Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Prog Urol ; 21(12): 859-65, 2011 Nov.
Article in French | MEDLINE | ID: mdl-22035912

ABSTRACT

PURPOSE: Cell therapy for urinary incontinence management has been experienced in animals with encouraging results, but studies in human beings are lacking. Our primary objective was to assess the safety of intrasphincteric injections of autologous muscular cells in patients with postprostatectomy incontinence (PPI). Secondary objectives focused on complications efficacy. METHODS: We conducted an open, prospective study in a single center on 12 patients presenting PPI. Patients underwent intrasphincteric injections of autologous muscular cells isolated from a biopsy of deltoid muscle. The primary endpoint was the Q(max) variation at the three month visit in order to assess potential bladder outlet obstruction. Secondary endpoints assessed side effects and efficacy parameters based on symptoms, quality of life score, voiding diary, pad-test, and urethral pressure profile at one, two, three, six and 12 months after injection. RESULTS: No immediate complication occurred and no significant variation was noted on Q(max). The only side effects possibly product-related were three cases of urinary tract infection treated by antibiotics. An acceptable safety and tolerability of the procedure whatever the injected dose of muscular cells was demonstrated. Results on efficacy after one year were heterogeneous, with 4/12 patients describing reduced urine leakage episodes, 1/12 patient presenting increased maximal closure pressure, and 8/12 patients showing improvement on pad-test. CONCLUSIONS: Cell therapy consisting of intrasphincteric injections of autologous muscular cells in patients with PPI was a feasible and safe procedure. The results point out that some subjects may positively respond to this procedure, but clinical efficacy remains to be confirmed.


Subject(s)
Muscle Cells/transplantation , Prostatectomy/adverse effects , Urethra , Urinary Incontinence, Stress/etiology , Urinary Incontinence, Stress/surgery , Aged , Deltoid Muscle , Feasibility Studies , Follow-Up Studies , Humans , Injections, Intralesional , Male , Middle Aged , Prospective Studies , Quality of Life , Risk Assessment , Transplantation, Autologous , Treatment Outcome
2.
Sci Rep ; 10(1): 9139, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499563

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is a lethal muscle disorder, caused by mutations in the DMD gene and affects approximately 1:5000-6000 male births. In this report, we identified dysregulation of members of the Dlk1-Dio3 miRNA cluster in muscle biopsies of the GRMD dog model. Of these, we selected miR-379 for a detailed investigation because its expression is high in the muscle, and is known to be responsive to glucocorticoid, a class of anti-inflammatory drugs commonly used in DMD patients. Bioinformatics analysis predicts that miR-379 targets EIF4G2, a translational factor, which is involved in the control of mitochondrial metabolic maturation. We confirmed in myoblasts that EIF4G2 is a direct target of miR-379, and identified the DAPIT mitochondrial protein as a translational target of EIF4G2. Knocking down DAPIT in skeletal myotubes resulted in reduced ATP synthesis and myogenic differentiation. We also demonstrated that this pathway is GC-responsive since treating mice with dexamethasone resulted in reduced muscle expression of miR-379 and increased expression of EIF4G2 and DAPIT. Furthermore, miR-379 seric level, which is also elevated in the plasma of DMD patients in comparison with age-matched controls, is reduced by GC treatment. Thus, this newly identified pathway may link GC treatment to a mitochondrial response in DMD.


Subject(s)
Glucocorticoids/therapeutic use , MicroRNAs/metabolism , Mitochondria/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Dexamethasone/pharmacology , Disease Models, Animal , Dogs , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Gene Expression Regulation/drug effects , Humans , Mice , MicroRNAs/chemistry , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myoblasts, Skeletal/metabolism , RNA Interference , RNA, Small Interfering/metabolism
3.
Gene Ther ; 16(6): 815-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19282846

ABSTRACT

Transplantation of muscle precursor cells (MPCs) is a promising approach for the treatment of muscular dystrophies. However, preclinical and clinical results have shown that the technology is not yet efficient enough for most therapeutic applications. Among the problems that remain unsolved are low cellular survival, poor proliferation and lack of migration of the transplanted cells. One major technical hurdle for the optimization of transplantation protocols is how to follow precisely the fate of the cells after transplantation. In this study, we examined the use of a secreted form of the mouse alkaline phosphatase (mSeAP) enzyme as the reporter system transduced into MPCs using a retroviral vector. We show that circulating mSeAP could be detected in the serum of the transplanted mice at different time points after MPC transplantation. We also found that the level of circulating mSeAP is highly correlated with the number of transplanted cells and that mSeAP is an excellent histological marker. Further, studying the levels of circulating mSeAP compared with the number of muscle fibers positive to mSeAP and to dystrophin, enabled detailed analyses of bottleneck steps for successful transplantation. Taken together, our results show that mSeAP is an excellent quantitative 'real-time' reporter gene for cell therapy preclinical studies.


Subject(s)
Alkaline Phosphatase/metabolism , Alkaline Phosphatase/pharmacokinetics , Genes, Reporter , Myoblasts/transplantation , Alkaline Phosphatase/genetics , Animals , Cell Survival , Cells, Cultured , Dystrophin/metabolism , Half-Life , Hindlimb , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal , Myoblasts/metabolism , Staining and Labeling , Transduction, Genetic , Transgenes
4.
J Cell Biol ; 140(1): 111-8, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9425159

ABSTRACT

Myf5 is the earliest-known muscle-specific factor to be expressed in vivo and its expression is associated with determination of the myoblast lineage. In C2 cells, we show by immunocytolocalization that Myf5 disappears rapidly from cells in which the differentiation program has been initiated. In proliferating myoblasts, the levels of Myf5 and MyoD detected from cell to cell are very heterogeneous. We find that some of the heterogeneity of Myf5 expression arises from a posttranscriptional regulation of Myf5 by the cell cycle. Immunoblotting of extracts from synchronized cultures reveals that Myf5 undergoes periodic fluctuations during the cell cycle and is absent from cells blocked early in mitosis by use of nocodazole. The disappearance of Myf5 from mitotic cells involves proteolytic degradation of a phosphorylated form of Myf5 specific to this phase of the cell cycle. In contrast, MyoD levels are not depleted in mitotic C2 cells. The mitotic destruction of Myf5 is the first example of a transcription factor showing cell cycle-regulated degradation. These results may be significant in view of the possible role of Myf5 in maintaining the determination of proliferating cells and in timing the onset of differentiation.


Subject(s)
Cell Cycle/physiology , DNA-Binding Proteins , Gene Expression Regulation , Muscle Proteins/biosynthesis , Muscle, Skeletal/cytology , Trans-Activators , Animals , Cell Differentiation , Cell Division , Cell Line , Fluorescent Antibody Technique, Indirect , Mice , Mitosis , Muscle, Skeletal/metabolism , Myogenic Regulatory Factor 5 , Transcription Factors/biosynthesis
5.
Mol Cell Biol ; 20(23): 8923-32, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11073992

ABSTRACT

Transcription factors Myf5 and MyoD play critical roles in controlling myoblast identity and differentiation. In the myogenic cell line C2, we have found that Myf5 expression, unlike that of MyoD, is restricted to cycling cells and regulated by proteolysis at mitosis. In the present study, we have examined Myf5 proteolysis through stable transfection of myogenically convertible U20S cells with Myf5 derivatives under the control of a tetracycline-sensitive promoter. A motif within the basic helix-loop-helix domain of Myf5 (R93 to Q101) resembles the "destruction box" characteristic of substrates of mitotic proteolysis and thought to be recognized by the anaphase-promoting complex or cyclosome (APC). Mutation of this motif in Myf5 stabilizes the protein at mitosis but does not affect its constitutive turnover. Conversely, mutation of a serine residue (S158) stabilizes Myf5 in nonsynchronized cultures but not at mitosis. Thus, at least two proteolytic pathways control Myf5 levels in cycling cells. The mitotic proteolysis of Myf5 is unlike that which has been described for other destruction box-dependent substrates: down-regulation of Myf5 at mitosis appears to precede that of known targets of the APC and is not affected by a dominant-negative version of the ubiquitin carrier protein UbcH10, implicated in the APC-mediated pathway. Finally, we find that induction of Myf5 perturbs the passage of cells through mitosis, suggesting that regulation of Myf5 levels at mitosis may influence cell cycle progression of Myf5-expressing muscle precursor cells.


Subject(s)
DNA-Binding Proteins , Helix-Loop-Helix Motifs , Mitosis/physiology , Muscle Proteins/metabolism , Muscles/cytology , Myogenic Regulatory Factors/metabolism , Trans-Activators , Ubiquitin-Protein Ligase Complexes , Anaphase-Promoting Complex-Cyclosome , Cell Differentiation , Ligases , Myogenic Regulatory Factor 5 , Stem Cells , Ubiquitin-Protein Ligases
6.
Mol Cell Biol ; 20(22): 8560-70, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11046151

ABSTRACT

The BCL6 proto-oncogene, frequently alterated in non-Hodgkin lymphoma, encodes a POZ/zinc finger protein that localizes into discrete nuclear subdomains. Upon prolonged BCL6 overexpression in cells bearing an inducible BCL6 allele (UTA-L cells), these subdomains apparently coincide with sites of DNA synthesis. Here, we explore the relationship between BCL6 and replication by both electron and confocal laser scanning microscopy. First, by electron microscope analyses, we found that endogenous BCL6 is associated with replication foci. Moreover, we show that a relatively low expression level of BCL6 reached after a brief induction in UTA-L cells is sufficient to observe its targeting to mid, late, and at least certain early replication foci visualized by a pulse-labeling with bromodeoxyuridine (BrdU). In addition, when UTA-L cells are simultaneously induced for BCL6 expression and exposed to BrdU for a few hours just after the release from a block in mitosis, a nuclear diffuse BCL6 staining indicates cells in G(1), while cells in S show a more punctate nuclear BCL6 distribution associated with replication foci. Finally, ultrastructural analyses in UTA-L cells exposed to BrdU for various times reveal that replication progresses just around, but not within, BCL6 subdomains. Thus, nascent DNA is localized near, but not colocalized with, BCL6 subdomains, suggesting that they play an architectural role influencing positioning and/or assembly of replication foci. Together with its previously function as transcription repressor recruiting a histone deacetylase complex, BCL6 may therefore contribute to link nuclear organization, replication, and chromatin-mediated regulation.


Subject(s)
Cell Nucleus Structures/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Cell Line , DNA/metabolism , DNA/ultrastructure , DNA-Binding Proteins/genetics , G1 Phase , Humans , Microscopy, Confocal , Microscopy, Electron , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-6 , S Phase , Transcription Factors/genetics
7.
Mol Cell Biol ; 19(6): 4047-55, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330145

ABSTRACT

Calpain 3 is a nonlysosomal cysteine protease whose biological functions remain unknown. We previously demonstrated that this protease is altered in limb girdle muscular dystrophy type 2A patients. Preliminary observations suggested that its gene is subjected to alternative splicing. In this paper, we characterize transcriptional and posttranscriptional events leading to alterations involving the NS, IS1, and IS2 regions and/or the calcium binding domains of the mouse calpain 3 gene (capn3). These events can be divided into three groups: (i) splicing of exons that preserve the translation frame, (ii) inclusion of two distinct intronic sequences between exons 16 and 17 that disrupt the frame and would lead, if translated, to a truncated protein lacking domain IV, and (iii) use of an alternative first exon specific to lens tissue. In addition, expression of these isoforms seems to be regulated. Investigation of the proteolytic activities and titin binding abilities of the translation products of some of these isoforms clearly indicated that removal of these different protein segments affects differentially the biochemical properties examined. In particular, removal of exon 6 impaired the autolytic but not fodrinolytic activity and loss of exon 16 led to an increased titin binding and a loss of fodrinolytic activity. These results are likely to impact our understanding of the pathophysiology of calpainopathies and the development of therapeutic strategies.


