Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Anat ; 223(5): 525-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24010829

ABSTRACT

Costameres are mechano-sensory sites of focal adhesion in the sarcolemma that provide a structural anchor for myofibrils. Their turnover is regulated by integrin-associated focal adhesion kinase (FAK). We hypothesized that changes in content of costamere components (beta 1 integrin, FAK, meta-vinculin, gamma-vinculin) with increased and reduced loading of human anti-gravity muscle would: (i) relate to changes in muscle size and molecular parameters of muscle size regulation [p70S6K, myosin heavy chain (MHC)1 and MHCIIA]; (ii) correspond to adjustments in activity and expression of FAK, and its negative regulator, FRNK; and (iii) reflect the temporal response to reduced and increased loading. Unloading induced a progressive decline in thickness of human vastus lateralis muscle after 8 and 34 days of bedrest (-4% and -14%, respectively; n = 9), contrasting the increase in muscle thickness after 10 and 27 days of resistance training (+5% and +13%; n = 6). Changes in muscle thickness were correlated with changes in cross-sectional area of type I muscle fibers (r = 0.66) and beta 1 integrin content (r = 0.76) at the mid-point of altered loading. Changes in meta-vinculin and FAK-pY397 content were correlated (r = 0.85) and differed, together with the changes of beta 1 integrin, MHCI, MHCII and p70S6K, between the mid- and end-point of resistance training. By contrast, costamere protein level changes did not differ between time points of bedrest. The findings emphasize the role of FAK-regulated costamere turnover in the load-dependent addition and removal of myofibrils, and argue for two phases of muscle remodeling with resistance training, which do not manifest at the macroscopic level.


Subject(s)
Costameres/physiology , Exercise/physiology , Quadriceps Muscle/physiology , Rest/physiology , Adult , Analysis of Variance , Cytoskeletal Proteins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Male , Muscle Fibers, Skeletal/physiology , Quadriceps Muscle/anatomy & histology , Young Adult
2.
Cardiovasc Pathol ; 22(3): 219-27, 2013.
Article in English | MEDLINE | ID: mdl-23266222

ABSTRACT

INTRODUCTION: The mechanical environment is a key regulator of function in cardiomyocytes. We studied the role of substrate stiffness on the organization of sarcomeres and costameres in adult rat cardiomyocytes and further examined the resulting changes in cell shortening and calcium dynamics. METHODS: Cardiomyocytes isolated from adult rats were plated on laminin-coated polydimethylsiloxane substrates of defined stiffness (255 kPa, 117 kPa, 27 kPa, and 7 kPa) for 48 h. Levels of α-actinin and ß1 integrins were determined by immunofluoresence imaging and immunoblotting, both in the absence and presence of the phosphatase inhibitor calyculin A. Quantitative reverse transcriptase polymerase chain reaction was used to measure message levels of key structural proteins (α-actinin, α7 integrin, ß1 integrin, vinculin). Sarcomere shortening and calcium dynamics were measured at 2, 24, and 48 h. RESULTS: Overall cardiomyocyte morphology was similar on all substrates. However, well organized sarcomere structures were observed on only the stiffest (255 kPa) and the most compliant (7 kPa) substrates. Levels of α-actinin in cells were the same on all substrates, while message levels of structural proteins were up-regulated on substrates of intermediate stiffness. Inhibition of phosphatase activity blocked the degradation of contractile structures, but altered overall cardiomyocyte morphology. Shortening and calcium dynamics also were dependent on substrate stiffness; however, there was no clear causative relationship between the phenomena. CONCLUSIONS: Extracellular matrix stiffness can affect structural remodeling by adult cardiomyocytes, and the resulting contractile activity. These findings illuminate changes in cardiomyocyte function in cardiac fibrosis, and may suggest cardiac-specific phosphatases as a target for therapeutic intervention.


Subject(s)
Costameres/physiology , Extracellular Matrix , Mechanical Phenomena , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Adaptation, Physiological/physiology , Animals , Cells, Cultured , Costameres/ultrastructure , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Immunoblotting , Myocytes, Cardiac/ultrastructure , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sarcomeres/ultrastructure
3.
Circ Res ; 109(3): 262-71, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21659647

ABSTRACT

RATIONALE: The proper function of cardiac muscle requires the precise assembly and interactions of numerous cytoskeletal and regulatory proteins into specialized structures that orchestrate contraction and force transmission. Evidence suggests that posttranscriptional regulation is critical for muscle function, but the mechanisms involved remain understudied. OBJECTIVE: To investigate the molecular mechanisms and targets of the muscle-specific fragile X mental retardation, autosomal homolog 1 (FXR1), an RNA binding protein whose loss leads to perinatal lethality in mice and cardiomyopathy in zebrafish. METHODS AND RESULTS: Using RNA immunoprecipitation approaches we found that desmoplakin and talin2 mRNAs associate with FXR1 in a complex. In vitro assays indicate that FXR1 binds these mRNA targets directly and represses their translation. Fxr1 KO hearts exhibit an up-regulation of desmoplakin and talin2 proteins, which is accompanied by severe disruption of desmosome as well as costamere architecture and composition in the heart, as determined by electron microscopy and deconvolution immunofluorescence analysis. CONCLUSIONS: Our findings reveal the first direct mRNA targets of FXR1 in striated muscle and support translational repression as a novel mechanism for regulating heart muscle development and function, in particular the assembly of specialized cytoskeletal structures.


Subject(s)
Desmoplakins/genetics , Myocytes, Cardiac/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Talin/genetics , Animals , COS Cells , Chlorocebus aethiops , Costameres/pathology , Costameres/physiology , Costameres/ultrastructure , Desmoplakins/metabolism , Desmosomes/pathology , Desmosomes/physiology , Desmosomes/ultrastructure , Humans , In Situ Hybridization, Fluorescence , Intermediate Filaments/pathology , Intermediate Filaments/physiology , Intermediate Filaments/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Myofibrils/pathology , Myofibrils/physiology , Myofibrils/ultrastructure , Protein Biosynthesis/physiology , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/metabolism , Sarcomeres/pathology , Sarcomeres/physiology , Sarcomeres/ultrastructure , Talin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...