Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Cell Mol Life Sci ; 74(19): 3439-3449, 2017 10.
Article in English | MEDLINE | ID: mdl-28444416

ABSTRACT

Skeletal muscle is a highly oxygen-consuming tissue that ensures body support and movement, as well as nutrient and temperature regulation. DNA damage induced by reactive oxygen species is present in muscles and tends to accumulate with age. Here, we present a summary of data obtained on DNA damage and its implication in muscle homeostasis, myogenic differentiation and neuromuscular disorders. Controlled and transient DNA damage appears to be essential for muscular homeostasis and differentiation while uncontrolled and chronic DNA damage negatively affects muscle health.


Subject(s)
DNA Damage , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Neuromuscular Diseases/genetics , Oxidative Stress , Aging , Animals , Antioxidants/therapeutic use , DNA/genetics , DNA Damage/drug effects , DNA Repair/drug effects , Humans , Muscle Development/drug effects , Muscle, Skeletal/metabolism , Neuromuscular Diseases/drug therapy , Neuromuscular Diseases/pathology , Oxidative Stress/drug effects
2.
J Cell Physiol ; 231(1): 62-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26218298

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is a neuromuscular disease with a prevalence that could reach 1 in 8,000 characterized by progressive asymmetric muscle weakness. Myoblasts isolated from FSHD muscles exhibit morphological differentiation defects and show a distinct transcription profile. These abnormalities may be linked to the muscle weakness in FSHD patients. We have tested whether fusion of FSHD myoblasts with primary myoblasts isolated from healthy individuals could correct the differentiation defects. Our results show that the number of hybrid myotubes with normal phenotype increased with the percentage of normal myoblasts initially cultured. We demonstrated that a minimum of 50% of normal nuclei is required for a phenotypic correction of the FSHD phenotype. Moreover, transcriptomic profiles of phenotypically corrected hybrid myotubes showed that the expression of deregulated genes in FSHD myotubes became almost normal. The number of deregulated pathways also decreased from 39 in FSHD myotubes to one in hybrid myotubes formed with 40% FSHD and 60% normal myoblasts. We thus propose that while phenotypical and functional correction of FSHD is feasible, it requires more than 50% of normal myoblasts, it creates limitations for cell therapy in the FSHD context.


Subject(s)
Cell Differentiation/physiology , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts/cytology , Adult , Cell Differentiation/genetics , Cells, Cultured , Female , Humans , Male , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Phenotype , Young Adult
3.
Histochem Cell Biol ; 145(4): 475-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26860865

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is a progressive muscular dystrophy linked to a deletion of a subset of D4Z4 macrosatellite repeats accompanied by a chromatin relaxation of the D4Z4 array on chromosome 4q. In vitro, FSHD primary myoblasts show altered expression of oxidative-related genes and are more susceptible to oxidative stress. Double homeobox 4 (DUX4) gene, encoded within each D4Z4 unit, is normally transcriptionally silenced but is found aberrantly expressed in skeletal muscles of FSHD patients. Its expression leads to a deregulation of DUX4 target genes including those implicated in redox balance. Here, we assessed DNA repair efficiency of oxidative DNA damage in FSHD myoblasts and DUX4-transfected myoblasts. We have shown that the DNA repair activity is altered neither in FSHD myoblasts nor in immortalized human myoblasts transiently expressing DUX4. DNA damage caused by moderate doses of an oxidant is efficiently repaired while FSHD myoblasts exposed for 24 h to high levels of oxidative stress accumulated more DNA damage than normal myoblasts, suggesting that FSHD myoblasts remain more vulnerable to oxidative stress at high doses of oxidants.


Subject(s)
DNA Damage , DNA Repair , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts, Skeletal/metabolism , Oxidative Stress , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/pathology , Oxidative Stress/drug effects
4.
Mol Ther Methods Clin Dev ; 5: 43-50, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28480303

ABSTRACT

Despite rapid progress, many problems and limitations persist and limit the applicability of gene-editing techniques. Making use of meganucleases, TALENs, or CRISPR/Cas9-based tools requires an initial step of pre-screening to determine the efficiency and specificity of the designed tools. This step remains time consuming and material consuming. Here we propose a simple, cheap, reliable, time-saving, and highly sensitive method to evaluate a given gene-editing tool based on its capacity to induce chromosomal translocations when combined with a reference engineered nuclease. In the proposed technique, designated engineered nuclease-induced translocations (ENIT), a plasmid coding for the DNA-editing tool to be tested is co-transfected into carefully chosen target cells along with that for an engineered nuclease of known specificity and efficiency. If the new enzyme efficiently cuts within the desired region, then specific chromosomal translocations will be generated between the two targeted genomic regions and be readily detectable by a one-step PCR or qPCR assay. The PCR product thus obtained can be directly sequenced, thereby determining the exact position of the double-strand breaks induced by the gene-editing tools. As a proof of concept, ENIT was successfully tested in different cell types and with different meganucleases, TALENs, and CRISPR/Cas9-based editing tools.

5.
PeerJ ; 5: e4144, 2017.
Article in English | MEDLINE | ID: mdl-29250470

ABSTRACT

BACKGROUND AND PURPOSE: Oxidative stress is involved in normal and pathological functioning of skeletal muscle. Protection of myoblasts from oxidative stress may improve muscle contraction and delay aging. Here we studied the effect of R. coriaria sumac fruit extract on human myoblasts and zebrafish embryos in conditions of hydrogen peroxide-induced oxidative stress. STUDY DESIGN AND METHODS: Crude ethanolic 70% extract (CE) and its fractions was obtained from sumac fruits. The composition of sumac ethyl acetate EtOAc fraction was studied by 1H NMR. The viability of human myoblasts treated with CE and the EtOAc fraction was determined by trypan blue exclusion test. Oxidative stress, cell cycle and adhesion were analyzed by flow cytometry and microscopy. Gene expression was analyzed by qPCR. RESULTS: The EtOAc fraction (IC50 2.57 µg/mL) had the highest antioxidant activity and exhibited the best protective effect against hydrogen peroxide-induced oxidative stress. It also restored cell adhesion. This effect was mediated by superoxide dismutase 2 and catalase. Pre-treatment of zebrafish embryos with low concentrations of the EtOAc fraction protected them from hydrogen peroxide-induced death in vivo. 1H NMR analysis revealed the presence of gallic acid in this fraction. CONCLUSION: Rhus coriaria extracts inhibited or slowed down the progress of skeletal muscle atrophy by decreasing oxidative stress via superoxide dismutase 2 and catalase-dependent mechanisms.

6.
Free Radic Biol Med ; 99: 244-258, 2016 10.
Article in English | MEDLINE | ID: mdl-27519269

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is one of the three most common muscular dystrophies in the Western world, however, its etiology remains only partially understood. Here, we provide evidence of constitutive DNA damage in in vitro cultured myoblasts isolated from FSHD patients and demonstrate oxidative DNA damage implication in the differentiation of these cells into phenotypically-aberrant myotubes. Double homeobox 4 (DUX4), the major actor in FSHD pathology induced DNA damage accumulation when overexpressed in normal human myoblasts, and RNAi-mediated DUX4 inhibition reduced the level of DNA damage in FSHD myoblasts. Addition of tempol, a powerful antioxidant, to the culture medium of proliferating DUX4-transfected myoblasts and FSHD myoblasts reduced the level of DNA damage, suggesting that DNA alterations are mainly due to oxidative stress. Antioxidant treatment during the myogenic differentiation of FSHD myoblasts significantly reduced morphological defects in myotube formation. We propose that the induction of DNA damage is a novel function of the DUX4 protein affecting myogenic differentiation of FSHD myoblasts.


Subject(s)
Homeodomain Proteins/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Myoblasts/metabolism , Oxidative Stress , Antioxidants/pharmacology , Case-Control Studies , Cell Differentiation , Cyclic N-Oxides/pharmacology , DNA Damage , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/metabolism , Humans , Molecular Sequence Annotation , Multigene Family , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spin Labels , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL