Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 295(13): 4194-4211, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32071079

ABSTRACT

Protein phosphatase 2A (PP2A) critically regulates cell signaling and is a human tumor suppressor. PP2A complexes are modulated by proteins such as cancerous inhibitor of protein phosphatase 2A (CIP2A), protein phosphatase methylesterase 1 (PME-1), and SET nuclear proto-oncogene (SET) that often are deregulated in cancers. However, how they impact cellular phosphorylation and how redundant they are in cellular regulation is poorly understood. Here, we conducted a systematic phosphoproteomics screen for phosphotargets modulated by siRNA-mediated depletion of CIP2A, PME-1, and SET (to reactivate PP2A) or the scaffolding A-subunit of PP2A (PPP2R1A) (to inhibit PP2A) in HeLa cells. We identified PP2A-modulated targets in diverse cellular pathways, including kinase signaling, cytoskeleton, RNA splicing, DNA repair, and nuclear lamina. The results indicate nonredundancy among CIP2A, PME-1, and SET in phosphotarget regulation. Notably, PP2A inhibition or reactivation affected largely distinct phosphopeptides, introducing a concept of nonoverlapping phosphatase inhibition- and activation-responsive sites (PIRS and PARS, respectively). This phenomenon is explained by the PPP2R1A inhibition impacting primarily dephosphorylated threonines, whereas PP2A reactivation results in dephosphorylation of clustered and acidophilic sites. Using comprehensive drug-sensitivity screening in PP2A-modulated cells to evaluate the functional impact of PP2A across diverse cellular pathways targeted by these drugs, we found that consistent with global phosphoproteome effects, PP2A modulations broadly affect responses to more than 200 drugs inhibiting a broad spectrum of cancer-relevant targets. These findings advance our understanding of the phosphoproteins, pharmacological responses, and cellular processes regulated by PP2A modulation and may enable the development of combination therapies.


Subject(s)
Autoantigens/genetics , Carboxylic Ester Hydrolases/genetics , DNA-Binding Proteins/genetics , Histone Chaperones/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Protein Phosphatase 2/antagonists & inhibitors , Apoptosis/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Nuclear Lamina/drug effects , Nuclear Lamina/genetics , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation/drug effects , Protein Phosphatase 2/genetics , Proteome/drug effects , Proto-Oncogene Mas , RNA, Small Interfering/genetics , Systems Biology
2.
Nucleus ; 10(1): 7-20, 2019 12.
Article in English | MEDLINE | ID: mdl-30663495

ABSTRACT

Immunosuppressive drugs such as cyclosporin A (CsA) can elicit hepatotoxicity by affecting gene expression. Here, we address the link between CsA and large-scale chromatin organization in HepG2 hepatocarcinoma cells. We show the existence of lamina-associated domains (LADs) interacting with lamin A, lamin B, or both. These 'A-B', 'A-only' and 'B-only' LADs display distinct fates after CsA treatment: A-B LADs remain constitutive or lose A, A-only LADs mainly lose A or switch to B, and B-only LADs remain B-only or acquire A. LAD rearrangement is overall uncoupled from changes in gene expression. Three-dimensional (3D) genome modeling predicts changes in radial positioning of LADs as LADs switch identities, which are corroborated by fluorescence in situ hybridization. Our results reveal interplay between A- and B-type lamins on radial locus positioning, suggesting complementary contributions to large-scale genome architecture. The data also unveil a hitherto unsuspected impact of cytotoxic drugs on genome conformation.Abbreviations: ChIP-seq: chromatin immunoprecipitation sequencing; CsA: cyclosporin A; FISH; fluorescence in situ hybridization; ICMT: isoprenylcysteine methyltransferase; LAD: lamina-associated domain; TAD: topologically-associated domain.


Subject(s)
Chromatin/metabolism , Lamin Type A/metabolism , Lamin Type B/metabolism , Nuclear Lamina/metabolism , Chromatin/drug effects , Cyclosporine/pharmacology , Hep G2 Cells , Humans , In Situ Hybridization, Fluorescence , Lamin Type A/antagonists & inhibitors , Lamin Type B/antagonists & inhibitors , Models, Genetic , Nuclear Lamina/drug effects , Tumor Cells, Cultured
3.
Aging Cell ; 18(1): e12851, 2019 02.
Article in English | MEDLINE | ID: mdl-30565836

ABSTRACT

The Ran GTPase regulates nuclear import and export by controlling the assembly state of transport complexes. This involves the direct action of RanGTP, which is generated in the nucleus by the chromatin-associated nucleotide exchange factor, RCC1. Ran interactions with RCC1 contribute to formation of a nuclear:cytoplasmic (N:C) Ran protein gradient in interphase cells. In previous work, we showed that the Ran protein gradient is disrupted in fibroblasts from Hutchinson-Gilford progeria syndrome (HGPS) patients. The Ran gradient disruption in these cells is caused by nuclear membrane association of a mutant form of Lamin A, which induces a global reduction in heterochromatin marked with Histone H3K9me3 and Histone H3K27me3. Here, we have tested the hypothesis that heterochromatin controls the Ran gradient. Chemical inhibition and depletion of the histone methyltransferases (HMTs) G9a and GLP in normal human fibroblasts reduced heterochromatin levels and caused disruption of the Ran gradient, comparable to that observed previously in HGPS fibroblasts. HMT inhibition caused a defect in nuclear localization of TPR, a high molecular weight protein that, owing to its large size, displays a Ran-dependent import defect in HGPS. We reasoned that pathways dependent on nuclear import of large proteins might be compromised in HGPS. We found that nuclear import of ATM requires the Ran gradient, and disruption of the Ran gradient in HGPS causes a defect in generating nuclear γ-H2AX in response to ionizing radiation. Our data suggest a lamina-chromatin-Ran axis is important for nuclear transport regulation and contributes to the DNA damage response.


Subject(s)
Chromatin/metabolism , DNA Damage , Nuclear Lamina/metabolism , Signal Transduction , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Azepines/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Histones/metabolism , Humans , Interphase/drug effects , Lamin Type A/metabolism , Lysine/metabolism , Methylation/drug effects , Nuclear Lamina/drug effects , Progeria/pathology , Quinazolines/pharmacology , Signal Transduction/drug effects
4.
J Virol ; 88(18): 10982-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24965476

ABSTRACT

Human cytomegalovirus (HCMV) kinase UL97 is required for efficient nuclear lamina disruption during nuclear egress. However, cellular protein kinase C (PKC) has been implicated in this process in other systems. Comparing the effects of UL97 and cellular kinase inhibitors on HCMV nuclear egress confirms a role for UL97 in lamina disruption and nuclear egress. A pan-PKC inhibitor did not affect lamina disruption but did reduce the number of cytoplasmic capsids more than the number of nuclear capsids.


Subject(s)
Cell Nucleus/virology , Cytomegalovirus Infections/enzymology , Cytomegalovirus/enzymology , Nuclear Lamina/virology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Virus Release/drug effects , Capsid/metabolism , Cell Line , Cell Nucleus/drug effects , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Humans , Nuclear Lamina/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , Virus Assembly/drug effects
5.
Virology ; 406(1): 127-37, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20674954

ABSTRACT

The nuclear lamina is thought to be a steric barrier to the herpesvirus capsid. Disruption of the lamina accompanied by phosphorylation of lamina proteins is a conserved feature of herpesvirus infection. In HSV-1-infected cells, protein kinase C (PKC) alpha and delta isoforms are recruited to the nuclear membrane and PKC delta has been implicated in phosphorylation of emerin and lamin B. We tested two critical hypotheses about the mechanism and significance of lamina disruption. First, we show that chemical inhibition of all PKC isoforms reduced viral growth five-fold and inhibited capsid egress from the nucleus. However, specific inhibition of either conventional PKCs or PKC delta does not inhibit viral growth. Second, we show hyperphosphorylation of emerin by viral and cellular kinases is required for its disassociation from the lamina. These data support hypothesis that phosphorylation of lamina components mediates lamina disruption during HSV nuclear egress.


Subject(s)
Herpesvirus 1, Human/physiology , Host-Pathogen Interactions/physiology , Nuclear Lamina/virology , Protein Kinase C/physiology , Virus Release/physiology , Animals , Base Sequence , Capsid/drug effects , Capsid/physiology , Capsid/ultrastructure , Cell Line , Chlorocebus aethiops , DNA Primers/genetics , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/ultrastructure , Host-Pathogen Interactions/drug effects , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/physiology , Microscopy, Electron, Transmission , Models, Biological , Nuclear Lamina/drug effects , Nuclear Lamina/enzymology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Vero Cells , Virus Assembly/drug effects , Virus Assembly/physiology , Virus Release/drug effects , Virus Replication/drug effects , Virus Replication/physiology
6.
Nucleic Acids Res ; 37(7): 2238-48, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19237397

ABSTRACT

In this article, we study how intercalation-induced changes in chromatin and DNA topology affect chromosomal DNA replication using Xenopus egg extracts. Unexpectedly, intercalation by ethidium or doxorubicin prevents formation of a functional nucleus: although nucleosome formation occurs, DNA decondensation is arrested, membranous vesicles accumulate around DNA but do not fuse to form a nuclear membrane, active transport is abolished and lamins are found on chromatin, but do not assemble into a lamina. DNA replication is inhibited at the stage of initiation complex activation, as shown by molecular combing of DNA and by the absence of checkpoint activation. Replication of single-stranded DNA is not prevented. Surprisingly, in spite of the absence of nuclear function, DNA-replication proteins of pre-replication and initiation complexes are loaded onto chromatin. This is a general phenomenon as initiation complexes could also be seen without ethidium in membrane-depleted extracts which do not form nuclei. These results suggest that DNA or chromatin topology is required for generation of a functional nucleus, and activation, but not formation, of initiation complexes.


Subject(s)
Cell Nucleus/genetics , DNA Replication , Animals , Chromatin/metabolism , DNA Replication/drug effects , DNA-Binding Proteins/metabolism , Ethidium/pharmacology , Intercalating Agents/pharmacology , Nuclear Envelope/drug effects , Nuclear Lamina/drug effects , Xenopus
7.
Exp Cell Res ; 314(6): 1392-405, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18294630

ABSTRACT

Mutation R453W in A-type lamins, that are major nuclear envelope proteins, generates Emery-Dreifuss muscular dystrophy. We previously showed that mouse myoblasts expressing R453W-lamin A incompletely exit the cell cycle and differentiate into myocytes with a low level of multinucleation. Here we attempted to improve differentiation by treating these cells with a mixture of PD98059, an extracellular-regulated kinase (ERK) kinase (also known as mitogen-activated kinase, MEK) inhibitor, and insulin-like growth factor-II, an activator of phosphoinositide 3-kinase. We show that mouse myoblasts expressing R453W-lamin A were sensitive to the drug treatment as shown by (i) an increase in multinucleation, (ii) downregulation of proliferation markers (cyclin D1, hyperphosphorylated Rb), (iii) upregulation of myogenin, and (iv) sustained activation of p21 and cyclin D3. However, nuclear matrix anchorage of p21 and cyclin D3 in a complex with hypophosphorylated Rb that is critical to trigger cell cycle arrest and myogenin induction was deficient and incompletely restored by drug treatment. As the turn-over of R453W-lamin A at the nuclear envelope was greatly enhanced, we propose that R453W-lamin A impairs the capacity of the nuclear lamina to serve as scaffold for substrates of the MEK-ERK pathway and for MyoD-induced proteins that play a role in the differentiation process.


Subject(s)
Cell Differentiation , Lamin Type A/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscular Dystrophy, Emery-Dreifuss/enzymology , Myoblasts/cytology , Myoblasts/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Antigens, CD1/metabolism , CD3 Complex/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Green Fluorescent Proteins/metabolism , Insulin-Like Growth Factor II/pharmacology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Muscular Dystrophy, Emery-Dreifuss/pathology , Mutant Proteins/metabolism , Mutation/genetics , Myoblasts/drug effects , Myogenin/metabolism , Nuclear Lamina/drug effects , Nuclear Lamina/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Retinoblastoma Protein/metabolism
8.
Exp Cell Res ; 314(3): 453-62, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18093584

ABSTRACT

Increasing interest in drugs acting on prelamin A has derived from the finding of prelamin A involvement in severe laminopathies. Amelioration of the nuclear morphology by inhibitors of prelamin A farnesylation has been widely reported in progeroid laminopathies. We investigated the effects on chromatin organization of two drugs inhibiting prelamin A processing by an ultrastructural and biochemical approach. The farnesyltransferase inhibitor FTI-277 and the non-peptidomimetic drug N-acetyl-S-farnesyl-l-cysteine methylester (AFCMe) were administered to cultured control human fibroblasts for 6 or 18 h. FTI-277 interferes with protein farnesylation causing accumulation of non-farnesylated prelamin A, while AFCMe impairs the last cleavage of the lamin A precursor and is expected to accumulate farnesylated prelamin A. FTI-277 caused redistribution of heterochromatin domains at the nuclear interior, while AFCMe caused loss of heterochromatin domains, increase of nuclear size and nuclear lamina thickening. At the biochemical level, heterochromatin-associated proteins and LAP2 alpha were clustered at the nuclear interior following FTI-277 treatment, while they were unevenly distributed or absent in AFCMe-treated nuclei. The reported effects show that chromatin is an immediate target of FTI-277 and AFCMe and that dramatic remodeling of chromatin domains occurs following treatment with the drugs. These effects appear to depend, at least in part, on the accumulation of prelamin A forms, since impairment of prelamin A accumulation, here obtained by 5-azadeoxycytidine treatment, abolishes the chromatin effects. These results may be used to evaluate downstream effects of FTIs or other prelamin A inhibitors potentially useful for the therapy of laminopathies.


Subject(s)
Cell Nucleus/drug effects , Chromatin Assembly and Disassembly/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Heterochromatin/drug effects , Nuclear Proteins/drug effects , Protein Precursors/drug effects , Protein Prenylation/drug effects , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Adult , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Decitabine , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Heterochromatin/genetics , Heterochromatin/ultrastructure , Humans , Lamin Type A/drug effects , Lamin Type A/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Methionine/analogs & derivatives , Methionine/pharmacology , Nuclear Lamina/drug effects , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Protein Prenylation/physiology
9.
J Cell Sci ; 120(Pt 9): 1673-80, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17430977

ABSTRACT

Many compounds in the cell nucleus are structurally organized. To assess the influence of structural organization on nuclear function, we investigated the physical mechanisms of structure formation by using molecular crowding as a parameter for nuclear integrity. Molecular crowding promotes compaction of macromolecular compounds depending on their size and shape without the need for site-specific interactions. HeLa and MCF7 cells were incubated with hypertonic medium to increase crowding of their macromolecular content as a result of the osmotic loss of water. Supplementation of sucrose, sorbitol or NaCl to the growth medium shifted nuclear organization, observed by fluorescence and electron microscopy, towards compaction of chromatin and segregation of other nuclear compounds. With increasing hypertonic load and incubation time, this nuclear re-organization proceeded gradually, irrespective of the substances used, and reversibly relaxed to a regular phenotype upon re-incubation of cells in isotonic growth medium. Gradual and reversible re-organization are major features of controlled de-mixing by molecular crowding. Of fundamental importance for nuclear function, we discuss how macromolecular crowding could account for the stabilization of processes that involve large, macromolecular machines.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/analysis , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Nucleus Structures/drug effects , Cell Nucleus Structures/metabolism , Cell Nucleus Structures/ultrastructure , Chromatin/drug effects , Chromatin/metabolism , Chromatin/ultrastructure , Dextrans/pharmacology , Digitonin/pharmacology , HeLa Cells , Histones/analysis , Humans , Hypertonic Solutions/pharmacology , Lamin Type A/analysis , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nuclear Lamina/drug effects , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure , Osmotic Pressure , Ribonucleoproteins/analysis , Serine-Arginine Splicing Factors , Sodium Chloride/pharmacology , Sorbitol/pharmacology , Sucrose/pharmacology
10.
J Cell Biol ; 168(2): 245-55, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15657395

ABSTRACT

Membrane blebbing during the apoptotic execution phase results from caspase-mediated cleavage and activation of ROCK I. Here, we show that ROCK activity, myosin light chain (MLC) phosphorylation, MLC ATPase activity, and an intact actin cytoskeleton, but not microtubular cytoskeleton, are required for disruption of nuclear integrity during apoptosis. Inhibition of ROCK or MLC ATPase activity, which protect apoptotic nuclear integrity, does not affect caspase-mediated degradation of nuclear proteins such as lamins A, B1, or C. The conditional activation of ROCK I was sufficient to tear apart nuclei in lamin A/C null fibroblasts, but not in wild-type fibroblasts. Thus, apoptotic nuclear disintegration requires actin-myosin contractile force and lamin proteolysis, making apoptosis analogous to, but distinct from, mitosis where nuclear disintegration results from microtubule-based forces and from lamin phosphorylation and depolymerization.


Subject(s)
Actins/metabolism , Apoptosis/physiology , Cell Nucleus/metabolism , Lamins/metabolism , Myosins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amides/pharmacology , Animals , Apoptosis/drug effects , Caspase Inhibitors , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cycloheximide/pharmacology , Cytochalasin D/pharmacology , Cytoskeletal Proteins , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Intracellular Signaling Peptides and Proteins , Lamins/genetics , Lim Kinases , Mice , Microscopy, Electron, Transmission , Microtubules/drug effects , Microtubules/metabolism , Mutation/physiology , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Myosins/antagonists & inhibitors , NIH 3T3 Cells , Nocodazole/pharmacology , Nuclear Lamina/drug effects , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyridines/pharmacology , Transfection , Tumor Necrosis Factor-alpha/pharmacology , rho-Associated Kinases
11.
J Cell Biol ; 167(6): 1051-62, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15611332

ABSTRACT

Resident integral proteins of the inner nuclear membrane (INM) are synthesized as membrane-integrated proteins on the peripheral endoplasmic reticulum (ER) and are transported to the INM throughout interphase using an unknown trafficking mechanism. To study this transport, we developed a live cell assay that measures the movement of transmembrane reporters from the ER to the INM by rapamycin-mediated trapping at the nuclear lamina. Reporter constructs with small (<30 kD) cytosolic and lumenal domains rapidly accumulated at the INM. However, increasing the size of either domain by 47 kD strongly inhibited movement. Reduced temperature and ATP depletion also inhibited movement, which is characteristic of membrane fusion mechanisms, but pharmacological inhibition of vesicular trafficking had no effect. Because reporter accumulation at the INM was inhibited by antibodies to the nuclear pore membrane protein gp210, our results support a model wherein transport of integral proteins to the INM involves lateral diffusion in the lipid bilayer around the nuclear pore membrane, coupled with active restructuring of the nuclear pore complex.


Subject(s)
Energy Metabolism/physiology , Membrane Proteins/physiology , Nuclear Pore/physiology , Nuclear Proteins/physiology , Adenosine Triphosphate/pharmacology , Endoplasmic Reticulum/physiology , HeLa Cells , Humans , Membrane Proteins/drug effects , Nuclear Lamina/drug effects , Nuclear Lamina/physiology , Nuclear Pore Complex Proteins/physiology , Protein Transport/drug effects , Protein Transport/physiology , Sirolimus/pharmacology , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...