Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Clin Rheumatol ; 39(3): 689-696, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31960207

ABSTRACT

INTRODUCTION: Liberia has no rheumatology providers for the nation's 4.7 million people. We proposed a short course format rheumatology curriculum to educate Liberian providers as an initial step in providing graduate medical education in musculoskeletal health. METHOD: A 1-week training curriculum in rheumatology encompassing introduction to musculoskeletal exam and approach to rheumatology diagnosis and management was designed. The curriculum used multiple education methods including interactive lectures, bedside training, and hands-on learning. RESULTS: A 1-week rheumatology training curriculum for 24 local physicians was feasible. The execution of the designed rheumatology curriculum in Liberia relied upon a mixed method format that was both didactic and case-based. A survey of the Liberian trainees revealed that the curriculum was salient to care of patients and barriers to optimal learning such as time and space limitations were identified. CONCLUSIONS: A 1-week rheumatology training education program is possible and relevant to local providers, but training length and setting may need to be optimized. Future training will aim to minimize barriers to education and expand the cohort of providers with rheumatologic knowledge in Liberia.Key Points• Liberia, like many nations in sub-Saharan Africa, has no trained rheumatologists to serve the nation's population.• Education and capacity building for rheumatologic care in short course format are relevant and feasible to local health-care providers.• Further efforts are needed to develop and evaluate continuing rheumatology education in Liberia.


Subject(s)
Capacity Building/methods , Curriculum , Rheumatologists/supply & distribution , Rheumatology/education , Education, Medical, Graduate/methods , Humans , Liberia , Surveys and Questionnaires
2.
Neurology ; 93(12): e1193-e1204, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31434690

ABSTRACT

OBJECTIVE: Activation of the type 1 interferon (IFN1) pathway is a prominent feature of dermatomyositis (DM) muscle and may play a role in the pathogenesis of this disease. However, the relevance of the IFN1 pathway in patients with other types of myositis such as the antisynthetase syndrome (AS), immune-mediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM) is largely unknown. Moreover, the activation of the type 2 interferon (IFN2) pathway has not been comprehensively explored in myositis. In this cross-sectional study, our objective was to determine whether IFN1 and IFN2 pathways are differentially activated in different types of myositis by performing RNA sequencing on muscle biopsy samples from 119 patients with DM, IMNM, AS, or IBM and on 20 normal muscle biopsies. METHODS: The expression of IFN1- and IFN2-inducible genes was compared between the different groups. RESULTS: The expression of IFN1-inducible genes was high in DM, moderate in AS, and low in IMNM and IBM. In contrast, the expression of IFN2-inducible genes was high in DM, IBM, and AS but low in IMNM. The expression of IFN-inducible genes correlated with the expression of genes associated with inflammation and muscle regeneration. Of note, ISG15 expression levels alone performed as well as composite scores relying on multiple genes to monitor activation of the IFN1 pathway in myositis muscle biopsies. CONCLUSIONS: IFN1 and IFN2 pathways are differentially activated in different forms of myositis. This observation may have therapeutic implications because immunosuppressive medications may preferentially target each of these pathways.


Subject(s)
Interferon Type I/genetics , Interferon-gamma/genetics , Muscle, Skeletal/metabolism , Myositis/genetics , Myositis/metabolism , Female , Gene Expression , Humans , Interferon Type I/biosynthesis , Interferon-gamma/biosynthesis , Male , Muscle, Skeletal/pathology , Myositis/pathology
3.
Arthritis Rheumatol ; 71(8): 1371-1376, 2019 08.
Article in English | MEDLINE | ID: mdl-30861336

ABSTRACT

OBJECTIVE: Although more than a dozen myositis-specific autoantibodies (MSAs) have been identified, most patients with myositis are positive for a single MSA. The specific overexpression of a given myositis autoantigen in myositis muscle has been proposed as initiating and/or propagating autoimmunity against that particular autoantigen. The present study was undertaken to test this hypothesis. METHODS: In order to quantify autoantigen RNA expression, RNA sequencing was performed on muscle biopsy samples from control subjects, MSA-positive patients with myositis, regenerating mouse muscles, and cultured human muscle cells. RESULTS: Muscle biopsy samples were available from 20 control subjects and 106 patients with autoantibodies recognizing hydroxymethylglutaryl-coenzyme A reductase (n = 40), signal recognition particles (n = 9), Jo-1 (n = 18), nuclear matrix protein 2 (n = 12), Mi-2 (n = 11), transcription intermediary factor 1γ (n = 11), or melanoma differentiation-associated protein 5 (n = 5). The increased expression of a given autoantigen in myositis muscle was not associated with autoantibodies recognizing that autoantigen (all q > 0.05). In biopsy specimens from both myositis muscle and regenerating mouse muscles, autoantigen expression correlated directly with the expression of muscle regeneration markers and correlated inversely with the expression of genes encoding mature muscle proteins. Myositis autoantigens were also expressed at high levels in cultured human muscle cells. CONCLUSION: Most myositis autoantigens are highly expressed during muscle regeneration, which may relate to the propagation of autoimmunity. However, factors other than overexpression of specific autoantigens are likely to govern the development of unique autoantibodies in individual patients with myositis.


Subject(s)
Autoantibodies/immunology , Autoantigens/metabolism , Muscle, Skeletal/immunology , Myositis/immunology , Regeneration/immunology , Animals , Autoantigens/immunology , Biopsy , Cells, Cultured , Humans , Mice , Myoblasts/immunology , Myoblasts/metabolism , Myositis/physiopathology , RNA/immunology , RNA/metabolism
4.
Semin Arthritis Rheum ; 47(6): 858-864, 2018 06.
Article in English | MEDLINE | ID: mdl-29174792

ABSTRACT

OBJECTIVE: To investigate in a pilot study the safety and efficacy of infliximab in patients with refractory dermatomyositis (DM) and polymyositis (PM). METHODS: A randomized, double-blind, placebo-controlled trial including subjects with active DM or PM. Participants had stable doses of immunosuppressive medication and prednisone (≤0.5mg/kg/day), and exhibited clinical signs of muscle weakness for at least 4 weeks prior to study entry. Participants received infusions of either placebo or infliximab 5mg/kg at 0, 2, 6, and 14 weeks in blinded manner. The primary outcome was a ≥15% manual muscle strength (MMT) improvement at week 16 compared to week 0. The secondary outcome measures were improvement defined by the International Myositis Assessment and Clinical Studies Group (IMACS) criteria. At week 16, responders in each arm had the option of either continuing the same treatment or changing to the non-responder treatment for that study arm. Non-responders in the 5mg/kg infliximab arm were increased to infliximab 7.5mg/kg for weeks 22, 30, and 38. Non-responders in the placebo arm at week 16 received infliximab 5mg/kg at weeks 16, 18, 22, 30, and 38. Outcomes were reassessed at week 40. RESULTS: Twelve subjects completed the study to week 16. Six of the 12 subjects received infliximab treatment at the dose of 5mg/kg with only one subject meeting the responder criteria at that dose. Of the remaining five subjects on infliximab, three crossed over to the infliximab 7.5mg/kg dose. One of those three subjects responded. All six patients in the placebo arm crossed over to the 5mg/kg dosing regimen after week 16, and two of those responded to infliximab. CONCLUSIONS: Infliximab therapy for patients with refractory PM and DM was well tolerated and may benefit a subset of patients.


Subject(s)
Dermatologic Agents/therapeutic use , Dermatomyositis/drug therapy , Immunosuppressive Agents/therapeutic use , Infliximab/therapeutic use , Adult , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Pilot Projects , Prednisone/therapeutic use , Treatment Outcome
6.
J Neuromuscul Dis ; 3(2): 209-225, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27854218

ABSTRACT

BACKGROUND: Molecular diagnostics in the genetic myopathies often requires testing of the largest and most complex transcript units in the human genome (DMD, TTN, NEB). Iteratively targeting single genes for sequencing has traditionally entailed high costs and long turnaround times. Exome sequencing has begun to supplant single targeted genes, but there are concerns regarding coverage and needed depth of the very large and complex genes that frequently cause myopathies. OBJECTIVE: To evaluate efficiency of next-generation sequencing technologies to provide molecular diagnostics for patients with previously undiagnosed myopathies. METHODS: We tested a targeted re-sequencing approach, using a 45 gene emulsion PCR myopathy panel, with subsequent sequencing on the Illumina platform in 94 undiagnosed patients. We compared the targeted re-sequencing approach to exome sequencing for 10 of these patients studied. RESULTS: We detected likely pathogenic mutations in 33 out of 94 patients with a molecular diagnostic rate of approximately 35%. The remaining patients showed variants of unknown significance (35/94 patients) or no mutations detected in the 45 genes tested (26/94 patients). Mutation detection rates for targeted re-sequencing vs. whole exome were similar in both methods; however exome sequencing showed better distribution of reads and fewer exon dropouts. CONCLUSIONS: Given that costs of highly parallel re-sequencing and whole exome sequencing are similar, and that exome sequencing now takes considerably less laboratory processing time than targeted re-sequencing, we recommend exome sequencing as the standard approach for molecular diagnostics of myopathies.


Subject(s)
Muscular Diseases/diagnosis , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Molecular Diagnostic Techniques , Muscular Diseases/genetics , Muscular Dystrophies/diagnosis , Muscular Dystrophies/genetics , Mutation , Polymerase Chain Reaction , Sequence Analysis, DNA
8.
J Neurol Sci ; 351(1-2): 120-123, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25772186

ABSTRACT

Rarely, inflammation can be present in genetic myopathies, such as dysferlinopathies, facioscapulohumeral muscular dystrophy and GNE-myopathy (hereditary inclusion body myopathy). This may lead to erroneous initial diagnosis and unnecessary therapy which bear serious side effects. We report on an unusual case of mutations in the TTN gene presenting with inflammatory infiltrates in the muscle biopsy. Only after intensive immune-modulating therapies failed, a genetic myopathy was considered. Exome sequencing and search for mutated muscle protein-encoding genes disclosed compound heterozygous mutations in TTN: K26320T and A6135G. The parents carry one each of the mutations. Titinopathy could be considered also in patients presenting with inflammatory infiltrates resistant to therapy.


Subject(s)
Connectin/genetics , Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/genetics , Adult , Age of Onset , Humans , Male , Myositis/diagnosis
10.
PLoS One ; 5(12): e15239, 2010 Dec 13.
Article in English | MEDLINE | ID: mdl-21179212

ABSTRACT

PGC-1α is a transcriptional co-activator that plays a central role in the regulation of energy metabolism. Our interest in this protein was driven by its ability to promote muscle remodeling. Conversion from fast glycolytic to slow oxidative fibers seemed a promising therapeutic approach in Pompe disease, a severe myopathy caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA) which is responsible for the degradation of glycogen. The recently approved enzyme replacement therapy (ERT) has only a partial effect in skeletal muscle. In our Pompe mouse model (KO), the poor muscle response is seen in fast but not in slow muscle and is associated with massive accumulation of autophagic debris and ineffective autophagy. In an attempt to turn the therapy-resistant fibers into fibers amenable to therapy, we made transgenic KO mice expressing PGC-1α in muscle (tgKO). The successful switch from fast to slow fibers prevented the formation of autophagic buildup in the converted fibers, but PGC-1α failed to improve the clearance of glycogen by ERT. This outcome is likely explained by an unexpected dramatic increase in muscle glycogen load to levels much closer to those observed in patients, in particular infants, with the disease. We have also found a remarkable rise in the number of lysosomes and autophagosomes in the tgKO compared to the KO. These data point to the role of PGC-1α in muscle glucose metabolism and its possible role as a master regulator for organelle biogenesis - not only for mitochondria but also for lysosomes and autophagosomes. These findings may have implications for therapy of lysosomal diseases and other disorders with altered autophagy.


Subject(s)
Glycogen Storage Disease Type II/metabolism , Lysosomes/metabolism , Muscle, Skeletal/metabolism , Trans-Activators/genetics , Trans-Activators/physiology , Animals , Autophagy , Disease Models, Animal , Glucose/metabolism , Glycogen/metabolism , Golgi Apparatus/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Muscles/cytology , Muscles/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors
11.
Autophagy ; 6(8): 1078-89, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20861693

ABSTRACT

Autophagy, an intracellular system for delivering portions of cytoplasm and damaged organelles to lysosomes for degradation/recycling, plays a role in many physiological processes and is disturbed in many diseases. We recently provided evidence for the role of autophagy in Pompe disease, a lysosomal storage disorder in which acid alphaglucosidase, the enzyme involved in the breakdown of glycogen, is deficient or absent. Clinically the disease manifests as a cardiac and skeletal muscle myopathy. The current enzyme replacement therapy (ERT) clears lysosomal glycogen effectively from the heart but less so from skeletal muscle. In our Pompe model, the poor muscle response to therapy is associated with the presence of pools of autophagic debris. To clear the fibers of the autophagic debris, we have generated a Pompe model in which an autophagy gene, Atg7, is inactivated in muscle. Suppression of autophagy alone reduced the glycogen level by 50­60%. Following ERT, muscle glycogen was reduced to normal levels, an outcome not observed in Pompe mice with genetically intact autophagy. The suppression of autophagy, which has proven successful in the Pompe model, is a novel therapeutic approach that may be useful in other diseases with disturbed autophagy.


Subject(s)
Autophagy , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/therapy , alpha-Glucosidases/therapeutic use , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Disease Models, Animal , Glycogen/metabolism , Glycogen Storage Disease Type II/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Integrases/metabolism , Mice , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Fast-Twitch/ultrastructure , Myosin Light Chains/metabolism , Phosphorylation , Ubiquitin/metabolism , alpha-Glucosidases/deficiency , alpha-Glucosidases/metabolism
12.
Mol Genet Metab ; 101(4): 324-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20801068

ABSTRACT

Pompe disease is a lysosomal storage disorder caused by the deficiency of acid alpha-glucosidase, the enzyme that degrades glycogen in the lysosomes. The disease manifests as a fatal cardiomyopathy and skeletal muscle myopathy in infants; in milder late-onset forms skeletal muscle is the major tissue affected. We have previously demonstrated that autophagic inclusions in muscle are prominent in adult patients and the mouse model. In this study we have evaluated the contribution of the autophagic pathology in infants before and 6 months after enzyme replacement therapy. Single muscle fibers, isolated from muscle biopsies, were stained for autophagosomal and lysosomal markers and analyzed by confocal microscopy. In addition, unstained bundles of fixed muscles were analyzed by second harmonic imaging. Unexpectedly, the autophagic component which is so prominent in juvenile and adult patients was negligible in infants; instead, the overwhelming characteristic was the presence of hugely expanded lysosomes. After 6 months on therapy, however, the autophagic buildup becomes visible as if unmasked by the clearance of glycogen. In most fibers, the two pathologies did not seem to coexist. These data point to the possibility of differences in the pathogenesis of Pompe disease in infants and adults.


Subject(s)
Autophagy/physiology , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/pathology , Lysosomes/pathology , Adult , Child , Child, Preschool , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/enzymology , Humans , Infant , Infant, Newborn , Lysosomes/enzymology , Muscle Fibers, Skeletal/pathology , alpha-Glucosidases/deficiency , alpha-Glucosidases/metabolism , alpha-Glucosidases/therapeutic use
13.
Arthritis Care Res (Hoboken) ; 62(4): 465-72, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20391500

ABSTRACT

OBJECTIVE: To validate manual muscle testing (MMT) for strength assessment in juvenile and adult dermatomyositis (DM) and polymyositis (PM). METHODS: Patients with PM/DM (73 children and 45 adults) were assessed at baseline and reevaluated 6-9 months later. We compared Total MMT (a group of 24 proximal, distal, and axial muscles) and Proximal MMT (7 proximal muscle groups) tested bilaterally on a 0-10 scale with 144 subsets of 6 and 96 subsets of 8 muscle groups tested unilaterally. Expert consensus was used to rank the best abbreviated MMT subsets for face validity and ease of assessment. RESULTS: The Total, Proximal, and best MMT subsets had excellent internal reliability (Total MMT r(s) = 0.91-0.98), and consistency (Cronbach's alpha = 0.78-0.97). Inter- and intrarater reliability were acceptable (Kendall's W 0.68-0.76, r(s) = 0.84-0.95). MMT subset scores correlated highly with Total and Proximal MMT scores and with the Childhood Myositis Assessment Scale, and correlated moderately with physician global activity, functional disability, magnetic resonance imaging, and axial and distal MMT scores, and, in adults, with creatine kinase level. The standardized response mean for Total MMT was 0.56 in juveniles and 0.75 in adults. Consensus was reached to use a subset of 8 muscles (neck flexors, deltoids, biceps, wrist extensors, gluteus maximus and medius, quadriceps, and ankle dorsiflexors) that performed as well as the Total and Proximal MMT, and had good face validity and ease of assessment. CONCLUSION: These findings aid in standardizing the use of MMT for assessing strength as an outcome measure for myositis.


Subject(s)
Dermatomyositis/diagnosis , Muscle Weakness/diagnosis , Physical Examination , Polymyositis/diagnosis , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Myositis/diagnosis , Observer Variation , Reproducibility of Results , Severity of Illness Index
14.
J Appl Physiol (1985) ; 108(5): 1383-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20223998

ABSTRACT

Pompe disease, a deficiency of lysosomal acid alpha-glucosidase, is a disorder of glycogen metabolism that can affect infants, children, or adults. In all forms of the disease, there is progressive muscle pathology leading to premature death. The pathology is characterized by accumulation of glycogen in lysosomes, autophagic buildup, and muscle atrophy. The purpose of the present investigation was to determine if myofibrillar dysfunction in Pompe disease contributes to muscle weakness beyond that attributed to atrophy. The study was performed on isolated myofibers dissected from severely affected fast glycolytic muscle in the alpha-glucosidase knockout mouse model. Psoas muscle fibers were first permeabilized, so that the contractile proteins could be directly relaxed or activated by control of the composition of the bathing solution. When normalized by cross-sectional area, single fibers from knockout mice produced 6.3 N/cm2 of maximum Ca2+-activated tension compared with 12.0 N/cm2 produced by wild-type fibers. The total protein concentration was slightly higher in the knockout mice, but concentrations of the contractile proteins myosin and actin remained unchanged. Structurally, X-ray diffraction showed that the actin and myosin filaments, normally arranged in hexagonal arrays, were disordered in the knockout muscle, and a lower fraction of myosin cross bridges was near the actin filaments in the relaxed muscle. The results are consistent with a disruption of actin and myosin interactions in the knockout muscles, demonstrating that impaired myofibrillar function contributes to weakness in the diseased muscle fibers.


Subject(s)
Actin Cytoskeleton/pathology , Glycogen Storage Disease Type II/enzymology , Muscle Contraction , Muscle Fibers, Fast-Twitch/enzymology , Muscle Strength , Muscle Weakness/enzymology , Psoas Muscles/enzymology , alpha-Glucosidases/deficiency , Actin Cytoskeleton/enzymology , Actins/metabolism , Animals , Calcium/metabolism , Disease Models, Animal , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/physiopathology , Mice , Mice, Knockout , Muscle Fibers, Fast-Twitch/pathology , Muscle Weakness/genetics , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Muscular Atrophy/enzymology , Muscular Atrophy/physiopathology , Myosins/metabolism , Psoas Muscles/pathology , Psoas Muscles/physiopathology , alpha-Glucosidases/genetics
15.
Autophagy ; 5(5): 729-31, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19571661

ABSTRACT

In Pompe disease, a lysosomal glycogen storage disorder, cardiac and skeletal muscle abnormalities are responsible for premature death and severe weakness. Swollen glycogen-filled lysosomes, the expected pathology, are accompanied in skeletal muscle by a secondary pathology-massive accumulation of autophagic debris-that appears to contribute greatly to the weakness. We have tried to reproduce these defects in murine, Pompe myotubes derived from either primary myoblasts or myoblasts with extended proliferative capacity. The cells accumulated large lysosomes filled with glycogen, but, to our disappointment, did not have autophagic buildup even though basal autophagy was intact. When we suppressed autophagy by knocking down Atg7, we found that glycogen uptake by lysosomes was not affected, suggesting that macroautophagy is not the major pathway for glycogen delivery to lysosomes. But two apparently incidental observations-a peculiar distribution of both microinjected dextran and of small acidic structures adjacent to the interior membrane of large alkalinized glycogen containing lysosomes-raised the possibility that glycogen traffics to the lysosomes by microautophagy or/and by the engulfment of small lysosomes by large ones. The cultured myotubes, therefore, appear to be a useful model for studying the mechanisms involved in glycogen accumulation in Pompe disease and to test substrate deprivation approaches.


Subject(s)
Glycogen Storage Disease Type II/pathology , Animals , Disease Models, Animal , Humans , Lysosomes/pathology , Mice , Muscle Fibers, Skeletal/pathology
16.
Methods Enzymol ; 453: 417-49, 2009.
Article in English | MEDLINE | ID: mdl-19216919

ABSTRACT

Lysosomes are the final destination of the autophagic pathway. It is in the acidic milieu of the lysosomes that autophagic cargo is metabolized and recycled. One would expect that diseases with primary lysosomal defects would be among the first systems in which autophagy would be studied. In reality, this is not the case. Lysosomal storage diseases, a group of more than 60 diverse inherited disorders, have only recently become a focus of autophagic research. Studies of these clinically severe conditions promise not only to clarify pathogenic mechanisms, but also to expand our knowledge of autophagy itself. In this chapter, we will describe the lysosomal storage diseases in which autophagy has been explored, and present the approaches used to evaluate this essential cellular pathway.


Subject(s)
Autophagy/physiology , Lysosomal Storage Diseases/metabolism , Animals , Cells, Cultured , Fluorescent Antibody Technique , Humans , Immunoblotting , Lysosomes/metabolism , Mice , Microscopy, Electron , Mitochondria/metabolism
17.
Mol Genet Metab ; 96(4): 208-17, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19167256

ABSTRACT

Lysosomes filled with glycogen are a major pathologic feature of Pompe disease, a fatal myopathy and cardiomyopathy caused by a deficiency of the glycogen-degrading lysosomal enzyme, acid alpha-glucosidase (GAA). To facilitate studies germane to this genetic disorder, we developed two in vitro Pompe models: myotubes derived from cultured primary myoblasts isolated from Pompe (GAA KO) mice, and myotubes derived from primary myoblasts of the same genotype that had been transduced with cyclin-dependent kinase 4 (CDK4). This latter model is endowed with extended proliferative capacity. Both models showed extremely large alkalinized, glycogen-filled lysosomes as well as impaired trafficking to lysosomes. Although both Pompe tissue culture models were derived from fast muscles and were fast myosin positive, they strongly resemble slow fibers in terms of their pathologic phenotype and their response to therapy with recombinant human GAA (rhGAA). Autophagic buildup, a hallmark of Pompe disease in fast muscle fibers, was absent, but basal autophagy was functional. To evaluate substrate deprivation as a strategy to prevent the accumulation of lysosomal glycogen, we knocked down Atg7, a gene essential for autophagosome formation, via siRNA, but we observed no effect on the extent of glycogen accumulation, thus confirming our recent observation in autophagy-deficient Pompe mice [N. Raben, V. Hill, L. Shea, S. Takikita, R. Baum, N. Mizushima, E. Ralston, P. Plotz, Suppression of autophagy in skeletal muscle uncovers the accumulation of ubiquitinated proteins and their potential role in muscle damage in Pompe disease, Hum. Mol. Genet. 17 (2008) 3897-3908] that macroautophagy is not the major route of glycogen transport to lysosomes. The in vitro Pompe models should be useful in addressing fundamental questions regarding the pathway of glycogen to the lysosomes and testing panels of small molecules that could affect glycogen biosynthesis or speed delivery of the replacement enzyme to affected lysosomes.


Subject(s)
Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/therapy , Muscle Cells/pathology , Animals , Autophagy , Autophagy-Related Protein 7 , Cathepsin B/metabolism , Cathepsin D/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 4/metabolism , Disease Models, Animal , Endocytosis , Humans , Hydrogen-Ion Concentration , Lysosomes/pathology , Mice , Microtubule-Associated Proteins/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Myoblasts/enzymology , Myoblasts/pathology , RNA, Small Interfering/metabolism , Transduction, Genetic , alpha-Glucosidases/deficiency , alpha-Glucosidases/metabolism
18.
Autophagy ; 5(1): 111-3, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001870

ABSTRACT

The role of autophagy, a catabolic lysosome-dependent pathway, has recently been recognized in a variety of disorders, including Pompe disease, which results from a deficiency of the glycogen-degrading lysosomal hydrolase acid-alpha glucosidase (GAA). Skeletal and cardiac muscle are most severely affected by the progressive expansion of glycogen-filled lysosomes. In both humans and an animal model of the disease (GAA KO), skeletal muscle pathology also involves massive accumulation of autophagic vesicles and autophagic buildup in the core of myofibers, suggesting an induction of autophagy. Only when we suppressed autophagy in the skeletal muscle of the GAA KO mice did we realize that the excess of autophagy manifests as a functional deficiency. This failure of productive autophagy is responsible for the accumulation of potentially toxic aggregate-prone ubiquitinated proteins, which likely cause profound muscle damage in Pompe mice. Also, by generating muscle-specific autophagy-deficient wild-type mice, we were able to analyze the role of autophagy in healthy skeletal muscle.


Subject(s)
Autophagy , Glycogen Storage Disease Type II/pathology , Muscle, Skeletal/pathology , Animals , Glycogen Storage Disease Type II/enzymology , Humans , Mice , Mice, Knockout , Muscle, Skeletal/enzymology , Muscle, Skeletal/ultrastructure , Organ Specificity , alpha-Glucosidases/deficiency , alpha-Glucosidases/metabolism
19.
Hum Mol Genet ; 17(24): 3897-908, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18782848

ABSTRACT

The role of autophagy, a catabolic lysosome-dependent pathway, has recently been recognized in a variety of disorders, including Pompe disease, the genetic deficiency of the glycogen-degrading lysosomal enzyme acid-alpha glucosidase. Accumulation of lysosomal glycogen, presumably transported from the cytoplasm by the autophagic pathway, occurs in multiple tissues, but pathology is most severe in skeletal and cardiac muscle. Skeletal muscle pathology also involves massive autophagic buildup in the core of myofibers. To determine if glycogen reaches the lysosome via autophagy and to ascertain whether autophagic buildup in Pompe disease is a consequence of induction of autophagy and/or reduced turnover due to defective fusion with lysosomes, we generated muscle-specific autophagy-deficient Pompe mice. We have demonstrated that autophagy is not required for glycogen transport to lysosomes in skeletal muscle. We have also found that Pompe disease involves induction of autophagy but manifests as a functional deficiency of autophagy because of impaired autophagosomal-lysosomal fusion. As a result, autophagic substrates, including potentially toxic aggregate-prone ubiquitinated proteins, accumulate in Pompe myofibers and may cause profound muscle damage.


Subject(s)
Autophagy , Glycogen Storage Disease Type II/pathology , Muscle, Skeletal/pathology , Muscular Diseases/etiology , Muscular Diseases/pathology , Proteins/adverse effects , Proteins/metabolism , Ubiquitination , Animals , Autophagy/genetics , Female , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Proteins/genetics , Ubiquitination/genetics , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics
20.
Am J Pathol ; 172(3): 774-85, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18276788

ABSTRACT

Dysferlin deficiency causes limb-girdle muscular dystrophy type 2B (LGMD2B; proximal weakness) and Miyoshi myopathy (distal weakness). Muscle inflammation is often present in dysferlin deficiency, and patients are frequently misdiagnosed as having polymyositis. Because monocytes normally express dysferlin, we hypothesized that monocyte/macrophage dysfunction in dysferlin-deficient patients might contribute to disease onset and progression. We therefore examined phagocytic activity, in the presence and absence of cytokines, in freshly isolated peripheral blood monocytes from LGMD2B patients and in the SJL dysferlin-deficient mouse model. Dysferlin-deficient monocytes showed increased phagocytic activity compared with control cells. siRNA-mediated inhibition of dysferlin expression in the J774 macrophage cell line resulted in significantly enhanced phagocytosis, both at baseline and in response to tumor necrosis factor-alpha. Immunohistochemical analysis revealed positive staining for several mononuclear cell activation markers in LGMD2B human muscle and SJL mouse muscle. SJL muscle showed strong up-regulation of endocytic proteins CIMPR, clathrin, and adaptin-alpha, and LGMD2B muscle exhibited decreased expression of decay accelerating factor, which was not dysferlin-specific. We further showed that expression levels of small Rho family GTPases RhoA, Rac1, and Cdc 42 were increased in dysferlin-deficient murine immune cells compared with control cells. Therefore, we hypothesize that mild myofiber damage in dysferlin-deficient muscle stimulates an inflammatory cascade that may initiate, exacerbate, and possibly perpetuate the underlying myofiber-specific dystrophic process.


Subject(s)
Inflammation/genetics , Membrane Proteins/genetics , Monocytes/physiology , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Phagocytosis/genetics , Adolescent , Adult , Aged , Animals , Cells, Cultured , Disease Models, Animal , Dysferlin , Female , Humans , Inflammation/complications , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Monocytes/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/physiology , Muscular Dystrophies, Limb-Girdle/etiology , RNA, Small Interfering/pharmacology , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...