Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 29(10): 1607-1623, 2020 06 27.
Article in English | MEDLINE | ID: mdl-32227114

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked disease characterized by progressive muscle degeneration. The condition is driven by nonsense and missense mutations in the dystrophin gene, leading to instability of the sarcolemma and skeletal muscle necrosis and atrophy. Resulting changes in muscle-specific gene expression that take place in dystrophin's absence remain largely uncharacterized, as they are potentially obscured by the chronic inflammation elicited by muscle damage in humans. Caenorhabditis elegans possess a mild inflammatory response that is not active in the muscle, and lack a satellite cell equivalent. This allows for the characterization of the transcriptome rearrangements affecting disease progression independently of inflammation and regeneration. In effort to better understand these dynamics, we have isolated and sequenced body muscle-specific transcriptomes from C. elegans lacking functional dystrophin at distinct stages of disease progression. We have identified an upregulation of genes involved in mitochondrial function early in disease progression, and an upregulation of genes related to muscle repair in later stages. Our results suggest that in C. elegans, dystrophin may have a signaling role early in development, and its absence may activate compensatory mechanisms that counteract muscle degradation caused by loss of dystrophin. We have also developed a temperature-based screening method for synthetic paralysis that can be used to rapidly identify genetic partners of dystrophin. Our results allow for the comprehensive identification of transcriptome changes that potentially serve as independent drivers of disease progression and may in turn allow for the identification of new therapeutic targets for the treatment of DMD.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Transcriptome/genetics , Animals , Caenorhabditis elegans/genetics , Codon, Nonsense/genetics , Disease Models, Animal , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/genetics , Sarcolemma/pathology
2.
Proc Natl Acad Sci U S A ; 116(31): 15716-15724, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31315980

ABSTRACT

In adult skeletal muscles, 2 junctophilin isoforms (JPH1 and JPH2) tether the sarcoplasmic reticulum (SR) to transverse tubule (T-tubule) membranes, generating stable membrane contact sites known as triads. JPHs are anchored to the membrane of the SR by a C-terminal transmembrane domain (TMD) and bind the T-tubule membrane through their cytosolic N-terminal region, which contains 8 lipid-binding (MORN) motifs. By combining expression of GFP-JPH1 deletion mutants in skeletal muscle fibers with in vitro biochemical experiments, we investigated the molecular determinants of JPH1 recruitment at triads in adult skeletal muscle fibers. We found that MORN motifs bind PI(4,5)P2 in the sarcolemma, but do not mediate the selective localization of JPH1 at the T-tubule compartment of triads. On the contrary, fusion proteins containing only the TMD of JPH1 were able to localize at the junctional SR compartment of the triad. Bimolecular fluorescence complementation experiments indicated that the TMD of JPH1 can form dimers, suggesting that the observed localization at triads may result from dimerization with the TMDs of resident JPH1. A second domain, capable of mediating homo- and heterodimeric interactions between JPH1 and JPH2 was identified in the cytosolic region. FRAP experiments revealed that removal of either one of these 2 domains in JPH1 decreases the association of the resulting mutant proteins with triads. Altogether, these results suggest that the ability to establish homo- and heterodimeric interactions with resident JPHs may support the recruitment and stability of newly synthesized JPHs at triads in adult skeletal muscle fibers.


Subject(s)
Membrane Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Sarcolemma/metabolism , Amino Acid Motifs , Animals , Humans , Membrane Proteins/genetics , Mice , Muscle Proteins/genetics , Mutation , Protein Domains , Rats , Rats, Sprague-Dawley , Sarcolemma/genetics
3.
J Cell Sci ; 132(2)2019 01 23.
Article in English | MEDLINE | ID: mdl-30630895

ABSTRACT

Skeletal muscle (SKM) differentiation is a highly regulated process leading to the formation of specialised cells with reorganised compartments and organelles, such as those of the early secretory pathway. During SKM differentiation the Golgi complex (GC) redistributes close to the nuclear envelope and in small distinct peripheral structures distributed throughout the myotube. Concurrently, GC elements closely associate with endoplasmic reticulum-exit sites (ERES). The mechanisms underlying this reorganisation and its relevance for SKM differentiation are poorly understood. Here, we show, by time-lapse imaging studies, that the changes in GC organisation involve GC fragmentation and redistribution of ERES with the formation of tightly associated GC-ERES units. We show that knockdown of GM130 (also known as GOLGA2) or p115 (also known as USO1), two regulators of the early secretory pathway, impairs GC and ERES reorganisation. This in turn results in inhibition of myotube fusion and M-cadherin (also known as CDH15) transport to the sarcolemma. Taken together, our data suggest that the correct reorganisation of the early secretory pathway components plays an important role in SKM differentiation and, thus, associated pathologies.


Subject(s)
Autoantigens/metabolism , Cell Differentiation , Golgi Matrix Proteins/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Sarcolemma/metabolism , Secretory Pathway , Vesicular Transport Proteins/metabolism , Animals , Autoantigens/genetics , Cell Line , Golgi Matrix Proteins/genetics , Membrane Proteins/genetics , Mice , Muscle, Skeletal/cytology , Sarcolemma/genetics , Vesicular Transport Proteins/genetics
4.
Proc Natl Acad Sci U S A ; 115(17): 4507-4512, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29632175

ABSTRACT

Close physical association of CaV1.1 L-type calcium channels (LTCCs) at the sarcolemmal junctional membrane (JM) with ryanodine receptors (RyRs) of the sarcoplasmic reticulum (SR) is crucial for excitation-contraction coupling (ECC) in skeletal muscle. However, the molecular mechanism underlying the JM targeting of LTCCs is unexplored. Junctophilin 1 (JP1) and JP2 stabilize the JM by bridging the sarcolemmal and SR membranes. Here, we examined the roles of JPs in localization and function of LTCCs. Knockdown of JP1 or JP2 in cultured myotubes inhibited LTCC clustering at the JM and suppressed evoked Ca2+ transients without disrupting JM structure. Coimmunoprecipitation and GST pull-down assays demonstrated that JPs physically interacted with 12-aa residues in the proximal C terminus of the CaV1.1. A JP1 mutant lacking the C terminus including the transmembrane domain (JP1ΔCT) interacted with the sarcolemmal/T-tubule membrane but not the SR membrane. Expression of this mutant in adult mouse muscles in vivo exerted a dominant-negative effect on endogenous JPs, impairing LTCC-RyR coupling at triads without disrupting JM morphology, and substantially reducing Ca2+ transients without affecting SR Ca2+ content. Moreover, the contractile force of the JP1ΔCT-expressed muscle was dramatically reduced compared with the control. Taken together, JPs recruit LTCCs to the JM through physical interaction and ensure robust ECC at triads in skeletal muscle.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Membrane Proteins/metabolism , Muscle Contraction/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Cell Line , Membrane Proteins/genetics , Mice , Muscle Proteins/genetics , Protein Domains , Sarcolemma/genetics , Sarcolemma/metabolism
5.
J Struct Biol ; 209(1): 107411, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31689503

ABSTRACT

Dystrophin is a large intracellular protein that prevents sarcolemmal ruptures by providing a mechanical link between the intracellular actin cytoskeleton and the transmembrane dystroglycan complex. Dystrophin deficiency leads to the severe muscle wasting disease Duchenne Muscular Dystrophy and the milder allelic variant, Becker Muscular Dystrophy (DMD and BMD). Previous work has shown that concomitant interaction of the actin binding domain 2 (ABD2) comprising spectrin like repeats 11 to 15 (R11-15) of the central domain of dystrophin, with both actin and membrane lipids, can greatly increase membrane stiffness. Based on a combination of SAXS and SANS measurements, mass spectrometry analysis of cross-linked complexes and interactive low-resolution simulations, we explored in vitro the molecular properties of dystrophin that allow the formation of ABD2-F-actin and ABD2-membrane model complexes. In dystrophin we identified two subdomains interacting with F-actin, one located in R11 and a neighbouring region in R12 and another one in R15, while a single lipid binding domain was identified at the C-terminal end of R12. Relative orientations of the dystrophin central domain with F-actin and a membrane model were obtained from docking simulation under experimental constraints. SAXS-based models were then built for an extended central subdomain from R4 to R19, including ABD2. Overall results are compatible with a potential F-actin/dystrophin/membrane lipids ternary complex. Our description of this selected part of the dystrophin associated complex bridging muscle cell membrane and cytoskeleton opens the way to a better understanding of how cell muscle scaffolding is maintained through this essential protein.


Subject(s)
Dystrophin/ultrastructure , Muscular Dystrophy, Duchenne/genetics , Sarcolemma/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Actins/genetics , Actins/ultrastructure , Dystrophin/genetics , Humans , Lipids/chemistry , Lipids/genetics , Muscular Dystrophy, Duchenne/pathology , Protein Binding , Sarcolemma/ultrastructure , Scattering, Small Angle , Ternary Complex Factors/genetics , Ternary Complex Factors/ultrastructure , X-Ray Diffraction
6.
Hum Mol Genet ; 26(8): 1458-1464, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28175314

ABSTRACT

Peptidyl-tRNA hydrolase 2 (PTRH2) regulates integrin-mediated pro-survival and apoptotic signaling. PTRH2 is critical in muscle development and regulates myogenic differentiation. In humans a biallelic mutation in the PTRH2 gene causes infantile-onset multisystem disease with progressive muscle weakness. We report here that the Ptrh2 knockout mouse model recapitulates the progressive congenital muscle pathology observed in patients. Ptrh2 null mice demonstrate multiple degenerating and regenerating muscle fibers, increased central nuclei, elevated creatine kinase activity and endomysial fibrosis. This progressive muscle pathology resembles the muscular dystrophy phenotype in humans and mice lacking the α7 integrin. We demonstrate that in normal muscle Ptrh2 associates in a complex with the α7ß1 integrin at the sarcolemma and Ptrh2 expression is decreased in α7 integrin null muscle. Furthermore, Ptrh2 expression is altered in skeletal muscle of classical congenital muscular dystrophy mouse models. Ptrh2 levels were up-regulated in dystrophin deficient mdx muscle, which correlates with the elevated levels of the α7ß1 integrin observed in mdx muscle and Duchenne muscular dystrophy patients. Similar to the α7 integrin, Ptrh2 expression was decreased in laminin-α2 dyW null gastrocnemius muscle. Our data establishes a PTRH2 mutation as a novel driver of congenital muscle degeneration and identifies a potential novel target to treat muscle myopathies.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Integrins/genetics , Mitochondrial Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Animals , Carboxylic Ester Hydrolases/biosynthesis , Dystrophin/genetics , Dystrophin/metabolism , Gene Expression Regulation, Developmental , Humans , Integrins/biosynthesis , Mice , Mice, Inbred mdx , Mice, Knockout , Mitochondrial Proteins/biosynthesis , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/genetics , Sarcolemma/pathology
7.
Mol Med ; 25(1): 31, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266455

ABSTRACT

BACKGROUND: Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. METHODS: Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. RESULTS: Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. CONCLUSIONS: Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton.


Subject(s)
Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type I/metabolism , Sarcolemma/metabolism , Utrophin/metabolism , Animals , Humans , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutation/genetics , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/pharmacology , Protein Binding/genetics , Sarcolemma/genetics , Sarcolemma/pathology , Utrophin/genetics
8.
FASEB J ; 32(6): 2950-2965, 2018 06.
Article in English | MEDLINE | ID: mdl-29401588

ABSTRACT

Stimulation of AMPK induces the expression of dystrophin-associated protein complex (DAPC) components in skeletal muscle, whereas reductions in AMPK are associated with DAPC dysfunction. We sought to determine whether AMPK was necessary for the maintenance of DAPC expression in skeletal muscle. Fast, glycolytic extensor digitorum longus (EDL) and slow, oxidative soleus (Sol) muscles from wild-type mice and from littermates with skeletal muscle-specific knockout of the AMPK ß1 and ß2 subunits (AMPK ß1 ß2M-KO; MKO) were analyzed. DAPC mRNA and protein expression were similar between genotypes, with the exception of elevated neuronal nitric oxide synthase expression at the sarcolemma in MKO muscles. The content of transcriptional and post-transcriptional regulators of the DAPC was also not affected by the loss of AMPK. However, MyoD and myogenin expression was diminished in MKO muscles, consistent with previous reports of myopathy in these animals. Furthermore, we observed decrements in extrasynaptic utrophin expression selectively in MKO Sol muscles, likely due to the adaptive accumulation of peroxisome proliferator-activated receptor γ coactivator-1α at the sarcolemma of MKO EDL muscles. Collectively, the evidence indicates that AMPK is sufficient but not essential for the maintenance of DAPC expression in skeletal muscle, yet it is required for preserving extrasynaptic utrophin levels in slow oxidative muscles.-Dial, A. G., Rooprai, P., Lally, J. S., Bujak, A. L., Steinberg, G. R., Ljubicic, V. The role of AMP-activated protein kinase in the expression of the dystrophin-associated protein complex in skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Dystrophin-Associated Proteins/biosynthesis , Gene Expression Regulation , Muscle, Skeletal/metabolism , Sarcolemma/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Dystrophin-Associated Proteins/genetics , Mice , Mice, Knockout , MyoD Protein/genetics , MyoD Protein/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Sarcolemma/genetics
9.
FASEB J ; 32(3): 1613-1625, 2018 03.
Article in English | MEDLINE | ID: mdl-29133341

ABSTRACT

ATP-sensitive K+ (KATP) channels uniquely link cellular energy metabolism to membrane excitability and are expressed in diverse cell types that range from the endocrine pancreas to neurons and smooth, skeletal, and cardiac muscle. A decrease in the surface expression of KATP channels has been linked to various disorders, including dysregulated insulin secretion, abnormal blood pressure, and impaired resistance to cardiac injury. In contrast, up-regulation of KATP channel surface expression may be protective, for example, by mediating the beneficial effect of ischemic preconditioning. Molecular mechanisms that regulate KATP channel trafficking are poorly understood. Here, we used cellular assays with immunofluorescence, surface biotinylation, and patch clamping to demonstrate that Eps15 homology domain-containing protein 2 (EHD2) is a novel positive regulator of KATP channel trafficking to increase surface KATP channel density. EHD2 had no effect on cardiac Na+ channels (Nav1.5). The effect is specific to EHD2 as other members of the EHD family-EHD1, EHD3, and EHD4-had no effect on KATP channel surface expression. EHD2 did not directly affect KATP channel properties as unitary conductance and ATP sensitivity were unchanged. Instead, we observed that the mechanism by which EHD2 increases surface expression is by stabilizing KATP channel-containing caveolar structures, which results in a reduced rate of endocytosis. EHD2 also regulated KATP channel trafficking in isolated cardiomyocytes, which validated the physiologic relevance of these observations. Pathophysiologically, EHD2 may be cardioprotective as a dominant-negative EHD2 mutant sensitized cardiomyocytes to ischemic damage. Our findings highlight EHD2 as a potential pharmacologic target in the treatment of diseases with KATP channel trafficking defects.-Yang, H. Q., Jana, K., Rindler, M. J., Coetzee, W. A. The trafficking protein, EHD2, positively regulates cardiac sarcolemmal KATP channel surface expression: role in cardioprotection.


Subject(s)
Carrier Proteins/metabolism , KATP Channels/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , Animals , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , HEK293 Cells , Humans , KATP Channels/genetics , Mice , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Protein Transport , Rats , Sarcolemma/genetics
10.
J Biol Chem ; 292(40): 16653-16664, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28808062

ABSTRACT

TBC1 domain family member 1 (TBC1D1), a Rab GTPase-activating protein and paralogue of Akt substrate of 160 kDa (AS160), has been implicated in both insulin- and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase-mediated glucose transporter type 4 (GLUT4) translocation. However, the role of TBC1D1 in contracting muscle remains ambiguous. We therefore explored the metabolic consequence of ablating TBC1D1 in both resting and contracting skeletal muscles, utilizing a rat TBC1D1 KO model. Although insulin administration rapidly increased (p < 0.05) plasma membrane GLUT4 content in both red and white gastrocnemius muscles, the TBC1D1 ablation did not alter this response nor did it affect whole-body insulin tolerance, suggesting that TBC1D1 is not required for insulin-induced GLUT4 trafficking events. Consistent with findings in other models of altered TBC1D1 protein levels, whole-animal and ex vivo skeletal muscle fat oxidation was increased in the TBC1D1 KO rats. Although there was no change in mitochondrial content in the KO rats, maximal ADP-stimulated respiration was higher in permeabilized muscle fibers, which may contribute to the increased reliance on fatty acids in resting KO animals. Despite this increase in mitochondrial oxidative capacity, run time to exhaustion at various intensities was impaired in the KO rats. Moreover, contraction-induced increases in sarcolemmal GLUT4 content and glucose uptake were lower in the white gastrocnemius of the KO animals. Altogether, our results highlight a critical role for TBC1D1 in exercise tolerance and contraction-mediated translocation of GLUT4 to the plasma membrane in skeletal muscle.


Subject(s)
Exercise Tolerance/physiology , Glucose Transporter Type 4/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Proteins/metabolism , Sarcolemma/metabolism , Animals , Glucose Transporter Type 4/genetics , Insulin/genetics , Insulin/metabolism , Oxidation-Reduction , Oxygen Consumption/physiology , Protein Transport/physiology , Proteins/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Sarcolemma/genetics
11.
Hum Mol Genet ; 25(24): 5395-5406, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798107

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic disorder that causes progressive muscle weakness, ultimately leading to early mortality in affected teenagers and young adults. Previous work from our lab has shown that a small transmembrane protein called sarcospan (SSPN) can enhance the recruitment of adhesion complex proteins to the cell surface. When human SSPN is expressed at three-fold levels in mdx mice, this increase in adhesion complex abundance improves muscle membrane stability, preventing many of the histopathological changes associated with DMD. However, expressing higher levels of human SSPN (ten-fold transgenic expression) causes a severe degenerative muscle phenotype in wild-type mice. Since SSPN-mediated stabilization of the sarcolemma represents a promising therapeutic strategy in DMD, it is important to determine whether SSPN can be introduced at high levels without toxicity. Here, we show that mouse SSPN (mSSPN) can be overexpressed at 30-fold levels in wild-type mice with no deleterious effects. In mdx mice, mSSPN overexpression improves dystrophic pathology and sarcolemmal stability. We show that these mice exhibit increased resistance to eccentric contraction-induced damage and reduced fatigue following exercise. mSSPN overexpression improved pulmonary function and reduced dystrophic histopathology in the diaphragm. Together, these results demonstrate that SSPN overexpression is well tolerated in mdx mice and improves sarcolemma defects that underlie skeletal muscle and pulmonary dysfunction in DMD.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Neoplasm Proteins/genetics , Sarcolemma/genetics , Animals , Carrier Proteins/biosynthesis , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Lung Diseases/genetics , Lung Diseases/pathology , Membrane Proteins/biosynthesis , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Neoplasm Proteins/biosynthesis , Sarcolemma/pathology
12.
Biochem J ; 474(1): 149-162, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27827305

ABSTRACT

Leptin stimulates fatty acid oxidation in muscle and heart; but, the mechanism by which these tissues provide additional intracellular fatty acids for their oxidation remains unknown. We examined, in isolated muscle and cardiac myocytes, whether leptin, via AMP-activated protein kinase (AMPK) activation, stimulated fatty acid translocase (FAT/CD36)-mediated fatty acid uptake to enhance fatty acid oxidation. In both mouse skeletal muscle and rat cardiomyocytes, leptin increased fatty acid oxidation, an effect that was blocked when AMPK phosphorylation was inhibited by adenine 9-ß-d-arabinofuranoside or Compound C. In wild-type mice, leptin induced the translocation of FAT/CD36 to the plasma membrane and increased fatty acid uptake into giant sarcolemmal vesicles and into cardiomyocytes. In muscles of FAT/CD36-KO mice, and in cardiomyocytes in which cell surface FAT/CD36 action was blocked by sulfo-N-succinimidyl oleate, the leptin-stimulated influx of fatty acids was inhibited; concomitantly, the normal leptin-stimulated increase in fatty acid oxidation was also prevented, despite the normal leptin-induced increase in AMPK phosphorylation. Conversely, in muscle of AMPK kinase-dead mice, leptin failed to induce the translocation of FAT/CD36, along with a failure to stimulate fatty acid uptake and oxidation. Similarly, when siRNA was used to reduce AMPK in HL-1 cardiomyocytes, leptin failed to induce the translocation of FAT/CD36. Our studies have revealed a novel mechanism of leptin-induced fatty acid oxidation in muscle tissue; namely, this process is dependent on the activation of AMPK to induce the translocation of FAT/CD36 to the plasma membrane, thereby stimulating fatty acid uptake. Without increasing this leptin-stimulated, FAT/CD36-dependent fatty acid uptake process, leptin-stimulated AMPK phosphorylation does not enhance fatty acid oxidation.


Subject(s)
CD36 Antigens/metabolism , Fatty Acids/metabolism , Leptin/metabolism , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , CD36 Antigens/genetics , Cell Line , Fatty Acids/genetics , Leptin/genetics , Mice , Mice, Knockout , Oleic Acids/pharmacology , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Protein Transport/drug effects , Rats , Sarcolemma/genetics , Succinimides/pharmacology , Vidarabine/pharmacology
13.
J Biol Chem ; 291(19): 9920-8, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26966179

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and ß-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.


Subject(s)
Cathepsins/biosynthesis , Gene Expression Regulation, Developmental , Muscle Fibers, Skeletal/enzymology , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Duchenne/enzymology , Animals , Cytoskeleton/enzymology , Cytoskeleton/genetics , Cytoskeleton/pathology , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Necrosis , Proteolysis , Sarcolemma/enzymology , Sarcolemma/genetics , Sarcolemma/pathology
14.
Hum Mol Genet ; 24(15): 4212-24, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25935002

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD although various promising approaches are progressing through human clinical trials. By pharmacologically modulating the expression of the dystrophin-related protein utrophin, we have previously demonstrated in dystrophin-deficient mdx studies, daily SMT C1100 treatment significantly reduced muscle degeneration leading to improved muscle function. This manuscript describes the significant disease modifying benefits associated with daily dosing of SMT022357, a second-generation compound in this drug series with improved physicochemical properties and a more robust metabolism profile. These studies in the mdx mouse demonstrate that oral administration of SMT022357 leads to increased utrophin expression in skeletal, respiratory and cardiac muscles. Significantly, utrophin expression is localized along the length of the muscle fibre, not just at the synapse, and is fibre-type independent, suggesting that drug treatment is modulating utrophin transcription in extra-synaptic myonuclei. This results in improved sarcolemmal stability and prevents dystrophic pathology through a significant reduction of regeneration, necrosis and fibrosis. All these improvements combine to protect the mdx muscle from contraction induced damage and enhance physiological function. This detailed evaluation of the SMT C1100 drug series strongly endorses the therapeutic potential of utrophin modulation as a disease modifying therapeutic strategy for all DMD patients irrespective of their dystrophin mutation.


Subject(s)
Dystrophin/biosynthesis , Muscle Fibers, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Utrophin/biosynthesis , Animals , Dystrophin/genetics , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Fibers, Skeletal/pathology , Muscles/drug effects , Muscles/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/drug effects , Sarcolemma/genetics , Utrophin/genetics
15.
J Cell Sci ; 128(2): 219-24, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25413344

ABSTRACT

A fundamental requirement of cells is their ability to transduce and interpret their mechanical environment. This ability contributes to regulation of growth, differentiation and adaptation in many cell types. The intermediate filament (IF) system not only provides passive structural support to the cell, but recent evidence points to IF involvement in active biological processes such as signaling, mechanotransduction and gene regulation. However, the mechanisms that underlie these processes are not well known. Skeletal muscle cells provide a convenient system to understand IF function because the major muscle-specific IF, desmin, is expressed in high abundance and is highly organized. Here, we show that desmin plays both structural and regulatory roles in muscle cells by demonstrating that desmin is required for the maintenance of myofibrillar alignment, nuclear deformation, stress production and JNK-mediated stress sensing. Finite element modeling of the muscle IF system suggests that desmin immediately below the sarcolemma is the most functionally significant. This demonstration of biomechanical integration by the desmin IF system suggests that it plays an active biological role in muscle in addition to its accepted structural role.


Subject(s)
Desmin/metabolism , Intermediate Filaments/metabolism , Muscle, Skeletal/metabolism , Myofibrils/metabolism , Animals , Desmin/genetics , Humans , Intermediate Filaments/ultrastructure , Mechanotransduction, Cellular/genetics , Mice, Knockout , Muscle, Skeletal/ultrastructure , Myofibrils/ultrastructure , Sarcolemma/genetics , Sarcolemma/metabolism , Stress, Mechanical
16.
Biochim Biophys Acta ; 1852(4): 585-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25086336

ABSTRACT

Muscular dystrophies are heterogeneous genetic disorders that share progressive muscle wasting. This may generate partial impairment of motility as well as a dramatic and fatal course. Less than 30 years ago, the identification of the genetic basis of Duchenne muscular dystrophy opened a new era. An explosion of new information on the mechanisms of disease was witnessed, with many thousands of publications and the characterization of dozens of other genetic forms. Genes mutated in muscular dystrophies encode proteins of the plasma membrane and extracellular matrix, several of which are part of the dystrophin-associated complex. Other gene products localize at the sarcomere and Z band, or are nuclear membrane components. In the present review, we focus on muscular dystrophies caused by defects that affect the sarcolemmal and sub-sarcolemmal proteins. We summarize the nature of each disease, the genetic cause, and the pathogenic pathways that may suggest future treatment options. We examine X-linked Duchenne and Becker muscular dystrophies and the autosomal recessive limb-girdle muscular dystrophies caused by mutations in genes encoding sarcolemmal proteins. The mechanism of muscle damage is reviewed starting from disarray of the shock-absorbing dystrophin-associated complex at the sarcolemma and activation of inflammatory response up to the final stages of fibrosis. We trace only a part of the biochemical, physiopathological and clinical aspects of muscular dystrophy to avoid a lengthy list of different and conflicting observations. We attempt to provide a critical synthesis of what we consider important aspects to better understand the disease. In our opinion, it is becoming ever more important to go back to the bedside to validate and then translate each proposed mechanism. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Subject(s)
Genetic Diseases, Inborn , Muscle Proteins , Muscular Dystrophies , Sarcolemma , Animals , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Sarcolemma/genetics , Sarcolemma/metabolism , Sarcolemma/pathology
17.
Expert Rev Mol Med ; 17: e12, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26088790

ABSTRACT

Muscle fibres are very specialised cells with a complex structure that requires a high level of organisation of the constituent proteins. For muscle contraction to function properly, there is a need for not only sarcomeres, the contractile structures of the muscle fibre, but also costameres. These are supramolecular structures associated with the sarcolemma that allow muscle adhesion to the extracellular matrix. They are composed of protein complexes that interact and whose functions include maintaining cell structure and signal transduction mediated by their constituent proteins. It is important to improve our understanding of these structures, as mutations in various genes that code for costamere proteins cause many types of muscular dystrophy. In this review, we provide a description of costameres detailing each of their constituent proteins, such as dystrophin, dystrobrevin, syntrophin, sarcoglycans, dystroglycans, vinculin, talin, integrins, desmin, plectin, etc. We describe as well the diseases associated with deficiency thereof, providing a general overview of their importance.


Subject(s)
Desmin/genetics , Dystroglycans/genetics , Dystrophin/genetics , Muscular Diseases/genetics , Costameres/genetics , Costameres/metabolism , Costameres/ultrastructure , Desmin/chemistry , Desmin/metabolism , Dystroglycans/chemistry , Dystroglycans/metabolism , Dystrophin/chemistry , Dystrophin/metabolism , Dystrophin-Associated Proteins/chemistry , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Gene Expression , Humans , Integrins/chemistry , Integrins/genetics , Integrins/metabolism , Muscle Contraction , Muscular Diseases/metabolism , Muscular Diseases/pathology , Mutation , Plectin/chemistry , Plectin/genetics , Plectin/metabolism , Sarcolemma/genetics , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Sarcomeres/genetics , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Talin/chemistry , Talin/genetics , Talin/metabolism , Vinculin/chemistry , Vinculin/genetics , Vinculin/metabolism
18.
FASEB J ; 27(3): 991-1000, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23159934

ABSTRACT

Malignant hyperthermia (MH) susceptibility has been attributed to a leaky sarcoplasmic reticulum (SR) caused by missense mutations in RYR1 or CACNA1S, and the MH crisis has been attributed solely to massive self-sustaining release of Ca(2+) from SR stores elicited by triggering agents. Here, we show in muscle cells from MH-RyR1(R163C) knock-in mice that increased passive SR Ca(2+) leak causes an enlarged basal influx of sarcolemmal Ca(2+) that results in chronically elevated myoplasmic free Ca(2+) concentration ([Ca(2+)]i) at rest. We discovered that Gd(+3) and GsMTx-4 were more effective than BTP2 or expression of the dominant-negative Orai1(E190Q) in reducing both Ca(2+) entry and [Ca(2+)]i, implicating a non-STIM1/Orai1 SOCE pathway in resetting resting [Ca(2+)]i. Indeed, two nonselective cationic channels, TRPC3 and TRPC6, are overexpressed, and [Na]i is chronically elevated in MH-RyR1(R163C) muscle cells. [Ca(2+)]i and [Na(+)]i are persistently elevated in vivo and further increased by halothane in MH-RyR1(R163C/WT) muscle. These increases are markedly attenuated by local perfusion of Gd(+3) or GsMTx-4 and completely suppressed by dantrolene. These results contribute a new paradigm for understanding MH pathophysiology by demonstrating that nonselective sarcolemmal cation channel activity plays a critical role in causing myoplasmic Ca(2+) and Na(+) overload both at rest and during the MH crisis.-Eltit, J. M., Ding, X., Pessah, I. N., Allen, P. D., Lopez, J. R. Nonspecific sarcolemmal cation channels are critical for the pathogenesis of malignant hyperthermia.


Subject(s)
Calcium Channels/metabolism , Malignant Hyperthermia/metabolism , Muscle Cells/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcolemma/metabolism , Amino Acid Substitution , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels, L-Type , Cations, Divalent/metabolism , Cations, Monovalent/metabolism , Malignant Hyperthermia/genetics , Malignant Hyperthermia/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Muscle Cells/pathology , Mutation, Missense , ORAI1 Protein , Ryanodine Receptor Calcium Release Channel/genetics , Sarcolemma/genetics , Sarcolemma/pathology , Sodium/metabolism , Stromal Interaction Molecule 1
19.
Mol Ther ; 21(3): 520-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319059

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle disease caused by mutations in the DMD gene, with loss of its gene product, dystrophin. Dystrophin helps link integral membrane proteins to the actin cytoskeleton and stabilizes the sarcolemma during muscle activity. We investigated an alternative therapeutic approach to dystrophin replacement by overexpressing human α7 integrin (ITGA7) using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal and cardiac muscle that links the extracellular matrix (ECM) to the actin skeleton. It is modestly upregulated in DMD muscle and has been proposed to be an important modifier of dystrophic symptoms. We delivered rAAV8.MCK.ITGA7 to the lower limb of mdx mice through isolated limb perfusion (ILP) of the femoral artery. We demonstrated ~50% of fibers in the tibialis anterior (TA) and extensor digitorum longus (EDL) overexpressing α7 integrin at the sarcolemma following AAV gene transfer. The increase in ITGA7 in skeletal muscle significantly protected against loss of force following eccentric contraction-induced injury compared with untreated (contralateral) muscles while specific force following tetanic contraction was unchanged. Reversal of additional dystrophic features included reduced Evans blue dye (EBD) uptake and increased muscle fiber diameter. Taken together, this data shows that rAAV8.MCK.ITGA7 gene transfer stabilizes the sarcolemma potentially preserving mdx muscle from further damage. This therapeutic approach demonstrates promise as a viable treatment for DMD with further implications for other forms of muscular dystrophy.


Subject(s)
Antigens, CD/genetics , Dependovirus/genetics , Genetic Vectors , Integrin alpha Chains/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Antigens, CD/metabolism , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Extracellular Matrix/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Integrin alpha Chains/metabolism , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Mutation , Sarcolemma/genetics , Up-Regulation
20.
Proc Natl Acad Sci U S A ; 108(17): 7046-51, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21482776

ABSTRACT

The skeletal muscle dihydropyridine receptor (DHPR) and ryanodine receptor (RyR1) are known to engage a form of conformation coupling essential for muscle contraction in response to depolarization, referred to as excitation-contraction coupling. Here we use WT and Ca(V)1.1 null (dysgenic) myotubes to provide evidence for an unexplored RyR1-DHPR interaction that regulates the transition of the RyR1 between gating and leak states. Using double-barreled Ca(2+)-selective microelectrodes, we demonstrate that the lack of Ca(V)1.1 expression was associated with an increased myoplasmic resting [Ca(2+)] ([Ca(2+)](rest)), increased resting sarcolemmal Ca(2+) entry, and decreased sarcoplasmic reticulum (SR) Ca(2+) loading. Pharmacological control of the RyR1 leak state, using bastadin 5, reverted the three parameters to WT levels. The fact that Ca(2+) sparks are not more frequent in dysgenic than in WT myotubes adds support to the hypothesis that the leak state is a conformation distinct from gating RyR1s. We conclude from these data that this orthograde DHPR-to-RyR1 signal inhibits the transition of gated RyR1s into the leak state. Further, it suggests that the DHPR-uncoupled RyR1 population in WT muscle has a higher propensity to be in the leak conformation. RyR1 leak functions are to keep [Ca(2+)](rest) and the SR Ca(2+) content in the physiological range and thus maintain normal intracellular Ca(2+) homeostasis.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Muscle Fibers, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcolemma/metabolism , Animals , Calcium Channels, L-Type/genetics , Calcium Signaling/drug effects , Halogenated Diphenyl Ethers/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Mice , Mice, Mutant Strains , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Skeletal/cytology , Ryanodine Receptor Calcium Release Channel/genetics , Sarcolemma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL