Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nat Rev Immunol ; 3(1): 73-8, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12511877

RESUMEN

The spontaneously occurring autoantibodies that are associated with human diseases bear the hallmarks of a typical immune response. The repertoire of autoantibodies is surprisingly limited, however, and is the same in both humans and mice. Neither molecular mimicry nor immune dysregulation accounts for this unexpectedly narrow focus of specificities. Experimental data on the properties of the target autoantigens--such as their structure, catabolism, exposure to the immune system after cell death and recently described immunostimulatory effects on immature dendritic cells--indicate that these properties, in conjunction with the tissue microenvironment, help to select the autoantibody repertoire.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Animales , Especificidad de Anticuerpos/inmunología , Apoptosis/inmunología , Células Dendríticas/inmunología , Humanos , Tolerancia Inmunológica/inmunología
2.
J Exp Med ; 196(6): 781-91, 2002 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-12235211

RESUMEN

Autoantibodies to histidyl-tRNA synthetase (HisRS) or to alanyl-, asparaginyl-, glycyl-, isoleucyl-, or threonyl-tRNA synthetase occur in approximately 25% of patients with polymyositis or dermatomyositis. We tested the ability of several aminoacyl-tRNA synthetases to induce leukocyte migration. HisRS induced CD4(+) and CD8(+) lymphocytes, interleukin (IL)-2-activated monocytes, and immature dendritic cells (iDCs) to migrate, but not neutrophils, mature DCs, or unstimulated monocytes. An NH(2)-terminal domain, 1-48 HisRS, was chemotactic for lymphocytes and activated monocytes, whereas a deletion mutant, HisRS-M, was inactive. HisRS selectively activated CC chemokine receptor (CCR)5-transfected HEK-293 cells, inducing migration by interacting with extracellular domain three. Furthermore, monoclonal anti-CCR5 blocked HisRS-induced chemotaxis and conversely, HisRS blocked anti-CCR5 binding. Asparaginyl-tRNA synthetase induced migration of lymphocytes, activated monocytes, iDCs, and CCR3-transfected HEK-293 cells. Seryl-tRNA synthetase induced migration of CCR3-transfected cells but not iDCs. Nonautoantigenic aspartyl-tRNA and lysyl-tRNA synthetases were not chemotactic. Thus, autoantigenic aminoacyl-tRNA synthetases, perhaps liberated from damaged muscle cells, may perpetuate the development of myositis by recruiting mononuclear cells that induce innate and adaptive immune responses. Therefore, the selection of a self-molecule as a target for an autoantibody response may be a consequence of the proinflammatory properties of the molecule itself.


Asunto(s)
Aminoacil-ARNt Sintetasas/fisiología , Aspartato-ARNt Ligasa , Autoantígenos/fisiología , Células Dendríticas/fisiología , Histidina-ARNt Ligasa/fisiología , Miositis/inmunología , Aminoacil-ARN de Transferencia , Receptores de Quimiocina/fisiología , Linfocitos T/fisiología , Movimiento Celular , Humanos , Interleucina-2/farmacología , Receptores CCR2 , Receptores CCR3 , Receptores CCR5/fisiología , Factores de Virulencia de Bordetella/farmacología
3.
Mol Genet Metab ; 101(4): 324-31, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20801068

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Lisosomas/patología , Adulto , Niño , Preescolar , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Humanos , Lactante , Recién Nacido , Lisosomas/enzimología , Fibras Musculares Esqueléticas/patología , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/uso terapéutico
4.
Mol Genet Metab ; 96(4): 208-17, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19167256

RESUMEN

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.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Células Musculares/patología , Animales , Autofagia , Proteína 7 Relacionada con la Autofagia , Catepsina B/metabolismo , Catepsina D/metabolismo , Células Cultivadas , Quinasa 4 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Endocitosis , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/patología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Mioblastos/enzimología , Mioblastos/patología , ARN Interferente Pequeño/metabolismo , Transducción Genética , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo
5.
Semin Arthritis Rheum ; 47(6): 858-864, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29174792

RESUMEN

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.


Asunto(s)
Fármacos Dermatológicos/uso terapéutico , Dermatomiositis/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Infliximab/uso terapéutico , Adulto , Estudios Cruzados , Método Doble Ciego , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Prednisona/uso terapéutico , Resultado del Tratamiento
6.
Medicine (Baltimore) ; 85(2): 111-127, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16609350

RESUMEN

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.


Asunto(s)
Autoanticuerpos/análisis , Antígenos HLA/genética , Antígenos HLA/inmunología , Miositis/genética , Miositis/inmunología , Alelos , Secuencias de Aminoácidos , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Antígenos HLA/clasificación , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/genética , Antígenos HLA-C/inmunología , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/inmunología , Cadenas alfa de HLA-DQ , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Cadenas HLA-DRB1 , Haplotipos , Humanos , Inmunidad Innata , Masculino , Miositis/patología , Unión Proteica , Factores de Riesgo , Población Blanca/genética
7.
J Neuromuscul Dis ; 3(2): 209-225, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27854218

RESUMEN

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.


Asunto(s)
Enfermedades Musculares/diagnóstico , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Técnicas de Diagnóstico Molecular , Enfermedades Musculares/genética , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Mutación , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
8.
Medicine (Baltimore) ; 84(6): 338-349, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16267409

RESUMEN

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.


Asunto(s)
Miositis/genética , Población Blanca/genética , Adulto , Alelos , Biomarcadores , Estudios de Casos y Controles , Susceptibilidad a Enfermedades , Femenino , Variación Genética , Antígenos HLA-B , Antígenos HLA-C , Antígenos HLA-DQ , Antígenos HLA-DR , Humanos , Inflamación/genética , Inflamación/inmunología , Masculino , Miositis/inmunología , Polimorfismo Genético , Medición de Riesgo , Factores de Riesgo
9.
Rheum Dis Clin North Am ; 28(4): 917-33, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12506778

RESUMEN

Current animal models of human myositis include spontaneous, induced, and transgenic models. Although it is clear that none of these models possesses all the features of the human diseases, they may provide insight into the pathophysiologic mechanisms, and possibly the therapy, of inflammatory muscle disease. Because the human IIMs are phenotypically heterogeneous, but may be divided into more homogeneous subgroups based upon clinical or serologic features, it is possible that different pathogeneses are involved in different subgroups. It is unlikely that any single model would reproduce all features of the human disease. It may be possible, however, to gain insight into some subgroups of the human disease if certain animal models faithfully reproduce one or more subtypes or aspects of the IIMs. Because immunogenetic risk factors, and exposure to certain environmental agents important in triggering myositis in genetically susceptible persons, may be necessary components for human disease induction, transgenic approaches to humanizing murine immune systems and a better understanding of environmental risk factors will be productive avenues for future research. Additional investigations into the molecular basis of the human myositis syndromes and the pathogenesis of the spontaneous, induced, and transgenic animal models should ultimately allow for better understanding and therapy of these diseases.


Asunto(s)
Modelos Animales de Enfermedad , Miositis/etiología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Enfermedades de los Perros/inmunología , Enfermedades de los Perros/patología , Enfermedades de los Perros/fisiopatología , Perros , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Ratones Transgénicos , Miositis/patología , Miositis/fisiopatología
10.
Best Pract Res Clin Rheumatol ; 18(3): 331-44, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15158744

RESUMEN

The major inflammatory myopathies of adults-dermatomyositis, polymyositis and inclusion body myositis-are uncommon and can be difficult to distinguish from many conditions that mimic them clinically. They have a high morbidity; they are not infrequently the first sign of an associated malignancy; and they may be a part of another connective tissue disease. Their pathogenetic features suggest that they are different illnesses. Dermatomyositis and polymyositis are clearly inflammatory, both clinically and histologically, and both generally respond to therapy directed towards inflammation. Inclusion body myositis is now generally recognized as the most common myopathy presenting in patients over the age of 50 years, and it responds only modestly and sometimes not at all to immunosuppressive therapy. In this review, we have summarised the major newly recognized features of pathogenesis, the involvement of extramuscular organs, the differential diagnosis, diagnostic approaches and the main lines of therapy.


Asunto(s)
Miositis , Adulto , Humanos , Miositis/diagnóstico , Miositis/etiología , Miositis/terapia
14.
PLoS One ; 5(12): e15239, 2010 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-21179212

RESUMEN

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.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Lisosomas/metabolismo , Músculo Esquelético/metabolismo , Transactivadores/genética , Transactivadores/fisiología , Animales , Autofagia , Modelos Animales de Enfermedad , Glucosa/metabolismo , Glucógeno/metabolismo , Aparato de Golgi/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculos/citología , Músculos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción
15.
Autophagy ; 6(8): 1078-89, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20861693

RESUMEN

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.


Asunto(s)
Autofagia , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , alfa-Glucosidasas/uso terapéutico , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Modelos Animales de Enfermedad , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Integrasas/metabolismo , Ratones , Fibras Musculares de Contracción Rápida/enzimología , Fibras Musculares de Contracción Rápida/patología , Fibras Musculares de Contracción Rápida/ultraestructura , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Ubiquitina/metabolismo , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo
16.
Arthritis Care Res (Hoboken) ; 62(4): 465-72, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20391500

RESUMEN

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.


Asunto(s)
Dermatomiositis/diagnóstico , Debilidad Muscular/diagnóstico , Examen Físico , Polimiositis/diagnóstico , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miositis/diagnóstico , Variaciones Dependientes del Observador , Reproducibilidad de los Resultados , Índice de Severidad de la Enfermedad
18.
Autophagy ; 5(5): 729-31, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19571661

RESUMEN

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.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Animales , Modelos Animales de Enfermedad , Humanos , Lisosomas/patología , Ratones , Fibras Musculares Esqueléticas/patología
19.
Curr Neurol Neurosci Rep ; 7(1): 71-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17217857

RESUMEN

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.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , alfa-Glucosidasas/administración & dosificación , alfa-Glucosidasas/deficiencia , Animales , Ensayos Clínicos como Asunto/métodos , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Humanos
20.
Autophagy ; 3(6): 546-52, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17592248

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Adolescente , Adulto , Autofagia/genética , Biomarcadores/metabolismo , Línea Celular Transformada , Transformación Celular Viral , Niño , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Heterocigoto , Histocitoquímica , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Mioblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA