Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
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
3.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Autophagy ; 3(6): 546-52, 2007.
Article in English | MEDLINE | ID: mdl-17592248

ABSTRACT

Autophagy is a major pathway for delivery of proteins and organelles to lysosomes where they are degraded and recycled. We have previously shown excessive autophagy in a mouse model of Pompe disease (glycogen storage disease type II), a devastating myopathy caused by a deficiency of the glycogen-degrading lysosomal enzyme acid alpha-glucosidase. The autophagic buildup constituted a major pathological component in skeletal muscle and interfered with delivery of the therapeutic enzyme. To assess the role of autophagy in the pathogenesis of the human disease, we have analyzed vesicles of the lysosomal-degradative pathway in isolated single muscle fibers from Pompe patients. Human myofibers showed abundant autophagosome formation and areas of autophagic buildup of a wide range of sizes. In patients, as in the mouse model, the enormous autophagic buildup causes greater skeletal muscle damage than the enlarged, glycogenfilled lysosomes outside the autophagic regions. Clearing or preventing autophagic buildup seems, therefore, a necessary target of Pompe disease therapy.


Subject(s)
Autophagy/physiology , Glycogen Storage Disease Type II/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Adolescent , Adult , Autophagy/genetics , Biomarkers/metabolism , Cell Line, Transformed , Cell Transformation, Viral , Child , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Heterozygote , Histocytochemistry , Humans , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Middle Aged , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Myoblasts/metabolism
13.
Curr Neurol Neurosci Rep ; 7(1): 71-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17217857

ABSTRACT

The development and recent approval of recombinant acid alpha-glucosidase for enzyme replacement therapy have been major milestones in Pompe disease research. Acid alpha-glucosidase is the enzyme responsible for degradation of glycogen polymers to glucose in the acidic milieu of the lysosomes. Cardiac and skeletal muscles are the two major tissues affected by the accumulation of glycogen within the lysosomes. Both cardiomyopathy and skeletal muscle myopathy are observed in patients with complete enzyme deficiency; this form of the disease is fatal within the first year of life. Skeletal muscle myopathy eventually leading to respiratory insufficiency is the predominant manifestation of partial enzyme deficiency. The recombinant enzyme alglucosidase alfa is the first drug ever approved for this devastating disorder. This review discusses the benefits and the shortcomings of the new therapy.


Subject(s)
Glycogen Storage Disease Type II/physiopathology , alpha-Glucosidases/administration & dosage , alpha-Glucosidases/deficiency , Animals , Clinical Trials as Topic/methods , Disease Models, Animal , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/therapy , Humans
14.
Mol Ther ; 14(6): 831-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17008131

ABSTRACT

Enzyme replacement therapy (ERT) became a reality for patients with Pompe disease, a fatal cardiomyopathy and skeletal muscle myopathy caused by a deficiency of glycogen-degrading lysosomal enzyme acid alpha-glucosidase (GAA). The therapy, which relies on receptor-mediated endocytosis of recombinant human GAA (rhGAA), appears to be effective in cardiac muscle, but less so in skeletal muscle. We have previously shown a profound disturbance of the lysosomal degradative pathway (autophagy) in therapy-resistant muscle of GAA knockout mice (KO). Our findings here demonstrate a progressive age-dependent autophagic buildup in addition to enlargement of glycogen-filled lysosomes in multiple muscle groups in the KO. Trafficking and processing of the therapeutic enzyme along the endocytic pathway appear to be affected by the autophagy. Confocal microscopy of live single muscle fibers exposed to fluorescently labeled rhGAA indicates that a significant portion of the endocytosed enzyme in the KO was trapped as a partially processed form in the autophagic areas instead of reaching its target--the lysosomes. A fluid-phase endocytic marker was similarly mistargeted and accumulated in vesicular structures within the autophagic areas. These findings may explain why ERT often falls short of reversing the disease process and point toward new avenues for the development of pharmacological intervention.


