Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

País/Región como asunto
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 ; 201(4): 591-601, 2005 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-15728237

RESUMEN

Unique autoantibody specificities are strongly associated with distinct clinical phenotypes, making autoantibodies useful for diagnosis and prognosis. To investigate the mechanisms underlying this striking association, we examined autoantigen expression in normal muscle and in muscle from patients with autoimmune myositis. Although myositis autoantigens are expressed at very low levels in control muscle, they are found at high levels in myositis muscle. Furthermore, increased autoantigen expression correlates with differentiation state, such that myositis autoantigen expression is increased in cells that have features of regenerating muscle cells. Consistent with this, we found that cultured myoblasts express high levels of autoantigens, which are strikingly down-regulated as cells differentiate into myotubes in vitro. These data strongly implicate regenerating muscle cells rather than mature myotubes as the source of ongoing antigen supply in autoimmune myositis. Myositis autoantigen expression is also markedly increased in several cancers known to be associated with autoimmune myositis, but not in their related normal tissues, demonstrating that tumor cells and undifferentiated myoblasts are antigenically similar. We propose that in cancer-associated myositis, an autoimmune response directed against cancer cross-reacts with regenerating muscle cells, enabling a feed-forward loop of tissue damage and antigen selection. Regulating pathways of antigen expression may provide unrecognized therapeutic opportunities in autoimmune diseases.


Asunto(s)
Autoantígenos/biosíntesis , Enfermedades Autoinmunes/inmunología , Células Musculares/inmunología , Miositis/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/etiología , Neoplasias de la Mama/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Células Musculares/patología , Miositis/etiología
3.
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
4.
Hum Mol Genet ; 17(24): 3897-908, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18782848

RESUMEN

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.


Asunto(s)
Autofagia , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Músculo Esquelético/patología , Enfermedades Musculares/etiología , Enfermedades Musculares/patología , Proteínas/efectos adversos , Proteínas/metabolismo , Ubiquitinación , Animales , Autofagia/genética , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Proteínas/genética , Ubiquitinación/genética , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
5.
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
6.
Clin Rheumatol ; 39(3): 689-696, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31960207

RESUMEN

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.


Asunto(s)
Creación de Capacidad/métodos , Curriculum , Reumatólogos/provisión & distribución , Reumatología/educación , Educación de Postgrado en Medicina/métodos , Humanos , Liberia , Encuestas y Cuestionarios
7.
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
8.
Arthritis Rheumatol ; 71(8): 1371-1376, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30861336

RESUMEN

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.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/metabolismo , Músculo Esquelético/inmunología , Miositis/inmunología , Regeneración/inmunología , Animales , Autoantígenos/inmunología , Biopsia , Células Cultivadas , Humanos , Ratones , Mioblastos/inmunología , Mioblastos/metabolismo , Miositis/fisiopatología , ARN/inmunología , ARN/metabolismo
9.
Neurology ; 93(12): e1193-e1204, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31434690

RESUMEN

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.


Asunto(s)
Interferón Tipo I/genética , Interferón gamma/genética , Músculo Esquelético/metabolismo , Miositis/genética , Miositis/metabolismo , Femenino , Expresión Génica , Humanos , Interferón Tipo I/biosíntesis , Interferón gamma/biosíntesis , Masculino , Músculo Esquelético/patología , Miositis/patología
10.
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
11.
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
12.
J Leukoc Biol ; 77(6): 854-61, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15917448

RESUMEN

We have investigated the chemoattractant properties of self-antigens associated with autoimmune diseases and solid tumors. Many autoantigens induced leukocyte migration, especially by immature dendritic cells (iDC) by interacting with various chemoattractant Gi-protein-coupled receptors (GiPCR). Our initial observation that myositis-associated autoantigens, histidyl-tRNA synthetase and asparaginyl-tRNA synthetase, were chemotactic for CC chemokine receptor 5 (CCR5)- and CCR3-expressing leukocytes, while other nonautoantigenic aminoacyl-tRNA synthesases were not, suggested that only self-antigens capable of interacting with receptors on antigen-presenting cells were immunogenic. We next determined that self-antigens associated with autoimmune diseases, e.g., multiple sclerosis or experimental autoimmune encephalomyelitis, type I diabetes, scleroderma, systemic lupus erythematosus, autoimmune uveitis, or experimental autoimmune uveitis (EAU), were chemotactic for GiPCR expressed by iDC. The majority of autoantigens were DC chemoattractants at 10-100 ng/ml, but did not induce DC maturation until they reached 1000-fold higher concentrations. Interphotoreceptor retinoid-binding protein and retinal arrestin (S-antigen) are targets of autoantibodies in human uveitis and are chemotactic for CXC chemokine receptor 5 (CXCR5)- and/or CXCR3-expressing iDC. However, although S-antigen does not induce EAU in wild-type mice, it is nevertheless a chemoattractant for murine iDC. These unexpected observations suggested that the chemotactic activity of these tissue-specific self-antigens could be involved in promotion of tissue repair and restoration. Thus, the primary role of autoantigens may be to alert the immune system to danger signals from invaded and damaged tissues to facilitate repair, and autoimmune responses subsequently develop only in subjects with impaired immunoregulatory function.


Asunto(s)
Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Factores Quimiotácticos/inmunología , Neoplasias/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/inmunología , Humanos , Leucocitos Mononucleares/inmunología , Ratones
13.
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
14.
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
15.
Curr Mol Med ; 2(2): 145-66, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11949932

RESUMEN

Glycogenosis type II (GSDII, Pompe disease) is an autosomal recessive lysosomal storage disease caused by a deficiency of acid alpha-glucosidase (acid maltase, GAA). The enzyme degrades alpha -1,4 and alpha -1,6 linkages in glycogen, maltose, and isomaltose. Deficiency of the enzyme results in accumulation of glycogen within lysosomes and in cytoplasm eventually leading to tissue destruction. The discovery of the acid a-glucosidase gene has led to rapid progress in understanding the molecular basis of glycogenosis type II and the biological properties of the GAA protein. The last decade has seen several developments: 1) extensive mutational analysis in patients with different forms of the disease, 2) characterization of the enzyme biosynthesis, processing, and lysosomal targeting, 3) generation of knockout mouse models, 4) development of viral vectors for gene replacement therapy, 5) the production of recombinant human enzyme, and 6) a shift in the enzyme replacement therapy approach from theory to practice. It is anticipated that the enzyme replacement therapy will be widely available for human use in the near future. Several recent reviews (including the most comprehensive one by R. Hirschhorn and A. Reuser [1]), address clinical, biochemical and genetic aspects of the disease, as well as development of new therapies for GSDII [2, 3, 4]. In this article we will review recent findings in the area including rapidly accumulating molecular genetic data (more than 20 mutations need to be added to the list), transcriptional control of gene expression, studies in mouse models, and new approaches to gene therapy. We will also highlight some emerging questions following the introduction of enzyme replacement therapy.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Mutación , Adulto , Animales , Preescolar , Femenino , Genotipo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Humanos , Lactante , Masculino , Ratones , Modelos Genéticos , Fenotipo , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
16.
J Neurol Sci ; 351(1-2): 120-123, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25772186

RESUMEN

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.


Asunto(s)
Conectina/genética , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/genética , Adulto , Edad de Inicio , Humanos , Masculino , Miositis/diagnóstico
17.
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
18.
DNA Cell Biol ; 21(10): 717-25, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12443541

RESUMEN

Autosomal recessive deficiency of lysosomal acid maltase (GAA) or glycogen storage disease type II (GSDII) results in a spectrum of phenotypes including a rapidly fatal infantile disorder (Pompe's), juvenile, and a late-onset adult myopathy. The infantile onset form presents as hypotonia with massive accumulation of glycogen in skeletal and heart muscle, with death due to cardiorespiratory failure. Adult patients with the slowly progressive form develop severe skeletal muscle weakness and respiratory failure. Particle bombardment is a safe, efficient physical method in which high-density, subcellular-sized particles are accelerated to high velocity to carry DNA into cells. Because it does not depend on a specific ligand, receptor, or biochemical features on cell surfaces, particle-mediated gene transfer can be readily applied to a variety of systems. We evaluated particle bombardment as a delivery system for therapy of GSDII. We utilized a vector carrying the CMV promoter linked to the human GAA cDNA. Human GSDII cell lines (fibroblasts and lymphoid) as well as ex vivo with adult-onset peripheral blood cells (lymphocytes and monocytes) were transiently transfected by bombardment with a Helios gene gun delivering gold particles coated with the GAA expression plasmid. All cell types showed an increase in human GAA activity greater than 50% of normal activity. Subsequently, GAA -/- mice were treated every 2 weeks for 4 months by particle bombardment to the epidermis of the lower back and hind limbs. Muscle weakness in the hind and forelimbs was reversed. These data suggest that particle delivery of the GAA cDNA by the Helios gene gun may be a safe, effective treatment for GSDII.


Asunto(s)
Biolística , Glucano 1,4-alfa-Glucosidasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Adulto , Animales , Formación de Anticuerpos , Línea Celular , Citomegalovirus/genética , ADN Complementario/administración & dosificación , ADN Complementario/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Glucano 1,4-alfa-Glucosidasa/sangre , Glucano 1,4-alfa-Glucosidasa/inmunología , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Técnicas In Vitro , Recién Nacido , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , alfa-Glucosidasas
19.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA