Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Semin Cell Dev Biol ; 119: 23-31, 2021 11.
Article in English | MEDLINE | ID: mdl-34332886

ABSTRACT

While Fibro-Adipogenic Progenitors (FAPs) have been originally identified as muscle-interstitial mesenchymal cells activated in response to muscle injury and endowed with inducible fibrogenic and adipogenic potential, subsequent studies have expanded their phenotypic and functional repertoire and revealed their contribution to skeletal muscle response to a vast range of perturbations. Here we review the emerging contribution of FAPs to skeletal muscle responses to motor neuron injuries and to systemic physiological (e.g., exercise) or pathological metabolic (e.g., diabetes) perturbations. We also provide an initial blueprint of discrete sub-clusters of FAPs that are activated by specific perturbations and discuss their role in muscle adaptation to these conditions.


Subject(s)
Adipogenesis/physiology , Muscle, Skeletal/metabolism , Neuromuscular Junction/pathology , Animals , Cell Differentiation , Homeostasis , Humans , Mice , Rats
2.
Cell Death Differ ; 23(11): 1839-1849, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27447110

ABSTRACT

Autophagy is emerging as a key regulatory process during skeletal muscle development, regeneration and homeostasis, and deregulated autophagy has been implicated in muscular disorders and age-related muscle decline. We have monitored autophagy in muscles of mdx mice and human Duchenne muscular dystrophy (DMD) patients at different stages of disease. Our data show that autophagy is activated during the early, compensatory regenerative stages of DMD. A progressive reduction was observed during mdx disease progression, in coincidence with the functional exhaustion of satellite cell-mediated regeneration and accumulation of fibrosis. Moreover, pharmacological manipulation of autophagy can influence disease progression in mdx mice. Of note, studies performed in regenerating muscles of wild-type mice revealed an essential role of autophagy in the activation of satellite cells upon muscle injury. These results support the notion that regeneration-associated autophagy contributes to the early compensatory stage of DMD progression, and interventions that extend activation of autophagy might be beneficial in the treatment of DMD. Thus, autophagy could be a 'disease modifier' targeted by interventions aimed to promote regeneration and delay disease progression in DMD.


Subject(s)
Autophagy , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Regeneration , Satellite Cells, Skeletal Muscle/pathology , Animals , Biopsy , Child , Child, Preschool , Disease Progression , Humans , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology
3.
Cell Death Dis ; 6: e1830, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26203859

ABSTRACT

Recent evidence has revealed the importance of reciprocal functional interactions between different types of mononuclear cells in coordinating the repair of injured muscles. In particular, signals released from the inflammatory infiltrate and from mesenchymal interstitial cells (also known as fibro-adipogenic progenitors (FAPs)) appear to instruct muscle stem cells (satellite cells) to break quiescence, proliferate and differentiate. Interestingly, conditions that compromise the functional integrity of this network can bias muscle repair toward pathological outcomes that are typically observed in chronic muscular disorders, that is, fibrotic and fatty muscle degeneration as well as myofiber atrophy. In this review, we will summarize the current knowledge on the regulation of this network in physiological and pathological conditions, and anticipate the potential contribution of its cellular components to relatively unexplored conditions, such as aging and physical exercise.


Subject(s)
Eosinophils/metabolism , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Adipocytes/cytology , Adipocytes/immunology , Adipocytes/metabolism , Cell Communication , Cell Differentiation , Eosinophils/cytology , Eosinophils/immunology , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Homeostasis , Humans , Macrophages/cytology , Macrophages/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Muscle Development/physiology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/immunology
5.
Oncogene ; 33(32): 4173-84, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24213577

ABSTRACT

The Polycomb group (PcG) proteins regulate stem cell differentiation via the repression of gene transcription, and their deregulation has been widely implicated in cancer development. The PcG protein Enhancer of Zeste Homolog 2 (EZH2) works as a catalytic subunit of the Polycomb Repressive Complex 2 (PRC2) by methylating lysine 27 on histone H3 (H3K27me3), a hallmark of PRC2-mediated gene repression. In skeletal muscle progenitors, EZH2 prevents an unscheduled differentiation by repressing muscle-specific gene expression and is downregulated during the course of differentiation. In rhabdomyosarcoma (RMS), a pediatric soft-tissue sarcoma thought to arise from myogenic precursors, EZH2 is abnormally expressed and its downregulation in vitro leads to muscle-like differentiation of RMS cells of the embryonal variant. However, the role of EZH2 in the clinically aggressive subgroup of alveolar RMS, characterized by the expression of PAX3-FOXO1 oncoprotein, remains unknown. We show here that EZH2 depletion in these cells leads to programmed cell death. Transcriptional derepression of F-box protein 32 (FBXO32) (Atrogin1/MAFbx), a gene associated with muscle homeostasis, was evidenced in PAX3-FOXO1 RMS cells silenced for EZH2. This phenomenon was associated with reduced EZH2 occupancy and H3K27me3 levels at the FBXO32 promoter. Simultaneous knockdown of FBXO32 and EZH2 in PAX3-FOXO1 RMS cells impaired the pro-apoptotic response, whereas the overexpression of FBXO32 facilitated programmed cell death in EZH2-depleted cells. Pharmacological inhibition of EZH2 by either 3-Deazaneplanocin A or a catalytic EZH2 inhibitor mirrored the phenotypic and molecular effects of EZH2 knockdown in vitro and prevented tumor growth in vivo. Collectively, these results indicate that EZH2 is a key factor in the proliferation and survival of PAX3-FOXO1 alveolar RMS cells working, at least in part, by repressing FBXO32. They also suggest that the reducing activity of EZH2 could represent a novel adjuvant strategy to eradicate high-risk PAX3-FOXO1 alveolar RMS.


Subject(s)
Forkhead Transcription Factors/metabolism , Muscle Proteins/antagonists & inhibitors , Paired Box Transcription Factors/metabolism , Polycomb Repressive Complex 2/physiology , Rhabdomyosarcoma, Alveolar/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Adolescent , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Child , Enhancer of Zeste Homolog 2 Protein , Female , Forkhead Box Protein O1 , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Homeostasis , Humans , Male , Muscle Proteins/physiology , PAX3 Transcription Factor , SKP Cullin F-Box Protein Ligases/physiology
6.
Rare Dis ; 2(1): e974969, 2014.
Article in English | MEDLINE | ID: mdl-26942105

ABSTRACT

Recent studies have revealed the contribution of fibro-adipogenic progenitors (FAPs) to the pathogenesis and progression of Duchenne Muscular Dystrophy (DMD). While FAPs direct compensatory regeneration at early stages of disease, as the disease progresses they contribute to the progressive replacement of contractile myofibers with fibrotic scars and fatty infiltration. Using the mouse model of DMD - the mdx mice - we have recently reported that FAPs mediate the ability of HDAC inhibitors (HDACi) to promote muscle regeneration and prevent fibro-adipogenic degeneration at early stages of disease. This effect is mediated by the induction of myomiRs that, in turn, target the SWI/SNF components BAF60A and B, thereby favoring the formation of BAF60C-based SWI/SNF complex, which directs the switch from the fibro-adipogenic to the myogenic lineage. Here we show direct evidence of induction of miR-206 and BAF60C, and reduction of BAF60A, in FAPs isolated from mdx muscles exposed to the HDACi Trichostatin A (TSA). We also discuss how increased expression of myomiRs in dystrophic muscles can be integrated with circulating myomiRs to provide accurate biomarkers of disease progression and response to treatment.