Subject(s)
Calpain/genetics , Calpain/metabolism , Isoenzymes , RNA Processing, Post-Transcriptional , Transcription, Genetic , Alternative Splicing , Animals , Brain/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Cloning, Molecular , Connectin , DNA Primers , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/metabolism , Humans , In Situ Hybridization , Introns , Lens, Crystalline/anatomy & histology , Lens, Crystalline/metabolism , Mice , Mice, Inbred BALB C , Microfilament Proteins/metabolism , Models, Genetic , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Smooth/metabolism , Myocardium/metabolism , Peptide Fragments/metabolism , Protein Kinases/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Distribution
8.
Cell Mol Biol (Noisy-le-grand) ; 53(2): 44-50, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17531139

ABSTRACT

Counting cells in culture is a common task in biotechnology research and production. This process should be automated to provide fast and objective quantification. Flow cytometry is adapted to count cells in suspension. However, the morphological information and the spatial organisation of adherent cells are lost when cells are removed from culture. This paper proposes a methodology based on image analysis to quantify stained nuclei in culture. The protocol is composed of several steps: cell staining, automatic microscopy imaging, segmentation by an automatic algorithm including a classification approach, and computation of quantitative data that characterizes the growth of cells. An evaluation shows that the automatic process of counting provides results similar to human manual counting. The major interests of the proposed approach are the fully automated processing and preservation of cell shapes and positions in culture. More than two thousand culture conditions have been measured by this tool for various applications including optimization of cell culture media, improvement of the culture processes and measurement of drug toxicity.


Subject(s)
Cell Count/methods , Image Processing, Computer-Assisted/methods , Algorithms , Cells, Cultured , Staining and Labeling
9.
Oncogene ; 12(2): 411-21, 1996 Jan 18.
Article in English | MEDLINE | ID: mdl-8570219

ABSTRACT

In C2 muscle cells, retinoic acid (RA) induces growth arrest associated with terminal differentiation. These RA actions are presumed to be mediated through nuclear receptors (RARs and RXRs) that belong to the superfamily of ligand-dependent transcription factors. In this study, we have characterized a myogenic C2 subclone, that unlike parental cells, is resistant to growth inhibition and differentiation by RA. Examination of these RA-sensitive and resistant C2 cells for the expression of retinoid acid receptors revealed a lack of RXR alpha expression at the myoblast stage in resistant C2 cells. To determine the functions of RXR alpha, we introduced an RXR alpha expression vector into RA-resistant C2 cells by transient or stable transfections. Our results show that RXR alpha restores the response to RA in this subclone with respect to AP1 inhibition and growth arrest. These observations indicate that RXR alpha plays a crucial role in mediating RA induced growth arrest of C2 myogenic cells.


Subject(s)
Muscles/drug effects , Receptors, Retinoic Acid/physiology , Transcription Factors/physiology , Tretinoin/pharmacology , Cell Differentiation , Cell Division/drug effects , Cells, Cultured , Muscles/cytology , Retinoid X Receptors , Transcription Factor AP-1/antagonists & inhibitors
10.
Oncogene ; 18(36): 5063-75, 1999 Sep 09.
Article in English | MEDLINE | ID: mdl-10490843

ABSTRACT

One of the most frequent genetic abnormalities associated with non Hodgkin lymphoma is the structural alteration of the 5' non coding/regulatory region of the BCL6 (LAZ3) protooncogene. BCL6 encodes a POZ/Zn finger protein, a structure similar to that of many Drosophila developmental regulators and to another protein involved in a human hematopoietic malignancy, PLZF. BCL6 is a sequence specific transcriptional repressor controlling germinal center formation and T cell dependent immune response. Although the expression of BCL6 negatively correlates with cellular proliferation in different cell types, the influence of BCL6 on cell growth and survival is currently unknown so that the way its deregulation may contribute to cancer remains elusive. To directly address this issue, we used a tetracycline-regulated system in human U2OS osteosarcoma cells and thus found that BCL6 mediates growth suppression associated with impaired S phase progression and apoptosis. Interestingly, overexpressed BCL6 can colocalize with sites of ongoing DNA synthesis, suggesting that it may directly interfere with S phase initiation and/or progression. In contrast, the isolated Zn finger region of BCL6, which binds BCL6 target sequence but lacks transcriptional repression activity, slows, but does not suppress, U2OS cell growth, is less efficient at delaying S phase progression, and does not trigger apoptosis. Thus, for a large part, the effects of BCL6 overexpression on cell growth and survival depend on its ability to engage protein/protein interactions with itself and/or its transcriptional corepressors. That BCL6 restricts cell growth suggests that its deregulation upon structural alterations may alleviate negative controls on the cell cycle and cell survival.


Subject(s)
Apoptosis/physiology , DNA Replication/physiology , DNA-Binding Proteins/physiology , Proto-Oncogene Proteins/physiology , S Phase/physiology , Transcription Factors/physiology , DNA Replication/genetics , DNA-Binding Proteins/genetics , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-6 , Transcription Factors/genetics , Tumor Cells, Cultured
11.
J Mol Biol ; 213(4): 677-86, 1990 Jun 20.
Article in English | MEDLINE | ID: mdl-2162966

ABSTRACT

The influence of different CC Ar GG boxes derived from either muscle-specific or serum-responsive genes, on the specificity of different promoters has been investigated. Inserted upstream from an 85 base-pair long minimal promoter of the human cardiac alpha-actin gene, a single copy of both the cognate CC Ar GG element (HCA1) and the c-fos gene serum response element (SRE) stimulate transcription four- to fivefold more efficiently in C2 myogenic cells than in L fibroblastic cells, SRE being two- to threefold more active than HCA1. Inserted upstream from the ubiquitous Herpes simplex thymidine kinase (HSV-tk) promoter, multimerized CC Ar GG boxes behave as strong muscle-specific activating elements, about 20-fold more active in myogenic C2 cells than in L fibroblasts and hepatoma HepG2 cells. They also confer serum responsiveness on the HSV-tk promoter. Efficiency of HCA1 and SRE tetramers in conferring both muscle specificity and serum responsiveness is roughly similar. It appears, therefore, that regardless of their origin (either muscle-specific or serum-responsive genes) CC Ar GG boxes behave by themselves as both muscle-specific activating and serum-responsive elements.


Subject(s)
Actins/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription, Genetic , Animals , Base Sequence , Blood , Cell Line , Cytosine , Genes , Guanine , Humans , Mice , Molecular Sequence Data , Muscles/metabolism , Organ Specificity , Serum Response Factor , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics
12.
Mol Endocrinol ; 6(8): 1185-94, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1406697

ABSTRACT

Thyroid hormones are among the positive regulators of muscle development in vivo, but little is known about the way they work. We demonstrate here that MyoD1, one of the master genes controlling myogenesis, is a target of T3. After proliferating C2 myoblasts have been treated with T3 for 15 h, we observed a rise in MyoD1 expression at both the mRNA and protein levels. This is the first positive hormonal control of MyoD1 gene expression reported so far. We also provide data which suggest that T3 nuclear receptor(s) have a direct role on MyoD1 gene transcription: 1) C2 cells express the alpha 1 form of T3 nuclear receptors; 2) T3 up-regulates MyoD1 gene transcription and does not affect MyoD1 mRNA stability, as demonstrated by run-on and actinomycin D chase experiments, respectively; and 3) this transcriptional activation does not need the synthesis of intermediate protein(s) since it is not abolished by simultaneous treatment with cycloheximide. Moreover, in presence of T3, the increase of MyoD1 transcripts is associated with a faster terminal differentiation. Indeed we observed an earlier expression of various markers of myogenesis including myogenin (a regulatory gene of the MyoD1 family mainly involved in the triggering of terminal differentiation), myosin light chain 1A, and troponin T in T3-treated cells vs. untreated cells. We suggest that the regulation of a pivotal myogenic gene could be an important step in the control exerted by T3 on muscle development in vivo.


Subject(s)
Gene Expression Regulation/physiology , Muscles/cytology , Stem Cells/cytology , Transcription, Genetic , Triiodothyronine/physiology , Animals , Blotting, Northern , Blotting, Western , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Electrophoresis, Polyacrylamide Gel , Mice , Microscopy, Fluorescence , Muscle Proteins/metabolism , RNA, Messenger/metabolism
13.
Hum Gene Ther ; 7(1): 11-21, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8825864

ABSTRACT

Protein delivery from genetically modified skeletal muscle has been reported previously. However, a stable and prolonged secretion was obtained in immunocompromised or newborn animals only. To evaluate the clinical relevance of this approach, we have transduced myoblasts from an adult beta-glucuronidase-deficient (MPS VII) mouse with retroviral vectors carrying either the human beta-glucuronidase cDNA or the murine erythropoietin (Epo) cDNA. The cells were then grafted into the tibialis anterior muscle of adult immunocompetent MPS VII recipients. Protein expression was controlled either by ubiquitous or muscle-specific transcriptional regulatory elements. Animals were analyzed over an 8-month period. The in situ detection of beta-glucuronidase activity revealed up to 60% of genetically modified myofibers in the recipient muscles. The human desmin promoter and enhancer showed the highest in vivo expression. Secretion of beta-glucuronidase induced a disappearance of lysosomal storage lesions in the liver and spleen of recipient animals. Delivery of Epo led to a permanent increase of hematocrit values over 3 months. These results showed that the transplantation of genetically modified myoblasts allowed a sustained secretion of recombinant proteins at therapeutic levels in immunocompetent adult mice. They suggest that the approach may be considered for human applications.


Subject(s)
Erythropoietin/genetics , Gene Expression , Genetic Vectors/genetics , Glucuronidase/genetics , Muscle, Skeletal/metabolism , Animals , Erythropoietin/metabolism , Glucuronidase/deficiency , Glucuronidase/metabolism , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Retroviridae/genetics
14.
Endocrinology ; 139(4): 1487-93, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9528925

ABSTRACT

Skeletal myoblast differentiation is stimulated by insulin-like growth factors (IGFs). The autocrine action of IGFs is mediated through the type-1 IGF receptor (IGFR-1) and modulated by IGF binding proteins (IGFBPs) secreted by the cells. The mouse C2 myoblast cell line stably transfected with a vector producing IGF-II antisense RNA was used to show that specific IGFBP expression changes with the state of the cells: high levels of IGFBP-2 messenger RNA (mRNA) were found only in proliferating myoblasts, whereas IGFBP-3 mRNA was induced in quiescent cells. Secretion of IGFBP5 was strongly stimulated during differentiation. Insulin and IGF dose-response experiments showed that up-regulation of IGFBP-5 resulted from IGFR-1 activation. Drugs interfering with IGFR-1 signaling and inhibiting myoblast differentiation had different effects on IGFBP-5 up-regulation. Two phosphatidylinositol 3-kinase (PI 3-kinase) inhibitors, wortmaninn and LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one], failed to alter IGFBP-5 up-regulation, which persisted in the absence of differentiation. Rapamycin which indirectly prevents activation of the p70 ribosomal protein-S6 kinase (p70S6k), suppressed IGFBP-5 induction. Because the PI3-kinase inhibitors block p70S6k, neither kinase would be required for IGFR-1-dependent IGFBP-5 induction. In C2 anti-IGF-II myoblasts, IGFBP-5 induction is therefore rapamycin-sensitive and independent of differentiation.


Subject(s)
Androstadienes/pharmacology , Cell Differentiation , Chromones/pharmacology , Gene Expression Regulation/drug effects , Insulin-Like Growth Factor Binding Protein 5/genetics , Morpholines/pharmacology , Muscles/cytology , Polyenes/pharmacology , Animals , Cell Division , Cell Line , Enzyme Inhibitors/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor Binding Protein 5/metabolism , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice , Phosphoinositide-3 Kinase Inhibitors , Protein Biosynthesis , Receptor, IGF Type 1/physiology , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Sirolimus , Transfection , Wortmannin
15.
FEBS Lett ; 488(3): 174-8, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11163767

ABSTRACT

We have derived skeletal muscle cell lines from wild-type (wt) and insulin receptor (IR) knockout mice to unravel the metabolic potential of IGF-1 receptor (IGF-1R). Both wt and IR(-/-) myoblasts differentiated into myotubes with similar patterns of expression of muscle-specific genes such as MyoD, myogenin and MLC1A indicating that IR is not required for this process. Binding of 125I-IGF-1 on wt and IR(-/-) myotubes was similar showing that IGF-1R was not upregulated in the absence of IR. Stimulation of IR(-/-) myotubes with IGF-1 (10(-10) to 10(-7) M) increased glucose uptake and incorporation into glycogen, induced IRS-1 phosphorylation and activated PI 3-kinase and MAP kinase, two enzymes of major signaling pathways. These effects were comparable to those obtained with wt myotubes using insulin or IGF-1 or with IR(-/-) myotubes using insulin at higher concentrations. This study provides a direct evidence that IGF-1R can represent an alternative receptor for metabolic signaling in muscle cells.


Subject(s)
Gene Deletion , Muscle, Skeletal/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/deficiency , Animals , Animals, Newborn , Binding Sites , Biomarkers , Cells, Cultured , Culture Media, Serum-Free , Deoxyglucose/metabolism , Glucose/metabolism , Glycogen/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptor, Insulin/genetics , Signal Transduction/drug effects
16.
Neuromuscul Disord ; 3(5-6): 413-7, 1993.
Article in English | MEDLINE | ID: mdl-8186684

ABSTRACT

We have explored the use of myoblasts obtained from adult animals as a target for somatic gene therapy. Myoblasts from an adult beta-glucuronidase deficient (MPS VII) mouse were isolated and infected with a retroviral vector carrying the human beta-glucuronidase cDNA. Beta-glucuronidase was used as a reporter gene to follow the fate of genetically-modified myoblasts after transplantation into the tibialis anterior of MPS VII recipients. When experimental necrosis had been induced in the recipient muscle prior to cell injection, histological analysis demonstrated efficient engraftment of adult derived myoblasts following gene transfer. The reconstituted myofibres expressed the transgene for at least 10 weeks following transplantation.


Subject(s)
Gene Transfer Techniques , Glucuronidase/deficiency , Glucuronidase/genetics , Muscles/metabolism , Muscles/transplantation , Animals , DNA, Complementary/metabolism , Genetic Therapy/methods , Genetic Vectors , Glucuronidase/biosynthesis , Humans , Mice , Mice, Mutant Strains , Muscles/physiology , Restriction Mapping , Retroviridae
17.
Biochimie ; 69(3): 171-6, 1987 Mar.
Article in English | MEDLINE | ID: mdl-3111545
19.
C R Seances Acad Sci III ; 295(12): 727-32, 1982 Dec 06.
Article in French | MEDLINE | ID: mdl-6820306

ABSTRACT

When plated in plastic culture dishes, muscle cells of the L6 cell line are unable to proliferate or to differentiate in a synthetic medium composed of Dulbecco's Minimal Essential medium (DME), bovine fetuin (1 mg/ml) and bovine insulin (10 micrograms/ml). In contrast, these cells will proliferate and differentiate morphologically and biochemically in this medium if the culture dishes have been previously treated with DME containing fetal Calf serum. Pretreatment of dishes does not increase cell attachment in synthetic medium but seems to act as a factor increasing cell viability.


Subject(s)
Muscles/physiology , Animals , Cell Differentiation , Cell Division , Culture Media , Kinetics , L Cells/physiology , Mice
20.
Dev Biol ; 108(2): 284-9, 1985 Apr.
Article in English | MEDLINE | ID: mdl-3908188

ABSTRACT

Cells of the myogenic rat cell line L6 can be obtained as a confluent, quiescent population of undifferentiated myoblasts after growth in F12 medium supplemented with fetal calf serum. Myogenic differentiation can be induced in these cells by changing to Dulbecco's modified Eagle's (DME) medium containing insulin as the only protein component. Labeling of the cells with [3H]thymidine demonstrates that this induction of fusion occurs in the absence of DNA synthesis in about 85% of the cells. This result was confirmed using cytosine arabinoside: fusion of quiescent L6 cells was induced in the presence of this inhibitor of DNA synthesis. The myotubes formed in DME + insulin medium, with or without cytosine arabinoside, synthesize or accumulate proteins characteristic of differentiated muscle cells including myosin heavy and light chains, alpha-actin, alpha- and beta-tropomyosins, and the acetylcholine receptor. These experiments represent a direct demonstration that DNA synthesis is not required for the induction of myogenic differentiation in undifferentiated quiescent cells.


Subject(s)
DNA/biosynthesis , Muscles/cytology , Animals , Autoradiography , Blood , Cattle , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Culture Media , Immunoenzyme Techniques , Insulin/pharmacology , Muscles/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL