Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Commun ; 14(1): 5253, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37644036

ABSTRACT

Loss of the tumor suppressive activity of the protein phosphatase 2A (PP2A) is associated with cancer, but the underlying molecular mechanisms are unclear. PP2A holoenzyme comprises a heterodimeric core, a scaffolding A subunit and a catalytic C subunit, and one of over 20 distinct substrate-directing regulatory B subunits. Methylation of the C subunit regulates PP2A heterotrimerization, affecting B subunit binding and substrate specificity. Here, we report that the leucine carboxy methyltransferase (LCMT1), which methylates the L309 residue of the C subunit, acts as a suppressor of androgen receptor (AR) addicted prostate cancer (PCa). Decreased methyl-PP2A-C levels in prostate tumors is associated with biochemical recurrence and metastasis. Silencing LCMT1 increases AR activity and promotes castration-resistant prostate cancer growth. LCMT1-dependent methyl-sensitive AB56αCme heterotrimers target AR and its critical coactivator MED1 for dephosphorylation, resulting in the eviction of the AR-MED1 complex from chromatin and loss of target gene expression. Mechanistically, LCMT1 is regulated by S6K1-mediated phosphorylation-induced degradation requiring the ß-TRCP, leading to acquired resistance to anti-androgens. Finally, feedforward stabilization of LCMT1 by small molecule activator of phosphatase (SMAP) results in attenuation of AR-signaling and tumor growth inhibition in anti-androgen refractory PCa. These findings highlight methyl-PP2A-C as a prognostic marker and that the loss of LCMT1 is a major determinant in AR-addicted PCa, suggesting therapeutic potential for AR degraders or PP2A modulators in prostate cancer treatment.


Subject(s)
Prostatic Neoplasms , Protein Phosphatase 2 , Humans , Male , Androgen Antagonists , Leucine , Methyltransferases , Prostate , Prostatic Neoplasms/genetics , Protein Phosphatase 2/genetics
2.
Cell Rep ; 30(9): 3171-3182.e6, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130916

ABSTRACT

Protein phosphatase 2A (PP2A) is an important regulator of signal transduction pathways and a tumor suppressor. Phosphorylation of the PP2A catalytic subunit (PP2AC) at tyrosine 307 has been claimed to inactivate PP2A and was examined in more than 180 studies using commercial antibodies, but this modification was never identified using mass spectrometry. Here we show that the most cited pTyr307 monoclonal antibodies, E155 and F-8, are not specific for phosphorylated Tyr307 but instead are hampered by PP2AC methylation at leucine 309 or phosphorylation at threonine 304. Other pTyr307 antibodies are sensitive to PP2AC methylation as well, and some cross-react with pTyr residues in general, including phosphorylated hemagglutinin tags. We identify pTyr307 using targeted mass spectrometry after transient overexpression of PP2AC and Src kinase. Yet under such conditions, none of the tested antibodies show exclusive pTyr307 specificity. Thus, data generated using these antibodies need to be revisited, and the mechanism of PP2A inactivation needs to be redefined.


Subject(s)
Antibodies/metabolism , Antibody Specificity/immunology , Leucine/metabolism , Phosphotyrosine/metabolism , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antibody Specificity/drug effects , Cross Reactions/drug effects , Epidermal Growth Factor/pharmacology , HEK293 Cells , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Methylation , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Peptides/chemistry , Peptides/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vanadates/pharmacology , src-Family Kinases/metabolism
3.
Sci Signal ; 13(616)2020 01 28.
Article in English | MEDLINE | ID: mdl-31992581

ABSTRACT

The methyl-esterification of the C-terminal leucine of the protein phosphatase 2A (PP2A) catalytic (C) subunit is essential for the assembly of specific trimeric PP2A holoenzymes, and this region of the C subunit also contains two threonine and tyrosine phosphorylation sites. Most commercial antibodies-including the monoclonal antibody 1D6 that is part of a frequently used, commercial phosphatase assay kit-are directed toward the C terminus of the C subunit, raising questions as to their ability to recognize methylated and phosphorylated forms of the enzyme. Here, we tested several PP2A C antibodies, including monoclonal antibodies 1D6, 7A6, G-4, and 52F8 and the polyclonal antibody 2038 for their ability to specifically detect PP2A in its various modified forms, as well as to coprecipitate regulatory subunits. The tested antibodies preferentially recognized the nonmethylated form of the enzyme, and they did not coimmunoprecipitate trimeric holoenzymes containing the regulatory subunits B or B', an issue that precludes their use to monitor PP2A holoenzyme activity. Furthermore, some of the antibodies also recognized the phosphatase PP4, demonstrating a lack of specificity for PP2A. Together, these findings suggest that reinterpretation of the data generated by using these reagents is required.


Subject(s)
Antibodies/chemistry , Protein Multimerization , Protein Phosphatase 2 , Animals , Cell Line , HEK293 Cells , Holoenzymes , Humans , Mice , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Rabbits
4.
Sci Signal ; 13(616)2020 01 28.
Article in English | MEDLINE | ID: mdl-31992583

ABSTRACT

Epitope tags are short, linear antibody recognition sequences that enable detection of tagged fusion proteins by antibodies. Epitope tag position and neighboring sequences potentially affect its recognition by antibodies, and such context-dependent differences in tag binding may have a wide-ranging effect on data interpretation. We tested by Western blotting six antibodies that recognize the c-Myc epitope tag, including monoclonal antibodies 9E10, 4A6, 9B11, and 71D10 and polyclonal antibodies 9106 and A-14. All displayed context-dependent differences in their ability to detect N- or C-terminal Myc-tagged proteins. In particular, clone 9E10, the most cited Myc-tag antibody, displayed high context-dependent detection variability, whereas others, notably 4A6 and 9B11, showed much less context sensitivity in their detection of Myc-tagged proteins. The very high context sensitivity of 9E10 was further substantiated by peptide microarray analyses. We conclude that recently developed, purpose-made monoclonal antibodies specific for Myc have much more uniform reactivity in diverse assays and are much less context sensitive than is the legacy antibody 9E10.


Subject(s)
Antibodies, Monoclonal , Epitopes , Proto-Oncogene Proteins c-myc , Single-Chain Antibodies , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Epitopes/chemistry , Epitopes/immunology , Humans , Mice , NIH 3T3 Cells , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology
5.
Sci Rep ; 6: 31363, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27531616

ABSTRACT

Western blotting is one of the most widely used techniques in molecular biology and biochemistry. Prestained proteins are used as molecular weight standards in protein electrophoresis. In the chemiluminescent Western blot analysis, however, these colored protein markers are invisible leaving researchers with the unsatisfying situation that the signal for the protein of interest and the signal for the markers are not captured simultaneously and have to be merged in an error-prone step. To allow the simultaneous detection of marker proteins we generated monoclonal antibodies specific for the protein dyes. To elicit a dye rather than protein specific immune response we immunized mice sequentially with dye-carrier protein complexes, in which a new carrier protein was used for each subsequent immunization. Moreover, by sequentially immunizing with dye-carrier protein complexes, in which different but structurally related dyes were used, we could also generate an antibody, termed anti-RAINBOW, that cross-reacted even with structurally related dyes not used in the immunizations. Our novel antibodies represent convenient tools for the simultaneous Western blot detection of commercially available prestained marker proteins in combination with the detection of any specific protein of interest. These antibodies will render obsolete the anachronistic tradition of manually charting marker bands on film.


Subject(s)
Antibodies/metabolism , Blotting, Western/methods , Fluorescent Dyes/administration & dosage , Proteins/chemistry , Animals , Biomarkers , Fluorescent Dyes/pharmacology , Immunization , Luminescence , Mice , Molecular Weight , Proteins/metabolism , Staining and Labeling
6.
PLoS One ; 10(12): e0145226, 2015.
Article in English | MEDLINE | ID: mdl-26678046

ABSTRACT

Protein phosphatase 2A (PP2A) is a conserved essential enzyme that is implicated as a tumor suppressor based on its central role in phosphorylation-dependent signaling pathways. Protein phosphatase methyl esterase (PME-1) catalyzes specifically the demethylation of the C-terminal Leu309 residue of PP2A catalytic subunit (PP2Ac). It has been shown that PME-1 affects the activity of PP2A by demethylating PP2Ac, but also by directly binding to the phosphatase active site, suggesting loss of PME-1 in cells would enhance PP2A activity. However, here we show that PME-1 knockout mouse embryonic fibroblasts (MEFs) exhibit lower PP2A activity than wild type MEFs. Loss of PME-1 enhanced poly-ubiquitination of PP2Ac and shortened the half-life of PP2Ac protein resulting in reduced PP2Ac levels. Chemical inhibition of PME-1 and rescue experiments with wild type and mutated PME-1 revealed methyl-esterase activity was necessary to maintain PP2Ac protein levels. Our data demonstrate that PME-1 methyl-esterase activity protects PP2Ac from ubiquitin/proteasome degradation.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Phosphatase 2/metabolism , Ubiquitin/metabolism , Animals , Carboxylic Ester Hydrolases/physiology , Immunoblotting , Immunoprecipitation , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/physiology , Protein Phosphatase 2/physiology , Proteolysis , Real-Time Polymerase Chain Reaction , Ubiquitin/physiology
7.
PLoS Genet ; 9(7): e1003575, 2013.
Article in English | MEDLINE | ID: mdl-23861665

ABSTRACT

Entry into mitosis is triggered by cyclinB/Cdk1, whose activity is abruptly raised by a positive feedback loop. The Greatwall kinase phosphorylates proteins of the endosulfine family and allows them to bind and inhibit the main Cdk1-counteracting PP2A-B55 phosphatase, thereby promoting mitotic entry. In contrast to most eukaryotic systems, Cdc14 is the main Cdk1-antagonizing phosphatase in budding yeast, while the PP2A(Cdc55) phosphatase promotes, instead of preventing, mitotic entry by participating to the positive feedback loop of Cdk1 activation. Here we show that budding yeast endosulfines (Igo1 and Igo2) bind to PP2A(Cdc55) in a cell cycle-regulated manner upon Greatwall (Rim15)-dependent phosphorylation. Phosphorylated Igo1 inhibits PP2A(Cdc55) activity in vitro and induces mitotic entry in Xenopus egg extracts, indicating that it bears a conserved PP2A-binding and -inhibitory activity. Surprisingly, deletion of IGO1 and IGO2 in yeast cells leads to a decrease in PP2A phosphatase activity, suggesting that endosulfines act also as positive regulators of PP2A in yeast. Consistently, RIM15 and IGO1/2 promote, like PP2A(Cdc55), timely entry into mitosis under temperature-stress, owing to the accumulation of Tyr-phosphorylated Cdk1. In addition, they contribute to the nuclear export of PP2A(Cdc55), which has recently been proposed to promote mitotic entry. Altogether, our data indicate that Igo proteins participate in the positive feedback loop for Cdk1 activation. We conclude that Greatwall, endosulfines, and PP2A are part of a regulatory module that has been conserved during evolution irrespective of PP2A function in the control of mitosis. However, this conserved module is adapted to account for differences in the regulation of mitotic entry in different organisms.


Subject(s)
Cell Cycle Proteins/genetics , Mitosis/genetics , Protein Kinases/genetics , Protein Phosphatase 2/genetics , Saccharomyces cerevisiae Proteins/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cyclin B/metabolism , Ovum/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tissue Extracts/genetics , Xenopus/genetics
8.
Nat Methods ; 9(6): 594-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22581371

ABSTRACT

We developed a protein-proximity assay in yeast based on fusing a histone lysine methyltransferase onto a bait and its substrate onto a prey. Upon binding, the prey is stably methylated and detected by methylation-specific antibodies. We applied this approach to detect varying interaction affinities among proteins in a mitogen-activated protein kinase pathway and to detect short-lived interactions between protein phosphatase 2A and its substrates that have so far escaped direct detection.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Protein Interaction Mapping/methods , Cell Cycle Proteins/metabolism , Humans , Methylation , Mitogen-Activated Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , TOR Serine-Threonine Kinases/metabolism , Tacrolimus Binding Proteins/metabolism
9.
PLoS Biol ; 5(6): e155, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17550305

ABSTRACT

Protein phosphatase 2A (PP2A) is a prime example of the multisubunit architecture of protein serine/threonine phosphatases. Until substrate-specific PP2A holoenzymes assemble, a constitutively active, but nonspecific, catalytic C subunit would constitute a risk to the cell. While it has been assumed that the severe proliferation impairment of yeast lacking the structural PP2A subunit, TPD3, is due to the unrestricted activity of the C subunit, we recently obtained evidence for the existence of the C subunit in a low-activity conformation that requires the RRD/PTPA proteins for the switch into the active conformation. To study whether and how maturation of the C subunit is coupled with holoenzyme assembly, we analyzed PP2A biogenesis in yeast. Here we show that the generation of the catalytically active C subunit depends on the physical and functional interaction between RRD2 and the structural subunit, TPD3. The phenotype of the tpd3Delta strain is therefore caused by impaired, rather than increased, PP2A activity. TPD3/RRD2-dependent C subunit maturation is under the surveillance of the PP2A methylesterase, PPE1, which upon malfunction of PP2A biogenesis, prevents premature generation of the active C subunit and holoenzyme assembly by counteracting the untimely methylation of the C subunit. We propose a novel model of PP2A biogenesis in which a tightly controlled activation cascade protects cells from untargeted activity of the free catalytic PP2A subunit.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Cell Cycle Proteins/metabolism , Enzyme Activation/physiology , Holoenzymes/metabolism , Methylation , Models, Biological , Phosphoprotein Phosphatases/biosynthesis , Protein Phosphatase 2 , Substrate Specificity
10.
J Neurochem ; 101(4): 959-71, 2007 May.
Article in English | MEDLINE | ID: mdl-17394530

ABSTRACT

Carboxymethylation and phosphorylation of protein phosphatase 2A (PP2A) catalytic C subunit are evolutionary conserved mechanisms that critically control PP2A holoenzyme assembly and substrate specificity. Down-regulation of PP2A methylation and PP2A enzymes containing the B alpha regulatory subunit occur in Alzheimer's disease. In this study, we show that expressed wild-type and methylation- (L309 Delta) and phosphorylation- (T304D, T304A, Y307F, and Y307E) site mutants of PP2A C subunit differentially bind to B, B', and B''-type regulatory subunits in NIH 3T3 fibroblasts and neuro-2a (N2a) neuroblastoma cells. They also display distinct binding affinity for microtubules (MTs). Relative to controls, expression of the wild-type, T304A and Y307F C subunits in N2a cells promotes the accumulation of acetylated and detyrosinated MTs. However, expression of the Y307E, L309 Delta, and T304D mutants, which are impaired in their ability to associate with the B alpha subunit, induces their loss. Silencing of B alpha subunit in N2a and NIH 3T3 cells is sufficient to induce a similar breakdown of acetylated and detyrosinated MTs. It also confers increased sensitivity to nocodazole-induced MT depolymerization. Our findings suggest that changes in intracellular PP2A subunit composition can modulate MT dynamics. They support the hypothesis that reduced amounts of neuronal B alpha-containing PP2A heterotrimers contribute to MT destabilization in Alzheimer's disease.


Subject(s)
Gene Expression Regulation/genetics , Microtubules/metabolism , Mutation/physiology , Phosphoprotein Phosphatases/metabolism , Acetylation , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Mice , Neuroblastoma , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2 , RNA Interference/physiology , Transfection/methods , Tyrosine/metabolism
11.
Nature ; 441(7089): 53-61, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16541024

ABSTRACT

Segregation of homologous maternal and paternal centromeres to opposite poles during meiosis I depends on post-replicative crossing over between homologous non-sister chromatids, which creates chiasmata and therefore bivalent chromosomes. Destruction of sister chromatid cohesion along chromosome arms due to proteolytic cleavage of cohesin's Rec8 subunit by separase resolves chiasmata and thereby triggers the first meiotic division. This produces univalent chromosomes, the chromatids of which are held together by centromeric cohesin that has been protected from separase by shugoshin (Sgo1/MEI-S332) proteins. Here we show in both fission and budding yeast that Sgo1 recruits to centromeres a specific form of protein phosphatase 2A (PP2A). Its inactivation causes loss of centromeric cohesin at anaphase I and random segregation of sister centromeres at the second meiotic division. Artificial recruitment of PP2A to chromosome arms prevents Rec8 phosphorylation and hinders resolution of chiasmata. Our data are consistent with the notion that efficient cleavage of Rec8 requires phosphorylation of cohesin and that this is blocked by PP2A at meiosis I centromeres.


Subject(s)
Centromere/metabolism , Chromatids/metabolism , Chromosome Pairing , Meiosis , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae/cytology , Schizosaccharomyces/cytology , Animals , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Conserved Sequence , Humans , Mice , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Phosphatase 2 , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Cohesins
12.
J Neuropathol Exp Neurol ; 63(10): 1080-91, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15535135

ABSTRACT

ABalphaC, a major protein phosphatase 2A (PP2A) heterotrimeric enzyme, binds to and regulates the microtubule cytoskeleton and tau. We have shown that ABalphaC protein expression levels are selectively reduced in Alzheimer disease (AD). Notably, the carboxyl methylation of PP2A catalytic subunit (PP2A(C)) is critically required for ABalphaC holoenzyme assembly, and catalyzed by a specific methyltransferase (PPMT). Here, we provide the first analysis of human PPMT and methylated PP2A(C) in brain regions from AD, non-AD demented, and aged control autopsy cases. Immunoblotting analyses revealed that PPMT protein expression and PP2A(C) methylation levels were quantitatively decreased in AD-affected brain regions. Immunohistochemical studies showed that PPMT was abundant in neurons throughout the cortex in normal control and non-AD demented cases. However, in AD, there was a regional loss of PPMT immunoreactivity that closely paralleled the severity of tau pathology, but not amyloid plaque burden. We propose that the deregulation of PPMT and PP2A methylation/demethylation cycles contributes to AD pathogenesis, by inducing changes in PP2A heteromultimeric composition and substrate specificity. In turn, PP2A dysfunction compromises the mechanisms that control tau, neuronal plasticity, and survival.


Subject(s)
Alzheimer Disease/etiology , Carbon Dioxide/metabolism , Methyltransferases/metabolism , Phosphoprotein Phosphatases/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Brain/enzymology , Cadaver , Case-Control Studies , Dementia/enzymology , Dementia/metabolism , Dementia/pathology , Down-Regulation , Humans , Methylation , Middle Aged , Phosphorylation , Protein Phosphatase 2 , Tissue Distribution , tau Proteins/metabolism
13.
J Neuropathol Exp Neurol ; 63(4): 287-301, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15099019

ABSTRACT

The formation of amyloid-containing senile plaques and tau-rich neurofibrillary tangles are central events in Alzheimer disease (AD) pathogenesis. Significantly, ABalphaC, a major protein phosphatase 2A (PP2A) holoenzyme, specifically binds to and dephosphorylates tau. Deregulation of PP2A results in tau hyperphosphorylation in vivo. Here, we compared the expression levels and distribution of PP2A subunits in various brain regions from autopsy cases of AD and aged controls with or without histological evidence of age-related neurofibrillary degeneration. Immunoblotting analyses revealed that there was a significant reduction in the total amounts of ABalphaC in AD frontal and temporal cortices that matched the decrease in PP2A activity measured in the same brain homogenates. Immunohistochemical studies showed that neuronal ABalphaC expression levels were significantly and selectively decreased in AD-affected regions and in tangle-bearing neurons, but not in AD cerebellum and in non-AD dementias. Reduced neuronal ABalphaC immunoreactivity closely correlated with tangle load, but not plaque burden, suggesting that ABalphaC dysfunction contributes to AD tau pathology. Glial cells within senile plaques were also positive for ABalphaC. Increased glial PP2A immunoreactivity was observed in both AD and non-AD cases and may play a role in the brain's response to general inflammatory processes and amyloidogenesis.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Brain/enzymology , Phosphoprotein Phosphatases/biosynthesis , Adult , Aged , Aged, 80 and over , Blotting, Western , Brain/pathology , Dementia/enzymology , Dementia/pathology , Female , Humans , Immunohistochemistry , Isoenzymes/biosynthesis , Male , Middle Aged , Neurofibrillary Tangles/enzymology , Neuroglia/enzymology , Neuroglia/pathology , Neurons/enzymology , Neurons/pathology , Plaque, Amyloid/pathology , Protein Phosphatase 2
14.
Curr Biol ; 13(23): 2058-64, 2003 Dec 02.
Article in English | MEDLINE | ID: mdl-14653995

ABSTRACT

Protein phosphatase 2A (PP2A) holoenzymes consist of a catalytic C subunit, a scaffolding A subunit, and one of several regulatory B subunits that recruit the AC dimer to substrates. PP2A is required for chromosome segregation, but PP2A's substrates in this process remain unknown. To identify PP2A substrates, we carried out a two-hybrid screen with the regulatory B/PR55 subunit. We isolated a human homolog of C. elegans HCP6, a protein distantly related to the condensin subunit hCAP-D2, and we named this homolog hHCP-6. Both C. elegans HCP-6 and condensin are required for chromosome organization and segregation. HCP-6 binding partners are unknown, whereas condensin is composed of the structural maintenance of chromosomes proteins SMC2 and SMC4 and of three non-SMC subunits. Here we show that hHCP-6 becomes phosphorylated during mitosis and that its dephosphorylation by PP2A in vitro depends on B/PR55, suggesting that hHCP-6 is a B/PR55-specific substrate of PP2A. Unlike condensin, hHCP-6 is localized in the nucleus in interphase, but similar to condensin, hHCP-6 associates with chromosomes during mitosis. hHCP-6 is part of a complex that contains SMC2, SMC4, kleisin-beta, and the previously uncharacterized HEAT repeat protein FLJ20311. hHCP-6 is therefore part of a condensin-related complex that associates with chromosomes in mitosis and may be regulated by PP2A.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation/physiology , DNA-Binding Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Chromosome Mapping , Electrophoresis, Polyacrylamide Gel , Humans , Immunoblotting , Multiprotein Complexes , Phosphorylation , Precipitin Tests , Protein Phosphatase 2 , Substrate Specificity
15.
Genes Dev ; 17(17): 2138-50, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12952889

ABSTRACT

Protein phosphatase 2A (PP2A) is an essential intracellular serine/threonine phosphatase containing a catalytic subunit that possesses the potential to dephosphorylate promiscuously tyrosine-phosphorylated substrates in vitro. How PP2A acquires its intracellular specificity and activity for serine/threonine-phosphorylated substrates is unknown. Here we report a novel and phylogenetically conserved mechanism to generate active phospho-serine/threonine-specific PP2A in vivo. Phosphotyrosyl phosphatase activator (PTPA), a protein of so far unknown intracellular function, is required for the biogenesis of active and specific PP2A. Deletion of the yeast PTPA homologs generated a PP2A catalytic subunit with a conformation different from the wild-type enzyme, as indicated by its altered substrate specificity, reduced protein stability, and metal dependence. Complementation and RNA-interference experiments showed that PTPA fulfills an essential function conserved from yeast to man.


Subject(s)
Phosphoprotein Phosphatases/metabolism , 3T3 Cells , Animals , Apoptosis/physiology , Binding Sites , HeLa Cells , Humans , Metals/metabolism , Mice , Mutation , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2 , RNA Interference/physiology , Substrate Specificity/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...