Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
FASEB J ; 28(5): 2047-61, 2014 May.
Article in English | MEDLINE | ID: mdl-24500923

ABSTRACT

Duchenne muscular dystrophy in boys progresses rapidly to severe impairment of muscle function and death in the second or third decade of life. Current supportive therapy with corticosteroids results in a modest increase in strength as a consequence of a general reduction in inflammation, albeit with potential untoward long-term side effects and ultimate failure of the agent to maintain strength. Here, we demonstrate that alternative approaches that rescue defective autophagy in mdx mice, a model of Duchenne muscular dystrophy, with the use of rapamycin-loaded nanoparticles induce a reproducible increase in both skeletal muscle strength and cardiac contractile performance that is not achievable with conventional oral rapamycin, even in pharmacological doses. This increase in physical performance occurs in both young and adult mice, and, surprisingly, even in aged wild-type mice, which sets the stage for consideration of systemic therapies to facilitate improved cell function by autophagic disposal of toxic byproducts of cell death and regeneration.


Subject(s)
Autophagy/drug effects , Immunosuppressive Agents/administration & dosage , Myocardium/metabolism , Nanoparticles/chemistry , Sirolimus/administration & dosage , Adrenal Cortex Hormones/therapeutic use , Animals , Cell Death , Creatine Kinase/metabolism , Drug Delivery Systems , Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Strength , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Myocardial Contraction , Regeneration , Tissue Distribution
2.
Autophagy ; 9(12): 2115-25, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24184927

ABSTRACT

Colchicine treatment is associated with an autophagic vacuolar myopathy in human patients. The presumed mechanism of colchicine-induced myotoxicity is the destabilization of the microtubule system that leads to impaired autophagosome-lysosome fusion and the accumulation of autophagic vacuoles. Using the MTOR inhibitor rapamycin we augmented colchicine's myotoxic effect by increasing the autophagic flux; this resulted in an acute myopathy with muscle necrosis. In contrast to myonecrosis induced by cardiotoxin, myonecrosis induced by a combination of rapamycin and colchicine was associated with accumulation of autophagic substrates such as LC3-II and SQSTM1; as a result, autophagic vacuoles accumulated in the center of myofibers, where LC3-positive autophagosomes failed to colocalize with the lysosomal protein marker LAMP2. A similar pattern of central LC3 accumulation and myonecrosis is seen in human patients with colchicine myopathy, many of whom have been treated with statins (HMGCR/HMG-CoA reductase inhibitors) in addition to colchicine. In mice, cotreatment with colchicine and simvastatin also led to muscle necrosis and LC3 accumulation, suggesting that, like rapamycin, simvastatin activates autophagy. Consistent with this, treatment of mice with four different statin medications enhanced autophagic flux in skeletal muscle in vivo. Polypharmacy is a known risk factor for toxic myopathies; our data suggest that some medication combinations may simultaneously activate upstream autophagy signaling pathways while inhibiting the degradation of these newly synthesized autophagosomes, resulting in myotoxicity.


Subject(s)
Autophagy/physiology , Colchicine/pharmacology , Lysosomal Storage Diseases/chemically induced , Muscular Diseases/chemically induced , Animals , Autophagy/drug effects , Disease Progression , Humans , Lysosomal Storage Diseases/pathology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/pathology , Phagosomes/drug effects , Phagosomes/pathology , Simvastatin/pharmacology , Up-Regulation/drug effects , Vacuoles/drug effects , Vacuoles/pathology
3.
PLoS One ; 8(6): e66027, 2013.
Article in English | MEDLINE | ID: mdl-23776597

ABSTRACT

OBJECTIVE: The glucose and dehydroascorbic acid (DHA) transporter GLUT1 contains a phosphorylation site, S490, for ataxia telangiectasia mutated (ATM). The objective of this study was to determine whether ATM and GLUT1-S490 regulate GLUT1. RESEARCH DESIGN AND METHODS: L6 myoblasts and mouse skeletal muscles were used to study the effects of ATM inhibition, ATM activation, and S490 mutation on GLUT1 localization, trafficking, and transport activity. RESULTS: In myoblasts, inhibition of ATM significantly diminished cell surface GLUT1, glucose and DHA transport, GLUT1 externalization, and association of GLUT1 with Gα-interacting protein-interacting protein, C-terminus (GIPC1), which has been implicated in recycling of endosomal proteins. In contrast, ATM activation by doxorubicin (DXR) increased DHA transport, cell surface GLUT1, and the GLUT1/GIPC1 association. S490A mutation decreased glucose and DHA transport, cell surface GLUT1, and interaction of GLUT1 with GIPC1, while S490D mutation increased transport, cell surface GLUT1, and the GLUT1/GIPC1 interaction. ATM dysfunction or ATM inhibition reduced DHA transport in extensor digitorum longus (EDL) muscles and decreased glucose transport in EDL and soleus. In contrast, DXR increased DHA transport in EDL. CONCLUSIONS: These results provide evidence that ATM and GLUT1-S490 promote cell surface GLUT1 and GLUT1-mediated transport in skeletal muscle associated with upregulation of the GLUT1/GIPC1 interaction.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Glucose Transporter Type 1/metabolism , Muscle, Skeletal/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line , Glucose Transporter Type 1/genetics , Immunoprecipitation , In Vitro Techniques , Mice , Phosphorylation/genetics , Phosphorylation/physiology , Protein Binding , Protein Transport/genetics , Protein Transport/physiology , Rats
4.
Autophagy ; 9(5): 799-800, 2013 May.
Article in English | MEDLINE | ID: mdl-23439279

ABSTRACT

Pathological phenotypes in inclusion body myopathy (IBM) associated with Paget disease of the bone (PDB), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) (IBMPFD/ALS) include defective autophagosome and endosome maturation that result in vacuolation, weakness and muscle atrophy. The link between autophagy and IBMPFD/ALS pathobiology has been poorly understood. We examined the AKT-FOXO3 and MTOR pathways to characterize the regulation of autophagy in IBMPFD/ALS mouse muscle. We identified a defect in MTOR signaling that results in enhanced autophagosome biogenesis. Modulating MTOR signaling may therefore be a viable therapeutic target in IBMPFD/ALS.