Subject(s)
Autophagy/physiology , Glucan 1,4-alpha-Glucosidase/genetics , Glycogen Storage Disease Type II/therapy , Muscle, Skeletal/metabolism , Age Factors , Animals , Autophagy/genetics , CHO Cells , Cricetinae , Cricetulus , Endocytosis/genetics , Endocytosis/physiology , Glucan 1,4-alpha-Glucosidase/metabolism , Glycogen/metabolism , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/metabolism , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Receptor, IGF Type 2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Autophagy ; 2(4): 318-20, 2006.
Article in English | MEDLINE | ID: mdl-16874053

ABSTRACT

In Pompe disease, a deficiency of lysosomal acid alpha-glucosidase, intralysosomal glycogen accumulates in multiple tissues, with skeletal and cardiac muscle most severely affected.(1) Complete enzyme deficiency results in rapidly progressive infantile cardiomyopathy and skeletal muscle myopathy that is fatal within the first two years of life. Patients with partial enzyme deficiency suffer from skeletal muscle myopathy and experience shortened lifespan due to respiratory failure. The major advance has been the development of enzyme replacement therapy, which recently became available for Pompe patients. However, the effective clearance of skeletal muscle glycogen, as shown by both clinical and preclinical studies, has proven more difficult than anticipated.(2-4) Our recent work published in Annals of Neurology(5) was designed to cast light on the problem, and was an attempt to look beyond the lysosomes by analyzing the downstream events affected by the accumulation of undigested substrate in lysosomes. We have found that the cellular pathology in Pompe disease spreads to affect both endocytic (the route of the therapeutic enzyme) and autophagic (the route of glycogen) pathways, leading to excessive autophagic buildup in therapy-resistant skeletal muscle fibers of the knockout mice.


Subject(s)
Autophagy/physiology , Glycogen Storage Disease Type II/physiopathology , Lysosomes/metabolism , Animals , Glycogen Storage Disease Type II/pathology , Humans , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
16.
Medicine (Baltimore) ; 85(2): 111-127, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16609350

ABSTRACT

The idiopathic inflammatory myopathies (IIM) are systemic connective tissue diseases defined by chronic muscle inflammation and weakness associated with autoimmunity. We have performed low to high resolution molecular typing to assess the genetic variability of major histocompatibility complex loci (HLA-A, -B, -Cw, -DRB1, and -DQA1) in a large population of European American patients with IIM (n = 571) representing the major myositis autoantibody groups. We established that alleles of the 8.1 ancestral haplotype (8.1 AH) are important risk factors for the development of IIM in patients producing anti-synthetase/anti-Jo-1, -La, -PM/Scl, and -Ro autoantibodies. Moreover, a random forests classification analysis suggested that 8.1 AH-associated alleles B*0801 and DRB1*0301 are the principal HLA risk markers. In addition, we have identified several novel HLA susceptibility factors associated distinctively with particular myositis-specific (MSA) and myositis-associated autoantibody (MAA) groups of the IIM. IIM patients with anti-PL-7 (anti-threonyl-tRNA synthetase) autoantibodies have a unique HLA Class I risk allele, Cw*0304 (pcorr = 0.046), and lack the 8.1 AH markers associated with other anti-synthetase autoantibodies (for example, anti-Jo-1 and anti-PL-12). In addition, HLA-B*5001 and DQA1*0104 are novel potential risk factors among anti-signal recognition particle autoantibody-positive IIM patients (pcorr = 0.024 and p = 0.010, respectively). Among those patients with MAA, HLA DRB1*11 and DQA1*06 alleles were identified as risk factors for myositis patients with anti-Ku (pcorr = 0.041) and anti-La (pcorr = 0.023) autoantibodies, respectively. Amino acid sequence analysis of the HLA DRB1 third hypervariable region identified a consensus motif, 70D (hydrophilic)/71R (basic)/74A (hydrophobic), conferring protection among patients producing anti-synthetase/anti-Jo-1 and -PM/Scl autoantibodies. Together, these data demonstrate that HLA signatures, comprising both risk and protective alleles or motifs, distinguish IIM patients with different myositis autoantibodies and may have diagnostic and pathogenic implications. Variations in associated polymorphisms for these immune response genes may reflect divergent pathogenic mechanisms and/or responses to unique environmental triggers in different groups of subjects resulting in the heterogeneous syndromes of the IIM.


Subject(s)
Autoantibodies/analysis , HLA Antigens/genetics , HLA Antigens/immunology , Myositis/genetics , Myositis/immunology , Alleles , Amino Acid Motifs , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Case-Control Studies , Female , Genetic Predisposition to Disease , HLA Antigens/classification , HLA-A Antigens/genetics , HLA-A Antigens/immunology , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-C Antigens/genetics , HLA-C Antigens/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DQ alpha-Chains , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , HLA-DRB1 Chains , Haplotypes , Humans , Immunity, Innate , Male , Myositis/pathology , Protein Binding , Risk Factors , White People/genetics
17.
Ann Neurol ; 59(4): 700-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16532490

ABSTRACT

OBJECTIVE: To understand the mechanisms of skeletal muscle destruction and resistance to enzyme replacement therapy in Pompe disease, a deficiency of lysosomal acid alpha-glucosidase (GAA), in which glycogen accumulates in lysosomes primarily in cardiac and skeletal muscles. METHODS: We have analyzed compartments of the lysosomal degradative pathway in GAA-deficient myoblasts and single type I and type II muscle fibers isolated from wild-type, untreated, and enzyme replacement therapy-treated GAA knock-out mice. RESULTS: Studies in myoblasts from GAA knock-out mice showed a dramatic expansion of vesicles of the endocytic/autophagic pathways, decreased vesicular movement in overcrowded cells, and an acidification defect in a subset of late endosomes/lysosomes. Analysis by confocal microscopy of isolated muscle fibers demonstrated that the consequences of the lysosomal glycogen accumulation are strikingly different in type I and II muscle fibers. Only type II fibers, which are the most resistant to therapy, contain large regions of autophagic buildup that span the entire length of the fibers. INTERPRETATION: The vastly increased autophagic buildup may be responsible for skeletal muscle damage and prevent efficient trafficking of replacement enzyme to lysosomes.


Subject(s)
Autophagy/physiology , Endocytosis/physiology , Lysosomal Storage Diseases/physiopathology , Age Factors , Animals , Blotting, Western/methods , Cells, Cultured , Disease Models, Animal , Fluorescent Antibody Technique/methods , Glucan 1,4-alpha-Glucosidase/deficiency , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Myoblasts/metabolism , Myoblasts/ultrastructure , Time Factors , Transfection/methods , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure , Tubulin/metabolism
18.
J Autoimmune Dis ; 3: 2, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16504012

ABSTRACT

BACKGROUND: While vascular and immune abnormalities are common in juvenile and adult dermatomyositis (DM), the molecular changes that contribute to these abnormalities are not clear. Therefore, we investigated pathways that facilitate new blood vessel formation and dendritic cell migration in dermatomyositis. METHODS: Muscle biopsies from subjects with DM (9 children and 6 adults) and non-myositis controls (6 children and 7 adults) were investigated by immunohistochemistry using antibodies that recognize existing (anti-CD146) and newly formed blood vessels (anti-alphaVbeta3) and mature dendritic cells (anti-DC-LAMP). Blood vessel quantification was performed by digitalized image analysis. Additional muscle biopsies from subjects with adult DM and non-myositis controls were used for global gene expression profiling experiments. RESULTS: A significant increase in neovascularization was found in muscle biopsies of DM patients; neovascularization (alphaVbeta3 positive capillaries and vessels per muscle fiber) was much higher in juvenile than in adult DM patients (control vs juvenile DM: Mean +/- SE: 0.06 +/- 0.01 vs 0.6 +/- 0.05; p < 0.0001 and control vs adult DM: Mean +/- SE: 0.60 +/- 0.1 vs 0.75 +/- 0.1; p = 0.051). Gene expression analysis demonstrated that genes that participate not only in angiogenesis but also in leukocyte trafficking and the complement cascade were highly up regulated in DM muscle in comparison to age matched controls. DC-LAMP positive dendritic cells were highly enriched at perivascular inflammatory sites in juvenile and adult DM patients along with molecules that facilitate dendritic cell transmigration and reverse transmigration (CD142 and CD31). CONCLUSION: These results suggest active neovascularization and endothelial cell activation in both juvenile and adult DM. It is likely that close association of monocytes with endothelial cells initiate rapid dendritic cell maturation and an autoimmune response in DM.

19.
Medicine (Baltimore) ; 84(6): 338-349, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16267409

ABSTRACT

The idiopathic inflammatory myopathies (IIM) are systemic connective tissue diseases in which autoimmune pathology is suspected to promote chronic muscle inflammation and weakness. We have performed low to high resolution genotyping to characterize the allelic profiles of HLA-A, -B, -Cw, -DRB1, and -DQA1 loci in a large population of North American Caucasian patients with IIM representing the major clinicopathologic groups (n = 571). We confirmed that alleles of the 8.1 ancestral haplotype were important risk markers for the development of IIM, and a random forests classification analysis suggested that within this haplotype, HLA-B*0801, DRB1*0301 and/ or closely linked genes are the principal HLA risk factors. In addition, we identified several novel HLA factors associated distinctly with 1 or more clinicopathologic groups of IIM. The DQA1*0201 allele and associated peptide-binding motif (KLPLFHRL) were exclusive protective factors for the CD8+ T cell-mediated IIM forms of polymyositis (PM) and inclusion body myositis (IBM) (pc < 0.005). In contrast, HLA-A*68 alleles were significant risk factors for dermatomyositis (DM) (pc = 0.0021), a distinct clinical group thought to involve a humorally mediated immunopathology. While the DQA1*0301 allele was detected as a possible risk factor for IIM, PM, and DM patients (p < 0.05), DQA1*03 alleles were protective factors for IBM (pc = 0.0002). Myositis associated with malignancies was the most distinctive group of IIM wherein HLA Class I alleles were the only identifiable susceptibility factors and a shared HLA-Cw peptide-binding motif (AGSHTLQWM) conferred significant risk (pc = 0.019). Together, these data suggest that HLA susceptibility markers distinguish different myositis phenotypes with divergent pathogenetic mechanisms. These variations in associated HLA polymorphisms may reflect responses to unique environmental triggers resulting in the tissue pathospecificity and distinct clinicopathologic syndromes of the IIM.


Subject(s)
Myositis/genetics , White People/genetics , Adult , Alleles , Biomarkers , Case-Control Studies , Disease Susceptibility , Female , Genetic Variation , HLA-B Antigens , HLA-C Antigens , HLA-DQ Antigens , HLA-DR Antigens , Humans , Inflammation/genetics , Inflammation/immunology , Male , Myositis/immunology , Polymorphism, Genetic , Risk Assessment , Risk Factors
20.
Arthritis Rheum ; 52(8): 2433-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16052581

ABSTRACT

OBJECTIVE: To assess possible seasonal patterns in the onset of polymyositis (PM) and dermatomyositis (DM). METHODS: The study group comprised 503 patients who met the criteria for probable or definite PM or DM and for whom detailed data on the time of myositis onset were available. Statistical analyses were performed using a Poisson model that assessed associations of ethnicity, sex, autoantibody presence, and month of onset of muscle weakness. RESULTS: There were no significant seasonal patterns of disease onset in myositis patients as a whole or in the total PM or DM populations. Significant seasonal associations were present, however, in the serologically defined groups. In the 131 patients with antisynthetase autoantibodies who were categorized as non-black, myositis onset peaked in March-April (P = 0.03). Among the antisynthetase-positive patients, the association was predominantly in those with PM (n = 85; P = 0.05) and in men (n = 51; P = 0.042). Patients with anti-signal recognition particle autoantibodies, however, did not have a significant seasonal onset, which is in contrast to previous findings. Patients without myositis-specific autoantibodies showed a significant peak in summer, with myositis onset in June-July (n = 252; P = 0.03); this seasonal association was significant in women (n = 182; P = 0.005), whereas there was no seasonal pattern in men (P = 0.9). CONCLUSION: These findings, in conjunction with other data, suggest that diverse environmental agents, acting upon different immunogenetic backgrounds, result in distinct immune responses and clinical syndromes in the idiopathic inflammatory myopathies. Our results emphasize the importance of considering more homogeneous disease groups, based on clinicopathologic features, immune responses, ethnicity, and sex, when attempting to decipher the pathogeneses of autoimmune disorders.


Subject(s)
Autoantibodies/blood , Dermatomyositis/etiology , Dermatomyositis/immunology , Polymyositis/etiology , Polymyositis/immunology , Seasons , Cross-Sectional Studies , Female , Humans , Ligases/immunology , Male , Retrospective Studies , Sex Factors , Signal Recognition Particle/immunology
SELECTION OF CITATIONS
SEARCH DETAIL