7.
Cell Death Differ ; 20(12): 1664-74, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24056763

ABSTRACT

Previous works have established a unique function of MyoD in the control of muscle gene expression during DNA damage response in myoblasts. Phosphorylation by DNA damage-activated ABL tyrosine kinase transiently inhibits MyoD-dependent activation of transcription in response to genotoxic stress. We show here that ABL-MyoD signaling is also an essential component of the DNA repair machinery in myoblasts exposed to genotoxic stress. DNA damage promoted the recruitment of MyoD to phosphorylated Nbs1 (pNbs1)-containing repair foci, and this effect was abrogated by either ABL knockdown or the ABL kinase inhibitor imatinib. Upon DNA damage, MyoD and pNbs1 were detected on the chromatin to MyoD target genes without activating transcription. DNA damage-mediated tyrosine phosphorylation was required for MyoD recruitment to target genes, as the ABL phosphorylation-resistant MyoD mutant (MyoD Y30F) failed to bind the chromatin following DNA damage, while retaining the ability to activate transcription in response to differentiation signals. Moreover, MyoD Y30F exhibited an impaired ability to promote repair in a heterologous system, as compared with MyoD wild type (WT). Consistently, MyoD-null satellite cells (SCs) displayed impaired DNA repair that was rescued by reintroduction of MyoD WT but not by MyoD Y30F. In addition, inhibition of ABL kinase prevented MyoD WT-mediated rescue of DNA repair in MyoD-null SCs. These results identify an unprecedented contribution of MyoD to DNA repair and suggest that ABL-MyoD signaling coordinates DNA repair and transcription in myoblasts.


Subject(s)
DNA Damage , DNA Repair , MyoD Protein/metabolism , Myoblasts, Skeletal/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Animals , Cell Nucleus/metabolism , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mutation/genetics , Myoblasts, Skeletal/cytology , Polymerase Chain Reaction , S Phase , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Transfection
8.
Nat Med ; 12(10): 1147-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980968

ABSTRACT

Pharmacological interventions that increase myofiber size counter the functional decline of dystrophic muscles. We show that deacetylase inhibitors increase the size of myofibers in dystrophin-deficient (MDX) and alpha-sarcoglycan (alpha-SG)-deficient mice by inducing the expression of the myostatin antagonist follistatin in satellite cells. Deacetylase inhibitor treatment conferred on dystrophic muscles resistance to contraction-coupled degeneration and alleviated both morphological and functional consequences of the primary genetic defect. These results provide a rationale for using deacetylase inhibitors in the pharmacological therapy of muscular dystrophies.


Subject(s)
Enzyme Inhibitors/pharmacology , Muscles/enzymology , Muscles/pathology , Muscular Dystrophy, Animal/drug therapy , Animals , Dystrophin/genetics , Fibrosis/pathology , Follistatin/metabolism , Hydroxamic Acids/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscles/drug effects , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Phenylbutyrates/pharmacology , Sarcoglycans/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/enzymology , Valproic Acid/pharmacology
9.
Mol Cell ; 8(4): 885-97, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11684023

ABSTRACT

We describe a functional and biochemical link between the myogenic activator MyoD, the deacetylase HDAC1, and the tumor suppressor pRb. Interaction of MyoD with HDAC1 in undifferentiated myoblasts mediates repression of muscle-specific gene expression. Prodifferentiation cues, mimicked by serum removal, induce both downregulation of HDAC1 protein and pRb hypophosphorylation. Dephosphorylation of pRb promotes the formation of pRb-HDAC1 complex in differentiated myotubes. pRb-HDAC1 association coincides with disassembling of MyoD-HDAC1 complex, transcriptional activation of muscle-restricted genes, and cellular differentiation of skeletal myoblasts. A single point mutation introduced in the HDAC1 binding domain of pRb compromises its ability to disrupt MyoD-HDAC1 interaction and to promote muscle gene expression. These results suggest that reduced expression of HDAC1 accompanied by its redistribution in alternative nuclear protein complexes is critical for terminal differentiation of skeletal muscle cells.


Subject(s)
Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Muscle Development/physiology , Muscle, Skeletal/growth & development , MyoD Protein/metabolism , Retinoblastoma Protein/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Culture Media, Serum-Free , DNA-Binding Proteins/metabolism , Genes, Reporter , Histone Deacetylase 1 , Histone Deacetylases/genetics , MEF2 Transcription Factors , Microscopy, Fluorescence , Models, Biological , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , MyoD Protein/genetics , Myogenic Regulatory Factors , Phosphorylation , Precipitin Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/genetics , Transcription Factors/metabolism
10.
Front Biosci ; 6: D1024-47, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11532612

