1, appeared to be significantly associated with a higher risk for BCR (HR 3.96, 3.44, 2.80 and 2.85, correspondingly) after correcting for established risk factors. When DNA methylation was treated as a continuous variable, a two-gene model PITX2 × 0.020677 + HOXD3 × 0.0043132 proved to be the best predictor of BCR (HR 4.85) compared with the individual markers. This finding was confirmed in an independent set of 52 high-risk PCa tumor samples (HR 11.89). CONCLUSIONS: Differential promoter methylation of HOXD3, PITX2, RASSF1 and TDRD1 emerges as an independent predictor of BCR in high-risk PCa patients. A two-gene continuous DNA methylation model "PITX2 × 0.020677 + HOXD3 × 0.0043132" is a better predictor of BCR compared with individual markers.


Subject(s)
Carrier Proteins/genetics , DNA Methylation , Homeodomain Proteins/genetics , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Aged , Aged, 80 and over , Cell Cycle Proteins , Cell Line, Tumor , Humans , Kallikreins/blood , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Promoter Regions, Genetic , Proportional Hazards Models , Prostate-Specific Antigen/blood , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Homeobox Protein PITX2
SELECTION OF CITATIONS
SEARCH DETAIL
...