Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Biol Reprod ; 78(2): 218-33, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17687118

ABSTRACT

Maturation of immature bovine oocytes requires cytoplasmic polyadenylation and synthesis of a number of proteins involved in meiotic progression and metaphase-II arrest. Aurora serine-threonine kinases--localized in centrosomes, chromosomes, and midbody--regulate chromosome segregation and cytokinesis in somatic cells. In frog and mouse oocytes, Aurora A regulates polyadenylation-dependent translation of several mRNAs such as MOS and CCNB1, presumably by phosphorylating CPEB, and Aurora B phosphorylates histone H3 during meiosis. We analyzed the expression of three Aurora kinase genes--AURKA, AURKB, and AURKC--in bovine oocytes during meiosis by reverse transcription followed by quantitative real-time PCR and immunodetection. Aurora A was the most abundant form in oocytes, both at mRNA and protein levels. AURKA protein progressively accumulated in the oocyte cytoplasm during antral follicle growth and in vitro maturation. AURKB associated with metaphase chromosomes. AURKB, AURKC, and Thr-phosphorylated AURKA were detected at a contractile ring/midbody during the first polar body extrusion. CPEB, localized in oocyte cytoplasm, was hyperphosphorylated during prophase/metaphase-I transition. Most CPEB degraded in metaphase-II oocytes and remnants remained localized in a contractile ring. Roscovitine, U0126, and metformin inhibited meiotic divisions; they all induced a decrease of CCNB1 and phospho-MAPK3/1 levels and prevented CPEB degradation. However, only metformin depleted AURKA. The Aurora kinase inhibitor VX680 at 100 nmol/L did not inhibit meiosis but led to multinuclear oocytes due to the failure of the polar body extrusion. Thus, in bovine oocyte meiosis, massive destruction of CPEB accompanies metaphase-I/II transition, and Aurora kinases participate in regulating segregation of the chromosomes, maintenance of metaphase-II, and formation of the first polar body.


Subject(s)
Meiosis , Oocytes/growth & development , Protein Serine-Threonine Kinases/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Aurora Kinase A , Aurora Kinase B , Aurora Kinase C , Aurora Kinases , Butadienes/pharmacology , CDC2 Protein Kinase/analysis , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cattle , Cyclin B/analysis , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1 , Cytoplasm/chemistry , Cytoplasm/metabolism , Embryo, Mammalian/metabolism , Female , Fertilization , Meiosis/drug effects , Metformin/pharmacology , Nitriles/pharmacology , Oocytes/chemistry , Oocytes/metabolism , Piperazines/pharmacology , Polyadenylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-mos/analysis , Proto-Oncogene Proteins c-mos/genetics , Proto-Oncogene Proteins c-mos/metabolism , Purines/pharmacology , RNA, Messenger/metabolism , Roscovitine , mRNA Cleavage and Polyadenylation Factors/analysis , mRNA Cleavage and Polyadenylation Factors/genetics
2.
Reproduction ; 131(5): 861-74, 2006 May.
Article in English | MEDLINE | ID: mdl-16672351