Subject(s)
Adenosine Triphosphatases/metabolism , Autophagy/drug effects , Cell Cycle Proteins/metabolism , Muscle Weakness/metabolism , Muscle Weakness/pathology , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Sirolimus/pharmacology , Animals , Disease Models, Animal , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Humans , Mice , Mutant Proteins/metabolism , Osteitis Deformans/metabolism , Osteitis Deformans/pathology , Phagosomes/drug effects , Phagosomes/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Valosin Containing Protein
5.
Hum Mol Genet ; 22(6): 1167-79, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23250913

ABSTRACT

Autophagy is dysfunctional in many degenerative diseases including myopathies. Mutations in valosin-containing protein (VCP) cause inclusion body myopathy (IBM) associated with Paget's disease of the bone, fronto-temporal dementia and amyotrophic lateral sclerosis (IBMPFD/ALS). VCP is necessary for protein degradation via the proteasome and lysosome. IBMPFD/ALS mutations in VCP disrupt autophagosome and endosome maturation resulting in vacuolation, weakness and muscle atrophy. To understand the regulation of autophagy in VCP-IBM muscle, we examined the AKT/FOXO3 and mammalian target of rapamycin (mTOR) pathways. Basal Akt and FOXO3 phosphorylation was normal. In contrast, the phosphorylation of mTOR targets was decreased. Consistent with this, global protein translation was diminished and autophagosome biogenesis was increased in VCP-IBM muscle. Further mTORC1 inhibition with rapamycin hastened weakness, atrophy and vacuolation in VCP-IBM mice. This was accompanied by the accumulation of autophagic substrates such as p62, LC3II and ubiquitinated proteins. The decrease in mTOR signaling was partially rescued by insulin and to a lesser extent by amino acid (AA) stimulation in VCP-IBM muscle. Cells expressing catalytically inactive VCP or treated with a VCP inhibitor also failed to activate mTOR upon nutrient stimulation. Expression of a constitutively active Rheb enhanced mTOR activity and increased the fiber size in VCP-IBM mouse skeletal muscle. These studies suggest that VCP mutations may disrupt mTOR signaling and contribute to IBMPFD/ALS disease pathogenesis. Treatment of some autophagic disorders with mTOR inhibitors such as rapamycin may worsen disease.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Myositis, Inclusion Body/metabolism , TOR Serine-Threonine Kinases/metabolism , Vacuoles/metabolism , Adenosine Triphosphatases/genetics , Animals , Autophagy , Cell Cycle Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/pathology , Myositis, Inclusion Body/physiopathology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Vacuoles/pathology , Valosin Containing Protein
6.
Am J Physiol Endocrinol Metab ; 303(8): E983-93, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22895779

ABSTRACT

Hyperammonemia and sarcopenia (loss of skeletal muscle) are consistent abnormalities in cirrhosis and portosystemic shunting. We have shown that muscle ubiquitin-proteasome components are not increased with hyperammonemia despite sarcopenia. This suggests that an alternative mechanism of proteolysis contributes to sarcopenia in cirrhosis. We hypothesized that autophagy could be this alternative pathway since we observed increases in classic autophagy markers, increased LC3 lipidation, beclin-1 expression, and p62 degradation in immunoblots of skeletal muscle protein in cirrhotic patients. We observed similar changes in these autophagy markers in the portacaval anastamosis (PCA) rat model. To determine the mechanistic relationship between hyperammonemia and autophagy, we exposed murine C(2)C(12) myotubes to ammonium acetate. Significant increases in LC3 lipidation, beclin-1 expression, and p62 degradation occurred by 1 h, whereas autophagy gene expression (LC3, Atg5, Atg7, beclin-1) increased at 24 h. C(2)C(12) cells stably expressing GFP-LC3 or GFP-mCherry-LC3 constructs showed increased formation of mature autophagosomes supported by electron microscopic studies. Hyperammonemia also increased autophagic flux in mice, as quantified by an in vivo autophagometer. Because hyperammonemia induces nitration of proteins in astrocytes, we quantified global muscle protein nitration in cirrhotic patients, in the PCA rat, and in C(2)C(12) cells treated with ammonium acetate. Increased protein nitration was observed in all of these systems. Furthermore, colocalization of nitrated proteins with GFP-LC3-positive puncta in hyperammonemic C(2)C(12) cells suggested that autophagy is involved in degradation of nitrated proteins. These observations show that increased skeletal muscle autophagy in cirrhosis is mediated by hyperammonemia and may contribute to sarcopenia of cirrhosis.


Subject(s)
Autophagy/physiology , Hyperammonemia/pathology , Liver Cirrhosis/pathology , Muscle, Skeletal/pathology , Sarcopenia/pathology , Animals , Cell Line , Cells, Cultured , Fluorescent Antibody Technique , Humans , Hydrogen-Ion Concentration , Male , Mice , Microscopy, Confocal , Microscopy, Electron , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Muscle Proteins/metabolism , Portacaval Shunt, Surgical , Proteasome Endopeptidase Complex/metabolism , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Transfection , Tyrosine/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...