Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Cell ; 21(3): 506-19, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21871841

ABSTRACT

In vertebrates, unfertilized eggs are arrested at metaphase of meiosis II by Mos and Emi2, an inhibitor of the APC/C ubiquitin ligase. In Xenopus, Cdk1 phosphorylates Emi2 and both destabilizes and inactivates it, whereas Mos recruits PP2A phosphatase to antagonize the Cdk1 phosphorylation. However, how Cdk1 phosphorylation inhibits Emi2 is largely unknown. Here we show that multiple N-terminal Cdk1 phosphorylation motifs bind cyclin B1-Cdk1 itself, Plk1, and CK1δ/ε to inhibit Emi2. Plk1, after rebinding to other sites by self-priming phosphorylation, partially destabilizes Emi2. Cdk1 and CK1δ/ε sequentially phosphorylate the C-terminal APC/C-docking site, thereby cooperatively inhibiting Emi2 from binding the APC/C. In the presence of Mos, however, PP2A-B56ß/ε bind to Emi2 and keep dephosphorylating it, particularly at the APC/C-docking site. Thus, Emi2 stability and activity are dynamically regulated by Emi2-bound multiple kinases and PP2A phosphatase. Our data also suggest a general role for Cdk1 substrate phosphorylation motifs in M phase regulation.


Subject(s)
CDC2 Protein Kinase/metabolism , Casein Kinase I/metabolism , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Meiosis , Ovum/cytology , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Xenopus Proteins/metabolism , Animals , CDC2 Protein Kinase/chemistry , Casein Kinase I/chemistry , Cell Cycle Proteins/chemistry , Cell Division , F-Box Proteins/chemistry , Female , Ovum/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Phosphatase 2/chemistry , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-mos/metabolism , Xenopus Proteins/chemistry , Xenopus laevis , Polo-Like Kinase 1
2.
Gene ; 429(1-2): 104-11, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18977421

ABSTRACT

In all vertebrates, mature oocytes arrest at the metaphase of the II meiotic division, while some invertebrates arrest at metaphase-I, others at prophase-I. Fertilization induces completion of meiosis and entry into the first mitotic division. Several experimental models have been considered from both vertebrates and invertebrates in order to shed light on the peculiar aspects of meiotic division, such as the regulation of the cytostatic factor (CSF) and the maturation promoting factor (MPF) in metaphase I or II. Recently, we proposed the oocytes of ascidian Ciona intestinalis as a new model to study the meiotic division. Here, taking advantage of the recent publication of the C. intestinalis genome, we presented a phylogenetic analysis of key molecular components of the CSF-related machinery. We showed that the Mos/MAP kinase pathway is perfectly conserved in ascidians. We demonstrated the presence of a CSF-like activity in metaphase-I arrested C. intestinalis oocytes able to block cell division in two-cell embryos. We further investigated the regulation of CSF by demonstrating that both CSF and MPF inactivation, at the exit of metaphase-I, are independent from protein synthesis, indicating the absence of short-lived factors that regulate metaphase stability, as in other invertebrate species. The results obtained suggest that meiotic regulation in C. intestinalis resembles that of vertebrates, such as Xenopus accordingly to the position of this organism in the evolutionary tree.


Subject(s)
Ciona intestinalis/genetics , Conserved Sequence , Phylogeny , Proto-Oncogene Proteins c-mos/genetics , Amino Acid Sequence , Animals , Ciona intestinalis/cytology , Ciona intestinalis/enzymology , Genome/genetics , Molecular Sequence Data , Oocytes/cytology , Oocytes/enzymology , Protein Biosynthesis , Proto-Oncogene Proteins c-mos/chemistry , Sequence Homology, Amino Acid
3.
BMC Biochem ; 8: 5, 2007 Apr 11.
Article in English | MEDLINE | ID: mdl-17425806

ABSTRACT

BACKGROUND: BIR family proteins are evolutionarily conserved anti-apoptotic molecules. One member of Xenopus BIR family proteins, xEIAP/XLX, is a weak apoptosis inhibitor and rapidly degraded in a cell-free apoptotic execution system derived from interphase egg extracts. However, unfertilized eggs are naturally arrested at the metaphase of meiosis II by the concerted activities of Mos-MEK-p42MAPK-p90Rsk kinase cascade (cytostatic factor pathway) and many mitotic kinases. Previous studies suggest that cytostatic factor-arrested egg extracts are more resistant to spontaneous apoptosis than interphase egg extracts in a p42MAPK-dependent manner. We tested whether xEIAP/XLX might be phosphorylated in cytostatic factor-arrested egg extracts, and also examined whether xEIAP/XLX could be functionally regulated by phosphorylation. RESULTS: We found that p42MAPK was the major kinase phosphorylating xEIAP/XLX in cytostatic factor-arrested egg extracts, and three Ser residues (Ser 235/251/254) were identified as p42MAPK-mediated phosphorylation sites. We characterized the behaviors of various xEIAP/XLX mutants that could not be phosphorylated by p42MAPK. However, neither protein stability nor anti-apoptotic ability of xEIAP/XLX was significantly altered by the substitution of Ser with either Ala or Asp at these three sites. CONCLUSION: xEIAP/XLX is physiologically phosphorylated by p42MAPK in Xenopus unfertilized eggs. However, this protein may not serve as an essential mediator of p42MAPK-dependent anti-apoptotic activity.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Oocytes/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Xenopus Proteins/metabolism , Xenopus laevis , Amino Acid Sequence , Animals , Apoptosis , Cell Extracts , Electrophoretic Mobility Shift Assay , Molecular Sequence Data , Oocytes/cytology , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Phosphoserine/metabolism , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-mos/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus laevis/metabolism
4.
Mol Cell Biol ; 26(14): 5300-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809767

ABSTRACT

The protein kinase Mos is responsible for the activation of MEK1 and p42 mitogen-activated protein kinase during Xenopus oocyte maturation and during mitosis in Xenopus egg extracts. Here we show that the activation of Mos depends upon the phosphorylation of Ser 3, a residue previously implicated in the regulation of Mos stability; the dephosphorylation of Ser 105, a previously unidentified phosphorylation site conserved in Mos proteins; and the regulated dissociation of Mos from CK2beta. Mutation of Ser 3 to alanine and/or mutation of Ser 105 to glutamate produces a Mos protein that is defective for M-phase activation, as assessed by in vitro kinase assays, and defective for induction of oocyte maturation and maintenance of the spindle assembly checkpoint in extracts. Interestingly, Ser 105 is situated at the beginning of helix alphaC in the N-terminal lobe of the Mos kinase domain. Changes in the orientation of this helix have been previously implicated in the activation of Cdk2 and Src family tyrosine kinases. Our work suggests that Ser 105 dephosphorylation represents a novel mechanism for reorienting helix alphaC.


Subject(s)
Mitosis/physiology , Proto-Oncogene Proteins c-mos/metabolism , Xenopus Proteins/metabolism , Animals , Binding Sites , Casein Kinase II/metabolism , Cyclin B/metabolism , Enzyme Activation , Female , In Vitro Techniques , Mutagenesis, Site-Directed , Oocytes/cytology , Oocytes/enzymology , Phosphorylation , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-mos/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Xenopus , Xenopus Proteins/chemistry , Xenopus Proteins/genetics
5.
EMBO J ; 21(22): 6061-71, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12426378

ABSTRACT

c-MOS, a MAP kinase kinase kinase, is a regulator of oocyte maturation. The concentration of c-MOS is controlled in part through its conditional degradation. Previous studies proposed the "second-codon rule", according to which the N-terminal proline (Pro) of c-MOS is a destabilizing residue that targets c-MOS for degradation. We analyzed the degradation signal (degron) of c-MOS in Xenopus oocytes, found it to be a portable degron, and demonstrated that, contrary to the model above, the N-terminal Pro residue of c-MOS is entirely dispensable for its degradation if Ser-2 (encoded Ser-3) of c-MOS is replaced by a small non-phosphorylatable residue such as Gly. The dependence of c-MOS degradation on N-terminal Pro is shown to be caused by a Pro-mediated downregulation of the net phosphorylation of Ser-2, a modification that halts c-MOS degradation in oocytes. Thus, the N-terminal Pro residue of c-MOS is not a recognition determinant for a ubiquitin ligase, in agreement with earlier evidence that Pro is a stabilizing residue in the N-end rule.


Subject(s)
Oocytes/metabolism , Proto-Oncogene Proteins c-mos/metabolism , 3T3 Cells , Amino Acid Sequence , Amino Acid Substitution , Animals , Endopeptidases/metabolism , Fibroblasts/metabolism , Genes, mos , Ligases/metabolism , Mice , Microinjections , Molecular Sequence Data , Phosphorylation , Phosphoserine/chemistry , Progesterone , Proline/chemistry , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-mos/genetics , Rabbits , Recombinant Fusion Proteins/metabolism , Reticulocytes/metabolism , Species Specificity , Xenopus laevis
6.
BMC Biochem ; 3: 6, 2002 Apr 04.
Article in English | MEDLINE | ID: mdl-12022922

ABSTRACT

BACKGROUND: The mos proto-oncogene encodes a cytoplasmic serine/threonine-specific protein kinase with crucial function during meiotic cell division in vertebrates. Based on oncogenic amino acid substitutions the viral derivative, 124-v-Mos, displays constitutive protein kinase activity and functions independent of unknown upstream effectors of mos protein kinase. We have utilized this property of 124-v-Mos and screened for novel mos substrates in immunocomplex kinase assays in vitro. RESULTS: We generated recombinant 124-v-Mos using the baculovirus expression system in Spodoptera frugiperda cells and demonstrated constitutive kinase activity by the ability of 124-v-Mos to auto-phosphorylate and to phosphorylate vimentin, a known substrate of c-Mos. Using this approach we analyzed a panel of acidic and basic substrates in immunocomplex protein kinase assays and identified novel in vitro substrates for 124-v-Mos, the protein tyrosine phosphatase 1B (PTP1B), alpha-casein and beta-casein. We controlled mos-specific phosphorylation of PTP1B and casein in comparative assays using a synthetic kinase-inactive 124-v-Mos mutant and further, tryptic digests of mos-phosphorylated beta-casein identified a phosphopeptide specifically targeted by wild-type 124-v-Mos. Two-dimensional phosphoamino acid analyses showed that 124-v-mos targets serine and threonine residues for phosphorylation in casein at a 1:1 ratio but auto-phosphorylation occurs predominantly on serine residues. CONCLUSION: The mos substrates identified in this study represent a basis to approach the identification of the mos-consensus phosphorylation motif, important for the development of specific inhibitors of the Mos protein kinase.


Subject(s)
Caseins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Animals , Binding Sites , Cell Line , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-mos/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Tubulin/metabolism , Vimentin/metabolism
7.
Prog Cell Cycle Res ; 3: 251-9, 1997.
Article in English | MEDLINE | ID: mdl-9552420

ABSTRACT

The mos proto-oncogene-encoded serine/threonine protein kinase plays a key cell cycle-regulatory role during meiosis. The Mos protein is required for the activation and stabilisation of M phase-promoting factor MPF. As a component of a large multiprotein complex known as the cytostatic factor (CSF), Mos is involved in causing metaphase II arrest of eggs in vertebrates. Upon expression in somatic cells, Mos causes cell cycle perturbations resulting in cytotoxicity and neoplastic transformation. All the known biological activities of Mos are mediated through activation of the mitogen activated protein (MAP) kinase pathway. Here we discuss the interrelationship between Mos and other cell cycle regulators.


Subject(s)
Cell Cycle/physiology , Proto-Oncogene Proteins c-mos/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Female , Humans , Maturation-Promoting Factor/metabolism , Microtubules/metabolism , Models, Biological , Neoplasms/etiology , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction
8.
Mol Cell Biol ; 16(7): 3472-9, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668163

ABSTRACT

The Mos protein is a serine/threonine protein kinase which acts to regulate progression through meiosis in vertebrate oocytes. Although Mos function is dependent on its ability to act as a protein kinase, little is known about the factors which regulate Mos kinase activity. To understand the mechanism by which Mos kinase activity is regulated, we have used molecular modeling to construct a three-dimensional model of Mos based on the crystallographic coordinates of cyclic AMP-dependent kinase (PKA). This model identified a loop in Mos which is positioned near the active site and appears capable of blocking substrate access to the active site. Mutagenesis was used to construct altered forms of the Mos protein with deletions of parts or all of the loop. In vitro kinase assays showed that Mos proteins with the loop removed had up to a fourfold increase in kinase activity compared with the wild-type protein, indicating that the loop acts in an autoinhibitory manner for Mos kinase activity. Point mutations were also made on individual residues of the loop which were determined from the molecular model to be capable of reaching the active site. Determination of the kinase activities of these mutants showed that individual mutations in the loop region are capable of either increasing or decreasing kinase activity with regard to the wild-type protein. These data suggest that the loop identified in Mos acts as an autoinhibitor of kinase activity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Proto-Oncogene Proteins c-mos/chemistry , Proto-Oncogene Proteins c-mos/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Female , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-mos/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Restriction Mapping , Sequence Deletion , Sequence Homology, Amino Acid , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...