ABSTRACT

RNA interference (RNAi) has been used for selective degradation of an mRNA transcript or inhibiting its translation to a functional protein in various species. Here, we applied the RNAi approach to suppress the expression of the maternal transcript C-mos and embryonic transcripts Oct-4 in bovine oocytes and embryos respectively, using microinjection of sequence-specific double-stranded RNA (dsRNA). For this, 435 bp C-mos and 341 bp Oct-4 dsRNA were synthesized and microinjected into the cytoplasm of immature oocytes and zygotes respectively. In experiment 1, immature oocytes were categorized into three groups: those injected with C-mos dsRNA, RNase-free water and uninjected controls. In experiment 2, in vitro produced zygotes were categorized into three groups: those injected with Oct-4 dsRNA, RNase-free water and uninjected controls. The developmental phenotypes, the level of mRNA and protein expression were investigated after treatment in both experiments. Microinjection of C-mos dsRNA has resulted in 70% reduction of C-mos transcript after maturation compared to the water-injected and uninjected controls (P<0.01). Microinjection of zygotes with Oct-4 dsRNA has resulted in 72% reduction in transcript abundance at the blastocyst stage compared to the uninjected control zygotes (P<0.01). Moreover, a significant reduction in the number of inner cell mass (ICM) cells was observed in Oct-4 dsRNA-injected embryos compared to the other groups. From oocytes injected with C-mos dsRNA, 60% showed the extrusion of the first polar body compared to 50% in water-injected and 44% in uninjected controls. Moreover, only oocytes injected with C-mos dsRNA showed spontaneous activation. In conclusion, our results demonstrated that sequence-specific dsRNA can be used to knockdown maternal or embryonic transcripts in bovine embryogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-mos/genetics , RNA Interference , RNA, Double-Stranded/administration & dosage , Animals , Base Sequence , Blastocyst/cytology , Blotting, Western/methods , Cattle , Cells, Cultured , DNA Primers/genetics , Embryonic Development , Female , Fertilization in Vitro , Microinjections , Molecular Sequence Data , Octamer Transcription Factor-3/analysis , Oogenesis , Proto-Oncogene Proteins c-mos/analysis , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling , Transcription, Genetic
3.
An. R. Acad. Farm ; 71(3): 525-534, jul. 2005. ilus
Article in En | IBECS | ID: ibc-042060

ABSTRACT

La familia del factor de crecimiento transformante-β (TGFβ) se destaca particularmente entre los factores de crecimiento como regulador de la proliferación y diferenciación celular. El TGFβ promueve el crecimiento de los tejidos y la morfogénesis durante la embriogénesis en organismos tan diversos como nematodos, mosca de la fruta y humanos. Para elucidar las bases de esta gran diversidad de respuestas del TGFβ en diferentes tipos celulares, hemos trazado la vía que comunica los receptores de membrana del TGFβ con los genes diana. El TGFβ forma un complejo receptor en el cual la quinasa del receptor tipo II fosforila y activa el receptor tipo I. Este receptor tipo I fosforila y activa los factores de transcripción Smad. Una vez activados, los factores Smad entran en el núcleo con el fin de formar complejos para el reconocimiento y regulación (activación o represión) de genes específicos. La señalización del TGFβ hace disminuir la actividad CDK y provoca la represión de distintos genes promotores del crecimiento. La vía del TGFβ, cuando se altera, juega un papel esencial en la tumorigénesis y en la metástasis


The transforming growth factor-β (TGFβ) family is particularly prominent among growth factors controlling cell proliferation and differentiation, and fosters tissue growth and morphogenesis during embryogenesis in organisms as diverse as the nematode, fruit fly, and human. To elucidate the basis for the great diversity of TGFβ responses in different cell types, we delineated the pathway linking membrane TGFβ receptors to target genes. TGFβ assembles a receptor complex in which the type II receptor kinase phosphorylates and activates the type I receptor. This type I receptor phosphorylates and activates Smad transcription factors. A receptor-activated Smad complex enters the nucleus to find partners for the recognition and regulation (activation of repression) of specific genes. TGFβ signaling decreases CDK activity and causes the repression of several growthpromoting genes. The TGFβ pathway, when altered, plays an essential role in tumorigenesis and metastasis


Subject(s)
Humans , Cyclin-Dependent Kinases/analysis , Transforming Growth Factor beta/analysis , Oncogenes , Cyclins/analysis , Transcription Factors/analysis , Neoplasm Metastasis/pathology , Proto-Oncogene Proteins c-mos/analysis
4.
Am J Clin Pathol ; 120(5): 699-705, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14608895

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 our previous studies on lung carcinomas and astrocytic gliomas, little has been published about its role in human neoplasia. The aim of this study was to investigate the expression of mos in ependymal neoplasms and to correlate it with tumor grade, proliferative fraction, and clinical behavior. We studied mos expression in biopsy specimens from 34 patients with ependymomas. Intracytoplasmic immunopositivity for mos was found in 16 (47%) and was associated significantly with tumor grade: 5 (24%) of 21 grade II ependymomas; 11 (85%) of 13 grade III anaplastic ependymomas (P < .01). Tumors with an MIB-1 labeling index of more than 4% were significantly more likely than those with a lower proliferative fraction to be immunopositive for mos (P = .012). Expression of mos showed a significant negative association with recurrence-free interval (P = .05) but not with overall survival. Our results suggest that overexpression of mos identifies a biologically aggressive subgroup of ependymal tumors and may be involved in their neoplastic progression.


Subject(s)
Brain Neoplasms/chemistry , Ependymoma/chemistry , Proto-Oncogene Proteins c-mos/analysis , Adolescent , Adult , Aged , Astrocytoma/chemistry , Astrocytoma/pathology , Brain Neoplasms/pathology , Child , Child, Preschool , Ependymoma/pathology , Female , Follow-Up Studies , Glioma , Humans , Immunohistochemistry , Infant , MAP Kinase Signaling System , Male , Middle Aged , Neoplasm Staging
5.
Exp Cell Res ; 253(2): 413-21, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10585264

ABSTRACT

In Xenopus eggs, metaphase II arrest is due to the cytostatic factor that maintains a high level of MPF activity. Kinases are important in this phenomenon since p39(mos) and MAPK play a part in the cytostatic activity whereas p34(cdc2) is the catalytic subunit of MPF. Fertilization induces a rise in intracellular calcium leading to egg activation that can be mimicked by calcium-increasing agents such as calcium ionophore. We have performed on Xenopus eggs a biochemical comparison of the effects of the kinase inhibitor 6-DMAP and the calcium ionophore. Both drugs were able to induce pronucleus formation but the underlying molecular events were different. The inactivation of MAPK occurred earlier in eggs exposed to 6-DMAP. Cyclins B1 and B2 were stable and p39(mos) was proteolysed in 6-DMAP-treated eggs while the three proteins underwent degradation in A23187-treated ones. These results suggest a differential regulation of ubiquitin-dependent proteolysis of cyclin B and p39(mos).


Subject(s)
Adenine/analogs & derivatives , Cyclin B/metabolism , Enzyme Inhibitors/pharmacology , Oocytes/enzymology , Proto-Oncogene Proteins c-mos/metabolism , Adenine/pharmacology , Animals , CDC2 Protein Kinase/analysis , CDC2 Protein Kinase/metabolism , Calcimycin/pharmacology , Calcium/metabolism , Cell Nucleus/enzymology , Cyclin B/analysis , Female , Ionophores/pharmacology , Metaphase/physiology , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 1/metabolism , Oocytes/cytology , Oocytes/drug effects , Proto-Oncogene Proteins c-mos/analysis , Xenopus laevis
6.
FEBS Lett ; 457(2): 175-8, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10471773

ABSTRACT

We have studied the effect of a protein tyrosine phosphatases (PTP) inhibitor on calcium-induced activation of Xenopus laevis oocytes arrested at metaphase II. Ammonium molybdate microinjection blocked pronucleus formation following A23187 treatment while cortical granules still underwent exocytosis. Pronuclei still occurred in ammonium molybdate-injected oocytes following 6-DMAP addition. Changes that usually occurred following A23187 exposure were inhibited in the presence of ammonium molybdate in the oocyte: MAPK dephosphorylation, p34(cdc2) rephosphorylation and cyclin B2 and p39(mos) proteolysis. These results suggest that a PTP is involved in the activation of the ubiquitin-dependent degradation machinery.


Subject(s)
Calcium/metabolism , Metaphase/physiology , Protein Tyrosine Phosphatases/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Calcimycin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Maturation-Promoting Factor/analysis , Metaphase/drug effects , Molybdenum/pharmacology , Oocytes/cytology , Oocytes/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Proto-Oncogene Proteins c-mos/analysis , Xenopus laevis
7.
Biol Reprod ; 56(1): 260-5, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9002658

ABSTRACT

The c-mos proto-oncogene product Mos is believed to be an active component of the cytostatic factor that stabilizes and sustains the activity of maturation-promoting factor. Mos has been found to be responsible for the metaphase arrest of oocytes at the second meiotic division in both Xenopus and the mouse. In this study, we have demonstrated, by Western blot and immunoprecipitation analysis, that an approximately 39-kDa protein, identified as Mos, was present in in vitro-matured (metaphase II stage) bovine oocytes but disappeared in parthenogenetically activated oocytes. The oocytes actively synthesized p39mos at the metaphase II stage (between 22 and 26 h of in vitro maturation [IVM]), whereas little p39mos synthesis was detected during the first 4 h of IVM and it was nondetectable during aging at 44-48 h of IVM, when oocytes lose the capability of normal development after fertilization. Ethanol activation of mature oocytes led to the disappearance of p39mos. beta-Tubulin, but not p34cdc2, was co-precipitated with Mos when extracts of metaphase II-stage bovine oocytes were incubated with Mos antiserum. These results demonstrated that Mos is present and actively synthesized in mature bovine oocytes and that oocytes aged beyond the optimal time for fertilization seem to lose the ability to synthesize the Mos protein. beta-Tubulin was found to be associated with Mos, which suggests a possible role for the cytoskeletal protein in maintaining the meiotic arrest in mature bovine oocytes.


Subject(s)
Oocytes/metabolism , Proto-Oncogene Proteins c-mos/biosynthesis , Amino Acid Sequence , Animals , Blotting, Western , Cattle , Cells, Cultured , Ethanol/pharmacology , Female , Humans , Immunosorbent Techniques , Mice , Molecular Sequence Data , Oocytes/drug effects , Oocytes/growth & development , Proto-Oncogene Mas , Proto-Oncogene Proteins c-mos/analysis , Xenopus
8.
Mol Reprod Dev ; 42(4): 397-406, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8607968

ABSTRACT

In mice, expression of the transcription factor Oct-3 and the proto-oncogene c-mos is limited to germ cells, suggesting a specific role for these factors in gamete physiology and early embryonic development. We have studied the expression pattern of Oct-3 and c-mos in various reproductive as well as control tissues in the cynomolgus monkey, using reverse transcriptase polymerase chain reaction (RT-PCR) and Northern analysis. Analogously with the data from the mouse model, strong expression of Oct-3 and c-mos could be detected in monkey ovary and oocytes. Unexpectedly, strong expression of c-mos was demonstrable in the pituitary gland and the amount of mRNA expression in the pituitary was roughly equal to that found in the ovary. Of the tissues examined, the testicular expression of c-mos was the most intense. Weak signal for c-mos mRNA was also seen in hypothalamus and brain; however, all other tissue types examined were negative for c-mos expression. In addition to the oocytes, expression of Oct-3 mRNA was detected in the ovarian granulosa cells, fallopian tube, myometrium, cervix, breast, liver, adrenal gland, pituitary, hypothalamus, brain cortex, prostate, and in testis. Thus, in the cynomolgus monkey, Oct-3 is predominantly, but not specifically, expressed in reproductive tissues. In the female monkey reproductive organs, the expression of c-mos seems to be germ cell specific. Therefore, further characterization of c-mos and Oct-3 functions in primate reproductive physiology, especially in gametogenesis and early embryonic development, is highly warranted.


Subject(s)
DNA-Binding Proteins/analysis , Germ Cells/metabolism , Proto-Oncogene Proteins c-mos/analysis , RNA, Messenger/analysis , Transcription Factors/analysis , Animals , Base Sequence , DNA-Binding Proteins/biosynthesis , Female , Gene Expression Regulation , Macaca fascicularis , Male , Molecular Sequence Data , Octamer Transcription Factor-3 , Organ Specificity , Proto-Oncogene Proteins c-mos/biosynthesis , Transcription Factors/biosynthesis
9.
Gen Comp Endocrinol ; 99(2): 127-36, 1995 Aug.
Article in English | MEDLINE | ID: mdl-8536921

ABSTRACT

Proto-oncogenes are said to influence the regulation of cellular growth and differentiation. Myc, Fos, Jun, and Mos protein localization has been studied by immunocytochemistry in the testis of the frog, Rana esculenta, during the annual reproductive cycle. Oncoproteins have been localized in the primary and secondary (I and II) spermatogonia (SPG). Myc and Mos also appear in I and II spermatocytes (SPC) while Jun appears in II SPC. Myc, Fos, and Jun in SPG translocate in the nucleus during the periods of active spermatogenesis. Myc, Fos, and Jun are also localized in Sertoli cells. Fos is present in interstitial cells during the period characterized by the androgen peak which precedes the sharp increase of estradiol. It is suggested that proto-oncogene activity exerts a regulatory role in steroidogenesis and spermatogenesis.


Subject(s)
Proto-Oncogenes/physiology , Rana esculenta/genetics , Reproduction/physiology , Testis/physiology , Androgens/metabolism , Animals , Estradiol/metabolism , Genes, fos/physiology , Genes, jun/physiology , Genes, mos/physiology , Genes, myc/physiology , Immunohistochemistry , Male , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/analysis , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-mos/analysis , Proto-Oncogene Proteins c-mos/genetics , Proto-Oncogene Proteins c-mos/metabolism , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rana esculenta/metabolism , Rana esculenta/physiology , Seasons , Sertoli Cells/chemistry , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatogenesis/physiology , Spermatozoa/chemistry , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/chemistry , Testis/cytology
10.
Mol Cell Endocrinol ; 109(2): 127-32, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7664974

ABSTRACT

The objective of the present paper was to do a comparative study to assess somatic versus germ cell localization of c-mos products in the testis. In mouse and amphibian oocytes, c-mos activity is necessary for meiotic maturation. Lack of c-mos expression has been reported in somatic cells of male and female gonads while transcripts have been found in germ cells of testis and ovary. Using a v-mos probe, we report here the detection of a c-mos related transcript (1.7 kb) in the dogfish Scyliorhinus canicula testis. Western blot analysis detects two proteins of 106 and 32 kDa. A specific immunostaining was exclusively localized in the interstitial tissue while the germinal compartment was completely negative. In conclusion, our results indicate for the first time the presence of c-mos products in an elasmobranch species and, moreover, their presence in somatic testicular cells rather than germ cells. Therefore, this finding in an ancient vertebrate indicates that c-mos activity does not have a direct universal role in the regulation of spermatogenesis.


Subject(s)
Dogfish/metabolism , Proto-Oncogene Proteins c-mos/analysis , Sertoli Cells/chemistry , Testis/cytology , Animals , Blotting, Western , Male , Molecular Weight , Proto-Oncogene Proteins c-mos/genetics , Proto-Oncogene Proteins c-mos/immunology , Proto-Oncogene Proteins c-mos/physiology , RNA, Messenger/analysis , Spermatocytes/chemistry , Spermatogenesis , Testis/chemistry
11.
Cancer ; 73(6): 1721-7, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-7908857

ABSTRACT

BACKGROUND: It has been suggested that tumorigenesis of the germ cell tumor of the testis includes abnormal and developmentlike differentiation of primordial germ cells to several mature type tumors. METHODS: To clarify roles of protooncogenes in the unique tumorigenic mechanism in the human germ cell tumor, the authors examined the expression of 15 protooncogenes in human primary germ cell tumors of the testis with Northern blot analyses. RESULTS: Fifteen (94%) of 16 seminomas and 5 (83%) of 6 embryonal carcinomas had a significant levels of N-myc expression, whereas they did not express two receptor type protooncogenes, c-erbB-1 and c-erbB-2. In contrast, some immature teratomas had a high level of c-erbB-1 expression, and an advanced case showed a significant level of c-erbB-2 expression. Immature teratomas did not show N-myc expression. Higher levels of c-mos expression were observed in several cases of seminomas and embryonal carcinomas. Expression of c-Ki-ras or N-ras was observed in all histologic subgroups and normal testes. CONCLUSION: A significant level of N-myc expression may be essential for undifferentiated tumors including seminoma and embryonal carcinoma, whereas c-erbB-1 and possibly c-erbB-2 may have important roles in the differentiated tumors such as immature teratoma. These results suggest that some of the protooncogene expression may be switched critically during the differentiation from seminomas or embryonal carcinomas to the more differentiated-type tumor.


