Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 49(12): 1038-1046, 2021 12.
Article in English | MEDLINE | ID: mdl-34548392

ABSTRACT

Most drugs are administered to children orally. An information gap remains on the protein abundance of small intestinal drug-metabolizing enzymes (DMEs) and drug transporters (DTs) across the pediatric age range, which hinders precision dosing in children. To explore age-related differences in DMEs and DTs, surgical leftover intestinal tissues from pediatric and adult jejunum and ileum were collected and analyzed by targeted quantitative proteomics for apical sodium-bile acid transporter, breast cancer resistance protein (BCRP), monocarboxylate transporter 1 (MCT1), multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein (MRP) 2, MRP3, organic anion-transporting polypeptide 2B1, organic cation transporter 1, peptide transporter 1 (PEPT1), CYP2C19, CYP3A4, CYP3A5, UDP glucuronosyltransferase (UGT) 1A1, UGT1A10, and UGT2B7. Samples from 58 children (48 ileums, 10 jejunums, age range: 8 weeks to 17 years) and 16 adults (8 ileums, 8 jejunums) were analyzed. When comparing age groups, BCRP, MDR1, PEPT1, and UGT1A1 abundance was significantly higher in adult ileum as compared with the pediatric ileum. Jejunal BCRP, MRP2, UGT1A1, and CYP3A4 abundance was higher in the adults compared with children 0-2 years of age. Examining the data on a continuous age scale showed that PEPT1 and UGT1A1 abundance was significantly higher, whereas MCT1 and UGT2B7 abundance was lower in adult ileum as compared with the pediatric ileum. Our data contribute to the deeper understanding of the ontogeny of small intestinal drug-metabolizing enzymes and drug transporters and shows DME-, DT-, and intestinal location-specific, age-related changes. SIGNIFICANCE STATEMENT: This is the first study that describes the ontogeny of small intestinal DTs and DMEs in human using liquid chromatography with tandem mass spectrometry-based targeted quantitative proteomics. The current analysis provides a detailed picture about the maturation of DT and DME abundances in the human jejunum and ileum. The presented results supply age-related DT and DME abundance data for building more accurate PBPK models that serve to support safer and more efficient drug dosing regimens for the pediatric population.


Subject(s)
Inactivation, Metabolic/physiology , Intestine, Small , Membrane Transport Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Age Factors , Biological Transport, Active , Child , Chromatography, Liquid/methods , Cytochrome P-450 CYP3A/metabolism , Enzyme Assays/methods , Gene Ontology , Glucuronosyltransferase/metabolism , Humans , Intestine, Small/drug effects , Intestine, Small/enzymology , Intestine, Small/metabolism , Metabolic Clearance Rate , Multidrug Resistance-Associated Protein 2/metabolism , Neoplasm Proteins/metabolism , Peptide Transporter 1/metabolism , Tandem Mass Spectrometry/methods
2.
Immunity ; 47(6): 1083-1099.e6, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246442

ABSTRACT

The immunosuppressive protein PD-L1 is upregulated in many cancers and contributes to evasion of the host immune system. The relative importance of the tumor microenvironment and cancer cell-intrinsic signaling in the regulation of PD-L1 expression remains unclear. We report that oncogenic RAS signaling can upregulate tumor cell PD-L1 expression through a mechanism involving increases in PD-L1 mRNA stability via modulation of the AU-rich element-binding protein tristetraprolin (TTP). TTP negatively regulates PD-L1 expression through AU-rich elements in the 3' UTR of PD-L1 mRNA. MEK signaling downstream of RAS leads to phosphorylation and inhibition of TTP by the kinase MK2. In human lung and colorectal tumors, RAS pathway activation is associated with elevated PD-L1 expression. In vivo, restoration of TTP expression enhances anti-tumor immunity dependent on degradation of PD-L1 mRNA. We demonstrate that RAS can drive cell-intrinsic PD-L1 expression, thus presenting therapeutic opportunities to reverse the innately immunoresistant phenotype of RAS mutant cancers.


Subject(s)
B7-H1 Antigen/immunology , Colorectal Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/immunology , Proto-Oncogene Proteins p21(ras)/immunology , Tristetraprolin/immunology , Tumor Escape , Animals , B7-H1 Antigen/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Proteins p21(ras)/genetics , RNA Cleavage , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/immunology , Signal Transduction , Tristetraprolin/genetics
3.
Cell Rep ; 17(12): 3319-3332, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009299

ABSTRACT

Receptor tyrosine kinases exhibit a variety of activation mechanisms despite highly homologous catalytic domains. Such diversity arises through coupling of extracellular ligand-binding portions with highly variable intracellular sequences flanking the tyrosine kinase domain and specific patterns of autophosphorylation sites. Here, we show that the juxtamembrane (JM) segment enhances RET catalytic domain activity through Y687. This phospho-site is also required by the JM region to rescue an otherwise catalytically deficient RET activation-loop mutant lacking tyrosines. Structure-function analyses identified interactions between the JM hinge, αC helix, and an unconventional activation-loop serine phosphorylation site that engages the HRD motif and promotes phospho-tyrosine conformational accessibility and regulatory spine assembly. We demonstrate that this phospho-S909 arises from an intrinsic RET dual-specificity kinase activity and show that an equivalent serine is required for RET signaling in Drosophila. Our findings reveal dual-specificity and allosteric components for the mechanism of RET activation and signaling with direct implications for drug discovery.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Proto-Oncogene Proteins c-ret/chemistry , Proto-Oncogene Proteins c-ret/metabolism , Receptor Protein-Tyrosine Kinases/chemistry , Structure-Activity Relationship , Allosteric Regulation/genetics , Amino Acid Sequence/genetics , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Enzyme Activation/genetics , Phosphorylation , Proto-Oncogene Proteins c-ret/genetics , Receptor Protein-Tyrosine Kinases/genetics , Serine/metabolism , Signal Transduction/genetics
4.
J Proteome Res ; 15(12): 4612-4623, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27654267

ABSTRACT

Long wavelength ultraviolet radiation (UVA, 320-400 nm) interacts with chromophores present in human cells to induce reactive oxygen species (ROS) that damage both DNA and proteins. ROS levels are amplified, and the damaging effects of UVA are exacerbated if the cells are irradiated in the presence of UVA photosensitizers such as 6-thioguanine (6-TG), a strong UVA chromophore that is extensively incorporated into the DNA of dividing cells, or the fluoroquinolone antibiotic ciprofloxacin. Both DNA-embedded 6-TG and ciprofloxacin combine synergistically with UVA to generate high levels of ROS. Importantly, the extensive protein damage induced by these photosensitizer+UVA combinations inhibits DNA repair. DNA is maintained in intimate contact with the proteins that effect its replication, transcription, and repair, and DNA-protein cross-links (DPCs) are a recognized reaction product of ROS. Cross-linking of DNA metabolizing proteins would compromise these processes by introducing physical blocks and by depleting active proteins. We describe a sensitive and statistically rigorous method to analyze DPCs in cultured human cells. Application of this proteomics-based analysis to cells treated with 6-TG+UVA and ciprofloxacin+UVA identified proteins involved in DNA repair, replication, and gene expression among those most vulnerable to cross-linking under oxidative conditions.


Subject(s)
DNA Repair/radiation effects , DNA Replication/radiation effects , DNA/metabolism , Photosensitizing Agents/pharmacology , Proteomics/methods , Ultraviolet Rays , Cells, Cultured , Ciprofloxacin/pharmacology , Gene Expression , Humans , Oxidation-Reduction , Proteins/metabolism , Thioguanine/pharmacology
5.
Dev Cell ; 38(4): 384-98, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27554858

ABSTRACT

Atypical protein kinase C (aPKC) is a key apical-basal polarity determinant and Par complex component. It is recruited by Par3/Baz (Bazooka in Drosophila) into epithelial apical domains through high-affinity interaction. Paradoxically, aPKC also phosphorylates Par3/Baz, provoking its relocalization to adherens junctions (AJs). We show that Par3 conserved region 3 (CR3) forms a tight inhibitory complex with a primed aPKC kinase domain, blocking substrate access. A CR3 motif flanking its PKC consensus site disrupts the aPKC kinase N lobe, separating P-loop/αB/αC contacts. A second CR3 motif provides a high-affinity anchor. Mutation of either motif switches CR3 to an efficient in vitro substrate by exposing its phospho-acceptor site. In vivo, mutation of either CR3 motif alters Par3/Baz localization from apical to AJs. Our results reveal how Par3/Baz CR3 can antagonize aPKC in stable apical Par complexes and suggests that modulation of CR3 inhibitory arms or opposing aPKC pockets would perturb the interaction, promoting Par3/Baz phosphorylation.


Subject(s)
Adherens Junctions/metabolism , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Kinase C/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Polarity/physiology , Drosophila , Drosophila Proteins/genetics , Epithelium/growth & development , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Phosphorylation , Protein Binding/genetics , Protein Structure, Tertiary
6.
EMBO Rep ; 17(3): 326-37, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26755742

ABSTRACT

Centrioles are the major constituents of the animal centrosome, in which Plk4 kinase serves as a master regulator of the duplication cycle. Many eukaryotes also contain numerous peripheral particles known as centriolar satellites. While centriolar satellites aid centriole assembly and primary cilium formation, it is unknown whether Plk4 plays any regulatory roles in centriolar satellite integrity. Here we show that Plk4 is a critical determinant of centriolar satellite organisation. Plk4 depletion leads to the dispersion of centriolar satellites and perturbed ciliogenesis. Plk4 interacts with the satellite component PCM1, and its kinase activity is required for phosphorylation of the conserved S372. The nonphosphorylatable PCM1 mutant recapitulates phenotypes of Plk4 depletion, while the phosphomimetic mutant partially rescues the dispersed centriolar satellite patterns and ciliogenesis in cells depleted of PCM1. We show that S372 phosphorylation occurs during the G1 phase of the cell cycle and is important for PCM1 dimerisation and interaction with other satellite components. Our findings reveal that Plk4 is required for centriolar satellite function, which may underlie the ciliogenesis defects caused by Plk4 dysfunction.


Subject(s)
Autoantigens/metabolism , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Cilia/metabolism , G1 Phase , HeLa Cells , Humans , Phosphorylation , Protein Binding , Protein Multimerization , Protein Serine-Threonine Kinases/genetics
7.
Genes Cells ; 20(12): 1046-58, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26525038

ABSTRACT

Fission yeast undergoes growth polarity transition from monopolar to bipolar during G2 phase, designated NETO (New End Take Off). It is known that NETO onset involves two prerequisites, the completion of DNA replication and attainment of a certain cell size. However, the molecular mechanism remains unexplored. Here, we show that casein kinase 1γ, Cki3 is a critical determinant of NETO onset. Not only did cki3∆ cells undergo NETO during G1- or S-phase, but they also displayed premature NETO under unperturbed conditions with a smaller cell size, leading to cell integrity defects. Cki3 interacted with the polarity factor Tea1, of which phosphorylation was dependent on Cki3 kinase activity. GFP nanotrap of Tea1 by Cki3 led to Tea1 hyperphosphorylation with monopolar growth, whereas the same entrapment by kinase-dead Cki3 resulted in converse bipolar growth. Intriguingly, the Tea1 interactor Tea4 was dissociated from Tea1 by Cki3 entrapment. Mass spectrometry identified four phosphoserine residues within Tea1 that were hypophosphorylated in cki3∆ cells. Phosphomimetic Tea1 mutants showed compromised binding to Tea4 and NETO defects, indicating that these serine residues are critical for protein-protein interaction and NETO onset. Our findings provide significant insight into the mechanism by which cell polarization is regulated in a spatiotemporal manner.


Subject(s)
Casein Kinase I/metabolism , Microtubule-Associated Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/growth & development , Binding Sites , Cell Polarity , G2 Phase , Microtubule-Associated Proteins/chemistry , Phosphorylation , Phosphoserine/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/ultrastructure , Schizosaccharomyces pombe Proteins/chemistry
8.
Mol Cell ; 53(5): 738-51, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24560924

ABSTRACT

To decipher the molecular basis for RET kinase activation and oncogenic deregulation, we defined the temporal sequence of RET autophosphorylation by label-free quantitative mass spectrometry. Early autophosphorylation sites map to regions flanking the kinase domain core, while sites within the activation loop only form at later time points. Comparison with oncogenic RET kinase revealed that late autophosphorylation sites become phosphorylated much earlier than wild-type RET, which is due to a combination of an enhanced enzymatic activity, increased ATP affinity, and surprisingly, by providing a better intermolecular substrate. Structural analysis of oncogenic M918T and wild-type RET kinase domains reveal a cis-inhibitory mechanism involving tethering contacts between the glycine-rich loop, activation loop, and αC-helix. Tether mutations only affected substrate presentation but perturbed the autophosphorylation trajectory similar to oncogenic mutations. This study reveals an unappreciated role for oncogenic RET kinase mutations in promoting intermolecular autophosphorylation by enhancing substrate presentation.


Subject(s)
Gene Expression Regulation, Enzymologic , Mutation , Proto-Oncogene Proteins c-ret/chemistry , Proto-Oncogene Proteins c-ret/genetics , Sequence Homology, Amino Acid , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Humans , Insecta , Ligands , Mass Spectrometry , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Substrate Specificity , Time Factors , Tyrosine/chemistry
9.
Mol Oncol ; 8(3): 633-41, 2014 May.
Article in English | MEDLINE | ID: mdl-24529480

ABSTRACT

The pivotal role of LYRIC/AEG-1 in malignant transformation, tumourigenesis and chemo-resistance has previously been demonstrated in different cell types and sub-cellular compartments. The localisation of LYRIC/AEG-1 appears crucial to its function and is regulated by three lysine-rich nuclear localisation signal regions, one of which was previously demonstrated to be modified by ubiquitin. Here we show that mutation of LYRIC/AEG-1 at K486 and K491 results in a loss of ubiquitination. A K486/491R double mutant that is incapable of ubiquitination shows reduced binding to the NFκB subunit p65 or importin-ß resulting in a distinctive peri-nuclear localisation of LYRIC/AEG-1. We also provide evidence to suggest that TOPORS, an E3 ligase that also regulates p53 modification may be responsible for LYRIC/AEG-1 ubiquitin modification. Overall we demonstrate that specific sites of LYRIC/AEG-1 ubiquitination are essential for regulating LYRIC/AEG-1 localisation and functionally interacting proteins.


Subject(s)
Cell Adhesion Molecules/metabolism , Ubiquitination , Animals , COS Cells , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Chlorocebus aethiops , Humans , Membrane Proteins , Point Mutation , Protein Interaction Maps , RNA-Binding Proteins , Ubiquitin/metabolism
11.
J Cell Sci ; 125(Pt 16): 3733-8, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22553211

ABSTRACT

In migrating NRK cells, aPKCs control the dynamics of turnover of paxillin-containing focal adhesions (FA) determining migration rate. Using a proteomic approach (two-dimensional fluorescence difference gel electrophoresis), dynein intermediate chain 2 (dynein IC2) was identified as a protein that is phosphorylated inducibly during cell migration in a PKC-regulated manner. By gene silencing and co-immunoprecipitation studies, we show that dynein IC2 regulates the speed of cell migration through its interaction with paxillin. This interaction is controlled by serine 84 phosphorylation, which lies on the aPKC pathway. The evidence presented thus links aPKC control of migration to the dynein control of FA turnover through paxillin.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Dyneins/metabolism , Paxillin/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cytoplasmic Dyneins , Kidney/cytology , Kidney/enzymology , Molecular Sequence Data , Phosphorylation , Rats
13.
Proteomics ; 5(17): 4376-88, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16294313

ABSTRACT

IMAC can be used to selectively enrich phosphopeptides from complex peptide mixtures, but co-retention of acidic peptides together with the failure to retain some phosphopeptides restricts the general utility of the method. In this study Fe(III)-IMAC was qualitatively and quantitatively assessed using a panel of phosphopeptides, both synthetic and derived from proteolysis of known phosphoproteins, to identify the causes of success and failure in the application of this technique. Here we demonstrate that, as expected, peptides with a more acidic amino acid content are generally more efficiently purified and detected by MALDI-MS after Fe(III)-IMAC than those with a more basic content. Modulating the loading buffer used for Fe(III)-IMAC significantly affects phosphopeptide binding and suggests that conformational factors that lead to steric hindrance and reduced accessibility to the phosphate are important. The use of 1,1,1,3,3,3-hexafluoroisopropanol is shown here to significantly improve Fe(III)-IMAC enrichment and subsequent detection of phosphopeptides by MALDI-MS.


Subject(s)
Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Propanols , Amino Acid Sequence , Caseins/chemistry , DNA, Complementary , Glutathione Transferase/chemistry , Humans , Imidazoles , Indicators and Reagents , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Phosphopeptides/chemical synthesis , Phosphoproteins/chemistry , Phosphorylation , Recombinant Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Methods Mol Biol ; 261: 479-98, 2004.
Article in English | MEDLINE | ID: mdl-15064477

ABSTRACT

Advances in genomic and proteomic technologies combined with molecular and cell biology have together enabled the identification of numerous genes and their products. Two-dimensional gel electrophoresis (2DE) is especially useful in the study of protein-protein interactions as it permits an improved separation of proteins as well as the detection of specific interacting protein isoform(s) of a protein resulting from post-translational modification. The investigation of interacting proteins using 2DE can be complemented by identification of the proteins by mass spectrometry. Here, I describe how protein complexes, isolated by methods such as immunoprecipitation, can be analyzed by 2DE using either isoelectric focusing (tube gels or immobilized pH gradient strips) or nonequilibrium pH gradient electrophoresis (NEPHGE) in the first dimension, SDS-PAGE in the second dimension, and gel staining (silver and Coomassie) or Western blotting for the final detection of the interacting proteins.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Protein Interaction Mapping/methods , Proteins/chemistry , Proteins/metabolism , Isoelectric Focusing , Precipitin Tests , Protein Isoforms , Proteins/genetics
15.
Methods Mol Biol ; 261: 499-510, 2004.
Article in English | MEDLINE | ID: mdl-15064478

ABSTRACT

The identification and characterization of binding partners from protein complexes is increasingly undertaken by mass spectrometry because of its high sensitivity and expedient elucidation of protein structure by accurate mass measurement. A variety of affinity purification methods including immunoprecipitation and glutathione-S-transferase (GST) pull-downs are commonly employed for the isolation of protein complexes and coupled to gel electrophoresis for further separation and basic information with regard to their constituents. For the successful analysis of gel-separated proteins by mass spectrometry, additional sample preparation steps involving sample clean-up, proteolysis, and peptide recovery are essential. This chapter describes the important procedure of in-gel digestion with particular emphasis on maximum peptide recovery and compatibility for subsequent mass spectrometric analysis.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Mass Spectrometry/methods , Proteins/isolation & purification , Proteins/metabolism , Protein Binding , Proteomics/methods
16.
Anal Chem ; 75(13): 3232-43, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12964774

ABSTRACT

A protocol combining immobilized metal ion affinity chromatography and beta-elimination with concurrent Michael addition has been developed for enhanced analysis of protein phosphorylation. Immobilized metal ion affinity chromatography was initially used to enrich for phosphorylated peptides. Beta-elimination, with or without concurrent Michael addition, was then subsequently used to simultaneously elute and derivatize phosphopeptides bound to the chromatography resin. Derivatization of the phosphate facilitated the precise determination of phosphorylation sites by MALDI-PSD/LIFT tandem mass spectrometry, avoiding complications due to ion suppression and phosphate lability in mass spectrometric analysis of phosphopeptides. Complementary use of immobilized metal ion affinity chromatography and beta-elimination with concurrent Michael addition in this manner circumvented several inherent disadvantages of the individual methods. In particular, (i) the protocol discriminated O-linked glycosylated peptides from phosphopeptides prior to beta-elimination/Michael addition and (ii) the elution of peptides from the chromatography resin as derivatized phosphopeptides distinguished them from unphosphorylated species that were also retained. The chemical derivatization of phosphopeptides greatly increased the information obtained during peptide sequencing by mass spectrometry. The combined protocol enabled the detection and sequencing of phosphopeptides from protein digests at low femtomole concentrations of initial sample and was employed to identify novel phosphorylation sites on the cell adhesion protein p120 catenin and the glycoprotein fetuin.


Subject(s)
Ferric Compounds/chemistry , Nickel/chemistry , Phosphoproteins/analysis , Proteins/analysis , Amino Acid Sequence , Animals , COS Cells , Catenins , Cattle , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chromatography, Affinity/methods , Mice , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Proteins/chemistry , Proteins/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , alpha-Fetoproteins/analysis , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/metabolism , Delta Catenin
17.
J Biol Chem ; 277(16): 13761-70, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-11827966

ABSTRACT

To defend against the potential damages induced by reactive oxygen species, proliferating cells enter a transient cell cycle arrest. We treated mouse fibroblasts with H(2)O(2) and found that sublethal doses of H(2)O(2) induced a transient multi-phase cell cycle arrest at the G(1), S, and G(2) phases but not the M phase. Western blot analysis demonstrated that this transient cell cycle arrest is associated with the down-regulation of cyclins D1 and D3 and up-regulation of the CKI p21(Cip1) expression. We also demonstrate that the induction in p21(Cip1) expression by H(2)O(2) is at least partially mediated at the transcriptional level and can occur in the absence of p53 function. Further immunoprecipitation kinase and immunodepletion assays indicated that in response to H(2)O(2) treatment, the down-regulation of cyclin Ds expression are associated with repression of cyclin D-CDK4, whereas the accumulation of p21(Cip1) is responsible for the inhibition of cyclin E and A-CDK2 activity and associated with the down-regulation of cyclin B-CDC2 activity. These data could account for the cell cycle arrest at the G(1), S, and G(2) phases following H(2)O(2) stimulation. Deletion of p21(Cip1), restoration of cyclin D expression, or overexpression of cyclin E alone is insufficient to effectively overcome the cell cycle arrest caused by sublethal doses of H(2)O(2). By contrast, overexpression of the human Herpesvirus 8 K cyclin, which can mimic the function of cyclin D and E, is enough to override this transient cell cycle arrest. On the basis of our findings, we propose a model in which moderate levels of H(2)O(2) induce a transient multi-phase cell cycle arrest at least partially through up-regulation of p21(Cip1) and down-regulation of cyclin D expression.


Subject(s)
Cell Cycle/drug effects , Cyclins/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Hydrogen Peroxide/pharmacology , 3T3 Cells , Animals , Blotting, Northern , Blotting, Western , Cell Cycle Proteins/metabolism , Cyclin D , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cytoplasm/metabolism , Down-Regulation , Genes, Reporter , Immunoblotting , Isopropyl Thiogalactoside/pharmacology , Mice , Microscopy, Fluorescence , Oxidative Stress , Oxygen/metabolism , Precipitin Tests , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
18.
Oncogene ; 21(7): 981-9, 2002 Feb 07.
Article in English | MEDLINE | ID: mdl-11850815

ABSTRACT

P53 activity plays a key role in mammalian cells when they undergo replicative senescence at their Hayflick limit. To determine whether p63 proteins, members of the family of p53-related genes, are also involved in this process, we examined their expression in serially passaged rat embryo fibroblasts. Upon senescence, two truncated DeltaNp63 proteins decreased in abundance whereas two TAp63 isoforms accumulated. 2-D gel analysis showed that the DeltaNp63 proteins underwent post-translational modifications in both proliferating and senescent cells. Direct binding of DeltaNp63 proteins to a p53 consensus motif was greater in proliferating cells than senescent cells. In contrast p63alpha isoforms bound to DNA in a p53 dependent manner and this was higher in senescent cells than proliferating cells. An interaction of p63alpha proteins with SV40 large tumour antigen was also detected and ectopic expression of DeltaNp63alpha can extend the lifespan of rat embryo fibroblasts. Taken together the results indicate that p63 proteins may play a role in replicative senescence either by competition for p53 DNA binding sites or by direct interaction with p53 protein bound to DNA.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cellular Senescence , Membrane Proteins , Phosphoproteins/metabolism , Phosphoproteins/physiology , Proteins , Trans-Activators/metabolism , Trans-Activators/physiology , Tumor Suppressor Protein p53 , Animals , Binding Sites , Cell Culture Techniques/methods , Cells, Cultured , Consensus Sequence , DNA/metabolism , DNA-Binding Proteins/analysis , DNA-Binding Proteins/immunology , Electrophoresis, Gel, Two-Dimensional , Electrophoretic Mobility Shift Assay , Fibroblasts/physiology , Genes, p53 , NADPH Oxidases , Phosphoproteins/genetics , Phosphoproteins/immunology , Protein Isoforms/immunology , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Sequence Deletion , Trans-Activators/genetics , Trans-Activators/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...