Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Hum Reprod ; 37(3): 612-620, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34997960

ABSTRACT

STUDY QUESTION: Are mutations in MOS (MOS proto-oncogene, serine/threonine kinase) involved in early embryonic arrest in infertile women? SUMMARY ANSWER: We identified mutations in MOS that may cause human female infertility characterized by preimplantation embryonic arrest (PREMBA), and the effects of the mutations in human embryonic kidney 293T (HEK293T cells) and mouse oocytes provided evidence for a causal relation between MOS and female infertility. WHAT IS KNOWN ALREADY: MOS, an activator of mitogen-activated protein kinase, mediates germinal vesicle breakdown and metaphase II arrest. Female MOS knockout mice are viable but sterile. Thus, MOS seems to be an important part of the mammalian cell cycle mechanism that regulates female meiosis. STUDY DESIGN, SIZE, DURATION: Whole-exome sequencing, bioinformatics filtering analysis and genetic analysis were performed to identify two different biallelic mutations in MOS in two independent families. The infertile patients presenting with early embryonic arrest were recruited from October 2018 to June 2020. PARTICIPANTS/MATERIALS, SETTING, METHODS: The female patients diagnosed with primary infertility were recruited from the reproduction centres of local hospitals. Genomic DNA from the affected individuals, their family members and healthy controls was extracted from peripheral blood. We performed whole-exome sequencing in patients diagnosed with PREMBA. Functional effects of the mutations were investigated in HEK293T cells by western blotting and in mouse oocytes by microinjection and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE: We identified the homozygous missense mutation c.285C>A (p.(Asn95Lys)) and the compound heterozygous mutations c.467delG (p.(Gly156Alafs*18)) and c.956G>A (p.(Arg319His)) in MOS in two independent patients. The mutations c.285C>A (p.(Asn95Lys)) and c.467delG (p.(Gly156Alafs*18)) reduced the protein level of MOS, and all mutations reduced the ability of MOS to phosphorylate its downstream target, extracellular signal-regulated kinase1/2. In addition, the identified mutations reduced the capacity of exogenous human MOS to rescue the metaphase II exit phenotype, and the F-actin cytoskeleton of mouse oocytes was affected by the patient-derived mutations. LIMITATIONS, REASONS FOR CAUTION: Owing to the lack of in vivo data from patient oocytes, the exact molecular mechanism affected by MOS mutations and leading to PREMBA is still unknown and should be further investigated using knock-out or knock-in mice. WIDER IMPLICATIONS OF THE FINDINGS: We identified recessive mutations in MOS in two independent patients with the PREMBA phenotype. Our findings reveal the important role of MOS during human oocyte meiosis and embryonic development and suggest that mutations in MOS may be precise diagnostic markers for clinical genetic counselling. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the National Natural Science Foundation of China (81725006, 81822019, 81771581, 81971450, 81971382,82001538 and 82071642), the project supported by the Shanghai Municipal Science and Technology Major Project (2017SHZDZX01), the Project of the Shanghai Municipal Science and Technology Commission (19JC1411001), the Natural Science Foundation of Shanghai (19ZR1444500 and 21ZR1404800), the Shuguang Program of the Shanghai Education Development Foundation and the Shanghai Municipal Education Commission (18SG03), the Foundation of the Shanghai Health and Family Planning Commission (20154Y0162), the Capacity Building Planning Program for Shanghai Women and Children's Health Service and the collaborative innovation centre project construction for Shanghai Women and Children's Health. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Infertility, Female , Oncogene Proteins v-mos/genetics , Animals , China , Female , HEK293 Cells , Humans , Infertility, Female/genetics , Infertility, Female/metabolism , Mammals , Mice , Mutation , Oocytes/metabolism , Pregnancy
2.
J Cell Sci ; 127(Pt 12): 2749-60, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24741069

ABSTRACT

Egg activation at fertilization in mammals is initiated by prolonged Ca(2+) oscillations that trigger the completion of meiosis and formation of pronuclei. A fall in mitogen-activated protein kinase (MAPK) activity is essential for pronuclear formation, but the precise timing and mechanism of decline are unknown. Here, we have measured the dynamics of MAPK pathway inactivation during fertilization of mouse eggs using novel chemiluminescent MAPK activity reporters. This reveals that the MAPK activity decrease begins during the Ca(2+) oscillations, but MAPK does not completely inactivate until after pronuclear formation. The MAPKs present in eggs are Mos, MAP2K1 and MAP2K2 (MEK1 and MEK2, respectively) and MAPK3 and MAPK1 (ERK1 and ERK2, respectively). Notably, the MAPK activity decline at fertilization is not explained by upstream destruction of Mos, because a decrease in the signal from a Mos-luciferase reporter is not associated with egg activation. Furthermore, Mos overexpression does not affect the timing of MAPK inactivation or pronuclear formation. However, the late decrease in MAPK could be rapidly reversed by the protein phosphatase inhibitor, okadaic acid. These data suggest that the completion of meiosis in mouse zygotes is driven by an increased phosphatase activity and not by a decline in Mos levels or MEK activity.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Ovum/enzymology , Animals , Calcium Signaling , Enzyme Inhibitors/pharmacology , Female , Fertilization , Genes, Reporter , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Okadaic Acid/pharmacology , Oncogene Proteins v-mos/genetics , Oncogene Proteins v-mos/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Spermatozoa/physiology
3.
Mol Reprod Dev ; 76(3): 289-300, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18671273

ABSTRACT

Resumption of meiosis from diplotene arrest during the first meiotic prophase in vertebrate oocytes is universally controlled by MPF, a heterodimer of Cdk1 and cyclin B. Activation of MPF depends on the withdrawal of Cdk1 inhibition by Wee1/Myt1 kinase on the one hand and the activation of Cdk1 by Cdc25 phosphatase on the other. It is relevant to know whether both these pathways are necessary to rescue diplotene arrest or if either one of them is sufficient. In MIH (17alpha, 20beta dihydroxy-4-pregnen-3-one) incubated perch (Anabas testudineus) oocytes we have examined these possibilities. Perch oocyte extract following MIH incubation showed a significant increase in Myt1 phosphorylation from 12 to 16 hr indicating its progressive deactivation. MIH induced Mos expression markedly increased at 16 hr effecting 95% GVBD. Cycloheximide inhibited MIH induced Mos expression and its phosphorylation, which in turn reduced Myt1 phosphorylation and GVBD. Myt1 phosphorylation was blocked in Mos immunodepleted oocytes. All these suggest the involvement of Mos in Myt1 phosphorylation. Oocytes incubated in MIH for 16 hr activated Cdc25, but such activation could not rescue the inhibition of GVBD due to Myt1 in Mos immunodepleted oocytes. Blocking Cdc25 with an antisense oligo significantly inhibited GVBD even though Myt1 remained deactivated during this period. Taken together, our findings indicate that MIH requires both pathways for perch oocyte maturation: the expression and activation of Mos, which is linked to Myt1 deactivation on the one hand, and the activation of Cdc25 on the other, as blocking either pathway compromised G2-M transition in perch oocytes.


Subject(s)
Cell Cycle Proteins/metabolism , G2 Phase/physiology , Oncogene Proteins v-mos/metabolism , Oocytes/growth & development , Perches/physiology , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Hydroxyprogesterones/metabolism , Maturation-Promoting Factor/genetics , Maturation-Promoting Factor/metabolism , Oncogene Proteins v-mos/genetics , Oogenesis/drug effects , Oogenesis/physiology , Phosphorylation/drug effects , Phosphorylation/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , cdc25 Phosphatases/metabolism
4.
Reprod Toxicol ; 23(1): 20-31, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17055699

ABSTRACT

The widely used hormonal herbicide, 2,4-dichlorophenoxyacetic acid, blocks meiotic maturation in vitro and is thus a potential environmental endocrine disruptor with early reproductive effects. To test whether maturation inhibition was dependent on protein kinase A, an endogenous maturation inhibitor, oocytes were microinjected with PKI, a specific PKA inhibitor, and exposed to 2,4-D. Oocytes failed to mature, suggesting that 2,4-D is not dependent on PKA activity and likely acts on a downstream target, such as Mos. De novo synthesis of Mos, which is triggered by mRNA poly(A) elongation, was examined. Oocytes were microinjected with radiolabelled in vitro transcripts of Mos RNA and exposed to progesterone and 2,4-D. RNA analysis showed progesterone-induced polyadenylation as expected but none with 2,4-D. 2,4-D-activated MAPK was determined to be cytoplasmic in localization studies but poorly induced Rsk2 phosphorylation and activation. In addition to inhibition of the G2/M transition, 2,4-D caused abrupt reduction of H1 kinase activity in MII phase oocytes. Attempts to rescue maturation in oocytes transiently exposed to 2,4-D failed, suggesting that 2,4-D induces irreversible dysfunction of the meiotic signaling mechanism.


Subject(s)
2,4-Dichlorophenoxyacetic Acid/toxicity , Herbicides/toxicity , Meiosis/drug effects , Oocytes/drug effects , Protein Biosynthesis/drug effects , Xenopus laevis , Animals , Cell Cycle/drug effects , Drug Therapy, Combination , Female , MAP Kinase Signaling System/drug effects , Meiosis/physiology , Oncogene Proteins v-mos/biosynthesis , Oncogene Proteins v-mos/genetics , Oocytes/growth & development , Oocytes/metabolism , Progesterone/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , RNA, Messenger/metabolism , RNA, Messenger/pharmacology
5.
Beijing Da Xue Xue Bao Yi Xue Ban ; 37(6): 575-8, 2005 Dec 18.
Article in Chinese | MEDLINE | ID: mdl-16378104

ABSTRACT

OBJECTIVE: To establish a stable cell line, which can express P37 protein of mycoplasma hyorhinis and be regulated by tetracycline, for investigating the effect of p37 on phenotype of cells and its mechanism. METHODS: Recombinant plasmid PcDNA5/FRT/TO-p37 was constructed and cotransfected with pOG44 into Flp-In-T-REx-293 cells by lipofectamine. Positive clones were screened with Hygromycin and Blasticidin. RT-PCR and Western blot were used to exam the mRNA and protein expression in selected clones. The expression level at different inducing times and concentrations of tetracycline were examined. MTT assay was used to observe the effect of P37 on proliferation of 293 cells. RESULTS: P37 protein, which is 43.5x10(3), was expressed in the selected clone as well as secreted from cells. Tetracycline showed a good regulation on the expression of P37 protein, which was not detectable without tetracycline induction. When induced with 2 mg/L tetracycline for 60 hours, the P37 protein expression reached maximum level. Cell growth was promoted after being transfected with p37. CONCLUSION: A stable cell line expressing P37 regularly was established, which provides a good cell model for studying p37 function and its molecular mechanism.


Subject(s)
Bacterial Proteins/metabolism , Mycoplasma hyorhinis/metabolism , Oncogene Proteins v-mos/metabolism , Bacterial Proteins/genetics , Cell Line , Cell Proliferation , Cinnamates/pharmacology , Gene Expression/drug effects , Humans , Hygromycin B/analogs & derivatives , Hygromycin B/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mycoplasma hyorhinis/genetics , Nucleosides/pharmacology , Oncogene Proteins v-mos/genetics , Plasmids , Tetracycline/pharmacology , Transfection
6.
J Cell Biol ; 163(6): 1231-42, 2003 Dec 22.
Article in English | MEDLINE | ID: mdl-14691134

ABSTRACT

In cells containing disrupted spindles, the spindle assembly checkpoint arrests the cell cycle in metaphase. The budding uninhibited by benzimidazole (Bub) 1, mitotic arrest-deficient (Mad) 1, and Mad2 proteins promote this checkpoint through sustained inhibition of the anaphase-promoting complex/cyclosome. Vertebrate oocytes undergoing meiotic maturation arrest in metaphase of meiosis II due to a cytoplasmic activity termed cytostatic factor (CSF), which appears not to be regulated by spindle dynamics. Here, we show that microinjection of Mad1 or Mad2 protein into early Xenopus laevis embryos causes metaphase arrest like that caused by Mos. Microinjection of antibodies to either Mad1 or Mad2 into maturing oocytes blocks the establishment of CSF arrest in meiosis II, and immunodepletion of either protein blocked the establishment of CSF arrest by Mos in egg extracts. A Mad2 mutant unable to oligomerize (Mad2 R133A) did not cause cell cycle arrest in blastomeres or in egg extracts. Once CSF arrest has been established, maintenance of metaphase arrest requires Mad1, but not Mad2 or Bub1. These results suggest a model in which CSF arrest by Mos is mediated by the Mad1 and Mad2 proteins in a manner distinct from the spindle checkpoint.


Subject(s)
Calcium-Binding Proteins/metabolism , Meiosis/physiology , Metaphase/physiology , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Repressor Proteins/metabolism , Animals , Antibodies/pharmacology , Calcium-Binding Proteins/pharmacology , Cell Cycle Proteins , Cell Differentiation/drug effects , Cell Differentiation/physiology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Female , Genes, cdc/drug effects , Genes, cdc/physiology , Mad2 Proteins , Meiosis/drug effects , Metaphase/drug effects , Mutation/genetics , Nuclear Proteins , Oncogene Proteins v-mos/genetics , Oncogene Proteins v-mos/metabolism , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Oogenesis/physiology , Phosphoproteins/pharmacology , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Repressor Proteins/pharmacology , Xenopus laevis
7.
Dev Biol ; 260(2): 438-48, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12921744

ABSTRACT

The molecular events regulating hormone-induced oocyte activation and meiotic maturation are probably best understood in Xenopus laevis. In X. laevis, progesterone activates the G2-arrested oocyte, induces entry into M phase of meiosis I (MI) and resumption of the meiotic cell cycles, and leads to the formation of a mature, fertilizable egg. Oocytes of Xenopus tropicalis offer several practical advantages over those of X. laevis, including faster and more synchronous meiotic cell cycle progression, less seasonal variability, and the availability of transgenic approaches. Previous work found several similarities in the pathways regulating oocyte maturation in the two species. Here, we report several additional ones that are conserved in X. tropicalis. (1). Injection of Mos mRNA into G2-arrested oocytes activates the MAP kinase cascade and induces the G2/MI transition. (2). Injection of the beta subunit of the kinase CK2 (a negative regulator of Mos and oocyte activation) delays the G2/MI transition. (3). Elevating PKA activity blocks progesterone-induced maturation; repressing PKA activity induces entry into MI in the absence of progesterone. (4). LF (anthrax lethal factor), which cleaves certain MAP kinase kinases, strongly reduces both the rate and extent of entry into MI. In contrast to the one previously reported major difference between oocytes of the two species, we find that injection of egg cytoplasm ("MPF activity") into G2-arrested X. tropicalis oocytes induces entry into meiosis I even when protein synthesis is blocked, just as it does in oocytes of X. laevis. These results indicate that much of what we have learned from studies of X. laevis oocytes holds for those of X. tropicalis, and suggest that X. tropicalis oocytes offer a good experimental system for investigating certain questions that require a rapid, synchronous progression through the G2/meiosis I transition.


Subject(s)
Antigens, Bacterial , G2 Phase/physiology , Mitosis/physiology , Oocytes/cytology , Animals , Bacterial Toxins/pharmacology , Casein Kinase II , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Female , G2 Phase/drug effects , Mitosis/drug effects , Oncogene Proteins v-mos/genetics , Oncogene Proteins v-mos/metabolism , Oocytes/drug effects , Oocytes/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Xenopus , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
8.
Hum Pathol ; 33(7): 703-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12196921

ABSTRACT

The c-mos gene and its protein product mos, components of the mitogen-activated protein kinase transduction pathway, are known to be involved in the control of meiosis and mitosis. Apart from a study on lung carcinomas, there is little information about its role in human neoplasia. The aim of this study was to investigate expression of mos in astrocytic tumors and to correlate it with accumulation of p53. We studied expression of mos in 62 cases of supratentorial astrocytic tumor. Intracytoplasmic immunostaining for mos was found in 28 (45%) cases: 3 of 20 (15%) grade 2 astrocytomas, 9 of 20 (45%) grade 3 anaplastic astrocytomas, and 16 of 22 (73%) glioblastomas. Immunopositivity for mos correlated significantly (P < 0.01) with tumor grade but not with p53 expression. In contrast to the findings in relation to lung tumors, immunopositivity for mos in astrocytic tumors did not predict recurrence-free or overall survival time. Cytoplasmic immunostaining was observed in scattered large cortical neurons adjacent to tumors, possibly due to stress-induced abortive entry into the cell cycle. The correlation of mos immunopositivity with tumor grade may reflect the expansion of more malignant mos-positive clones. This study provides evidence that mos may be involved in the neoplastic progression of a proportion of astrocytic tumors.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Oncogene Proteins v-mos/metabolism , Adolescent , Adult , Aged , Astrocytoma/pathology , Astrocytoma/surgery , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Disease-Free Survival , Female , Fluorescent Antibody Technique, Indirect , Genes, mos , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Recurrence, Local , Oncogene Proteins v-mos/genetics , Tumor Suppressor Protein p53/metabolism
9.
J Cell Biol ; 157(4): 603-13, 2002 May 13.
Article in English | MEDLINE | ID: mdl-12011110

ABSTRACT

Vertebrate oocytes arrest in the second metaphase of meiosis (metaphase II [MII]) by an activity called cytostatic factor (CSF), with aligned chromosomes and stable spindles. Segregation of chromosomes occurs after fertilization. The Mos/.../MAPK (mitogen-activated protein kinases) pathway mediates this MII arrest. Using a two-hybrid screen, we identified a new MAPK partner from a mouse oocyte cDNA library. This protein is unstable during the first meiotic division and accumulates only in MII, where it localizes to the spindle. It is a substrate of the Mos/.../MAPK pathway. The depletion of endogenous RNA coding for this protein by three different means (antisense RNA, double-stranded [ds] RNA, or morpholino oligonucleotides) induces severe spindle defects specific to MII oocytes. Overexpressing the protein from an RNA not targeted by the morpholino rescues spindle destabilization. However, dsRNA has no effect on the first two mitotic divisions. We therefore have discovered a new MAPK substrate involved in maintaining spindle integrity during the CSF arrest of mouse oocytes, called MISS (for MAP kinase-interacting and spindle-stabilizing protein).


Subject(s)
Carrier Proteins/isolation & purification , Cell Cycle Proteins/isolation & purification , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/physiology , Meiosis/physiology , Nuclear Proteins/isolation & purification , Oocytes/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cloning, Molecular , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Genes, myc/genetics , Mice , Mice, Knockout , Mitosis/genetics , Molecular Sequence Data , Mutation/genetics , Nuclear Proteins/genetics , Oligonucleotide Probes/pharmacology , Oncogene Proteins v-mos/genetics , Oocytes/cytology , Phenotype , Protein Structure, Tertiary/genetics , RNA, Messenger/genetics , RNA, Messenger/isolation & purification
10.
Oncogene ; 20(25): 3193-205, 2001 May 31.
Article in English | MEDLINE | ID: mdl-11423969

ABSTRACT

The p53 tumor suppressor protein plays a critical role in the regulation of the cell cycle and apoptosis. The importance of p53's functions is underscored by the high incidence of p53 mutations in human cancers. Recently, two p53-related proteins, p73 and p63, were identified as members of the p53 gene family. Multiple isoforms of p73 have been found, including DeltaN variants in which the N-termini are truncated. p63 is expressed as three major forms, p63alpha, p63beta and p63gamma, each of which differ in their C-termini. All three forms can be alternatively transcribed from a cryptic promoter located within intron 3, producing DeltaNp63alpha, DeltaNp63beta and DeltaNp63gamma. The high degree of similarity of p73 and p63 to evolutionarily conserved regions of p53 suggests that these proteins play an important and potentially redundant role in regulating cell cycle arrest and apoptosis. Here we describe the characterization of cell lines generated to inducibly express p63alpha and DeltaNp63alpha. We have found that p63alpha and DeltaNp63alpha can differentially regulate endogenous p53 target genes and induce cell cycle arrest and apoptosis. Deletion of the N-terminal 26 amino acids of DeltaNp63alpha abolished its ability to transactivate p53 target genes and induce cell cycle arrest and apoptosis. This indicates that a putative transactivation domain exists within the N-terminus of the DeltaN variants of p63. Furthermore, the differential regulation of p53 target genes by p63alpha and DeltaNp63alpha suggests that p63 and p53 utilize both similar and different signaling pathways to execute their cellular functions.


Subject(s)
Apoptosis/genetics , Biomarkers, Tumor , Cell Cycle/genetics , Exonucleases , Genes, Tumor Suppressor , Membrane Proteins , Neoplasm Proteins , Phosphoproteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Trans-Activators , 14-3-3 Proteins , DNA-Binding Proteins/genetics , Exoribonucleases , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/genetics , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/genetics , Oncogene Proteins v-mos/genetics , Phosphoproteins/metabolism , Protein Isoforms , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Sequence Deletion , Transcription Factors , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins , bcl-2-Associated X Protein , GADD45 Proteins
11.
Oncogene ; 20(6): 692-703, 2001 Feb 08.
Article in English | MEDLINE | ID: mdl-11314003

ABSTRACT

We previously reported that R7Delta447, a 2954-base-pair (bp) laboratory-generated Moloney murine sarcoma virus, induced subcutaneous tumors in about 14% of infected mice but did not induce brain lesions. We now report that R7Delta447K, a spontaneous mutant of R7Delta447, induced brain lesions as well as subcutaneous tumors in all injected mice. The genomes of the two viruses differ in a single base pair: the deduced Glu(62) of the Mos residue of the R7Delta447 Gag-tMos protein is changed to Lys(62). More R7Delta447 than R7Delta447K focus-forming units were detected in both NIH3T3 and mouse cerebral vascular endothelial (MCVE) cells. However, R7Delta447K transformed NIH3T3 and MCVE cells more acutely than did R7Delta447. A distinctive feature that distinguished the morphologic transformation of R7Delta447- and R7Delta447K-infected MCVE cells is the markedly prolonged spindle-shaped phase exhibited by R7Delta447-infected MCVE cells. In addition, R7Delta447K was more efficient in inducing the phosphorylation of ERK1/2 than R7Delta447 in both MCVE and NIH3T3 cells. Moreover morphologic transformation was inhibited, and levels of phosphorylated ERK1/2 were reduced when R7Delta447- or R7Delta447K-infected NIH3T3 or MCVE cells were grown in the presence of the MEK1/2-specific inhibitor PD98095. Thus, we have identified a key residue in the Gag-tMos protein that profoundly affects activation of the Mos/MEK/ERK pathway, virus and cell replication, morphologic transformation in vitro and pathogenicity in vivo.


Subject(s)
Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Moloney murine sarcoma virus/genetics , Moloney murine sarcoma virus/pathogenicity , Mutation , Oncogene Proteins v-mos/genetics , Oncogene Proteins, Fusion/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphorylation , Skin Neoplasms/genetics
12.
Oncogene ; 20(56): 8045-56, 2001 Dec 06.
Article in English | MEDLINE | ID: mdl-11781817

ABSTRACT

The interferon (IFN)-induced, double stranded RNA (dsRNA)-activated serine/threonine kinase, PKR, is a potent negative regulator of cell growth when overexpressed in yeast or mammalian cells. Paradoxically, while it can function as a tumor suppressor and inducer of apoptosis, it is overexpressed in a variety of human cancers. To resolve this enigma, we established cell-lines that overexpress PKR in non-transformed and in v-mos transformed CHO cells. Overexpression of PKR suppressed the proliferation of CHO cells by inducing a transient G0/G1 arrest, followed by a delayed G2/M arrest, which attenuated cell cycle progression. These effects were accompanied by early induction of p21/WAF-1 and delayed downregulation of CDC2 and cyclin B1. Induction of proapoptotic activity of the ectopic PKR paralleled the onset of G2/M arrest in CHO cells. However, while transiently inducing p21/WAF-1, PKR did not impose G2/M arrest or apoptosis in v-mos-transformed cells, nor was CDC2 or cyclin B1 down-regulated in those cells. These findings link the proapoptotic activity of PKR to the arrest of cell cycle at the G2/M phase. Consequently, the apoptotic activity of PKR could be counter-acted by an oncogene-like v-mos that overrides the G2/M arrest induced by PKR.


Subject(s)
Apoptosis , CDC2 Protein Kinase/metabolism , Cell Transformation, Neoplastic/metabolism , Cyclin B/metabolism , Ecdysterone/analogs & derivatives , Oncogene Proteins v-mos/pharmacology , eIF-2 Kinase/physiology , Animals , CHO Cells , Cell Cycle , Cell Division , Cell Line, Transformed , Cell Transformation, Neoplastic/pathology , Cricetinae , Cyclin B1 , Down-Regulation , Ecdysterone/pharmacology , Flow Cytometry , Kinetics , Oncogene Proteins v-mos/genetics , Transfection , Transformation, Genetic , eIF-2 Kinase/genetics
13.
J Biol Chem ; 274(53): 38083-90, 1999 Dec 31.
Article in English | MEDLINE | ID: mdl-10608877

ABSTRACT

The generation of micronuclei is a reflection of DNA damage, defective mitosis, and loss of genetic material. The involvement of the MAPK pathway in mediating v-ras-induced micronuclei in NIH 3T3 cells was examined by inhibiting MAPK activation. Conversely, the MAPK pathway was constitutively activated by infecting cells with a v-mos retrovirus. Micronucleus formation was inhibited by the MAPK kinase inhibitors PD98059 and U0126, but not by wortmannin, an inhibitor of the Ras/phosphatidylinositol 3-kinase pathway. Transduction of cells with v-mos resulted in an increase in micronucleus formation, also consistent with the involvement of the MAPK pathway. Staining with the anti-centromeric CREST antibody revealed that instability induced by constitutive activation of MAPK is due predominantly to aberrant mitotic segregation, since most of the micronuclei were CREST-positive, reflective of lost chromosomes. A significant fraction of the micronuclei were CREST-negative, reflective of lost acentric chromosome fragments. Some of the instability observed was due to mitotic events, consistent with the increased formation of bi-nucleated cells, which result from perturbations of the mitotic spindle and failure to undergo cytokinesis. This chromosome instability, therefore, is a consequence of mitotic aberrations, mediated by the MAPK pathway, including centrosome amplification and formation of mitotic chromosome bridges.


Subject(s)
Chromosome Deletion , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein p21(ras)/physiology , 3T3 Cells , Animals , Cell Transformation, Viral , Enzyme Activation , Enzyme Inhibitors/pharmacology , Leukemia Virus, Murine/physiology , MAP Kinase Signaling System , Mice , Micronuclei, Chromosome-Defective , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitosis/genetics , Oncogene Proteins v-mos/genetics , Phosphorylation
14.
FEBS Lett ; 461(1-2): 107-10, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10561505

ABSTRACT

When ectopically expressed, the serine/threonine kinase Mos can induce oncogenic transformation of somatic cells by direct phosphorylation of MAP kinase/ERK kinase (MEK1), activating the mitogen-activated protein kinases ERK1 and ERK2. On the other hand, overexpression of Mos in C2C12 myoblasts is not transforming. Mos activates myogenic differentiation by promoting heterodimerization of the MyoD/E12 proteins, increasing the expression of myogenic markers and the positive autoregulatory loop of MyoD. In this study, we show that in myogenic cells, the mitogenic and oncogenic signalling from the Mos/MEK/ERK pathway is suppressed by MyoD through the formation of a heterotrimeric complex.


Subject(s)
MAP Kinase Signaling System , MyoD Protein/metabolism , Oncogene Proteins v-mos/metabolism , Protein Serine-Threonine Kinases , Animals , Blotting, Western , Cell Differentiation , Cell Line , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , Luciferases/metabolism , MAP Kinase Kinase 1 , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , MyoD Protein/genetics , Oncogene Proteins v-mos/genetics , Phosphorylation , Plasmids/metabolism , Protein Binding , Signal Transduction , Transfection
15.
J Biol Chem ; 273(26): 15946-53, 1998 Jun 26.
Article in English | MEDLINE | ID: mdl-9632642

ABSTRACT

Recently, we identified the major in vivo phosphorylation site on v-Mos as Ser-56, which is phosphorylated by cyclic AMP dependent protein kinase (PKA). Others have shown that c-Mos phosphorylation at Ser-3 (equivalent to Ser-34 in v-Mos) is important for the interaction of c-Mos with its substrate MEK and for its stability and cytostatic factor activity in eggs. To investigate the role of Ser-56 phosphorylation, we generated site-directed mutants of v-Mos that would mimic phosphorylation in terms of charge at positions 56 and 34. After mutating serine (S) residues with alanine (A) or glutamic acid (E) in different combinations, various v-Mos mutants were expressed in a rabbit reticulocyte lysate in vitro translation system and in COS-1 or NIH/3T3 cells. The effect of mutations on Mos function was evaluated by in vitro protein kinase assays and by the ability of Mos to cause neoplastic transformation of NIH/3T3 cells. The S56E but not the S56A mutation inhibited v-Mos kinase activity suggesting that Ser-56 phosphorylation has an inhibitory role. As predicted from Xenopus c-Mos studies, S34A but not S34E mutation inhibited v-Mos activity. Studies with the double mutants showed that the S56E mutation but not S56A mutation inhibited v-Mos kinase activity of both S34A and S34E mutants. Interestingly, the S56A mutation blocked the inhibitory effect of the S34A mutation on v-Mos kinase suggesting that in c-Mos the corresponding serine (Ser-25) can influence the regulation of c-Mos by Ser-3. Results showing inhibition of v-Mos kinase activity of the S34E mutant by the S56E mutation is significant as it suggests that doubly phosphorylated Mos at these residues would be inactive. Because residues corresponding to both v-Mos Ser-34 and Ser-56 are evolutionarily conserved in c-Mos, the kinase activity of c-Mos during meiosis may also be regulated in the same manner as v-Mos kinase activity.


Subject(s)
Oncogene Proteins v-mos/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Serine/metabolism , 3T3 Cells , Animals , COS Cells , Cyclic AMP-Dependent Protein Kinases/metabolism , Mice , Mutagenesis, Site-Directed , Oncogene Proteins v-mos/genetics , Phosphorylation , Proto-Oncogene Proteins c-mos/genetics , Rabbits , Structure-Activity Relationship , Xenopus
16.
Oncogene ; 12(10): 2203-11, 1996 May 16.
Article in English | MEDLINE | ID: mdl-8668347

ABSTRACT

The function of the Xenopus c-mos proto-oncogene product (Mos(xe)) has been investigated during oocyte maturation. Experiments with a new antibody able to immunoblot Mos(xe) demonstrated the time course of MAP kinase (MAP K) activation in oocytes paralleled Mos(xe) accumulation, and in activated eggs the deactivation of MAP K paralleled the degradation of Mos(xe). Ablation of Mos synthesis by microinjection of antisense oligodeoxynucleotides abolished activation of MAP K by progesterone, but microinjection of GST-Mos fully restored both MAP K activation and germinal vesicle breakdown (GVBD). The Mos(xe) level at metaphase of Meiosis I (MI) was 2 - 3-fold less than that at metaphase of Meiosis II (MII), but MAP K activation was maximal at metaphase in both MI and MII. In the transition between MI and MII, both cyclin B and Mos(xe) levels rapidly declined in the presence of cycloheximide and injection of exogenous GST-Mos(xe) did not prevent degradation of either protein, although MAP K was activated. Microinjection of GST-Mos(xe) into oocytes was able to activate MAP K before GVBD and H1 kinase activation, and microinjection of constitutively-activated thiophosphorylated MAP K induced de novo synthesis of Mos(xe) before H1 kinase activation, suggesting the existence of a positive feedback loop between MAP K and Mos(xe) accumulation.


Subject(s)
Oncogene Proteins v-mos/physiology , Oocytes/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Activation , Female , Genes, mos , Meiosis/physiology , Metaphase/physiology , Oncogene Proteins v-mos/genetics , Oncogene Proteins v-mos/metabolism , Oocytes/cytology , Oocytes/enzymology , Xenopus laevis
17.
FEBS Lett ; 385(3): 131-7, 1996 May 06.
Article in English | MEDLINE | ID: mdl-8647237

ABSTRACT

PC12 pheochromocytoma cells possess four known MEK activators: A-, B-, c-Raf-1 and MEKK. In order to examine whether differentiation factors or growth factors have a Raf isozyme preference for activation of the mitogenic cytoplasmic Raf-MEK-MAPK protein kinase cascade, the activation kinetics of these enzymes in response to epidermal growth factor (EGF) and nerve growth factor (NGF) were compared. An initial activation of all three Raf kinases was noticed, but only A- and B-Raf showed sustained activation by NGF, which was not seen after EGF treatment. Furthermore, expression of oncogenic versions of all three Raf kinases as well, as a potentially Raf-independent MEK activator, v-Mos, leads to activation of MAPK and to differentiation of PC12 cells. These data suggest a differential regulation of Raf kinases and that probably no alternative Raf substrates are involved in differentiation processes of PC12 cells.


Subject(s)
Epidermal Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Cell Division , Enzyme Activation , Immunoblotting , Isoenzymes/metabolism , Kinetics , Mitogen-Activated Protein Kinase Kinases , Neurites/ultrastructure , Neurons/cytology , Oncogene Proteins v-mos/genetics , Oncogene Proteins v-mos/pharmacology , PC12 Cells , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Rats , Transfection
18.
Mol Cell Biol ; 16(3): 800-9, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8622681

ABSTRACT

We investigated the effect of cyclic AMP-dependent protein kinase (PKA ) on v-Mos kinase activity. Increase in PKA activity in vivo brought about either by forskolin treatment or by overexpression of PKA catalytic subunit resulted in a significant inhibition of v-Mos kinase activity. The purified PKA catalytic subunit was able to phosphorylate recombinant p37v-mos in vitro, suggesting that the mechanism of in vivo inhibition of v-Mos kinase involves direct phosphorylation by PKA. Combined tryptic phosphopeptide two-dimensional mapping analysis and in vitro mutagenesis studies indicated that Ser-56 is the major in vivo phosphorylation site on v-Mos. In vivo phosphorylation at Ser-56 correlated with slower migration of the v-Mos protein during sodium dodecyl sulfate-polyacrylamide gel electrophoresis. However, even though Ser-56 was phosphorylated by PKA, this phosphorylation was not involved in the inhibition of v-Mos kinase. The alanine-for-serine substitution at residue 56 did not affect the ability of v-Mos to autophosphorylate in vitro or, more importantly, to activate MEK1 in transformed NIH 3T3 cells. We identified Ser-263 phosphorylation, the Ala-263 mutant of v-Mos was not inhibited by forskolin treatment. From our results, we propose that the known inhibitory role of PKA in the initiation of oocyte maturation in mice could be explained at least in part by its inhibition of Mos kinase.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Oncogene Proteins v-mos/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Mice , Molecular Sequence Data , Mutation , Oncogene Proteins v-mos/antagonists & inhibitors , Oncogene Proteins v-mos/genetics , Phosphorylation , Serine/metabolism
19.
Oncogene ; 11(12): 2639-48, 1995 Dec 21.
Article in English | MEDLINE | ID: mdl-8545121

ABSTRACT

We undertook a study to determine if the serine-threonine kinase-encoding v-mos oncogene regulated the expression of the urokinase-type plasminogen activator. An expression vector encoding v-mos, but not a kinase-inactive mutant, stimulated urokinase promoter activity in CAT assays employing a squamous cell carcinoma cell line. The induction of urokinase promoter activity by v-mos was mediated, in part, via an increased AP-1 activity since (a) mutation of 2 AP-1 binding sites (at -1967 and -1885), or the co-expression of a transactivation domain-lacking c-jun mutant reduced the induction of the urokinase promoter by v-mos and (b) expression of v-mos increased the activity of a CAT reporter driven by three AP-1 tandem repeats. The stimulation of the urokinase promoter by v-mos was partially countered by co-expression of an ERK1/ERK2-inactivating phosphatase. Western blotting and zymographic analysis indicated that v-mos-transformed NIH3T3 cells (MSV NIH-3T3) secreted more urokinase compared with NIH3T3 cells and this was associated with a higher level of activated ERK1 and ERK2. Expression of a catalytically-inactive MAPKK mutant reduced the activity of a urokinase promoter-driven CAT reporter in the MSV NIH-3T3 cells. In conclusion, the data herein indicate that urokinase expression is regulated by v-mos through a MAPKK-dependent signaling pathway.


Subject(s)
Gene Expression Regulation, Enzymologic , Oncogene Proteins v-mos/genetics , Oncogenes , Urokinase-Type Plasminogen Activator/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites , Humans , Mice , Mitogen-Activated Protein Kinase Kinases , Molecular Sequence Data , Promoter Regions, Genetic , Protein Kinases/physiology , Transcription Factor AP-1/metabolism , Transfection , Tumor Cells, Cultured
20.
Biokhimiia ; 60(11): 1881-8, 1995 Nov.
Article in Russian | MEDLINE | ID: mdl-8590759

ABSTRACT

The effects of exogenous human p53 and its various mutants (Ala-141, His-175, His-194, Trp-248, His-273) on two key enzymes of purine uptake, adenosine deaminase (AD) and hypoxanthine phosphoribosyl transferase (HPRT), has been studied in Rat 1 immortalized fibroblasts and their sublines transformed by N-RAS or v-mos oncogenes. Introduction into Rat1 cells of both wild type (wt) and mutant p53 produced a 2- to 7.5-fold increase in the AD activity, p53 mutants having a stronger effect than p53wt. In contrast, the HPRT activity decreased 8- to 10-fold in cells containing exogenous p53wt, while p53 mutants partly lost their ability to inhibit HPRT. Transformation of Rat1 by ras or mos oncogenes was also accompanied by an increase in the AD activity (4-5-fold and 1.5-2-fold, respectively) as well as by suppression of HPRT (20-fold and 2-fold, respectively). However, simultaneous expression of exogenous p53 and ras or p53 and mos produced opposite effects, i.e., a dramatic decrease in the AD activity and complete (p53wt, His-273) or partial (His-175, Trp-248) restoration of the HPRT activity. Possible functional significance and mechanisms of AD and HPRT regulation by p53 as well as the role of modifications of activity of nucleotide synthesis enzymes in the cooperative effect of predominant oncogenes and mutant p53 oncogenes in tumour transformation are discussed.


Subject(s)
Adenosine Deaminase/metabolism , Genes, ras , Hypoxanthine Phosphoribosyltransferase/metabolism , Oncogene Proteins v-mos/genetics , Tumor Suppressor Protein p53/physiology , Animals , Cell Line, Transformed , Humans , Mutation , Rats , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...