Subject(s)
Gene Expression Regulation, Neoplastic , Germinoma/genetics , Proto-Oncogenes/genetics , Testicular Neoplasms/genetics , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma, Embryonal/genetics , ErbB Receptors/analysis , ErbB Receptors/genetics , Genes, fms/genetics , Genes, mos/genetics , Genes, myc/genetics , Genes, ras/genetics , Humans , Male , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mos/analysis , Proto-Oncogene Proteins c-mos/genetics , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-raf , Receptor, ErbB-2 , Receptor, Macrophage Colony-Stimulating Factor/analysis , Receptor, Macrophage Colony-Stimulating Factor/genetics , Seminoma/genetics , Teratoma/genetics , Testis/chemistry
12.
Oncogene ; 8(6): 1685-91, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8502488

ABSTRACT

Although the human c-mos proto-oncogene has been characterized for more than a decade, very little is known about its protein product and its expression in somatic cells. We generated three human c-mos-specific antisera and report here the detection of c-mos protein in a human neuroblastoma cell line, SK-N-BE2 (BE2). Both Western (immuno-) blot and immunoprecipitation analyses detected a p37 as the major form and p40 and p35 as minor forms of the c-mos protein. Using Northern blot analysis, 3.5- and 1.7-kb c-mos messages were detected. Using a highly sensitive method that combines reverse transcription and the polymerase chain reaction (RT-PCR), c-mos RNA was detected in all the human samples examined. With Western blot analysis, we further showed that c-mos proteins are expressed in cervical carcinoma-derived cell lines. This ubiquitous expression of low levels of c-mos suggests a fundamental role for the c-mos proto-oncogene.


Subject(s)
Genes, mos , Proto-Oncogene Proteins c-mos/analysis , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression , Humans , Mice , Molecular Sequence Data , Neuroblastoma , Oligodeoxyribonucleotides , Polymerase Chain Reaction , Proto-Oncogene Mas , Proto-Oncogene Proteins c-mos/biosynthesis , Proto-Oncogene Proteins c-mos/genetics , RNA, Neoplasm/analysis , RNA, Neoplasm/genetics , Tumor Cells, Cultured , Uterine Cervical Neoplasms
13.
Oncogene ; 7(12): 2489-98, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1461652

ABSTRACT

We have characterized the mouse Mos proto-oncogene product, pp39Mos, in murine fibroblasts. When expressed in NIH3T3 cells under the influence of the long terminal repeat regulatory element from Moloney murine sarcoma virus [NIH(pTS-1) cells], the Mos protein was present in low levels and had a half-life of about 30 min. In extracts from NIH(pTS-1) cells, we detected additional forms of Mos protein that apparently arose from internal initiation codons (p24Mos and p29Mos) or from upstream non-AUG initiation codons (p42Mos and p44Mos). The Mos protein was found to exist in these cells as a phosphoprotein, pp39Mos, and, when immunoprecipitated with an antiserum specific for the Mos N-terminus [anti-Mos(6-24)], had autophosphorylating kinase activity. We found that anti-Mos(6-24) also detected non-Mos protein kinase activity and non-Mos phosphoproteins in addition to p39Mos. We present evidence, on both the RNA and protein levels, that non-transformed mouse 3T3 cells do not express endogenous Mos.


Subject(s)
Genes, mos , Phosphoproteins/genetics , Proto-Oncogene Proteins c-mos/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle/drug effects , Codon , Cysteine/metabolism , Hydroxyurea/pharmacology , Kinetics , Methionine/metabolism , Mice , Molecular Sequence Data , Moloney murine sarcoma virus/genetics , Phosphoproteins/analysis , Phosphoproteins/biosynthesis , Phosphorylation , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-mos/analysis , Proto-Oncogene Proteins c-mos/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL