Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Bull Cancer ; 88(6): 545-8, 2001 Jun.
Article in French | MEDLINE | ID: mdl-11459700

ABSTRACT

The transcription factor MyoD, member of the myogenic regulators family, induces differentiation in precursor cells by its ability to arrest cell proliferation and to activate muscle specific genes. MyoD plays a key role in the antagonism between proliferation and differentiation. The withdrawal from the cell cycle and the activation of muscle differentiation are related to the level of MyoD protein. The cyclin E-cdk2 complex, one of the key regulators of the G1/S transition is directly implicated in the degradation of MyoD by the ubiquitin-proteasome pathway, leading the myoblasts to proliferate. The display of this control in normal myoblasts suggests that its deficiency in the muscle stem cells could lead to the formation of rhabdomyosarcomas which have lost both the control of cell proliferation and the transcriptional activity of MyoD.


Subject(s)
MyoD Protein/physiology , Rhabdomyosarcoma/etiology , Cell Cycle Proteins/physiology , Child , Humans , Muscle, Skeletal/embryology , Phosphorylation , Rhabdomyosarcoma/congenital
3.
Exp Cell Res ; 259(1): 300-7, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10942602

ABSTRACT

Proliferating myoblasts already express MyoD before the induction of differentiation. Overexpression of MyoD in normal and transformed cell lines was shown to block cells from entering S phase, suggesting that the MyoD growth suppressive effect must be tightly controlled in growing myoblasts. Here we show that during G1 phase, but not in G2, MyoD abundance is down-regulated by the ubiquitin-proteasome pathway through phosphorylation of serine 200. Roscovitine, a specific inhibitor of cyclin-Cdk2 complexes, prevents both phosphorylation and degradation of MyoD in G1. Inhibition of the ubiquitin-dependent proteasome pathway by MG132 results in stabilization of MyoD-wt, with little effect on a MyoD mutant where serine 200 is replaced by an alanine. Our results show that MyoD Ser200 is the substrate for phosphorylation by cyclin E-Cdk2 stimulating its degradation by the ubiquitin-proteasome system which controls MyoD levels in G1. Phosphorylation/degradation of MyoD at the end of G1 thus represents the regulatory checkpoint in growing myoblasts allowing progression into S phase in a manner similar to the recently examplified cdk2-phosphorylation/degradation of p27(Kip1).


Subject(s)
CDC2-CDC28 Kinases , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , G1 Phase/physiology , Muscle Fibers, Skeletal/cytology , MyoD Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cyclin-Dependent Kinase 2 , Cysteine Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , G1 Phase/drug effects , Mice , Multienzyme Complexes/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/cytology , Phosphorylation , Proteasome Endopeptidase Complex , Purines/pharmacology , Roscovitine , S Phase/drug effects , S Phase/physiology , Serine , Substrate Specificity , Ubiquitins/metabolism
4.
J Biol Chem ; 275(25): 18767-76, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10764802

ABSTRACT

Recent data have demonstrated the role of Cdk1- and Cdk2-dependent phosphorylation of MyoD(Ser200) in the regulation of MyoD activity and protein turnover. In the present study, we show that in presence of p57(Kip2), MyoD(Ala200), a MyoD mutant that cannot be phosphorylated by cyclin-Cdk complexes, displayed activity 2-5-fold higher than of MyoD(Ala200) alone in transactivation of muscle-specific genes myosin heavy chain, creatine kinase, and myosin light chain 1. Furthermore, p57(Kip2) increases the levels of MyoD(Ala200) in cotransfected cells. This result implies that p57(Kip2) may regulate MyoD through a process distinct from its function as a cyclin-dependent kinase inhibitors. We report that overexpression of p57(Kip2) increased the half-life of MyoD(Ala200). This increased half-life of MyoD involves a physical interaction between MyoD and p57(Kip2) but not with p16(Ink4a), as shown by cross-immunoprecipitation not only on overexpressed proteins from transfected cells, but also on endogenous MyoD and p57(Kip2) from C2C12 myogenic cells. Mutational and functional analyses of the two proteins show that the NH(2) domain of p57(Kip2) associates with basic region in the basic helix-loop-helix domain of MyoD. Competition/association assays and site-directed mutagenesis of the NH(2) terminus of p57(Kip2) identified the intermediate alpha-helix domain, located between the Cdk and the cyclin binding sites, as essential for MyoD interaction. These data show that the alpha-helix domain of p57(Kip2), which is conserved in the Cip/Kip proteins, is implicated in protein-protein interaction and confers a specific regulatory mechanism, outside of their Cdk-inhibitory activity, by which the p57(Kip2) family members positively act on myogenic differentiation.


Subject(s)
Fungal Proteins/metabolism , Microtubule-Associated Proteins/metabolism , MyoD Protein/metabolism , Proto-Oncogene Proteins , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , Cell Line , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , DNA Primers , Fungal Proteins/genetics , Humans , Mice , Mice, Inbred C3H , Microtubule-Associated Proteins/genetics , Molecular Motor Proteins , Molecular Sequence Data , Phosphorylation , Protein Binding , Sequence Homology, Amino Acid , Transcriptional Activation
5.
Oncogene ; 19(9): 1147-52, 2000 Feb 24.
Article in English | MEDLINE | ID: mdl-10713702

ABSTRACT

Previous studies have led to the proposal that a single molecule of Cki can associate with the cyclin/Cdk complex to repress its activity. On the other hand, multiple inhibitor molecules are required to inhibit Cdks. In the present work, by using differently tagged p57Kip2 proteins we demonstrate that p57Kip2 can bind to itself in vitro and in vivo. Mutational deletion analysis showed that the NH2 terminal domain of p57Kip2 is necessary and sufficient to dimerization. Using an in vitro competition/association assay, we demonstrate that cyclin D1 alone, Cdk4 alone and/or cyclin D1/Cdk4 complexes do not compete for the p57Kip2 homodimers formation. However, a mutation in the alpha-helix domain of p57Kip2 (R33L) strongly reduced homodimer formation but did not modify interaction with cyclin D1-Cdk4 complexes. Also, increasing amounts of p57Kip2 lead in vivo to a significant augmentation in the level of p57Kip2 homodimerization associated with cyclin D1-Cdk4 complexes and to a marked inhibition of the cyclin D1-Cdk4 kinase activity. Altogether, these data suggest a model whereby p57Kip2 associates with itself by using the NH2 domain to form a homodimeric species which interacts with and inhibits the cyclin D1-Cdk4 complexes.


Subject(s)
Cyclin D1/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins , Animals , Biopolymers/metabolism , Cell Line , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p57 , Cyclin-Dependent Kinases/metabolism , Dimerization , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Fibroblasts/metabolism , Mice , Nuclear Proteins/physiology , Peptide Fragments/physiology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Transfection
6.
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
7.
Mol Cell Biol ; 19(11): 7621-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10523650

ABSTRACT

We show that expression of p57(Kip2), a potent tight-binding inhibitor of several G(1) cyclin-cyclin-dependent kinase (Cdk) complexes, increases markedly during C2C12 myoblast differentiation. We examined the effect of p57(Kip2) on the activity of the transcription factor MyoD. In transient transfection assays, transcriptional transactivation of the mouse muscle creatine kinase promoter by MyoD was enhanced by the Cdk inhibitors. In addition, p57(Kip2), p21(Cip1), and p27(Kip1) but not p16(Ink4a) induced an increased level of MyoD protein, and we show that MyoD, an unstable nuclear protein, was stabilized by p57(Kip2). Forced expression of p57(Kip2) correlated with hypophosphorylation of MyoD in C2C12 myoblasts. A dominant-negative Cdk2 mutant arrested cells at the G(1) phase transition and induced hypophosphorylation of MyoD. Furthermore, phosphorylation of MyoD by purified cyclin E-Cdk2 complexes was inhibited by p57(Kip2). In addition, the NH2 domain of p57(Kip2) necessary for inhibition of cyclin E-Cdk2 activity was sufficient to inhibit MyoD phosphorylation and to stabilize it, leading to its accumulation in proliferative myoblasts. Taken together, our data suggest that repression of cyclin E-Cdk2-mediated phosphorylation of MyoD by p57(Kip2) could play an important role in the accumulation of MyoD at the onset of myoblast differentiation.


Subject(s)
CDC2-CDC28 Kinases , Cyclin E/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Fungal Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Saccharomyces cerevisiae Proteins , Animals , Cell Differentiation , Creatine Kinase/genetics , Cyclin-Dependent Kinase 2 , Fungal Proteins/genetics , Mice , Microtubule-Associated Proteins/genetics , Molecular Motor Proteins , Muscle, Skeletal/metabolism , Peptide Fragments/metabolism , Phosphorylation , Recombinant Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcriptional Activation
8.
J Cell Sci ; 110 ( Pt 9): 1083-9, 1997 May.
Article in English | MEDLINE | ID: mdl-9175704

ABSTRACT

Urokinase can form a tripartite complex binding urokinase receptor (uPAR) and plasminogen activator inhibitor type-1 (PAI-1), a component of the extracellular matrix (ECM). The components of the tripartite complex are modulated throughout the in vitro myogenic differentiation process. A series of experiments aimed at elucidating the role of the urokinase tripartite complex in the fusion of human myogenic cells were performed in vitro. Myogenic cell fusion was associated with increased cell-associated urokinase-type plasminogen activator (uPA) activity, cell-associated uPAR, and uPAR occupancy. Incubation of cultures with either uPA anticatalytic antibodies, or the amino-terminal fragment of uPA (ATF), which inhibits competitively uPA binding to its receptor, or anti-PAI-1 antibodies, which inhibit uPA binding to PAI-1, resulted in a 30 to 47% decrease in fusion. Incubation of cultures with the plasmin inhibitor aprotinin did not affect fusion. Decreased fusion rates induced by interfering with uPAR/uPA/PAI-1 interactions were not associated with significant changes in mRNA levels of both the myogenic regulatory factor myogenin and its inhibitor of DNA binding, Id. Incubation of cultures with purified uPA resulted in a decrease in fusion, likely due to a competitive inhibition of PAI-1 binding of endogenous uPA. We conclude that muscle cell fusion largely depends on interactions between the members of the urokinase complex (uPAR/uPA/PAI-1), but does not require proteolytic activation of plasmin. Since the intrinsic muscle cell differentiation program appears poorly affected by the state of integrity of the urokinase complex, and since cell migration is a prerequisite for muscle cell fusion in vitro, it is likely that the urokinase system is instrumental in fusion through its connection with the cell migration process. Our results suggest that the urokinase tripartite complex may be involved in cell migration in a non conventional way, playing the role of an adhesion system bridging cell membrane to ECM.


Subject(s)
Cell Fusion/physiology , Muscle, Skeletal/cytology , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Antibodies/pharmacology , Aprotinin/pharmacology , Blotting, Northern , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Fibrinolysin/antagonists & inhibitors , Humans , Immunoglobulin G/pharmacology , Multienzyme Complexes/physiology , Muscle, Skeletal/chemistry , Muscle, Skeletal/enzymology , Myogenin/genetics , Peptides/pharmacology , Plasminogen Activator Inhibitor 1/immunology , RNA, Messenger/analysis , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator , Serine Proteinase Inhibitors/pharmacology , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/pharmacology
9.
Mol Cell Biol ; 17(2): 584-93, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9001211

ABSTRACT

The activities of myogenic basic helix-loop-helix (bHLH) factors are regulated by a number of different positive and negative signals. Extensive information has been published about the molecular mechanisms that interfere with the process of myogenic differentiation, but little is known about the positive signals. We previously showed that overexpression of rat Mos in C2C12 myoblasts increased the expression of myogenic markers whereas repression of Mos products by antisense RNAs inhibited myogenic differentiation. In the present work, our results show that the rat mos proto-oncogene activates transcriptional activity of MyoD protein. In transient transfection assays, Mos promotes transcriptional transactivation by MyoD of the muscle creatine kinase enhancer and/or a reporter gene linked to MyoD-DNA binding sites. Physical interaction between Mos and MyoD, but not with E12, is demonstrated in vivo by using the two-hybrid approach with C3H10T1/2 cells and in vitro by using the glutathione S-transferase (GST) pull-down assays. Unphosphorylated MyoD from myogenic cell lysates and/or bacterially expressed MyoD physically interacts with Mos. This interaction occurs via the helix 2 region of MyoD and a highly conserved region in Mos proteins with 40% similarity to the helix 2 domain of the E-protein class of bHLH factors. Phosphorylation of MyoD by activated GST-Mos protein inhibits the DNA-binding activity of MyoD homodimers and promotes MyoD-E12 heterodimer formation. These data support a novel function for Mos as a mediator (coregulator) of muscle-specific gene(s) expression.


Subject(s)
DNA-Binding Proteins/metabolism , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Proto-Oncogene Proteins c-mos/physiology , Transcription Factors , Transcriptional Activation/physiology , Amino Acid Sequence , Animals , Cell Differentiation , Cell Line , Creatine Kinase/genetics , Dimerization , Enhancer Elements, Genetic/genetics , Helix-Loop-Helix Motifs , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-mos/metabolism , Rats , Recombinant Fusion Proteins , Sequence Homology, Amino Acid , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transfection
10.
Cell Biol Toxicol ; 12(3): 177-85, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8817063

ABSTRACT

We analysed the signaling pathways involved in myogenic differentiation of primary cultures of rat satellite cells using substances targeting the protein kinase C (PKC) and the cAMP protein kinase (PKA) pathways. We have previously shown that iso-H7, which putatively inhibits both PKC and PKA, strongly stimulates satellite cell differentiation, as well as the PKA inhibitor HA1004. In the study reported here, the effects of iso-H7 on satellite cell differentiation were compared to those observed in the presence of agents which reduce PKC activity. It was shown that treatments with the highly specific PKC inhibitor GF109203X or with 12-O-tetradecanoylphorbol 13-acetate (TPA) which induced a partial PKC downregulation, did not significantly alter myogenic differentiation. Northern blot analyses showed that iso-H7 activated the expression of myogenin but not that of MyoD mRNA. Concurrently, iso-H7 increased myosin light-chain mRNA expression. In contrast, TPA had no effect on these syntheses. Taken together, these results showed that iso-H7 did not act intracellularly as a PKC inhibitor but rather as a PKA inhibitor as previously suggested. Our results are compatible with the hypothesis that a reduction in PKA activity controls satellite cell myogenesis through an increased myogenin mRNA expression.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Muscle, Skeletal/drug effects , Myogenin/metabolism , Protein Kinase C/metabolism , Sulfonamides , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Blotting, Northern , Cell Differentiation/drug effects , Creatine Kinase/metabolism , Indoles/pharmacology , Isoquinolines/pharmacology , Maleimides/pharmacology , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Myosin Light Chains/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tetradecanoylphorbol Acetate/pharmacology
11.
J Biol Chem ; 271(9): 5258-64, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8617811

ABSTRACT

Terminal differentiation of muscle cells results in opposite effects on gene promoters: muscle-specific promoters, which are repressed during active proliferation of myoblasts, are turned on, whereas at least some proliferation-associated promoters, such as c-fos, which are active during cell division, are turned off. MyoD and myogenin, transcription factors from the basic-helix-loop-helix (bHLH) family, are involved in both processes, up-regulating muscle genes and down-regulating c-fos. On the other hand, the serum response factor (SRF) is involved in the activation of muscle-specific genes, such as c-fos, as well as in the up-regulation of a subset of genes that are responsive to mitogens. Upon terminal differentiation, the activity of these various transcription factors could be modulated by the formation of distinct protein-protein complexes. Here, we have investigated the hypothesis that the function of SRF and/or MyoD and myogenin could be modulated by a physical association between these transcription factors. We show that myogenin from differentiating myoblasts specifically binds to SRF. In vitro analysis, using the glutathione S-transferase pull-down assay, indicates that SRF-myogenin interactions occur only with myogenin-E12 heterodimers and not with isolated myogenin. A physical interaction between myogenin, E12, and SRF could also be demonstrated in vivo using a triple-hybrid approach in yeast. Glutathione S-transferase pull-down analysis of various mutants of the proteins demonstrated that the bHLH domain of myogenin and that of E12 were necessary and sufficient for the interaction to be observed. Specific binding to SRF was also seen with MyoD. In contrast, Id, a natural inhibitor of myogenic bHLH proteins, did not bind SRF in any of the situations tested. These data suggest that SRF, on one hand, and myogenic bHLH, on the other, could modulate each other's activity through the formation of a heterotrimeric complex.


Subject(s)
DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , Muscle, Skeletal/metabolism , Myogenin/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Base Sequence , Cell Differentiation , Cloning, Molecular , DNA Primers , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/isolation & purification , Glutathione Transferase/biosynthesis , Glutathione Transferase/isolation & purification , Mice , Molecular Sequence Data , Muscle, Skeletal/cytology , Mutagenesis , Myogenin/biosynthesis , Myogenin/isolation & purification , Nuclear Proteins/biosynthesis , Nuclear Proteins/isolation & purification , Plasmids , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Serum Response Factor , Transfection
12.
Cancer ; 75(7): 1694-9, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-8826929

ABSTRACT

BACKGROUND: Some genetic alterations have been shown to have prognostic implication for patients with neuroblastoma: MYCN oncogene amplification, deletion of the short arm of chromosome 1 and di- or tetraploidy. The goal of this study was to analyze these factors in children with neuroblastoma. METHODS: Twenty neuroblastoma samples were analyzed with morphologic cytogenetics, and each of them was compared with MYCN amplification status by Southern blot and fluorescent in situ hybridization (FISH) with a genomic probe. RESULTS: A complete karyotype was obtained for 14 children. A diploid or tetraploid mode and a 1p deletion were found in most children with advanced stages. MYCN amplification status was totally concordant with both methods in all patients, even in a case with low level amplification. A wide intercellular variation in the amplification level in each MYCN amplified sample was shown. CONCLUSION: The use of FISH to assess MYCN amplification rapidly in neuroblastoma is recommended. This method could be very useful in future therapeutic protocols in which treatment is based on MYCN status (and especially for infants and children with localized tumor).


Subject(s)
Chromosome Aberrations , Neuroblastoma/genetics , Adolescent , Child , Child, Preschool , Gene Amplification , Genes, myc , Humans , In Situ Hybridization, Fluorescence , Infant , Neuroblastoma/pathology
13.
Oncogene ; 10(2): 251-60, 1995 Jan 19.
Article in English | MEDLINE | ID: mdl-7838525

ABSTRACT

We have cloned and sequenced an almost complete c-DNA and the entire genomic sequence of rat the H19 gene, which is developmentally regulated in skeletal muscle. The data base comparison revealed a 92% homology with mouse gene H19. However the rat H19 ORFs do not display significant homology with the H19 ORFs from other species. In contrast to the mouse, the rat H19 mRNA is not easily detectable in fetal rat skeletal fibers. Its level increases after birth (up to 12 to 18 days) and remains stable thereafter. The pattern of H19 mRNA expression in rat muscle in vivo is very similar to the c-mos gene expression in this tissue, suggesting an interrelationship between H19 and c-mos products during muscle differentiation. Indeed, our results indicate that overexpression of c-mos protein in the muscle cell line C2C12 induces a concomitant increase of H19 mRNA expression. Furthermore, repression of c-mos protein expression by anti-sense RNAs extinguishes H19 mRNA expression and inhibits the differentiation process. These data suggest a relationship between c-mos and H19 expression and, in addition, the involvement of both gene products in the process of myogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Genes, mos , Muscle, Skeletal/embryology , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation/genetics , Cell Line , Mice , Molecular Sequence Data , Open Reading Frames , RNA, Antisense/pharmacology , RNA, Messenger/metabolism , Rats , Sequence Homology, Nucleic Acid
14.
Neuromuscul Disord ; 3(5-6): 379-83, 1993.
Article in English | MEDLINE | ID: mdl-8186679

ABSTRACT

Satellite cells are involved in physiological growth and post-traumatic regeneration of adult skeletal muscle fibres. In this study, it is shown that differentiation of primary cultures of rat satellite cells is increased by inhibitors of ser/thr protein kinases such as iso-H7, which both inhibit cAMP-dependent protein kinase (PKA) and protein kinase C (PKC) activities, and HA1004, a PKA inhibitor. These results, showing a preponderant effect of PKA inhibition on myogenesis in vitro, prompted the effects of iso-H7 on muscular regeneration in vivo to be tested. Preliminary results showed that regeneration of rat muscle EDL was improved by iso-H7 treatment.


Subject(s)
Isoquinolines/pharmacology , Muscles/physiology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sulfonamides , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Alkaloids/pharmacology , Animals , Cells, Cultured , Creatine Kinase/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Isoenzymes , Kinetics , Male , Muscles/cytology , Muscles/drug effects , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Wistar , Regeneration/drug effects , Staurosporine
15.
Oncogene ; 8(9): 2361-9, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8395677

ABSTRACT

We have used fractionation of subcellular components of the skeletal muscle followed by Western blot analyses to study the localization of the c-mos protein in adult rat muscle. We find that p43c-mos is predominantly located in the KCl supernatant fraction. We show that immunoprecipitates of p43c-mos phosphorylate in vitro two polypeptides of about 34 kDa and 80 kDa respectively. Muscle fractionation and immunodetection studies showed that the p34 protein associated with p43c-mos is the cdc2 protein. p43c-mos is coprecipitated with p34cdc2 when using either anti PSTAIR antibody, antibody directed against the conserved COOH terminal region of the p34cdc2 and by binding to beads that contain cross-linked p13suc1, a protein known to bind p34cdc2. Likewise p34cdc2 coprecipitated with p43c-mos when using anti mos antibody. However p43c-mos is not present in histone H1 kinase active p34cdc2 complex precipitated with anti p34cdc2 COOH-terminal peptide antibody. In adult muscle tissue tubulin is not complexed with p34cdc2 and p43c-mos as previously observed in c-mos and v-mos transformed cells. Gel filtration and crosslinking experiments show that a 170 kDa complex contains c-mos and p34cdc2 proteins. In addition during postnatal development of skeletal muscle we observe modifications in the migration pattern of p34cdc2 correlated with the accumulation of p43c-mos. Our findings raise the possibility of a p43c-mos-p34cdc2 complex could play a role in the differentiation process and maintenance of myotubes in Go.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins , Muscles/enzymology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-mos/metabolism , Schizosaccharomyces pombe Proteins , Animals , Cell Compartmentation , Cytosol/enzymology , Fungal Proteins/metabolism , Muscle Proteins/metabolism , Phosphoproteins/metabolism , Protamine Kinase/chemistry , Protein Binding , Rats
16.
Leukemia ; 6(10): 1011-9, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1328775

ABSTRACT

The changes occurring in the hematopoietic extracellular matrix in an experimental myeloproliferative syndrome were explored by comparing the glycosaminoglycan (GAG) composition of normal mouse spleens and spleens infected with myeloproliferative sarcoma virus (MPSV). Large quantities of hyaluronate and of sulfated GAGs accumulated in the extracellular matrix of infected spleens, as shown by histoimmunoassay and alcian blue staining, respectively. The splenic GAGs were either labeled with 35S-sulfate injected in vivo or unlabeled. The spleens were fractionated to separate hematopoietic cells from the stromal component containing extracellular matrix material and fibroblasts, and the GAGs were extracted from each fraction. Specific degradative treatments and electrophoresis indicated that sulfated GAGs were mostly chondroitin sulfate and heparan sulfate. Three hours after in vivo injection of 35S-sulfate, the amount of 35S-GAGs was increased approximately fivefold per mg stromal proteins. The bulk of these 35S-GAGs (70%) was recovered in the stromal fraction. The higher amount of sulfated GAGs in leukemic spleen was due both to the presence of more producer cells (infected fibroblasts and hematopoietic cells) and to a stimulation of GAG synthesis per cell, as evidenced 35S-labeling in in vitro experiments. Chondroitin sulfate was the main sulfated GAG present in the culture medium of both hematopoietic and fibroblastic cells and in the pericellular material released by trypsin from fibroblastic cells. High amounts of chondroitin sulfate, which has a possible role in the detachment of hematopoietic cells from the stromal cells, may favour the release of hematopoietic cells from the spleen into the peripheral blood. Heparan sulfate was produced by fibroblastic cells and it was principally present in their pericellular material. Considering the capacity of heparan sulfate to retain cytokines, as demonstrated by others in vitro, large amounts of heparan sulfate may result in the retention of large amounts of the cytokines, which production is enhanced in the infected spleen. This phenomenon may contribute to promote the hematopoietic stem cell proliferation characteristic of the MPSV-induced myeloproliferative disease.


Subject(s)
Extracellular Matrix/metabolism , Glycosaminoglycans/biosynthesis , Myeloproliferative Disorders/metabolism , Animals , DNA, Viral/analysis , Hematopoiesis , Hyaluronic Acid/metabolism , Mice , Mice, Inbred DBA , Proteins/metabolism , Proviruses/chemistry , Sarcoma Viruses, Murine , Sarcoma, Experimental/metabolism , Spleen/metabolism , Sulfates/metabolism
17.
Growth Factors ; 6(3): 209-18, 1992.
Article in English | MEDLINE | ID: mdl-1389227

ABSTRACT

We have isolated and sequenced a cDNA (mrfms) encoding rat c-fms gene (CSF-1 receptor) from proliferating L6 alpha 1 myoblasts. The predicted amino acid sequence was highly identical with the c-fms protein found in monocytes and macrophages (98, 76 and 84% identity from mouse, cat and human c-fms proteins, respectively). The mechanisms responsible for the regulation of mrfms gene expression during myogenesis were examined. Mrfms products were observed during proliferation of L6 alpha 1 myoblasts and were downregulated during differentiation. Run-on transcription assays demonstrated that the mrfms gene was transcriptionally active only in undifferentiated myoblasts. These findings suggested that mrfms levels in L6 alpha 1 myoblasts are controlled by transcriptional mechanisms. The half-life of mrfms transcripts was found to be at least 5 hr while inhibition of protein synthesis with cycloheximide (CHX) decreased this half-life to 30 min without changes in the rate of mrfms gene transcription. In addition oncogenic transformation of L6 alpha 1 myoblasts by the v-fms induced constitutive upregulation of mrfms mRNAs, and nuclear run-on assays demonstrated that mrfms transcription was not growth-factor dependent. Furthermore, these findings with others previously published indicate that mrfms gene products may play a role in the normal and neoplastic growth of muscular cells.


Subject(s)
Gene Expression Regulation/physiology , Muscles/metabolism , Receptor, Macrophage Colony-Stimulating Factor/physiology , Transcription, Genetic/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cloning, Molecular , Cycloheximide/pharmacology , DNA , Dactinomycin/pharmacology , Mice , Molecular Sequence Data , Muscles/cytology , RNA, Messenger/metabolism , Rats , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/genetics , Transformation, Genetic/physiology
18.
Biochem Biophys Res Commun ; 180(3): 1241-50, 1991 Nov 14.
Article in English | MEDLINE | ID: mdl-1953776

ABSTRACT

A rat adult skeletal muscle probe (Asm15) originated from a rhabdomyosarcoma was used to isolate the human homologous sequence from a placenta cDNA library. Among several positive clones the longest EcoRI-EcoRI insert (ASM1) obtained was 1875 bp long with 72% homology with rat Asm15 cDNA sequence. Important variations of ASM1 RNA level were observed in different adult skeletal muscles. Expression of a 29kD ASM1 protein was demonstrated in human adult skeletal muscle lysates using an antiserum (PB1579) raised against the C terminal region of the rat Asm15 protein. The human ASM gene was assigned by somatic cell analysis with human (ASM1) and rat (Asm15) probes to chromosome 11, and by in situ hybridization with the human probe to 11p15, a chromosome region involved in human embryonal rhabdomyosarcomas. Except for the presence of a HindII restriction site, the results obtained for the restriction map and the sequence of ASM1 cDNA (data not shown) exhibited extensive homology with the human H19 DNA sequence which have been mapped with a mouse probe also in 11p15. This suggests that ASM/Asm and H19 may represent the same sequence (in this hypothesis the presence of the supplementary HindII site in our ASM1 probe is explained by polymorphic variability). However it was reported that human and mouse H19 mRNA did not encode for a protein but acted as an RNA molecule whereas in our present study ASM protein was detected in human adult skeletal muscle. This could be explained by important regulation of ASM protein expression during development and cell differentiation. However we cannot exclude for the different species studied (mouse, rat, and man) the hypothesis that H19 and ASM/Asm mRNA may represent two distinct messengers from the same gene or even from duplicated genes.


Subject(s)
Chromosomes, Human, Pair 11 , Muscles/physiology , Adult , Animals , Blotting, Southern , Cell Line , Chromosome Banding , Chromosome Mapping , DNA/genetics , DNA/isolation & purification , DNA Probes , Female , Humans , Mice , Placenta/physiology , Pregnancy , Rats , Restriction Mapping , Rhabdomyosarcoma/genetics
19.
Oncogene ; 6(9): 1617-22, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1833718

ABSTRACT

Previously we reported that c-mos proto-oncogene RNA was developmentally up-regulated during post-natal maturation of the rat skeletal muscle. Using two different site-directed affinity-purified antipeptide antibodies we can observe that c-mos product (p43 c-mos) accumulates increasingly during post-natal development of the skeletal muscle and exhibits protein kinase activity. We find that in adult rat p43 c-mos is 10-fold higher in skeletal muscle than in ovaries, and 20- to 40-fold higher than in heart, lung, testis and liver, and may represent about 0.005% of the total soluble proteins. In addition adult skeletal muscle from Xenopus, mouse and man was found to contain p43 c-mos. These data argue in favour of a novel muscle-specific function of c-mos.


Subject(s)
Muscle Development , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Aging , Animals , Cloning, Molecular , Escherichia coli/genetics , Female , Humans , Male , Muscles/enzymology , Ovary/enzymology , Ovary/growth & development , Protein Biosynthesis , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mos , Proto-Oncogenes , Rats , Recombinant Fusion Proteins/metabolism , Species Specificity , Transcription, Genetic , Xenopus
20.
Oncogene ; 5(8): 1149-57, 1990 Aug.
Article in English | MEDLINE | ID: mdl-1697408

ABSTRACT

We have isolated and sequenced a cDNA clone, homologous to the rat c-mos gene, from a cDNA library of rat skeletal muscles. The 3220 nucleotide cDNA clone codes for a protein of 339 amino acids (37.4 kDa). Both the nucleotide sequence and the deduced amino acid sequence show 60-90% overall homology to Xenopus, chicken, mouse and human mos. By Northern blot analysis, we detected two c-mos transcripts, one major of about 3.6 Kb long, and one minor of about 1.7 Kb long. These are differently regulated during the development of cardiac and skeletal muscles. By Western blot with two antibodies directed against two different portions of the mos protein, we observed in rat muscle two polypeptides of 43 kDa, and 75 kDa respectively.


Subject(s)
Cloning, Molecular , DNA/analysis , Gene Expression Regulation , Muscles/analysis , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Molecular Sequence Data , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-mos , RNA/analysis , Rats , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...