ABSTRACT

Chromatin remodeling and protein acetylation control gene expression and consequently regulate cellular growth and differentiation. Here we review the role of individual chromatin remodeling factors, acetyltransferases and deacetylases in the establishment and maintenance of different cell lineages and in the genesis of some human diseases.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Proteins/metabolism , Acetylation , Animals , Chromatin/metabolism , Gene Expression Regulation , Humans
11.
J Cell Physiol ; 185(2): 155-73, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11025438

ABSTRACT

Skeletal muscle differentiation is influenced by multiple pathways, which regulate the activity of myogenic regulatory factors (MRFs)-the myogenic basic helix-loop-helix proteins and the MEF2-family members-in positive or negative ways. Here we will review and discuss the network of signals that regulate MRF function during myocyte proliferation, differentiation, and post-mitotic growth. Elucidating the mechanisms governing muscle-specific transcription will provide important insight in better understanding the embryonic development of muscle at the molecular level and will have important implications in setting out strategies aimed at muscle regeneration. Since the activity of MRFs are compromised in tumors of myogenic derivation-the rhabdomyosarcomas-the studies summarized in this review can provide a useful tool to uncover the molecular basis underlying the formation of these tumors.


Subject(s)
DNA-Binding Proteins/physiology , Myogenic Regulatory Factors/physiology , Protein Processing, Post-Translational/physiology , Animals , Gene Expression Regulation/physiology , Humans , Muscles/physiology , MyoD Protein/genetics , MyoD Protein/physiology , Phosphorylation , Rhabdomyosarcoma/physiopathology
12.
Mol Cell Biol ; 20(11): 3951-64, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10805738

ABSTRACT

The extracellular signals which regulate the myogenic program are transduced to the nucleus by mitogen-activated protein kinases (MAPKs). We have investigated the role of two MAPKs, p38 and extracellular signal-regulated kinase (ERK), whose activities undergo significant changes during muscle differentiation. p38 is rapidly activated in myocytes induced to differentiate. This activation differs from those triggered by stress and cytokines, because it is not linked to Jun-N-terminal kinase stimulation and is maintained during the whole process of myotube formation. Moreover, p38 activation is independent of a parallel promyogenic pathway stimulated by insulin-like growth factor 1. Inhibition of p38 prevents the differentiation program in myogenic cell lines and human primary myocytes. Conversely, deliberate activation of endogenous p38 stimulates muscle differentiation even in the presence of antimyogenic cues. Much evidence indicates that p38 is an activator of MyoD: (i) p38 kinase activity is required for the expression of MyoD-responsive genes, (ii) enforced induction of p38 stimulates the transcriptional activity of a Gal4-MyoD fusion protein and allows efficient activation of chromatin-integrated reporters by MyoD, and (iii) MyoD-dependent myogenic conversion is reduced in mouse embryonic fibroblasts derived from p38alpha(-/-) embryos. Activation of p38 also enhances the transcriptional activities of myocyte enhancer binding factor 2A (MEF2A) and MEF2C by direct phosphorylation. With MEF2C, selective phosphorylation of one residue (Thr293) is a tissue-specific activating signal in differentiating myocytes. Finally, ERK shows a biphasic activation profile, with peaks of activity in undifferentiated myoblasts and postmitotic myotubes. Importantly, activation of ERK is inhibitory toward myogenic transcription in myoblasts but contributes to the activation of myogenic transcription and regulates postmitotic responses (i.e., hypertrophic growth) in myotubes.


Subject(s)
DNA-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , MyoD Protein/metabolism , Myogenic Regulatory Factors/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Enzyme Activation , Humans , Insulin-Like Growth Factor I/metabolism , Isoenzymes/metabolism , MADS Domain Proteins , MEF2 Transcription Factors , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases
13.
Genes Dev ; 14(5): 574-84, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10716945

ABSTRACT

MyoD inhibits cell proliferation and promotes muscle differentiation. A paradoxical feature of rhabdomyosarcoma (RMS), a tumor arising from muscle precursors, is the block of the differentiation program and the deregulated proliferation despite MyoD expression. A deficiency in RMS of a factor required for MyoD activity has been implicated by previous studies. We report here that p38 MAP kinase (MAPK) activation, which is essential for muscle differentiation, is deficient in RMS cells. Enforced induction of p38 MAPK by an activated MAPK kinase 6 (MKK6EE) restored MyoD function and enhanced MEF2 activity in RMS deficient for p38 MAPK activation, leading to growth arrest and terminal differentiation. Stress and cytokines could activate the p38 MAPK in RMS cells, however, these stimuli did not promote differentiation, possibly because they activated p38 MAPK only transiently and they also activated JNK, which could antagonize differentiation. Thus, the selective and sustained p38 MAPK activation, which is distinct from the stress-activated response, is required for differentiation and can be disrupted in human tumors.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Muscles/cytology , Rhabdomyosarcoma/pathology , Animals , Cell Division , Cell Line , Enzyme Activation , Enzyme Induction , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 6 , Mice , Mitogen-Activated Protein Kinases/metabolism , MyoD Protein/metabolism , Recombinant Proteins/metabolism , Rhabdomyosarcoma/enzymology , Transfection , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
14.
Mol Cell Biol ; 19(7): 5203-17, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373569

ABSTRACT

During the terminal differentiation of skeletal myoblasts, the activities of myogenic factors regulate not only tissue-specific gene expressions but also the exit from the cell cycle. The induction of cell cycle inhibitors such as p21 and pRb has been shown to play a prominent role in the growth arrest of differentiating myoblasts. Here we report that, at the onset of differentiation, activation by MyoD of the Rb, p21, and cyclin D3 genes occurs in the absence of new protein synthesis and with the requirement of the p300 transcriptional coactivator. In differentiated myocytes, cyclin D3 also becomes stabilized and is found nearly totally complexed with unphosphorylated pRb. The detection of complexes containing cyclin D3, cdk4, p21, and PCNA suggests that cdk4, along with PCNA, may get sequestered into high-order structures held together by pRb and cyclin D3. Cyclin D3 up-regulation and stabilization is inhibited by adenovirus E1A, and this correlates with the ability of E1A to promote pRb phosphorylation; conversely, the overexpression of cyclin D3 in differentiated myotubes counteracts the E1A-mediated reactivation of DNA synthesis. These results indicate that cyclin D3 critically contributes to the irreversible exit of differentiating myoblasts from the cell cycle.


Subject(s)
Cyclins/physiology , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Proto-Oncogene Proteins , Adenovirus E1A Proteins/metabolism , Animals , Cell Cycle , Cell Differentiation , Cyclin D3 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , E1A-Associated p300 Protein , Humans , Mice , MyoD Protein/genetics , Nuclear Proteins/metabolism , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Trans-Activators/metabolism
15.
Cell ; 96(3): 405-13, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-10025406

ABSTRACT

Histone acetyltransferases (HAT) play a critical role in transcriptional control by relieving repressive effects of chromatin, and yet how HATs themselves are regulated remains largely unknown. Here, it is shown that Twist directly binds two independent HAT domains of acetyltransferases, p300 and p300/CBP-associated factor (PCAF), and directly regulates their HAT activities. The N terminus of Twist is a primary domain interacting with both acetyltransferases, and the same domain is required for inhibition of p300-dependent transcription by Twist. Adenovirus E1A protein mimics the effects of Twist by inhibiting the HAT activities of p300 and PCAF. These findings establish a cogent argument for considering the HAT domains as a direct target for acetyltransferase regulation by both a cellular transcription factor and a viral oncoprotein.


Subject(s)
Acetyltransferases/metabolism , Adenovirus E1A Proteins/physiology , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Transcription Factors/physiology , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/physiology , Adenovirus E1A Proteins/metabolism , Animals , COS Cells , Cells, Cultured , E1A-Associated p300 Protein , Enzyme Activation , Histone Acetyltransferases , Mice , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Oncogene Proteins, Viral/metabolism , Oncogene Proteins, Viral/physiology , Peptide Fragments/metabolism , Peptide Fragments/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/metabolism , Transcription, Genetic , Twist-Related Protein 1
16.
Mol Cell ; 4(5): 725-34, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10619020

ABSTRACT

p300/CBP and PCAF coactivators have acetyltransferase activities and regulate transcription, cell cycle progression, and differentiation. They are both required for MyoD activity and muscle differentiation. Nevertheless, their roles must be different since the acetyltransferase activity of PCAF but not of p300 is involved in controlling myogenic transcription and differentiation. Here, we provide a molecular explanation of this phenomenon and report that MyoD is directly acetylated by PCAF at evolutionarily conserved lysines. Acetylated MyoD displays an increased affinity for its DNA target. Importantly, conservative substitutions of acetylated lysines with nonacetylatable arginines impair the ability of MyoD to stimulate transcription and to induce muscle conversion indicating that acetylation of MyoD is functionally critical.


Subject(s)
Acetyltransferases/metabolism , Muscles/cytology , Muscles/metabolism , MyoD Protein/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors , Transcriptional Activation/genetics , Acetylation , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Binding Sites , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , Conserved Sequence , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Fibroblasts/cytology , Fibroblasts/metabolism , Histone Acetyltransferases , Lysine/genetics , Lysine/metabolism , Mice , Muscle Development , Muscles/enzymology , MyoD Protein/chemistry , MyoD Protein/genetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein
17.
J Cell Biochem ; 71(4): 467-78, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9827693

ABSTRACT

CDK9 has been recently shown to have increased kinase activity in differentiated cells in culture and a differentiated tissue-specific expression in the developing mouse. In order to identify factors that contribute to CDK9's differentiation-specific function, we screened a mouse embryonic library in the yeast two-hybrid system and found a tumor necrosis factor signal transducer, TRAF2, to be an interacting protein. CDK9 interacts with a conserved domain in the TRAF-C region of TRAF2, a motif that is known to bind other kinases involved in TRAF-mediated signaling. Endogenous interaction between the two proteins appears to be specific to differentiated tissue. TRAF2-mediated signaling may incorporate additional kinases to signal cell survival in myotubes, a cell type that is severely affected in TRAF2 knockout mice.


Subject(s)
Muscle, Skeletal/cytology , Protein Kinases/metabolism , Proteins/metabolism , Animals , Binding Sites , Cell Differentiation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 9 , Humans , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , NF-kappa B/metabolism , Protein Kinases/genetics , Proteins/genetics , Proteins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Subcellular Fractions , TNF Receptor-Associated Factor 2 , Tumor Necrosis Factor-alpha/pharmacology
18.
Cancer Res ; 58(7): 1325-31, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9537223

ABSTRACT

E2F/DP heterodimers play a pivotal role in the regulation of cell growth and differentiation. A decrease in E2F/DP activity occurs during cell cycle arrest and differentiation. However, very little is known about the specific role of the various E2F/DP members along the transition from proliferation to terminal differentiation. We have previously shown that E2F4 accounts for the vast majority of the endogenous E2F in differentiating muscle cells. Here, we show that E2F4, which lacks a nuclear localization signal (nls), is distributed in both the nucleus and the cytoplasm, in either asynchronously growing myoblasts or differentiated myotubes. E2F4 nuclear accumulation is induced by the binding in the cytoplasm with specific partners p107, pRb2/p130, and DP3delta, an nls-containing spliced form of DP3, which provide the nls. Although overexpression of E2F4/DP3delta reactivates the cell cycle in quiescent cells, the E2F4 nuclear accumulation induced by pRb2/p130 and p107 correlates with cell growth arrest Moreover, E2F4/DP3delta-induced cell cycle reactivation is efficiently counteracted by either p107 or pRb2/p130 overexpression. Reinduction in quiescent cells of DNA synthesis by E2F1/DP1 overexpression is abrogated by coexpression of pRb and is hampered by MyoD overexpression. Both pRb2/p130 and pRb, as well as MyoD, are up-regulated in myotubes. Accordingly, multinucleated myotubes, which are induced to reenter the S-phase by oncoviral proteins, are refractory to cell cycle reactivation by forced expression of E2F4/DP3delta or E2F1/DP1. Thus, E2F/DP repression represents only one of multiple redundant circuits that control the postmitotic state in terminally differentiated cells and that are targeted by adenovirus E1A and SV40 large T antigen.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins , Growth Substances/physiology , Muscles/cytology , Muscles/metabolism , Transcription Factors/physiology , Translocation, Genetic/physiology , Animals , Cell Cycle/physiology , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , DNA/biosynthesis , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Dimerization , E2F4 Transcription Factor , Genetic Vectors , Growth Substances/biosynthesis , Growth Substances/metabolism , Mice , Microinjections , Muscles/physiology , Subcellular Fractions/metabolism , Trans-Activators/biosynthesis , Trans-Activators/metabolism , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Up-Regulation
19.
Proc Natl Acad Sci U S A ; 94(15): 8162-7, 1997 Jul 22.
Article in English | MEDLINE | ID: mdl-9223332

ABSTRACT

The human hepatitis B virus (HBV) protein pX is a multifunctional regulatory protein that is known to affect both transcription and cell growth. Here we describe induction of apoptosis in NIH 3T3 polyclonal cell lines upon stimulation of pX expression from a dexamethasone inducible mouse mammary tumor virus (MMTV)-X expression vector. The effect of long-term pX expression on the cell survival of mouse fibroblasts was confirmed in colony generation assays. This effect is not shared either by the other HBV products and it is c-myc mediated, as shown by the use of a dominant negative deletion mutant of c-myc. pX also sensitize cells to programmed cell death after exposure to DNA damaging agents. Taking advantage of stable transfectants carrying the p53val135 temperature-sensitive allele, we directly demonstrate that induction of apoptosis by pX requires p53. In p53 null mouse embryo fibroblasts pX activates transcription and confers an evident growth advantage without loss of cell viability. Although pX protein was not detectable in the experimental conditions we used, our results indicate that its expression affects both cell growth and cell death control.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/genetics , Hepatitis B Antigens/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , 3T3 Cells , Animals , Cell Survival/genetics , Culture Media, Serum-Free , DNA Replication/genetics , Mice , Mice, Inbred BALB C , Transcriptional Activation , Viral Regulatory and Accessory Proteins
20.
J Cell Biochem ; 66(1): 27-36, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9215525

ABSTRACT

Doxorubicin (Dox, Adriamicin), a potent broad spectrum anthracycline anticancer drug, selectively inhibits muscle specific gene expression in cardiac cells in vivo and prevents terminal differentiation of skeletal muscle cells in vitro. By inducing the expression of the helix-loop-helix (HLH) transcriptional inhibitor ld2, Dox represses the myogenic function of the MyoD family of muscle regulatory factors (MRFs). In many cell types, terminal differentiation is coupled to an irreversible exit from the cell cycle and MyoD plays a critical role in the permanent cell cycle arrest of differentiating myocytes by upregulating the cyclin dependent kinase inhibitor (cdki) p21. Here, we correlate Dox effects on cell cycle with changes of E2F/DP complexes and activity in differentiating C2C12 myocytes. In Dox-treated quiescent myoblasts, which fail to differentiate into myotubes under permissive culture conditions, serum re-stimulation induces cyclin/cdk re-association on the E2F/DP complexes and this correlates with an evident increase in E2F/DP driven transcription and re-entry of myoblasts into the cell cycle. Despite Dox ability to activate the DNA-damage dependent p53/p21 pathway, when induced in the absence of MyoD or other MRFs, p21 fails to maintain the postmitotic state in Dox-treated myocytes induced to differentiate. Thus, uncoupling p21 induction and MyoD activity results in a serum-reversible cell cycle arrest, indicating that MRF specific activation of cdki(s) is required for permanent cell cycle arrest in differentiating muscle cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carrier Proteins , Cell Cycle Proteins , Cell Cycle/drug effects , Cyclins/metabolism , Doxorubicin/pharmacology , Muscles/cytology , MyoD Protein/metabolism , Repressor Proteins , Animals , Cell Differentiation/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Inhibitor of Differentiation Protein 2 , Mice , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...