Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
PLoS One ; 12(11): e0186989, 2017.
Article in English | MEDLINE | ID: mdl-29095838

ABSTRACT

Reduced muscle mass due to pathological development can occur through several mechanisms, including the loss or reduced proliferation of muscle stem cells. Muscle-specific ablation of the α-thalassemia mental retardation syndrome mutant protein, Atrx, in transgenic mice results in animals with a severely reduced muscle mass at three weeks of age; yet this muscle mass reduction resolves by adult age. Here, we explore the cellular mechanism underlying this effect. Analysis of Atrx mutant mice included testing for grip strength and rotorod performance. Muscle fiber length, fiber volume and numbers of myofiber-associated nuclei were determined from individual EDL or soleus myofibers isolated at three, five, or eight weeks. Myofibers from three week old Atrx mutant mice are smaller with fewer myofiber-associated nuclei and reduced volume compared to control animals, despite similar fiber numbers. Nonetheless, the grip strength of Atrx mutant mice was comparable to control mice when adjusted for body weight. Myofiber volume remained smaller at five weeks, becoming comparable to controls by 8 weeks of age. Concomitantly, increased numbers of myofiber-associated nuclei and Ki67+ myoblasts indicated that the recovery of muscle mass likely arises from the prolonged accretion of new myonuclei. This suggests that under disease conditions the muscle satellite stem cell niche can remain in a prolonged active state, allowing for the addition of a minimum number of myonuclei required to achieve a normal muscle size.


Subject(s)
Cell Nucleus/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Animals , Mice , Mice, Mutant Strains
2.
J Clin Invest ; 122(12): 4412-23, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23114596

ABSTRACT

ATR-X syndrome is a severe intellectual disability disorder caused by mutations in the ATRX gene. Many ancillary clinical features are attributed to CNS deficiencies, yet most patients have muscle hypotonia, delayed ambulation, or kyphosis, pointing to an underlying skeletal muscle defect. Here, we identified a cell-intrinsic requirement for Atrx in postnatal muscle growth and regeneration in mice. Mice with skeletal muscle-specific Atrx conditional knockout (Atrx cKO mice) were viable, but by 3 weeks of age presented hallmarks of underdeveloped musculature, including kyphosis, 20% reduction in body mass, and 34% reduction in muscle fiber caliber. Atrx cKO mice also demonstrated a marked regeneration deficit that was not due to fewer resident satellite cells or their inability to terminally differentiate. However, activation of Atrx-null satellite cells from isolated muscle fibers resulted in a 9-fold reduction in myoblast expansion, caused by delayed progression through mid to late S phase. While in S phase, Atrx colocalized specifically to late-replicating chromatin, and its loss resulted in rampant signs of genomic instability. These observations support a model in which Atrx maintains chromatin integrity during the rapid developmental growth of a tissue.


Subject(s)
DNA Helicases/genetics , Genomic Instability , Muscle Development , Nuclear Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Helicases/physiology , DNA Replication , DNA-Binding Proteins/metabolism , Female , Histones/metabolism , Humans , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitosis , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Rad51 Recombinase/metabolism , Regeneration/genetics , S Phase Cell Cycle Checkpoints , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/physiology , Telomere/genetics , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , X-linked Nuclear Protein , alpha-Thalassemia/genetics , alpha-Thalassemia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL