Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 142(7): 1242-53, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25742797

RESUMEN

The satellite cells, which serve as adult muscle stem cells, are both located beneath myofiber basement membranes and closely associated with capillary endothelial cells. We observed that 90% of capillaries were associated with pericytes in adult mouse and human muscle. During post-natal growth, newly formed vessels with their neuroglial 2 proteoglycan (NG2)-positive pericytes became progressively associated with the post-natal muscle stem cells, as myofibers increased in size and satellite cells entered into quiescence. In vitro, human muscle-derived pericytes promoted myogenic cell differentiation through insulin-like growth factor 1 (IGF1) and myogenic cell quiescence through angiopoietin 1 (ANGPT1). Diphtheria toxin-induced ablation of muscle pericytes in growing mice led both to myofiber hypotrophy and to impaired establishment of stem cells quiescence. Similar effects were observed following conditional in vivo deletion of pericyte Igf1 and Angpt1 genes, respectively. Our data therefore demonstrate that, by promoting post-natal myogenesis and stem cell quiescence, pericytes play a key role in the microvascular niche of satellite cells.


Asunto(s)
Ciclo Celular , Fibras Musculares Esqueléticas/citología , Neovascularización Fisiológica , Pericitos/citología , Células Satélite del Músculo Esquelético/citología , Adolescente , Angiopoyetina 1/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Niño , Preescolar , Células Endoteliales/citología , Eliminación de Gen , Humanos , Lactante , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones Endogámicos C57BL , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Pericitos/metabolismo , Receptores de Superficie Celular/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Células Madre/citología , Adulto Joven
2.
Rheumatology (Oxford) ; 57(5): 873-879, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29474663

RESUMEN

Objectives: Myositis-specific autoantibodies (MSAs) are increasingly used to delineate distinct subgroups of JDM. The aim of our study was to explore without a priori hypotheses whether MSAs are associated with distinct clinical-pathological changes and severity in a monocentric JDM cohort. Methods: Clinical, biological and histological findings from 23 JDM patients were assessed. Twenty-six histopathological parameters were subjected to multivariate analysis. Results: Autoantibodies included anti-NXP2 (9/23), anti-TIF1γ (4/23), anti-MDA5 (2/23), no MSAs (8/23). Multivariate analysis yielded two histopathological clusters. Cluster 1 (n = 11) showed a more severe and ischaemic pattern than cluster 2 (n = 12) assessed by: total score severity ⩾ 20 (100.0% vs 25.0%); visual analogic score ⩾6 (100.0% vs 25.0%); the vascular domain score >1 (100.0% vs 41.7%); microinfarcts (100% vs 58.3%); ischaemic myofibrillary loss (focal punched-out vacuoles) (90.9 vs 25%); and obvious capillary loss (81.8% vs 16.7). Compared with cluster 2, patients in cluster 1 had strikingly more often anti-NXP2 antibodies (7/11 vs 2/12), more pronounced muscle weakness, more gastrointestinal involvement and required more aggressive treatment. Furthermore, patients with anti-NXP2 antibodies, mostly assigned in the first cluster, also displayed more severe muscular disease, requiring more aggressive treatment and having a lower remission rate during the follow-up period. Conclusion: Marked muscle ischaemic involvement and the presence of anti-NXP2 autoantibodies are associated with more severe forms of JDM.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Autoanticuerpos/inmunología , Proteínas de Unión al ADN/inmunología , Dermatomiositis/complicaciones , Isquemia/etiología , Músculo Esquelético/irrigación sanguínea , Adenosina Trifosfatasas/metabolismo , Biomarcadores/metabolismo , Biopsia , Proteínas de Unión al ADN/metabolismo , Dermatomiositis/inmunología , Dermatomiositis/metabolismo , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Isquemia/diagnóstico , Isquemia/inmunología , Masculino , Músculo Esquelético/diagnóstico por imagen , Índice de Severidad de la Enfermedad
3.
Muscle Nerve ; 56(3): 529-533, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27935078

RESUMEN

INTRODUCTION: Patients with renal failure who are being treated with dialysis frequently develop neuromuscular manifestations. Renal failure-associated calciphylaxis, also termed calcific uremic arteriolopathy (CUA), is a life-threatening condition usually observed in patients with end-stage renal disease on chronic dialysis or after renal transplantation. METHODS: We describe a hemodialyzed patient who presented with rapidly progressive unexplained systemic vasculopathy, muscle atrophy, and proximal weakness, that unexpectedly proved to be caused by calciphylaxis. RESULTS: Quadriceps muscle biopsy disclosed diffuse vascular calcific deposits on medium- and small-sized vessels, characteristic of CUA. Other changes included ischemic myopathy, focal intracellular calcium accumulation within myofibers, and calcium deposits in endomysial capillaries associated with marked complement activation and C5b9 formation. CONCLUSION: There are only a few descriptions of muscle involvement in the context of CUA, a condition with a prognosis that depends on early diagnosis and treatment. This report underscores the usefulness of muscle biopsy in the diagnosis of systemic calciphylaxis. Muscle Nerve 56: 529-533, 2017.


Asunto(s)
Calcifilaxia/diagnóstico , Isquemia/diagnóstico , Enfermedades Musculares/diagnóstico , Vasculitis Sistémica/diagnóstico , Calcifilaxia/complicaciones , Humanos , Isquemia/complicaciones , Trasplante de Riñón/tendencias , Masculino , Persona de Mediana Edad , Enfermedades Musculares/complicaciones , Músculo Cuádriceps/patología , Diálisis Renal/tendencias , Vasculitis Sistémica/complicaciones
4.
Am J Hum Genet ; 93(6): 1100-7, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24268661

RESUMEN

Congenital poikiloderma is characterized by a combination of mottled pigmentation, telangiectasia, and epidermal atrophy in the first few months of life. We have previously described a South African European-descent family affected by a rare autosomal-dominant form of hereditary fibrosing poikiloderma accompanied by tendon contracture, myopathy, and pulmonary fibrosis. Here, we report the identification of causative mutations in FAM111B by whole-exome sequencing. In total, three FAM111B missense mutations were identified in five kindreds of different ethnic backgrounds. The mutation segregated with the disease in one large pedigree, and mutations were de novo in two other pedigrees. All three mutations were absent from public databases and were not observed on Sanger sequencing of 388 ethnically matched control subjects. The three single-nucleotide mutations code for amino acid changes that are clustered within a putative trypsin-like cysteine/serine peptidase domain of FAM111B. These findings provide evidence of the involvement of FAM111B in congenital poikiloderma and multisystem fibrosis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Contractura/fisiopatología , Enfermedades Musculares/complicaciones , Mutación , Fibrosis Pulmonar/complicaciones , Síndrome Rothmund-Thomson/complicaciones , Síndrome Rothmund-Thomson/genética , Tendones/fisiopatología , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Fenotipo , Síndrome Rothmund-Thomson/diagnóstico , Adulto Joven
5.
Rheumatology (Oxford) ; 55(3): 470-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26424834

RESUMEN

OBJECTIVE: Outcome of JDM is highly heterogeneous. Our objective was to determine clinical and muscle biopsy features associated with poor outcome and response to treatment. METHODS: Clinical data and muscle biopsy were obtained from a monocentric cohort of 29 patients. Clinical subgroups were defined by latent class model analysis of initial and follow-up parameters. Myopathological features were analysed using validated scores. Capillary loss was determined on reconstructions of transversal sections and assessed in the different age groups to take into account variations of muscle capillarization during post-natal development. Regression models were used to identify initial predictors of therapeutic response. RESULTS: Two distinct homogeneous subgroups of patients were identified according to clinical severity and pathological findings. The smallest group of patients (7/29) presented with severe JDM. Compared with the other group (22/29), patients had more severe muscle weakness at disease onset, low remission rate at 12 months, frequent subcutaneous limb oedema or gastrointestinal (GI) involvement and higher myopathological scores (capillary dropout, perifascicular necrosis/regeneration, fibres with internal myonuclei and fibrosis subscores). Relevance of capillary dropout to JDM severity was substantiated by age-based analysis, confirming its major role in JDM pathophysiology. Most of these manifestations could be related to vasculopathy (limb oedema, GI involvement, capillary dropout). Furthermore, Childhood Myositis Assessment Scale <34 with either GI involvement or muscle endomysial fibrosis at disease onset were the best predictors of poor response to treatment. CONCLUSION: Vasculopathy is prominent in severe JDM. Simple criteria can be used at initial evaluation to identify patients requiring a more intensive therapy.


Asunto(s)
Dermatomiositis/patología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Enfermedades Vasculares/patología , Adolescente , Biopsia con Aguja , Capilares/patología , Niño , Preescolar , Estudios de Cohortes , Dermatomiositis/complicaciones , Dermatomiositis/fisiopatología , Progresión de la Enfermedad , Femenino , Francia , Humanos , Inmunohistoquímica , Masculino , Pronóstico , Valores de Referencia , Estudios Retrospectivos , Medición de Riesgo , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas , Enfermedades Vasculares/complicaciones , Enfermedades Vasculares/fisiopatología
6.
Brain ; 138(Pt 9): 2485-92, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26198592

RESUMEN

Idiopathic inflammatory myopathies can be classified as polymyositis, dermatomyositis, immune-mediated necrotizing myopathy, sporadic inclusion body myositis or non-specific myositis. Anti-Jo-1 antibody-positive patients are assigned to either polymyositis or dermatomyositis suggesting overlapping pathological features. We aimed to determine if anti-Jo-1 antibody-positive myopathy has a specific morphological phenotype. In a series of 53 muscle biopsies of anti-Jo-1 antibody-positive patients, relevant descriptive criteria defining a characteristic morphological pattern were identified. They were tested in a second series of anti-Jo-1 antibody-positive patients and compared to 63 biopsies from patients suffering from other idiopathic inflammatory myopathies. In anti-Jo-1 antibody-positive patients, necrotic fibres, which strongly clustered in perifascicular regions, were frequently observed. Sarcolemmal complement deposition was detected specifically in perifascicular areas. Inflammation was mainly located in the perimysium and around vessels in 90.6%. Perimysial fragmentation was observed in 90% of cases. Major histocompatibility complex class I staining was diffusely positive, with a perifascicular reinforcement. Multivariate analysis showed that criteria defining perifascicular pathology: perifascicular necrosis, atrophy, and perimysial fragmentation allow the distinction of anti-Jo-1 antibody-positive patients, among patients suffering from other idiopathic inflammatory myopathies. Anti-Jo-1 antibody-positive patients displayed perifascicular necrosis, whereas dermatomyositis patients exhibited perifascicular atrophy.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Autoanticuerpos/sangre , Dermatomiositis/patología , Músculo Esquelético/patología , Miositis por Cuerpos de Inclusión/patología , Miositis , Adulto , Femenino , Humanos , Ligasas/inmunología , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Músculo Esquelético/ultraestructura , Miositis/sangre , Miositis/inmunología , Miositis/patología , Estudios Retrospectivos , Estadísticas no Paramétricas
7.
BMC Med ; 13: 144, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-26082187

RESUMEN

BACKGROUND: Aluminum oxyhydroxide (alum) is a crystalline compound widely used as an immunologic adjuvant of vaccines. Concerns linked to alum particles have emerged following recognition of their causative role in the so-called macrophagic myofasciitis (MMF) lesion in patients with myalgic encephalomyelitis, revealing an unexpectedly long-lasting biopersistence of alum within immune cells and a fundamental misconception of its biodisposition. Evidence that aluminum-coated particles phagocytozed in the injected muscle and its draining lymph nodes can disseminate within phagocytes throughout the body and slowly accumulate in the brain further suggested that alum safety should be evaluated in the long term. However, lack of specific staining makes difficult the assessment of low quantities of bona fide alum adjuvant particles in tissues. METHODS: We explored the feasibility of using fluorescent functionalized nanodiamonds (mfNDs) as a permanent label of alum (Alhydrogel(®)). mfNDs have a specific and perfectly photostable fluorescence based on the presence within the diamond lattice of nitrogen-vacancy centers (NV centers). As the NV center does not bleach, it allows the microspectrometric detection of mfNDs at very low levels and in the long-term. We thus developed fluorescent nanodiamonds functionalized by hyperbranched polyglycerol (mfNDs) allowing good coupling and stability of alum:mfNDs (AluDia) complexes. Specificities of AluDia complexes were comparable to the whole reference vaccine (anti-hepatitis B vaccine) in terms of particle size and zeta potential. RESULTS: In vivo, AluDia injection was followed by prompt phagocytosis and AluDia particles remained easily detectable by the specific signal of the fND particles in the injected muscle, draining lymph nodes, spleen, liver and brain. In vitro, mfNDs had low toxicity on THP-1 cells and AluDia showed cell toxicity similar to alum alone. Expectedly, AluDia elicited autophagy, and allowed highly specific detection of small amounts of alum in autophagosomes. CONCLUSIONS: The fluorescent nanodiamond technology is able to overcome the limitations of previously used organic fluorophores, thus appearing as a choice methodology for studying distribution, persistence and long-term neurotoxicity of alum adjuvants and beyond of other types of nanoparticles.


Asunto(s)
Compuestos de Alumbre/efectos adversos , Colorantes Fluorescentes/farmacología , Nanodiamantes , Coloración y Etiquetado/métodos , Adyuvantes Inmunológicos/efectos adversos , Adulto , Fascitis/inducido químicamente , Femenino , Humanos , Miositis/inducido químicamente
8.
Neuropathol Appl Neurobiol ; 41(7): 952-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25711697

RESUMEN

OBJECTIVE: To analyse the paradox of a lack of giant cell formation and fibrosis in chronic lesions of macrophagic myofasciitis (MMF) in comparison with muscular sarcoidosis (MuS). METHODS: Inflammatory lesions and contiguous muscle regions from biopsy samples of 10 patients with MuS and 10 patients with MMF were cut out by laser microdissection. Mediators of the T helper cell (Th)1 inducing classical macrophage activation (e.g. STAT1, IFNγ and CXCR3), and Th2 inducing alternative activation of macrophages (e.g. CD206/MRC1, STAT6, SOCS1), molecules involved in development of fibrosis (e.g. TGFß) and giant cells (e.g. TYROBP), were assessed by immunohistochemistry and real-time polymerase chain reaction (PCR). RESULTS: STAT6-induced Th2 immunity was associated with up-regulated gene expression of MRC1, SOCS1 and TGFB in inflammatory foci, in comparison with adjacent tissue. TYROBP and TREM2, genes regulating giant cell formation, were more strongly expressed in lesions of MuS patients than in those of MMF. TGFß co-localized with CD206(+) macrophages in MuS but not in MMF. Conversely, Th1 immunity was illustrated by STAT1 staining both in macrophages and myofibres in MuS, but not in MMF. Also, STAT1-induced IFNG and CXCR3 expression in lesions and the surrounding tissue was elevated compared with normal controls, but without statistically significant differences. CONCLUSION: Giant cell and typical granuloma formations, including fibrogenesis, is dependent on two main mechanisms, both involving specific macrophage activation: a strong Th2-M2 polarization and a significant expression of TYROBP and TGFß in macrophages. The low-grade alternative activation of macrophages in MMF lesions and poor TYROBP and TGFßco-expression are obviously insufficient to produce giant cells.


Asunto(s)
Fascitis/inmunología , Macrófagos/inmunología , Músculo Esquelético/inmunología , Miositis/inmunología , Sarcoidosis/inmunología , Células Th2/inmunología , Adulto , Anciano , Fascitis/patología , Femenino , Humanos , Activación de Macrófagos , Macrófagos/patología , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Miositis/patología , Sarcoidosis/patología , Células Th2/patología , Adulto Joven
9.
Am J Pathol ; 182(6): 2298-309, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624156

RESUMEN

Dysferlinopathy refers to a group of autosomal recessive muscular dystrophies due to mutations in the dysferlin gene causing deficiency of a membrane-bound protein crucially involved in plasma membrane repair. The condition is characterized by marked clinical heterogeneity, the different phenotypes/modes of presentation being unrelated to the genotype. For unknown reasons, patients are often remarkably active before the onset of symptoms. Dysferlin deficiency-related persistence of mechanically induced sarcolemma disruptions causes myofiber damage and necrosis. We postulate that limited myodamage may initially remain hidden with well-preserved resistance to physical strains. By subjecting dysferlin-deficient B6.A/J-Dysf(prmd) mice to long-term swimming exercise, we observed that concentric/isometric strain improved muscle strength and alleviated muscular dystrophy by limiting the accumulation of membrane lesions. By contrast, eccentric strain induced by long-term running in a wheel worsened the dystrophic process. Myofiber damage induced by eccentric strain increased with age, reflecting the accumulation of non-necrotic membrane lesions up to a critical threshold. This phenomenon was modulated by daily spontaneous activity. Transposed to humans, our results may suggest that the past activity profile shapes the clinical phenotype of the myopathy and that patients with dysferlinopathy should likely benefit from concentric exercise-based physiotherapy.


Asunto(s)
Distrofia Muscular de Cinturas/rehabilitación , Condicionamiento Físico Animal/fisiología , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Membrana Celular/ultraestructura , Disferlina , Locomoción/fisiología , Proteínas de la Membrana/deficiencia , Ratones , Ratones Mutantes , Microscopía Electrónica , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/patología , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/etiología , Distrofia Muscular de Cinturas/patología , Distrofia Muscular de Cinturas/fisiopatología , Necrosis , Músculo Cuádriceps/ultraestructura , Carrera/fisiología , Natación/fisiología
10.
Bull Acad Natl Med ; 198(1): 37-48; discussion 49-53, 2014 Jan.
Artículo en Francés | MEDLINE | ID: mdl-26259285

RESUMEN

Aluminium oxyhydroxide (alum), a nanocrystalline compound that forms agglomerates, has been widely used as a vaccine adjuvant since 1927, but the mechanisms by which it stimulates immune responses remain poorly understood. Although generally well tolerated, alum may occasionally cause chronic health problems in presumably susceptible individuals. Some individuals may rarely develop delayed-onset diffuse myalgia, chronic exhaustion and cognitive dysfunction, associated with long-term persistence (up to 12 years) of alum-loaded macrophages at site of i.m. immunization, defining so-called macrophagic myofasciitis (MMF). Symptoms are consistent with the chronic fatigue/myalgic encephalomyelitis (CFS/ME) syndrome, and have been used as a paradigm of the "autoimmune/inflammatory syndrome induced by adjuvants" (ASIA). Cognitive dysfunction is reminiscent of that described in workers exposed to inhaled Al particles. Individual susceptibility may influence both alum biopersistence and difusion away from injection sites. Biopersistent particles such as fluorescent alum-coated nanohybrids, when injected into mouse muscle, are captured by monocyte-lineage cells and then carried to distant organs, draining lymph nodes and blood, probably via the thoracic duct, with delayed and accumulative translocation to the brain (microglial cells). Brain penetration occurs at extremely low levels in normal conditions, possibly explaining the good tolerance of alum despite its high neurotoxic potential. However, systemic diffusion is considerably enhanced by the potentiating effect of MCP-1, the main monocyte chemoattractant factor, the production of which is subject to marked variations linked to age and to genetic and environmental factors. Selective MCP-1 elevation is the only known circulating biomarker of MMF.


Asunto(s)
Adyuvantes Inmunológicos/farmacocinética , Compuestos de Alumbre/farmacocinética , Adyuvantes Inmunológicos/efectos adversos , Compuestos de Alumbre/efectos adversos , Quimiocina CCL2/análisis , Fascitis/inducido químicamente , Humanos , Inyecciones Intramusculares , Macrófagos/química , Miositis/inducido químicamente , Vacunas/química
11.
Toxics ; 12(7)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-39058143

RESUMEN

(1) Background: Macrophagic myofasciitis (MMF) is an inflammatory histopathological lesion demonstrating long-term biopersistence of vaccine-derived aluminum adjuvants within muscular phagocytic cells. Affected patients suffer from widespread myalgia and severe fatigue consistent with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), a poorly understood disorder suspected to result from chronic immune stimulation by infectious and inorganic particles. (2) Methods: In this study we determined the immuno-metabolic properties of MMF phagocytic cells compared to controls, at rest and upon exposure to aluminum oxyhydroxide adjuvant, with or without adsorbed antigens, using protein quantification and an oxygen consumption assay. (3) Results: MMF and control cells similarly internalized the adjuvant and vaccine but MMF cells specifically expressed Rubicon and Nox2, two molecules unique to the LC3-associated phagocytosis (LAP) machinery, a non-canonical autophagic pathway able to downregulate canonical autophagy. MMF cells exhibited an altered inflammatory secretome, producing more pain-inducing CXC chemokines and less TNF-α than controls, consistent with chronic myalgia and exhaustion of the immune system previously documented in ME/CFS. MMF cells exhibited mitochondrial metabolism dysfunction, with exacerbated reaction to adjuvanted vaccine, contrasting with limited spare respiratory capacity and marked proton leak weakening energy production. (4) Conclusions: MMF phagocytes seemingly use LAP to handle aluminum oxyhydroxide vaccine particles, secrete pain-inducing molecules, and exhibit exacerbated metabolic reaction to the vaccine with limited capacity to respond to ongoing energetic requests.

12.
Glia ; 61(10): 1645-58, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23893349

RESUMEN

As neuroinflammatory processes are involved in the pathogenesis of Parkinson's disease (PD), we provide several key data describing the time-course of microglial accumulation in relation with behavioral alterations and neurodegeneration in a murine model of PD induced by intrastriatal injection of 6-hydroxydopamine (6-OHDA). Our study argues for a major role of microglia which accumulation is somehow early and transient in spite of the neuronal loss progression. Moreover, we observed less 6-OHDA-induced neurodegeneration associated with less inflammatory reaction in DAP-12 Knock-In mice. The direct cell-to-cell contacts that may support physical interactions between microglia and altered dopaminergic neurons are ill-defined, while it is currently hypothesized that microglia support an immune-mediated amplification of neurodegeneration by establishing a molecular cross talk with neurons. Indeed, we sought to map microglia/neuron appositions in substantia nigra (SN) of 6-OHDA injected C57Bl/6 mice and CX3CR1/(GFP/+) mice. Confocal immunofluorescence analyses followed by 3D reconstitutions reveal close appositions between the soma of TH+ neurons and microglial cell bodies and ramifications. Interestingly, some microglial ramifications penetrated TH(+) somas and about 40% of GFP(+) microglial cells in the injured SN harbored TH(+) intracytoplasmic inclusions. These results suggest a direct cross talk between neurons and microglia that may exert a microphagocytic activity toward TH+ neurons. Altogether, these results obtained in a murine PD model may participate in the understanding of microglial cells' function in neurodegenerative diseases.


Asunto(s)
Adrenérgicos/toxicidad , Comunicación Celular/fisiología , Microglía/fisiología , Neuronas/fisiología , Oxidopamina/toxicidad , Enfermedad de Parkinson , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos de Diferenciación/metabolismo , Apomorfina , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Recuento de Células , Modelos Animales de Enfermedad , Agonistas de Dopamina , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Imagenología Tridimensional , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Microglía/efectos de los fármacos , Microscopía Confocal , Neuronas/citología , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Receptores de Interleucina-8A/deficiencia , Rotación , Sustancia Negra/patología , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
13.
J Exp Med ; 204(5): 1057-69, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17485518

RESUMEN

Macrophages (MPs) are important for skeletal muscle regeneration in vivo and may exert beneficial effects on myogenic cell growth through mitogenic and antiapoptotic activities in vitro. However, MPs are highly versatile and may exert various, and even opposite, functions depending on their activation state. We studied monocyte (MO)/MP phenotypes and functions during skeletal muscle repair. Selective labeling of circulating MOs by latex beads in CX3CR1(GFP/+) mice showed that injured muscle recruited only CX3CR1(lo)/Ly-6C(+) MOs from blood that exhibited a nondividing, F4/80(lo), proinflammatory profile. Then, within muscle, these cells switched their phenotype to become proliferating antiinflammatory CX3CR1(hi)/Ly-6C(-) cells that further differentiated into F4/80(hi) MPs. In vitro, phagocytosis of muscle cell debris induced a switch of proinflammatory MPs toward an antiinflammatory phenotype releasing transforming growth factor beta1. In co-cultures, inflammatory MPs stimulated myogenic cell proliferation, whereas antiinflammatory MPs exhibited differentiating activity, assessed by both myogenin expression and fusion into myotubes. Finally, depletion of circulating MOs in CD11b-diphtheria toxin receptor mice at the time of injury totally prevented muscle regeneration, whereas depletion of intramuscular F4/80(hi) MPs at later stages reduced the diameter of regenerating fibers. In conclusion, injured skeletal muscle recruits MOs exhibiting inflammatory profiles that operate phagocytosis and rapidly convert to antiinflammatory MPs that stimulate myogenesis and fiber growth.


Asunto(s)
Diferenciación Celular/inmunología , Macrófagos/inmunología , Monocitos/citología , Desarrollo de Músculos/fisiología , Músculo Esquelético/lesiones , Animales , Antígeno CD11b , Receptor 1 de Quimiocinas CX3C , Cartilla de ADN , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Ratones , Ratones Mutantes , Microesferas , Desarrollo de Músculos/inmunología , Músculo Esquelético/inmunología , Fagocitosis/inmunología , Receptores de Quimiocina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
BMC Med ; 11: 99, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23557144

RESUMEN

BACKGROUND: Long-term biodistribution of nanomaterials used in medicine is largely unknown. This is the case for alum, the most widely used vaccine adjuvant, which is a nanocrystalline compound spontaneously forming micron/submicron-sized agglomerates. Although generally well tolerated, alum is occasionally detected within monocyte-lineage cells long after immunization in presumably susceptible individuals with systemic/neurologic manifestations or autoimmune (inflammatory) syndrome induced by adjuvants (ASIA). METHODS: On the grounds of preliminary investigations in 252 patients with alum-associated ASIA showing both a selective increase of circulating CCL2, the major monocyte chemoattractant, and a variation in the CCL2 gene, we designed mouse experiments to assess biodistribution of vaccine-derived aluminum and of alum-particle fluorescent surrogates injected in muscle. Aluminum was detected in tissues by Morin stain and particle induced X-ray emission) (PIXE) Both 500 nm fluorescent latex beads and vaccine alum agglomerates-sized nanohybrids (Al-Rho) were used. RESULTS: Intramuscular injection of alum-containing vaccine was associated with the appearance of aluminum deposits in distant organs, such as spleen and brain where they were still detected one year after injection. Both fluorescent materials injected into muscle translocated to draining lymph nodes (DLNs) and thereafter were detected associated with phagocytes in blood and spleen. Particles linearly accumulated in the brain up to the six-month endpoint; they were first found in perivascular CD11b+ cells and then in microglia and other neural cells. DLN ablation dramatically reduced the biodistribution. Cerebral translocation was not observed after direct intravenous injection, but significantly increased in mice with chronically altered blood-brain-barrier. Loss/gain-of-function experiments consistently implicated CCL2 in systemic diffusion of Al-Rho particles captured by monocyte-lineage cells and in their subsequent neurodelivery. Stereotactic particle injection pointed out brain retention as a factor of progressive particle accumulation. CONCLUSION: Nanomaterials can be transported by monocyte-lineage cells to DLNs, blood and spleen, and, similarly to HIV, may use CCL2-dependent mechanisms to penetrate the brain. This occurs at a very low rate in normal conditions explaining good overall tolerance of alum despite its strong neurotoxic potential. However, continuously escalating doses of this poorly biodegradable adjuvant in the population may become insidiously unsafe, especially in the case of overimmunization or immature/altered blood brain barrier or high constitutive CCL-2 production.


Asunto(s)
Adyuvantes Inmunológicos/farmacocinética , Compuestos de Alumbre/farmacocinética , Encéfalo/inmunología , Quimiocina CCL2/metabolismo , Músculos/inmunología , Nanopartículas , Virión/patogenicidad , Animales , Asia , Barrera Hematoencefálica/inmunología , Encéfalo/metabolismo , Movimiento Celular , Quimiocina CCL2/sangre , Humanos , Inmunización/efectos adversos , Inyecciones Intramusculares/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Músculos/metabolismo , Distribución Tisular , Vacunas/administración & dosificación , Vacunas/efectos adversos
15.
Ann Rheum Dis ; 72(3): 445-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22962315

RESUMEN

OBJECTIVES: The pathophysiology of dermatomyositis (DM) remains unclear, combining immunopathological mechanisms with ischaemic changes regarded as a consequence of membranolytic attack complex (MAC)-induced capillary destruction. The study is a reappraisal of the microvascular involvement in light of the microvascular organisation in normal human muscle. METHODS: Muscle microvasculature organisation was analysed using 3D reconstructions of serial sections immunostained for CD31, and histoenzymatic detection of endogenous alkaline phosphatase activity of microvessels. An unbiased point pattern analysis-based method was used to evaluate focal capillary loss. Double immunostainings identified cell types showing MAC deposits. RESULTS: The normal arterial tree includes perimysial arcade arteries, transverse arteries penetrating perpendicularly into the endomysium and terminal arterioles feeding a microvascular unit (MVU) of six to eight capillaries contacting an average of five myofibres. Amyopathic DM cases (n=3) and non-necrotic fascicles of early DM cases (n=27), showed patchy capillary loss in the form of 6-by-6 capillary drop-out, corresponding to depletion of one or multiple MVUs. MAC deposits were also clustered (5-8 immunostained structures, including endothelial cells, but also pericytes, mesenchymal cells and myosatellite cells). CONCLUSIONS: Capillary loss may not be the primary cause of muscle ischaemia in DM. The primary event rather stands upstream, probably at the level of perimysial arcade arteries around which inflammatory infiltrates predominate and which lumen may show narrowing in chronic DM. Ischaemia-reperfusion injury, which is favoured by autoimmune backgrounds in experimental models and which activates the complement cascade in capillaries, could represent an hitherto unsuspected (and potentially preventable) mechanism of muscle damage in DM.


Asunto(s)
Dermatomiositis/patología , Microvasos/patología , Músculo Esquelético/irrigación sanguínea , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Imagenología Tridimensional , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Arthritis Rheum ; 64(10): 3441-51, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22674045

RESUMEN

OBJECTIVE: To investigate the influence of myoinjury on antigen presentation to T cells in draining lymph nodes (LNs). METHODS: Muscle crush was performed in mice injected with exogenous ovalbumin (OVA) and in transgenic SM-OVA mice expressing OVA as a muscle-specific self antigen. Antigen exposure and the resulting stimulation of T cell proliferation in draining LNs was assessed by transferring carboxyfluorescein succinimidyl ester (CFSE)-labeled OVA-specific CD8+ and CD4+ T cells from OT-I and OT-II mice and by measuring the dilution of CFSE, which directly reflects their proliferation. The role of monocyte-derived dendritic cells (DCs) in T cell priming was assessed using pharmacologic blockade of DC migration. Immunofluorescence was used to detect CD8+ T cells, inflammatory monocyte-derived DCs, and type I major histocompatibility complex (MHC)-expressing myofibers in crushed muscle, and to assess expression of perforin, interferon-γ (IFNγ), interleukin-2 (IL-2), IL-10, and transforming growth factor ß1 (TGFß1). RESULTS: OVA injection into intact muscle induced strong proliferation of CD4+ and CD8+ T cells, indicating efficient exposure of soluble antigens in draining LNs. OVA-specific CD8+ T cell proliferation in draining LNs of SM-OVA mice required myoinjury and was unaffected by pharmacologic inhibition of monocyte-derived DC migration. On day 7 postinjury, activated CD8+ T cells expressing perforin, IFNγ and IL-2 were transiently detected in crushed muscle, and these cells were in close contact with class I MHC-positive regenerating myofibers. Beginning on day 7, the immunosuppressive cytokines IL-10 and TGFß1 were conspicuously expressed by CD11b+ cells, and CD8+ T cells rapidly disappeared from the healing muscle. CONCLUSION: Myofiber damage induces an episode of muscle antigen-specific CD8+ T cell proliferation in draining LNs. Activated CD8+ T cells transiently infiltrate the injured muscle, with prompt control by immunosuppressive cues. Inadequate control might favor sustained autoimmune myositis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ganglios Linfáticos/citología , Activación de Linfocitos/inmunología , Músculo Esquelético/lesiones , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular/inmunología , Proliferación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo
17.
Muscle Nerve ; 45(6): 803-14, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22581532

RESUMEN

INTRODUCTION: Duchenne Muscular Dystrophy (DMD) is characterized by the lack of dystrophin that leads to severe myofiber degeneration. We have shown that endomysial fibrosis is correlated with age at ambulation loss in DMD patients. However, the dystrophin-deficient mdx mouse does not have fibrotic lesions in adult limb muscles. Here, we describe a model of chronic mechanical muscle injury that triggers chronic lesions in mdx hindlimb muscle. METHODS: Micromechanical injuries were performed daily in tibialis anterior muscles for 2 weeks. RESULTS: Endomysial fibrosis appeared beginning 1 week post-injury, remained stable for 3 months and was associated with loss of specific maximal force. Fibrosis was associated with an increased expression of factors involved in fibrogenesis including α-smooth muscle actin, connective tissue growth factor, and lysyl oxidase, which colocalized with collagen deposits. CONCLUSIONS: This induced fibrotic dystrophic model may be useful to study mechanisms of fibrosis in dystrophinopathies and to evaluate antifibrotic treatments.


Asunto(s)
Modelos Animales de Enfermedad , Miembro Posterior , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Distrofias Musculares/patología , Distrofia Muscular de Duchenne/patología , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Colágeno/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Distrofina/deficiencia , Distrofina/genética , Distrofina/metabolismo , Fibrosis , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Proteína-Lisina 6-Oxidasa/metabolismo
18.
Autophagy ; 18(11): 2519-2536, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35383530

RESUMEN

At a time when complex diseases affect globally 280 million people and claim 14 million lives every year, there is an urgent need to rapidly increase our knowledge into their underlying etiologies. Though critical in identifying the people at risk, the causal environmental factors (microbiome and/or pollutants) and the affected pathophysiological mechanisms are not well understood. Herein, we consider the variations of autophagy-related (ATG) genes at the heart of mechanisms of increased susceptibility to environmental stress. A comprehensive autophagy genomic resource is presented with 263 single nucleotide polymorphisms (SNPs) for 69 autophagy-related genes associated with 117 autoimmune, inflammatory, infectious, cardiovascular, neurological, respiratory, and endocrine diseases. We thus propose the term 'autophagopathies' to group together a class of complex human diseases the etiology of which lies in a genetic defect of the autophagy machinery, whether directly related or not to an abnormal flux in autophagy, LC3-associated phagocytosis, or any associated trafficking. The future of precision medicine for common diseases will lie in our ability to exploit these ATG SNP x environment relationships to develop new polygenetic risk scores, new management guidelines, and optimal therapies for afflicted patients.Abbreviations: ATG, autophagy-related; ALS-FTD, amyotrophic lateral sclerosis-frontotemporal dementia; ccRCC, clear cell renal cell carcinoma; CD, Crohn disease; COPD, chronic obstructive pulmonary disease; eQTL, expression quantitative trait loci; HCC, hepatocellular carcinoma; HNSCC, head and neck squamous cell carcinoma; GTEx, genotype-tissue expression; GWAS, genome-wide association studies; LAP, LC3-associated phagocytosis; LC3-II, phosphatidylethanolamine conjugated form of LC3; LD, linkage disequilibrium; LUAD, lung adenocarcinoma; MAF, minor allele frequency; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; NSCLC, non-small cell lung cancer; OS, overall survival; PtdIns3K CIII, class III phosphatidylinositol 3 kinase; PtdIns3P, phosphatidylinositol-3-phosphate; SLE, systemic lupus erythematosus; SNPs, single-nucleotide polymorphisms; mQTL, methylation quantitative trait loci; ULK, unc-51 like autophagy activating kinase; UTRs, untranslated regions; WHO, World Health Organization.


Asunto(s)
Esclerosis Amiotrófica Lateral , Carcinoma Hepatocelular , Carcinoma de Pulmón de Células no Pequeñas , Demencia Frontotemporal , Neoplasias de Cabeza y Cuello , Neoplasias Hepáticas , Neoplasias Pulmonares , Humanos , Autofagia/genética , Medicina de Precisión , Estudio de Asociación del Genoma Completo , Carcinoma de Células Escamosas de Cabeza y Cuello , Polimorfismo Genético
19.
Mol Biol Cell ; 18(4): 1397-409, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17287398

RESUMEN

Genetically engineered mice (Myf5nLacZ/+, Myf5GFP-P/+) allowing direct muscle satellite cell (SC) visualization indicate that, in addition to being located beneath myofiber basal laminae, SCs are strikingly close to capillaries. After GFP(+) bone marrow transplantation, blood-borne cells occupying SC niches previously depleted by irradiation were similarly detected near vessels, thereby corroborating the anatomical stability of juxtavascular SC niches. Bromodeoxyuridine pulse-chase experiments also localize quiescent and less quiescent SCs near vessels. SCs, and to a lesser extent myonuclei, were nonrandomly associated with capillaries in humans. Significantly, they were correlated with capillarization of myofibers, regardless to their type, in normal muscle. They also varied in paradigmatic physiological and pathological situations associated with variations of capillary density, including amyopathic dermatomyositis, a unique condition in which muscle capillary loss occurs without myofiber damage, and in athletes in whom capillaries increase in number. Endothelial cell (EC) cultures specifically enhanced SC growth, through IGF-1, HGF, bFGF, PDGF-BB, and VEGF, and, accordingly, cycling SCs remained mainly juxtavascular. Conversely, differentiating myogenic cells were both proangiogenic in vitro and spatiotemporally associated with neoangiogenesis in muscular dystrophy. Thus, SCs are largely juxtavascular and reciprocally interact with ECs during differentiation to support angio-myogenesis.


Asunto(s)
Capilares/citología , Células Endoteliales/fisiología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Adulto , Anciano , Animales , Bromodesoxiuridina/metabolismo , Capilares/fisiología , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Perros , Células Endoteliales/citología , Endotelio Vascular/citología , Humanos , Péptidos y Proteínas de Señalización Intercelular/aislamiento & purificación , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Neovascularización Fisiológica , Ratas , Células Satélite del Músculo Esquelético/efectos de los fármacos
20.
Biomedicines ; 8(3)2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-32121613

RESUMEN

Macroautophagy (hereafter referred to as autophagy) is an evolutionarily conserved catabolic process whose loss-of-function has been linked to a growing list of pathologies. Knockout mouse models of key autophagy genes have been instrumental in the demonstration of the critical functions of autophagy, but they display early lethality, neurotoxicity and unwanted autophagy-independent phenotypes, limiting their applications for in vivo studies. To avoid problems encountered with autophagy-null transgenic mice, we investigated the possibility of disturbing autophagy pharmacologically in the long term. Hydroxychloroquine (HCQ) ip injections were done in juvenile and adult C57bl/6j mice, at range doses adapted from the human malaria prophylactic treatment. The impact on autophagy was assessed by western-blotting, and juvenile neurodevelopment and adult behaviours were evaluated for four months. Quite surprisingly, our results showed that HCQ treatment in conditions used in this study neither impacted autophagy in the long term in several tissues and organs nor altered neurodevelopment, adult behaviour and motor capabilities. Therefore, we recommend for future long-term in vivo studies of autophagy, to use genetic mouse models allowing conditional inhibition of selected Atg genes in appropriate lineage cells instead of HCQ treatment, until it could be successfully revisited using higher HCQ doses and/or frequencies with acceptable toxicity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA