Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Skelet Muscle ; 8(1): 2, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29329560

ABSTRACT

BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD) is associated with DNA hypomethylation at the 4q35 D4Z4 repeat array. Both the causal gene DUX4 and its homolog DUX4c are induced. DUX4c is immunodetected in every myonucleus of proliferative cells, while DUX4 is present in only 1/1000 of myonuclei where it initiates a gene deregulation cascade. FSHD primary myoblasts differentiate into either atrophic or disorganized myotubes. DUX4 expression induces atrophic myotubes and associated FSHD markers. Although DUX4 silencing normalizes the FSHD atrophic myotube phenotype, this is not the case for the disorganized phenotype. DUX4c overexpression increases the proliferation rate of human TE671 rhabdomyosarcoma cells and inhibits their differentiation, suggesting a normal role during muscle differentiation. METHODS: By gain- and loss-of-function experiments in primary human muscle cells, we studied the DUX4c impact on proliferation, differentiation, myotube morphology, and FSHD markers. RESULTS: In primary myoblasts, DUX4c overexpression increased the staining intensity of KI67 (a proliferation marker) in adjacent cells and delayed differentiation. In differentiating cells, DUX4c overexpression led to the expression of some FSHD markers including ß-catenin and to the formation of disorganized myotubes presenting large clusters of nuclei and cytoskeletal defects. These were more severe when DUX4c was expressed before the cytoskeleton reorganized and myofibrils assembled. In addition, endogenous DUX4c was detected at a higher level in FSHD myotubes presenting abnormal clusters of nuclei and cytoskeletal disorganization. We found that the disorganized FSHD myotube phenotype could be rescued by silencing of DUX4c, not DUX4. CONCLUSION: Excess DUX4c could disturb cytoskeletal organization and nuclear distribution in FSHD myotubes. We suggest that DUX4c up-regulation could contribute to DUX4 toxicity in the muscle fibers by favoring the clustering of myonuclei and therefore facilitating DUX4 diffusion among them. Defining DUX4c functions in the healthy skeletal muscle should help to design new targeted FSHD therapy by DUX4 or DUX4c inhibition without suppressing DUX4c normal function.


Subject(s)
Homeodomain Proteins/physiology , Muscle Development/physiology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Transcription Factors/physiology , Cell Differentiation/physiology , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Gene Silencing , Homeodomain Proteins/genetics , Humans , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Muscular Dystrophy, Facioscapulohumeral/physiopathology , Myoblasts/metabolism , Phenotype , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transfection , Troponin T/metabolism , Up-Regulation/physiology , beta Catenin/metabolism
2.
J Cell Mol Med ; 17(1): 76-89, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23206257

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most frequent hereditary muscle disorders. It is linked to contractions of the D4Z4 repeat array in 4q35. We have characterized the double homeobox 4 (DUX4) gene in D4Z4 and its mRNA transcribed from the distal D4Z4 unit to a polyadenylation signal in the flanking pLAM region. It encodes a transcription factor expressed in FSHD but not healthy muscle cells which initiates a gene deregulation cascade causing differentiation defects, muscle atrophy and oxidative stress. PITX1 was the first identified DUX4 target and encodes a transcription factor involved in muscle atrophy. DUX4 was found expressed in only 1/1000 FSHD myoblasts. We have now shown it was induced upon differentiation and detected in about 1/200 myotube nuclei. The DUX4 and PITX1 proteins presented staining gradients in consecutive myonuclei which suggested a diffusion as known for other muscle nuclear proteins. Both protein half-lifes were regulated by the ubiquitin-proteasome pathway. In addition, we could immunodetect the DUX4 protein in FSHD muscle extracts. As a model, we propose the DUX4 gene is stochastically activated in a small number of FSHD myonuclei. The resulting mRNAs are translated in the cytoplasm around an activated nucleus and the DUX4 proteins diffuse to adjacent nuclei where they activate target genes such as PITX1. The PITX1 protein can further diffuse to additional myonuclei and expand the transcriptional deregulation cascade initiated by DUX4. Together the diffusion and the deregulation cascade would explain how a rare protein could cause the muscle defects observed in FSHD.


Subject(s)
Homeodomain Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts, Skeletal/metabolism , Paired Box Transcription Factors/metabolism , RNA, Messenger/metabolism , Animals , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cytoplasm/genetics , Cytoplasm/metabolism , Gene Expression Regulation , Half-Life , Homeodomain Proteins/genetics , Humans , Mice , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts, Skeletal/pathology , Paired Box Transcription Factors/genetics , Primary Cell Culture , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA, Messenger/genetics , Signal Transduction
3.
PLoS One ; 6(10): e26820, 2011.
Article in English | MEDLINE | ID: mdl-22053214

ABSTRACT

BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD) is linked to deletions in 4q35 within the D4Z4 repeat array in which we identified the double homeobox 4 (DUX4) gene. We found stable DUX4 mRNAs only derived from the most distal D4Z4 unit and unexpectedly extended to the flanking pLAM region that provided an intron and a polyadenylation signal. DUX4 encodes a transcription factor expressed in FSHD but not control primary myoblasts or muscle biopsies. The DUX4 protein initiates a large transcription deregulation cascade leading to muscle atrophy and oxidative stress, which are FSHD key features. METHODOLOGY/PRINCIPAL FINDINGS: We now show that transfection of myoblasts with a DUX4 expression vector leads to atrophic myotube formation associated with the induction of E3 ubiquitin ligases (MuRF1 and Atrogin1/MAFbx) typical of muscle atrophy. DUX4 induces expression of downstream targets deregulated in FSHD such as mu-crystallin and TP53. We developed specific siRNAs and antisense oligonucleotides (AOs) targeting the DUX4 mRNA. Addition of these antisense agents to primary FSHD myoblast cultures suppressed DUX4 protein expression and affected expression of the above-mentioned markers. CONCLUSIONS/SIGNIFICANCE: These results constitute a proof of concept for the development of therapeutic approaches for FSHD targeting DUX4 expression.


Subject(s)
Homeodomain Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Animals , Biomarkers/metabolism , Cells, Cultured , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Humans , Mice , Models, Biological , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Phenotype , RNA Interference/drug effects , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Transfection , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism
4.
J Biol Chem ; 286(52): 44620-31, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-21937448

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD), a dominant hereditary disease with a prevalence of 7 per 100,000 individuals, is associated with a partial deletion in the subtelomeric D4Z4 repeat array on chromosome 4q. The D4Z4 repeat contains a strong transcriptional enhancer that activates promoters of several FSHD-related genes. We report here that the enhancer within the D4Z4 repeat binds the Krüppel-like factor KLF15. KLF15 was found to be up-regulated during myogenic differentiation induced by serum starvation or by overexpression of the myogenic differentiation factor MYOD. When overexpressed, KLF15 activated the D4Z4 enhancer and led to overexpression of DUX4c (Double homeobox 4, centromeric) and FRG2 (FSHD region gene 2) genes, whereas its silencing caused inactivation of the D4Z4 enhancer. In immortalized human myoblasts, the D4Z4 enhancer was activated by the myogenic factor MYOD, an effect that was abolished upon KLF15 silencing or when the KLF15-binding sites within the D4Z4 enhancer were mutated, indicating that the myogenesis-related activation of the D4Z4 enhancer was mediated by KLF15. KLF15 and several myogenesis-related factors were found to be expressed at higher levels in myoblasts, myotubes, and muscle biopsies from FSHD patients than in healthy controls. We propose that KLF15 serves as a molecular link between myogenic factors and the activity of the D4Z4 enhancer, and it thus contributes to the overexpression of the DUX4c and FRG2 genes during normal myogenic differentiation and in FSHD.


Subject(s)
Chromosomes, Human, Pair 4/metabolism , Enhancer Elements, Genetic , Kruppel-Like Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Nuclear Proteins/metabolism , Animals , Chromosomes, Human, Pair 4/genetics , Cricetinae , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , HeLa Cells , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Muscle Development/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , MyoD Protein/genetics , MyoD Protein/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...