Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
Nature ; 629(8010): 219-227, 2024 May.
Article in English | MEDLINE | ID: mdl-38570683

ABSTRACT

The Integrator complex can terminate RNA polymerase II (Pol II) in the promoter-proximal region of genes. Previous work has shed light on how Integrator binds to the paused elongation complex consisting of Pol II, the DRB sensitivity-inducing factor (DSIF) and the negative elongation factor (NELF) and how it cleaves the nascent RNA transcript1, but has not explained how Integrator removes Pol II from the DNA template. Here we present three cryo-electron microscopy structures of the complete Integrator-PP2A complex in different functional states. The structure of the pre-termination complex reveals a previously unresolved, scorpion-tail-shaped INTS10-INTS13-INTS14-INTS15 module that may use its 'sting' to open the DSIF DNA clamp and facilitate termination. The structure of the post-termination complex shows that the previously unresolved subunit INTS3 and associated sensor of single-stranded DNA complex (SOSS) factors prevent Pol II rebinding to Integrator after termination. The structure of the free Integrator-PP2A complex in an inactive closed conformation2 reveals that INTS6 blocks the PP2A phosphatase active site. These results lead to a model for how Integrator terminates Pol II transcription in three steps that involve major rearrangements.


Subject(s)
Cryoelectron Microscopy , Models, Molecular , Protein Phosphatase 2 , RNA Polymerase II , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/ultrastructure , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/ultrastructure , Transcription Termination, Genetic , Humans , Transcription Factors/metabolism , Transcription Factors/chemistry , Protein Binding , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/chemistry , Nuclear Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/ultrastructure , Protein Subunits/metabolism , Protein Subunits/chemistry
2.
Nature ; 625(7993): 195-203, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123684

ABSTRACT

Progression through the cell cycle is controlled by regulated and abrupt changes in phosphorylation1. Mitotic entry is initiated by increased phosphorylation of mitotic proteins, a process driven by kinases2, whereas mitotic exit is achieved by counteracting dephosphorylation, a process driven by phosphatases, especially PP2A:B553. Although the role of kinases in mitotic entry is well established, recent data have shown that mitosis is only successfully initiated when the counterbalancing phosphatases are also inhibited4. Inhibition of PP2A:B55 is achieved by the intrinsically disordered proteins ARPP195,6 and FAM122A7. Despite their critical roles in mitosis, the mechanisms by which they achieve PP2A:B55 inhibition is unknown. Here, we report the single-particle cryo-electron microscopy structures of PP2A:B55 bound to phosphorylated ARPP19 and FAM122A. Consistent with our complementary NMR spectroscopy studies, both intrinsically disordered proteins bind PP2A:B55, but do so in highly distinct manners, leveraging multiple distinct binding sites on B55. Our extensive structural, biophysical and biochemical data explain how substrates and inhibitors are recruited to PP2A:B55 and provide a molecular roadmap for the development of therapeutic interventions for PP2A:B55-related diseases.


Subject(s)
Cryoelectron Microscopy , Intracellular Signaling Peptides and Proteins , Intrinsically Disordered Proteins , Phosphoproteins , Protein Phosphatase 2 , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/ultrastructure , Mitosis , Nuclear Magnetic Resonance, Biomolecular , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphoproteins/ultrastructure , Phosphorylation , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/ultrastructure
3.
Mol Syst Biol ; 19(12): e11782, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37916966

ABSTRACT

Phosphoprotein phosphatases (PPPs) regulate major signaling pathways, but the determinants of phosphatase specificity are poorly understood. This is because methods to investigate this at scale are lacking. Here, we develop a novel in vitro assay, MRBLE:Dephos, that allows multiplexing of dephosphorylation reactions to determine phosphatase preferences. Using MRBLE:Dephos, we establish amino acid preferences of the residues surrounding the dephosphorylation site for PP1 and PP2A-B55, which reveals common and unique preferences. To compare the MRBLE:Dephos results to cellular substrates, we focused on mitotic exit that requires extensive dephosphorylation by PP1 and PP2A-B55. We use specific inhibition of PP1 and PP2A-B55 in mitotic exit lysates coupled with phosphoproteomics to identify more than 2,000 regulated sites. Importantly, the sites dephosphorylated during mitotic exit reveal key signatures that are consistent with MRBLE:Dephos. Furthermore, integration of our phosphoproteomic data with mitotic interactomes of PP1 and PP2A-B55 provides insight into how binding of phosphatases to substrates shapes dephosphorylation. Collectively, we develop novel approaches to investigate protein phosphatases that provide insight into mitotic exit regulation.


Subject(s)
Mitosis , Protein Phosphatase 2 , Phosphorylation , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Signal Transduction , Substrate Specificity
4.
J Chem Phys ; 158(21)2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37260014

ABSTRACT

Mutations in protein phosphatase 2A (PP2A) are connected to intellectual disability and cancer. It has been hypothesized that these mutations might disrupt the autoinhibition and phosphorylation-induced activation of PP2A. Since they are located far from both the active and substrate binding sites, it is unclear how they exert their effect. We performed allosteric pathway analysis based on molecular dynamics simulations and combined it with biochemical experiments to investigate the autoinhibition of PP2A. In the wild type (WT), the C-arm of the regulatory subunit B56δ obstructs the active and substrate binding sites exerting a dual autoinhibition effect. We find that the disease mutant, E198K, severely weakens the allosteric pathways that stabilize the C-arm in the WT. Instead, the strongest allosteric pathways in E198K take a different route that promotes exposure of the substrate binding site. To facilitate the allosteric pathway analysis, we introduce a path clustering algorithm for lumping pathways into channels. We reveal remarkable similarities between the allosteric channels of E198K and those in phosphorylation-activated WT, suggesting that the autoinhibition can be alleviated through a conserved mechanism. In contrast, we find that another disease mutant, E200K, which is in spatial proximity of E198, does not repartition the allosteric pathways leading to the substrate binding site; however, it may still induce exposure of the active site. This finding agrees with our biochemical data, allowing us to predict the activity of PP2A with the phosphorylated B56δ and provide insight into how disease mutations in spatial proximity alter the enzymatic activity in surprisingly different mechanisms.


Subject(s)
Protein Phosphatase 2 , Protein Phosphatase 2/genetics , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , Phosphorylation/genetics , Protein Domains , Mutation , Protein Binding
5.
New Phytol ; 236(5): 1762-1778, 2022 12.
Article in English | MEDLINE | ID: mdl-36073540

ABSTRACT

The various combinations and regulations of different subunits of phosphatase PP2A holoenzymes underlie their functional complexity and importance. However, molecular mechanisms governing the assembly of PP2A complex in response to external or internal signals remain largely unknown, especially in Arabidopsis thaliana. We found that the phosphorylation status of Bß of PP2A acts as a switch to regulate the activity of PP2A. In the absence of ethylene, phosphorylated Bß leads to an inactivation of PP2A; the substrate EIR1 remains to be phosphorylated, preventing the EIR1-mediated auxin transport in epidermis, leading to normal root growth. Upon ethylene treatment, the dephosphorylated Bß mediates the formation of the A2-C4-Bß protein complex to activate PP2A, resulting in the dephosphorylation of EIR1 to promote auxin transport in epidermis of elongation zone, leading to root growth inhibition. Altogether, our research revealed a novel molecular mechanism by which the dephosphorylation of Bß subunit switches on PP2A activity to dephosphorylate EIR1 to establish EIR1-mediated auxin transport in the epidermis in elongation zone for root growth inhibition in response to ethylene.


Subject(s)
Arabidopsis , Phosphoric Monoester Hydrolases , Phosphorylation , Phosphoric Monoester Hydrolases/metabolism , Ethylenes/metabolism , Arabidopsis/metabolism , Indoleacetic Acids/pharmacology , Indoleacetic Acids/metabolism , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism
6.
Science ; 374(6569): 883-887, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34762484

ABSTRACT

Integrator and protein phosphatase 2A (PP2A) form a complex that dephosphorylates paused RNA polymerase II (Pol II), cleaves the nascent RNA, and terminates transcription. We report the structure of the pretermination complex containing the human Integrator-PP2A complex bound to paused Pol II. Integrator binds Pol II and the pausing factors DSIF and NELF to exclude binding of the elongation factors SPT6 and PAF1 complex. Integrator also binds the C-terminal domain of Pol II and positions PP2A to counteract Pol II phosphorylation and elongation. The Integrator endonuclease docks to the RNA exit site and opens to cleave nascent RNA about 20 nucleotides from the Pol II active site. Integrator does not bind the DNA clamps formed by Pol II and DSIF, enabling release of DNA and transcription termination.


Subject(s)
Gene Expression Regulation , Multiprotein Complexes/chemistry , Protein Phosphatase 2/chemistry , RNA Polymerase II/chemistry , Transcription, Genetic , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Humans , Models, Molecular , Multiprotein Complexes/metabolism , Protein Binding , Protein Conformation , Protein Domains , Protein Phosphatase 2/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA/metabolism , RNA Polymerase II/metabolism
7.
Clin Sci (Lond) ; 135(13): 1545-1556, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34192314

ABSTRACT

Tumor initiation is driven by oncogenes that activate signaling networks for cell proliferation and survival involving protein phosphorylation. Protein kinases in these pathways have proven to be effective targets for pharmaceutical inhibitors that have progressed to the clinic to treat various cancers. Here, we offer a narrative about the development of small molecule modulators of the protein Ser/Thr phosphatase 2A (PP2A) to reduce the activation of cell proliferation and survival pathways. These novel drugs promote the assembly of select heterotrimeric forms of PP2A that act to limit cell proliferation. We discuss the potential for the near-term translation of this approach to the clinic for cancer and other human diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Protein Phosphatase 2/antagonists & inhibitors , Allosteric Regulation , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/pathology , Phosphorylation , Protein Conformation , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , Signal Transduction , Structure-Activity Relationship
8.
J Biol Chem ; 297(1): 100908, 2021 07.
Article in English | MEDLINE | ID: mdl-34171357

ABSTRACT

The cAMP response element-binding protein (CREB) is an important regulator of cell growth, metabolism, and synaptic plasticity. CREB is activated through phosphorylation of an evolutionarily conserved Ser residue (S133) within its intrinsically disordered kinase-inducible domain (KID). Phosphorylation of S133 in response to cAMP, Ca2+, and other stimuli triggers an association of the KID with the KID-interacting (KIX) domain of the CREB-binding protein (CBP), a histone acetyl transferase (HAT) that promotes transcriptional activation. Here we addressed the mechanisms of CREB attenuation following bursts in CREB phosphorylation. We show that phosphorylation of S133 is reversed by protein phosphatase 2A (PP2A), which is recruited to CREB through its B56 regulatory subunits. We found that a B56-binding site located at the carboxyl-terminal boundary of the KID (BS2) mediates high-affinity B56 binding, while a second binding site (BS1) located near the amino terminus of the KID mediates low affinity binding enhanced by phosphorylation of adjacent casein kinase (CK) phosphosites. Mutations that diminished B56 binding to BS2 elevated both basal and stimulus-induced phosphorylation of S133, increased CBP interaction with CREB, and potentiated the expression of CREB-dependent reporter genes. Cells from mice harboring a homozygous CrebE153D mutation that disrupts BS2 exhibited increased S133 phosphorylation stoichiometry and elevated transcriptional bursts to cAMP. These findings provide insights into substrate targeting by PP2A holoenzymes and establish a new mechanism of CREB attenuation that has implications for understanding CREB signaling in cell growth, metabolism, synaptic plasticity, and other physiologic contexts.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Protein Phosphatase 2/chemistry , Animals , Binding Sites , Cells, Cultured , HeLa Cells , Humans , Mice , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Signal Transduction , Transcriptional Activation
9.
Mech Ageing Dev ; 197: 111496, 2021 07.
Article in English | MEDLINE | ID: mdl-33957218

ABSTRACT

We have reported that pseudoginsenoside-F11 (PF11) can significantly improve the cognitive impairments in several Alzheimer's disease (AD) models, but the mechanism has not been fully elucidated. In the present study, the effects of PF11 on AD, in particular the underlying mechanisms related with protein phosphatase 2A (PP2A), were investigated in a rat model induced by okadaic acid (OA), a selective inhibitor of PP2A. The results showed that PF11 treatment dose-dependently improved the learning and memory impairments in OA-induced AD rats. PF11 could significantly inhibit OA-induced tau hyperphosphorylation, suppress the activation of glial cells, alleviate neuroinflammation, thus rescue the neuronal and synaptic damage. Further investigation revealed that PF11 could regulate the protein expression of methyl modifying enzymes (leucine carboxyl methyltransferase-1 and protein phosphatase methylesterase-1) in the brain, thus increase methyl-PP2A protein expression and indirectly increase the activity of PP2A. Molecular docking analysis, structural alignment and in vitro results showed that PF11 was similar in the shape and electrostatic field feature to a known activator of PP2A, and could directly bind and activate PP2A. In conclusion, the present data indicate that PF11 can ameliorate OA-induced learning and memory impairment in rats via modulating PP2A.


Subject(s)
Enzyme Activators , Ginsenosides , Learning Disabilities , Memory Disorders , Molecular Docking Simulation , Okadaic Acid/toxicity , Protein Phosphatase 2 , Animals , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Ginsenosides/chemistry , Ginsenosides/pharmacology , Learning Disabilities/chemically induced , Learning Disabilities/drug therapy , Learning Disabilities/enzymology , Male , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Memory Disorders/enzymology , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , Rats , Rats, Sprague-Dawley
10.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836590

ABSTRACT

Reactive oxygen species (ROS) can cause cellular damage and promote cancer development. Besides such harmful consequences of overproduction of ROS, all cells utilize ROS for signaling purposes and stabilization of cell homeostasis. In particular, the latter is supported by the NADPH oxidase 4 (Nox4) that constitutively produces low amounts of H2O2 By that mechanism, Nox4 forces differentiation of cells and prevents inflammation. We hypothesize a constitutive low level of H2O2 maintains basal activity of cellular surveillance systems and is unlikely to be cancerogenic. Utilizing two different murine models of cancerogen-induced solid tumors, we found that deletion of Nox4 promotes tumor formation and lowers recognition of DNA damage. Nox4 supports phosphorylation of H2AX (γH2AX), a prerequisite of DNA damage recognition, by retaining a sufficiently low abundance of the phosphatase PP2A in the nucleus. The underlying mechanism is continuous oxidation of AKT by Nox4. Interaction of oxidized AKT and PP2A captures the phosphatase in the cytosol. Absence of Nox4 facilitates nuclear PP2A translocation and dephosphorylation of γH2AX. Simultaneously AKT is left phosphorylated. Thus, in the absence of Nox4, DNA damage is not recognized and the increased activity of AKT supports proliferation. The combination of both events results in genomic instability and promotes tumor formation. By identifying Nox4 as a protective source of ROS in cancerogen-induced cancer, we provide a piece of knowledge for understanding the role of moderate production of ROS in preventing the initiation of malignancies.


Subject(s)
Carcinogens/toxicity , NADPH Oxidase 4/genetics , Neoplasms/chemically induced , Animals , Cell Nucleus/metabolism , Cytosol/metabolism , DNA Damage , Genomic Instability , Mice , NADPH Oxidase 4/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Oxidation-Reduction , Phosphorylation , Protein Binding , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , Protein Subunits , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species , Signal Transduction
12.
Nat Struct Mol Biol ; 28(3): 290-299, 2021 03.
Article in English | MEDLINE | ID: mdl-33633399

ABSTRACT

The striatin-interacting phosphatase and kinase (STRIPAK) complex is a large, multisubunit protein phosphatase 2A (PP2A) assembly that integrates diverse cellular signals in the Hippo pathway to regulate cell proliferation and survival. The architecture and assembly mechanism of this critical complex are poorly understood. Using cryo-EM, we determine the structure of the human STRIPAK core comprising PP2AA, PP2AC, STRN3, STRIP1, and MOB4 at 3.2-Å resolution. Unlike the canonical trimeric PP2A holoenzyme, STRIPAK contains four copies of STRN3 and one copy of each the PP2AA-C heterodimer, STRIP1, and MOB4. The STRN3 coiled-coil domains form an elongated homotetrameric scaffold that links the complex together. An inositol hexakisphosphate (IP6) is identified as a structural cofactor of STRIP1. Mutations of key residues at subunit interfaces disrupt the integrity of STRIPAK, causing aberrant Hippo pathway activation. Thus, STRIPAK is established as a noncanonical PP2A complex with four copies of regulatory STRN3 for enhanced signal integration.


Subject(s)
Cryoelectron Microscopy , Multienzyme Complexes/metabolism , Multienzyme Complexes/ultrastructure , Protein Multimerization , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autoantigens/chemistry , Autoantigens/genetics , Autoantigens/metabolism , Autoantigens/ultrastructure , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Calmodulin-Binding Proteins/ultrastructure , Hippo Signaling Pathway , Humans , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutation , Phosphate-Binding Proteins/chemistry , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/ultrastructure , Phytic Acid/metabolism , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/ultrastructure , Protein Serine-Threonine Kinases/chemistry , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Signal Transduction
13.
Biochem Biophys Res Commun ; 534: 353-358, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33342519

ABSTRACT

Protein phosphatase 2A (PP2A) B″-family subunits have Ca2+-binding EF-hand motifs and can bind PP2A substrates. Arabidopsis thaliana PP2A B″-family subunits are encoded by six genes, and bind a transcription factor, VIP1. VIP1 is dephosphorylated and nuclear-localized by hypo-osmotic stress. However, whether PP2A B″-family subunits mediate the VIP1 dephosphorylation is unclear. Here, we show by yeast two-hybrid and in vitro pull down assays that Arabidopsis PP2A B″-family subunits bind Arabidopsis PP2A A (scaffold) subunits. We also show that VIP1 dephosphorylation in vitro can be induced by a PP2A B″-family subunit.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , Transcription Factors/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant , Genes, Plant , Osmotic Pressure , Phosphorylation , Plants, Genetically Modified , Protein Interaction Domains and Motifs , Protein Phosphatase 2/genetics , Protein Subunits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Two-Hybrid System Techniques
14.
Biomolecules ; 10(11)2020 11 22.
Article in English | MEDLINE | ID: mdl-33266510

ABSTRACT

Protein phosphorylation is a post-translational modification essential for the control of the activity of most enzymes in the cell. This protein modification results from a fine-tuned balance between kinases and phosphatases. PP2A is one of the major serine/threonine phosphatases that is involved in the control of a myriad of different signaling cascades. This enzyme, often misregulated in cancer, is considered a tumor suppressor. In this review, we will focus on PP2A-B55, a particular holoenzyme of the family of the PP2A phosphatases whose specific role in cancer development and progression has only recently been highlighted. The discovery of the Greatwall (Gwl)/Arpp19-ENSA cascade, a new pathway specifically controlling PP2A-B55 activity, has been shown to be frequently altered in cancer. Herein, we will review the current knowledge about the mechanisms controlling the formation and the regulation of the activity of this phosphatase and its misregulation in cancer.


Subject(s)
Neoplasms/enzymology , Neoplasms/genetics , Protein Phosphatase 2/pharmacokinetics , Animals , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
15.
Science ; 370(6520)2020 11 27.
Article in English | MEDLINE | ID: mdl-33243860

ABSTRACT

The 14-subunit metazoan-specific Integrator contains an endonuclease that cleaves nascent RNA transcripts. Here, we identified a complex containing Integrator and protein phosphatase 2A core enzyme (PP2A-AC), termed INTAC. The 3.5-angstrom-resolution structure reveals that nine human Integrator subunits and PP2A-AC assemble into a cruciform-shaped central scaffold formed by the backbone and shoulder modules, with the phosphatase and endonuclease modules flanking the opposite sides. As a noncanonical PP2A holoenzyme, the INTAC complex dephosphorylates the carboxy-terminal repeat domain of RNA polymerase II at serine-2, -5, and -7 and thus regulates transcription. Our study extends the function of PP2A to transcriptional regulation and reveals how dual enzymatic activities-RNA cleavage and RNA polymerase II dephosphorylation-are structurally and functionally integrated into the INTAC complex.


Subject(s)
Multienzyme Complexes/chemistry , Protein Phosphatase 2/chemistry , RNA Polymerase II/chemistry , Chromatin/chemistry , Cryoelectron Microscopy , Holoenzymes/chemistry , Humans , Protein Domains
16.
Biochem Soc Trans ; 48(5): 2015-2027, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33125487

ABSTRACT

Phosphoprotein Phosphatases (PPPs) are enzymes highly conserved from yeast and human and catalyze the majority of the serine and threonine dephosphorylation in cells. To achieve substrate specificity and selectivity, PPPs form multimeric holoenzymes consisting of catalytic, structural/scaffolding, and regulatory subunits. For the Protein Phosphatase 2A (PP2A)-subfamily of PPPs, holoenzyme assembly is at least in part regulated by an unusual carboxyl-terminal methyl-esterification, commonly referred to as 'methylation'. Carboxyl-terminal methylation is catalyzed by Leucine carboxyl methyltransferase-1 (LCMT1) that utilizes S-adenosyl-methionine (SAM) as the methyl donor and removed by protein phosphatase methylesterase 1 (PME1). For PP2A, methylation dictates regulatory subunit selection and thereby downstream phosphorylation signaling. Intriguingly, there are four families of PP2A regulatory subunits, each exhibiting different levels of methylation sensitivity. Thus, changes in PP2A methylation stoichiometry alters the complement of PP2A holoenzymes in cells and creates distinct modes of kinase opposition. Importantly, selective inactivation of PP2A signaling through the deregulation of methylation is observed in several diseases, most prominently Alzheimer's disease (AD). In this review, we focus on how carboxyl-terminal methylation of the PP2A subfamily (PP2A, PP4, and PP6) regulates holoenzyme function and thereby phosphorylation signaling, with an emphasis on AD.


Subject(s)
Enzymes/chemistry , Gene Expression Regulation , Phosphoproteins/chemistry , Protein Phosphatase 2/chemistry , Alzheimer Disease/metabolism , Animals , Catalysis , Catalytic Domain , Dimerization , Holoenzymes/chemistry , Humans , Methylation , Mice , Mutation , Phosphorylation , Protein Conformation , Protein Domains , Protein Processing, Post-Translational , Protein Subunits/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction , Substrate Specificity
17.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32847850

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Vif recruits a cellular ubiquitin ligase complex to degrade antiviral APOBEC3 enzymes (APOBEC3C-H) and PP2A phosphatase regulators (PPP2R5A to PPP2R5E). While APOBEC3 antagonism is the canonical function of HIV-1 Vif, this viral accessory protein is also known to trigger G2/M cell cycle arrest. Vif initiates G2/M arrest by degrading multiple PPP2R5 family members, an activity prevalent among diverse HIV-1 and simian immunodeficiency virus (SIV) isolates. Here, computational protein-protein docking was used to delineate a Vif/CBF-ß/PPP2R5 complex in which Vif is predicted to bind the same PPP2R5 surface as physiologic phosphatase targets. This model was tested using targeted mutagenesis of amino acid residues within or adjacent to the putative interface to show loss or retention, respectively, of Vif-induced PPP2R5 degradation activity. Additionally, expression of a peptide that mimics cellular targets of PPP2R5s robustly inhibited Vif-mediated degradation of PPP2R5A but not APOBEC3G. Moreover, live-cell imaging studies examining Vif-mediated degradation of PPP2R5A and APOBEC3G within the same cell revealed that PPP2R5A degradation kinetics are comparable to those of APOBEC3G with a half-life of roughly 6 h postinfection, demonstrating that Vif can concurrently mediate the degradation of distinct cellular substrates. Finally, experiments with a panel of patient-derived Vif isolates indicated that PPP2R5A degradation activity is common in patient-derived isolates. Taken together, these results support a model in which PPP2R5 degradation and global changes in the cellular phosphoproteome are likely to be advantageous for viral pathogenesis.IMPORTANCE A critical function of HIV-1 Vif is to counteract the family of APOBEC3 innate immune proteins. It is also widely accepted that Vif induces G2/M cell cycle arrest in several different cell types. Recently, it has been shown that Vif degrades multiple PPP2R5 phosphoregulators to induce the G2/M arrest phenotype. Here, computational approaches are used to test a structural model of the Vif/PPP2R5 complex. In addition, imaging studies are used to show that Vif degrades these PPP2R5 substrates in roughly the same time frame as APOBEC3 degradation and that this activity is prevalent in patient-derived Vif isolates. These studies are important by further defining PPP2R5 proteins as a bona fide substrate of HIV-1 Vif.


Subject(s)
APOBEC-3G Deaminase/chemistry , HIV-1/genetics , Protein Phosphatase 2/chemistry , vif Gene Products, Human Immunodeficiency Virus/chemistry , APOBEC-3G Deaminase/genetics , APOBEC-3G Deaminase/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , HIV Infections/virology , HIV-1/isolation & purification , HIV-1/metabolism , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Kinetics , Models, Molecular , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Structure, Secondary , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Substrate Specificity , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
18.
Nat Commun ; 11(1): 3583, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681005

ABSTRACT

The phosphatases PP1 and PP2A are responsible for the majority of dephosphorylation reactions on phosphoserine (pSer) and phosphothreonine (pThr), and are involved in virtually all cellular processes and numerous diseases. The catalytic subunits exist in cells in form of holoenzymes, which impart substrate specificity. The contribution of the catalytic subunits to the recognition of substrates is unclear. By developing a phosphopeptide library approach and a phosphoproteomic assay, we demonstrate that the specificity of PP1 and PP2A holoenzymes towards pThr and of PP1 for basic motifs adjacent to the phosphorylation site are due to intrinsic properties of the catalytic subunits. Thus, we dissect this amino acid specificity of the catalytic subunits from the contribution of regulatory proteins. Furthermore, our approach enables discovering a role for PP1 as regulator of the GRB-associated-binding protein 2 (GAB2)/14-3-3 complex. Beyond this, we expect that this approach is broadly applicable to detect enzyme-substrate recognition preferences.


Subject(s)
Protein Phosphatase 1/chemistry , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/metabolism , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Catalytic Domain , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Phosphorylation , Protein Binding , Protein Engineering , Protein Phosphatase 1/genetics , Protein Phosphatase 2/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Substrate Specificity
19.
Cell ; 181(3): 688-701.e16, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32315618

ABSTRACT

Impairment of protein phosphatases, including the family of serine/threonine phosphatases designated PP2A, is essential for the pathogenesis of many diseases, including cancer. The ability of PP2A to dephosphorylate hundreds of proteins is regulated by over 40 specificity-determining regulatory "B" subunits that compete for assembly and activation of heterogeneous PP2A heterotrimers. Here, we reveal how a small molecule, DT-061, specifically stabilizes the B56α-PP2A holoenzyme in a fully assembled, active state to dephosphorylate selective substrates, such as its well-known oncogenic target, c-Myc. Our 3.6 Å structure identifies molecular interactions between DT-061 and all three PP2A subunits that prevent dissociation of the active enzyme and highlight inherent mechanisms of PP2A complex assembly. Thus, our findings provide fundamental insights into PP2A complex assembly and regulation, identify a unique interfacial stabilizing mode of action for therapeutic targeting, and aid in the development of phosphatase-based therapeutics tailored against disease specific phospho-protein targets.


Subject(s)
Protein Phosphatase 2/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Enzyme Activators/metabolism , HEK293 Cells , Heterografts , Humans , Male , Mice , Mice, Nude , Models, Molecular , Multiprotein Complexes/metabolism , Protein Phosphatase 2/chemistry , Protein Subunits
20.
Cell Rep ; 30(9): 3164-3170.e3, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130915

ABSTRACT

Aberrant hyperphosphorylation of the protein phosphatase 2A catalytic subunit (PP2Ac) at Tyr307 has been associated with aggressive disease and poor clinical outcome in multiple cancers. However, the study of reversible phosphorylation at this site has relied entirely upon the use of antibodies-most prominently, the clone E155. Here, we provide evidence that the E155 and F-8 phospho-Tyr307 antibodies cannot differentiate between phosphorylated and unphosphorylated forms of PP2Ac. The form of PP2Ac bound by these antibodies in H358 cells is unphosphorylated at the C-terminal tail. Furthermore, these antibodies are sensitive to additional protein modifications that occur near Tyr307, including Thr304 phosphorylation and Leu309 methylation, when these post-translational modifications are present. Thus, studies that used these antibodies to report PP2Ac hyperphosphorylation require reinterpretation, as these antibodies cannot be reliably used as readouts for a single PP2Ac post-translational modification (PTM) change.


Subject(s)
Antibodies/metabolism , Phosphotyrosine/metabolism , Protein Phosphatase 2/metabolism , Research , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Humans , Leucine/metabolism , Methylation , Mutation/genetics , Peptides/chemistry , Peptides/metabolism , Phosphorylation/drug effects , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Processing, Post-Translational , Vanadates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...