Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 5: e1136, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24651439

ABSTRACT

Obesity is associated with chronic low-grade inflammation and oxidative stress that blunt insulin response in its target tissues, leading to insulin resistance (IR). IR is a characteristic feature of type 2 diabetes. Skeletal muscle is responsible for 75% of total insulin-dependent glucose uptake; consequently, skeletal muscle IR is considered to be the primary defect of systemic IR development. Interestingly, some obese people stay insulin-sensitive and metabolically healthy. With the aim of understanding this difference and identifying the mechanisms responsible for insulin sensitivity maintenance/IR development during obesity, we explored the role of the latent endoribonuclease (RNase L) in skeletal muscle cells. RNase L is a regulator of innate immunity, of double-stranded RNA sensors and of toll-like receptor (TLR) 4 signaling. It is regulated during inflammation by interferons and its activity is dependent on its binding to 2-5A, an oligoadenylate synthesized by oligoadenylate synthetases (OAS). Increased expression of RNase L or downregulation of its inhibitor (RLI) improved insulin response in mouse myogenic C2C12 cells and in primary human myotubes from normal-weight subjects treated with palmitate, a saturated free fatty acid (FFA) known to induce inflammation and oxidative stress via TLR4 activation. While RNase L and RLI levels remained unchanged, OAS level was decreased in primary myotubes from insulin-resistant obese subjects (OB-IR) compared with myotubes from insulin-sensitive obese subjects (OB-IS). TLR3 and mitochondrial manganese superoxide dismutase (MnSOD) were also underexpressed in OB-IR myotubes. Activation of RNase L by 2-5A transfection allowed to restore insulin response, OAS, MnSOD and TLR3 expression in OB-IR myotubes. Due to low expression of OAS, OB-IR myotubes present a defect in RNase L activation and TLR3 regulation. Consequently, MnSOD level is low and insulin sensitivity is reduced. These results support that RNase L activity limits FFA/obesity-induced impairment of insulin response in muscle cells via TLR3 and MnSOD expression.


Subject(s)
Endoribonucleases/metabolism , Insulin Resistance , Insulin/metabolism , Myoblasts, Skeletal/enzymology , Obesity/enzymology , Quadriceps Muscle/enzymology , Superoxide Dismutase/metabolism , Toll-Like Receptor 3/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Case-Control Studies , Down-Regulation , Endoribonucleases/genetics , Enzyme Activation , Female , HeLa Cells , Humans , Male , Mice , Middle Aged , Obesity/genetics , Palmitic Acid/metabolism , RNA Interference , Signal Transduction , Superoxide Dismutase/genetics , Toll-Like Receptor 3/genetics , Transfection
2.
Diabetologia ; 53(6): 1151-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20333349

ABSTRACT

AIMS/HYPOTHESIS: Intramyocellular lipids (IMCL) accumulation is a classical feature of metabolic diseases. We hypothesised that IMCL accumulate mainly as a consequence of increased adiposity and independently of type 2 diabetes. To test this, we examined IMCL accumulation in two different models and four different populations of participants: muscle biopsies and primary human muscle cells derived from non-obese and obese participants with or without type 2 diabetes. The mechanism regulating IMCL accumulation was also studied. METHODS: Muscle biopsies were obtained from ten non-obese and seven obese participants without type 2 diabetes, and from eight non-obese and eight obese type 2 diabetic patients. Mitochondrial respiration, citrate synthase activity and both AMP-activated protein kinase and acetyl-CoA carboxylase phosphorylation were measured in muscle tissue. Lipid accumulation in muscle and primary myotubes was estimated by Oil Red O staining and fatty acid translocase (FAT)/CD36 localisation by immunofluorescence. RESULTS: Obesity and type 2 diabetes are independently characterised by skeletal muscle IMCL accumulation and permanent FAT/CD36 relocation. Mitochondrial function is not reduced in type 2 diabetes. IMCL accumulation was independent of type 2 diabetes in cultured myotubes and was correlated with obesity markers of the donor. In obese participants, membrane relocation of FAT/CD36 is a determinant of IMCL accumulation. CONCLUSIONS/INTERPRETATION: In skeletal muscle, mitochondrial function is normal in type 2 diabetes, while IMCL accumulation is dependent upon obesity or type 2 diabetes and is related to sarcolemmal FAT/CD36 relocation. In cultured myotubes, IMCL content and FAT/CD36 relocation are independent of type 2 diabetes, suggesting that distinct factors in obesity and type 2 diabetes contribute to permanent FAT/CD36 relocation ex vivo.


Subject(s)
CD36 Antigens/metabolism , Diabetes Mellitus, Type 2/metabolism , Lipids/analysis , Muscle, Skeletal/chemistry , Obesity/metabolism , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Analysis of Variance , Blotting, Western , Body Fat Distribution , Cells, Cultured , Citrate (si)-Synthase/metabolism , Diabetes Mellitus, Type 2/complications , Fluorescent Antibody Technique , Humans , Male , Middle Aged , Mitochondria/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Obesity/complications , Phosphorylation/physiology , Waist Circumference
3.
Cell Mol Life Sci ; 58(4): 571-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11361092

ABSTRACT

During the early process of skeletal muscle differentiation, myogenic factors are not only involved in muscle-specific gene induction but also in regulating the transition from the proliferative stage, when MyoD and Myf5 are already expressed, to the orderly exit from the cell division cycle. This key step in skeletal muscle differentiation involves the down-regulation of cell cycle activators such as cyclins and cdks, and up-regulation of cell cycle inhibitors such as Rb, p21, p27, and p57. In particular, Rb and p21 have been shown to play an important role in the growth arrest of differentiating myoblasts. Their level and/or activity, while being negatively controlled by growth factors, appear to be positively linked with the myogenic factor MyoD, which plays a cooperative role in the induction of growth arrest. MyoD can block proliferation independently of its transcriptional activity. Therefore, the interplay between G1 cyclins and cdk inhibitors, on the one hand, and MyoD and its co-factors, on the other, plays a critical role in myoblast cell cycle withdrawal. Accurate synchronization of dividing myoblasts revealed that MyoD and Myf5 are themselves subject to specific cell cycle-dependent regulation, with MyoD at its highest level in early G1 and its lowest level at the G1 to S phase transition. The time-window when cells exit their cycle into differentiation is in G1, when MyoD is maximal and Myf5 is down. In contrast, quiescent non-differentiating myoblasts (i. e., in G0) present an opposite pattern for the two factors: high Myf5 and no MyoD. Several recent studies have focused on MyoD phosphorylation and its potential role in ubiquitination-mediated degradation of the protein. Linking this phosphorylation to the cell cycle-dependent drop in MyoD protein before S phase leads, to a mechanism implying cdk2-cyclin E and its inhibitors (p57kip and p21cip) in the tight control of MyoD levels and subsequent myoblast cell cycle progression or exit into differentiation.


Subject(s)
Cell Cycle Proteins/metabolism , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Animals , Cell Cycle , Cell Differentiation , Cell Division , Humans , Muscle, Skeletal/cytology , Signal Transduction/physiology
4.
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
5.
J Biol Chem ; 274(41): 29063-70, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10506158

ABSTRACT

The molecular mechanisms underlying the developmental regulation of L-type voltage-dependent Ca(2+) channels (VDCCs) are still unknown. In this study, we have characterized the expression patterns of skeletal (alpha(1S)) and cardiac (alpha(1C)) L-type VDCCs during cardiogenic differentiation in H9C2 cells that derived from embryonic rat heart. We report that chronic treatment of H9C2 cells with 10 nM all-trans-retinoic acid (all-trans-RA) enhanced cardiac Ca(2+) channel expression, as demonstrated by reverse transcription-polymerase chain reaction, immunoblotting, and indirect immunofluorescence studies, as well as patch-clamp experiments. In addition, RA treatment prevented expression of functional skeletal L-type VDCCs, which were restricted to myotubes that spontaneously appear in control H9C2 cultures undergoing myogenic transdifferentiation. The use of specific skeletal and cardiac markers indicated that RA, by preventing myogenic transdifferentiation, preserves cardiac differentiation of this cell line. Altogether, we provide evidence that cardiac and skeletal subtype-specific L-type Ca(2+) channels are relevant functional markers of differentiated cardiac and skeletal myocytes, respectively. In conclusion, our data demonstrate that in vitro RA stimulates cardiac (alpha(1C)) L-type Ca(2+) channel expression, therefore supporting the hypothesis that the RA pathway might be involved in the tissue specific expression of Ca(2+) channels in mature cardiac cells.


Subject(s)
Calcium Channels, L-Type/genetics , Myocardium/metabolism , Animals , Cell Differentiation , Cell Line , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Muscle, Skeletal/metabolism , Patch-Clamp Techniques , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/pharmacology
6.
Mol Cell Biol ; 19(4): 3167-76, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10082583

ABSTRACT

We have examined the role of protein phosphorylation in the modulation of the key muscle-specific transcription factor MyoD. We show that MyoD is highly phosphorylated in growing myoblasts and undergoes substantial dephosphorylation during differentiation. MyoD can be efficiently phosphorylated in vitro by either purified cdk1-cyclin B or cdk1 and cdk2 immunoprecipitated from proliferative myoblasts. Comparative two-dimensional tryptic phosphopeptide mapping combined with site-directed mutagenesis revealed that cdk1 and cdk2 phosphorylate MyoD on serine 200 in proliferative myoblasts. In addition, when the seven proline-directed sites in MyoD were individually mutated, only substitution of serine 200 to a nonphosphorylatable alanine (MyoD-Ala200) abolished the slower-migrating hyperphosphorylated form of MyoD, seen either in vitro after phosphorylation by cdk1-cyclin B or in vivo following overexpression in 10T1/2 cells. The MyoD-Ala200 mutant displayed activity threefold higher than that of wild-type MyoD in transactivation of an E-box-dependent reporter gene and promoted markedly enhanced myogenic conversion and fusion of 10T1/2 fibroblasts into muscle cells. In addition, the half-life of MyoD-Ala200 protein was longer than that of wild-type MyoD, substantiating a role of Ser200 phosphorylation in regulating MyoD turnover in proliferative myoblasts. Taken together, our data show that direct phosphorylation of MyoD Ser200 by cdk1 and cdk2 plays an integral role in compromising MyoD activity during myoblast proliferation.


Subject(s)
CDC2 Protein Kinase/metabolism , CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/metabolism , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Stem Cells/cytology , Alanine/genetics , Alanine/metabolism , Cell Differentiation , Cell Division , Cyclin-Dependent Kinase 2 , Half-Life , Muscle, Skeletal/metabolism , Phosphorylation , Serine/metabolism , Stem Cells/metabolism , Transcriptional Activation
7.
J Cell Biol ; 142(6): 1447-59, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9744876

ABSTRACT

The muscle regulators MyoD and Myf-5 control cell cycle withdrawal and induction of differentiation in skeletal muscle cells. By immunofluorescence analysis, we show that MyoD and Myf-5 expression patterns become mutually exclusive when C2 cells are induced to differentiate with Myf-5 staining present in cells which fail to differentiate. Isolation of these undifferentiated cells reveals that upon serum stimulation they reenter the cell cycle, express MyoD and downregulate Myf-5. Similar regulations of MyoD and Myf-5 were observed using cultured primary myoblasts derived from satellite cells. To further analyze these regulations of MyoD and Myf-5 expression, we synchronized proliferating myoblasts. Analysis of MyoD and Myf-5 expression during cell cycle progression revealed distinct and contrasting profiles of expression. MyoD is absent in G0, peaks in mid-G1, falls to its minimum level at G1/S and reaugments from S to M. In contrast, Myf-5 protein is high in G0, decreases during G1 and reappears at the end of G1 to remain stable until mitosis. These data demonstrate that the two myogenic factors MyoD and Myf-5 undergo specific and distinct cell cycle-dependent regulation, thus establishing a correlation between the cell cycle-specific ratios of MyoD and Myf-5 and the capacity of cells to differentiate: (a) in G1, when cells express high levels of MyoD and enter differentiation; (b) in G0, when cells express high levels of Myf-5 and fail to differentiate.


Subject(s)
Cell Cycle , DNA-Binding Proteins , Muscle Proteins/biosynthesis , Muscles/metabolism , MyoD Protein/biosynthesis , Trans-Activators , Animals , Cell Differentiation , Cell Division , Cell Line , Cells, Cultured , Methionine/metabolism , Mice , Mice, Inbred BALB C , Muscles/cytology , Myogenic Regulatory Factor 5
8.
Oncogene ; 16(26): 3369-78, 1998 Jul 02.
Article in English | MEDLINE | ID: mdl-9692544

ABSTRACT

The results reported here indicate that retinoic acid (RA) induces growth arrest and differentiation only in MyoD-expressing muscle cells. Transient transfection assays reveal a functional interaction between MyoD, a key myogenic regulator and RA-receptors, principal mediators of RA actions. Interestingly, we demonstrate that RXR-MyoD-containing complexes are recruited at specific MyoD DNA-binding sites in muscle cells. Furthermore, we also demonstrate that RA-receptors and the muscle basic helix-loop-helix (b-HLH) proteins interact physically. Mutational analysis suggests that this interaction occurs via the basic region of muscle b-HLH proteins and the DNA-binding domain of RA-receptors and is important for functional interactions between these two families of transcription factors. In conclusion, these results highlight novel interactions between two distinct groups of regulatory proteins that influence cell growth and differentiation.


Subject(s)
Helix-Loop-Helix Motifs , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Binding Sites , Cell Differentiation , DNA Mutational Analysis , Gene Expression Regulation , Muscle, Skeletal/drug effects , MyoD Protein/genetics , Protein Binding , Receptors, Retinoic Acid/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tretinoin/pharmacology , Troponin/analysis , Troponin T
9.
Mol Biol Cell ; 9(7): 1891-902, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658178

ABSTRACT

MyoD and Myf5 belong to the family of basic helix-loop-helix transcription factors that are key operators in skeletal muscle differentiation. MyoD and Myf5 genes are selectively activated during development in a time and region-specific manner and in response to different stimuli. However, molecules that specifically regulate the expression of these two genes and the pathways involved remain to be determined. We have recently shown that the serum response factor (SRF), a transcription factor involved in activation of both mitogenic response and muscle differentiation, is required for MyoD gene expression. We have investigated here whether SRF is also involved in the control of Myf5 gene expression, and the potential role of upstream regulators of SRF activity, the Rho family G-proteins including Rho, Rac, and CDC42, in the regulation of MyoD and Myf5. We show that inactivation of SRF does not alter Myf5 gene expression, whereas it causes a rapid extinction of MyoD gene expression. Furthermore, we show that RhoA, but not Rac or CDC42, is also required for the expression of MyoD. Indeed, blocking the activity of G-proteins using the general inhibitor lovastatin, or more specific antagonists of Rho proteins such as C3-transferase or dominant negative RhoA protein, resulted in a dramatic decrease of MyoD protein levels and promoter activity without any effects on Myf5 expression. We further show that RhoA-dependent transcriptional activation required functional SRF in C2 muscle cells. These data illustrate that MyoD and Myf5 are regulated by different upstream activation pathways in which MyoD expression is specifically modulated by a RhoA/SRF signaling cascade. In addition, our results establish the first link between RhoA protein activity and the expression of a key muscle regulator.


Subject(s)
Botulinum Toxins , DNA-Binding Proteins/physiology , GTP Phosphohydrolases/physiology , GTP-Binding Proteins/physiology , MyoD Protein/biosynthesis , Nuclear Proteins/physiology , 3T3 Cells , ADP Ribose Transferases/physiology , Animals , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , GTP Phosphohydrolases/antagonists & inhibitors , GTP-Binding Proteins/antagonists & inhibitors , Gene Expression Regulation , Genes, Dominant , Mice , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , MyoD Protein/antagonists & inhibitors , Myogenic Regulatory Factor 5 , Nuclear Proteins/antagonists & inhibitors , Promoter Regions, Genetic/physiology , Rats , Repressor Proteins/physiology , Serum Response Factor , Trans-Activators/genetics , rhoA GTP-Binding Protein
10.
J Cell Sci ; 110 ( Pt 10): 1251-60, 1997 May.
Article in English | MEDLINE | ID: mdl-9191048

ABSTRACT

We have examined the expression, activity and localization of cyclin dependent kinase 5 (cdk5), during myogenesis. Cdk5 protein was found expressed in adult mouse muscle. In murine C2 cells, both the protein level and kinase activity of cdk5 showed a marked increase during early myogenesis with a peak between 36 and 48 hours of differentiation, decreasing as myotubes fuse after 60 to 72 hours. This increase in cdk5 protein level was specific for differentiation and not simply related to cell cycle arrest since it was not observed in fibroblasts grown for 48 hours in low serum medium. Indirect immunofluorescence using monospecific purified anti-cdk5 antibodies showed a low level cytoplasmic staining in proliferative myoblasts, a rapid increase in nuclear staining during the initial 12 hours of differentiation and a predominant nuclear staining in myotubes. Microinjection of plasmids encoding wild-type cdk5 into C2 myoblasts enhanced differentiation as assessed by both myogenin and troponin T expression after 48 hours of differentiation. In contrast, microinjection of plasmids encoding a dominant negative mutant of cdk5 inhibited the onset of differentiation. These data imply a previously unsuspected role for cdk5 protein kinase as a positive modulator of early myogenesis.


Subject(s)
Cyclin-Dependent Kinases , Muscle Development , Muscle, Skeletal/enzymology , Muscle, Skeletal/growth & development , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Line , Cell Nucleus/enzymology , Cyclin-Dependent Kinase 5 , Cytoplasm/enzymology , DNA Primers/genetics , Gene Expression , Immunohistochemistry , Mice , Muscle, Skeletal/metabolism , Mutagenesis, Site-Directed , Myogenin/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Rats , Subcellular Fractions/enzymology , Troponin/metabolism , Troponin T
11.
Neurosci Lett ; 218(1): 21-4, 1996 Oct 25.
Article in English | MEDLINE | ID: mdl-8939471

ABSTRACT

We have examined the expression of cyclin dependent kinase (cdk) 5 protein kinase and p35nck5a, its activator subunit, during postnatal neurogenesis in rat cerebellum, using mono-specific antibodies. Both cdk5 and p35nck5a are present and associated in proliferative stages, although cdk5-p35 kinase activity is barely detectable. Cdk5-p35 activity, but not the expression of either subunit, increases up to 6-fold during neuronal differentiation. Since we observe that cdk5 is phosphorylated on tyrosine in proliferative, but not in post-mitotic stages, we suggest that post-translational regulatory mechanisms control cdk5-p35 protein kinase activity during neurogenesis.


Subject(s)
Cerebellum/metabolism , Cyclin-Dependent Kinases , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Tyrosine/metabolism , Animals , Blotting, Western , Cyclin-Dependent Kinase 5 , Immunohistochemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...