Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Neuropathol Commun ; 11(1): 167, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37858263

RESUMEN

Duchenne muscular dystrophy (DMD) is a devastating X-linked muscular disease, caused by mutations in the DMD gene encoding Dystrophin and affecting 1:5000 boys worldwide. Lack of Dystrophin leads to progressive muscle wasting and degeneration resulting in cardiorespiratory failure. Despite the absence of a definitive cure, innovative therapeutic avenues are emerging. Myopathologic studies are important to further understand the biological mechanisms of the disease and to identify histopathologic benchmarks for clinical evaluations. We conducted a myopathologic analysis on twenty-four muscle biopsies from DMD patients, with particular emphasis on regeneration, fibro-adipogenic progenitors and muscle stem cells behavior. We describe an increase in content of fibro-adipogenic progenitors, central orchestrators of fibrotic progression and lipid deposition, concurrently with a decline in muscle regenerative capacity. This regenerative impairment strongly correlates with compromised activation and expansion of muscle stem cells. Furthermore, our study uncovers an early acquisition of a senescence phenotype by DMD-afflicted muscle stem cells. Here we describe the myopathologic trajectory intrinsic to DMD and establish muscle stem cell senescence as a pivotal readout for future therapeutic interventions.


Asunto(s)
Distrofia Muscular de Duchenne , Células Satélite del Músculo Esquelético , Humanos , Masculino , Distrofina/genética , Fibrosis , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Regeneración/genética , Senescencia Celular/genética
2.
Sci Transl Med ; 15(685): eadd5275, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36857434

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe and progressive myopathy leading to motor and cardiorespiratory impairment. We analyzed samples from patients with DMD and a preclinical rat model of severe DMD and determined that compromised repair capacity of muscle stem cells in DMD is associated with early and progressive muscle stem cell senescence. We also found that extraocular muscles (EOMs), which are spared by the disease in patients, contain muscle stem cells with long-lasting regenerative potential. Using single-cell transcriptomics analysis of muscles from a rat model of DMD, we identified the gene encoding thyroid-stimulating hormone receptor (Tshr) as highly expressed in EOM stem cells. Further, TSHR activity was involved in preventing senescence. Forskolin, which activates signaling downstream of TSHR, was found to reduce senescence of skeletal muscle stem cells, increase stem cell regenerative potential, and promote myogenesis, thereby improving muscle function in DMD rats. These findings indicate that stimulation of adenylyl cyclase leads to muscle repair in DMD, potentially providing a therapeutic approach for patients with the disease.


Asunto(s)
Distrofia Muscular de Duchenne , Receptores de Tirotropina , Animales , Ratas , Receptores Acoplados a Proteínas G , Fibras Musculares Esqueléticas , Células Madre , Regeneración , Tirotropina
3.
Matrix Biol ; 112: 90-115, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35963565

RESUMEN

Obesity triggers skeletal muscle physio-pathological alterations. However, the crosstalk between adipose tissue and myogenic cells remains poorly understood during obesity. We identified NID-1 among the adipose tissue secreted factors impairing myogenic potential of human myoblasts and murine muscle stem cells in vitro. Mice under High Fat Diet (HFD) displayed increased NID-1 expression in the skeletal muscle endomysium associated with intramuscular fat adipose tissue expansion and compromised muscle stem cell function. We show that NID-1 is highly secreted by skeletal muscle fibro-adipogenic/mesenchymal progenitors (FAPs) during obesity. We demonstrate that increased muscle NID-1 impairs muscle stem cells proliferation and primes the fibrogenic differentiation of FAPs, giving rise to an excessive deposition of extracellular matrix. Finally, we propose a model in which obesity leads to skeletal muscle extracellular matrix remodeling by FAPs, mediating the alteration of myogenic function by adipose tissue and highlighting the key role of NID-1 in the crosstalk between adipose tissue and skeletal muscle.


Asunto(s)
Adipogénesis , Desarrollo de Músculos , Animales , Diferenciación Celular , Matriz Extracelular , Humanos , Ratones , Músculo Esquelético/metabolismo , Obesidad/genética , Obesidad/metabolismo
4.
Acta Neuropathol Commun ; 10(1): 60, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35468843

RESUMEN

Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disorder caused by mutations in the Dystrophin gene and for which there is currently no cure. To bridge the gap between preclinical and therapeutic evaluation studies, we have generated a rat model for DMD that carries an exon 52 deletion (R-DMDdel52) causing a complete lack of dystrophin protein. Here we show that R-DMDdel52 animals recapitulated human DMD pathophysiological trajectory more faithfully than the mdx mouse model. We report that R-DMDdel52 rats displayed progressive and severe skeletal muscle loss associated with fibrotic deposition, fat infiltration and fibre type switch. Early fibrosis was also apparent in the cardiac muscle. These histological modifications led to severe muscle, respiratory and cardiac functional impairments leading to premature death around 1 year. Moreover, DMD muscle exhibited systemic inflammation with a mixed M1/M2 phenotype. A comparative single cell RNAseq analysis of the diaphragm muscle was performed, revealing cellular populations alteration and molecular modifications in all muscle cell types. We show that DMD fibroadipogenic progenitors produced elevated levels of cartilage oligomeric matrix protein, a glycoprotein responsible for modulating homeostasis of extracellular matrix, and whose increased concentration correlated with muscle fibrosis both in R-DMDdel52 rats and human patients. Fibrosis is a component of tissue remodelling impacting the whole musculature of DMD patients, at the tissue level but most importantly at the functional level. We therefore propose that this specific biomarker can optimize the prognostic monitoring of functional improvement of patients included in clinical trials.


Asunto(s)
Distrofia Muscular de Duchenne , Animales , Biomarcadores , Proteína de la Matriz Oligomérica del Cartílago/uso terapéutico , Distrofina/metabolismo , Fibrosis , Humanos , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/terapia , Ratas
5.
STAR Protoc ; 2(3): 100650, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34278336

RESUMEN

Platelet preparations are commonly used in the clinic in combination with mesenchymal stem cells (MSCs) to improve their wound healing capacity and optimize their therapeutic efficacy following their delivery into diseased tissues. To investigate the mechanisms by which platelets enhance the repair properties of MSCs, we detail a protocol using a humanized mouse model for excisional wounds to study by reverse transcription real-time PCR whether human platelets alter the therapeutic efficacy of grafted human MSCs. For complete details on the use and execution of this protocol, please refer to Levoux et al. (2021).


Asunto(s)
Perfilación de la Expresión Génica/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Transfusión de Plaquetas/métodos , Heridas Penetrantes/terapia , Animales , Plaquetas , Xenoinjertos , Humanos , Masculino , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Heridas Penetrantes/genética
7.
Cell Metab ; 33(2): 283-299.e9, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33400911

RESUMEN

Platelets are known to enhance the wound-healing activity of mesenchymal stem cells (MSCs). However, the mechanism by which platelets improve the therapeutic potential of MSCs has not been elucidated. Here, we provide evidence that, upon their activation, platelets transfer respiratory-competent mitochondria to MSCs primarily via dynamin-dependent clathrin-mediated endocytosis. We found that this process enhances the therapeutic efficacy of MSCs following their engraftment in several mouse models of tissue injury, including full-thickness cutaneous wound and dystrophic skeletal muscle. By combining in vitro and in vivo experiments, we demonstrate that platelet-derived mitochondria promote the pro-angiogenic activity of MSCs via their metabolic remodeling. Notably, we show that activation of the de novo fatty acid synthesis pathway is required for increased secretion of pro-angiogenic factors by platelet-preconditioned MSCs. These results reveal a new mechanism by which platelets potentiate MSC properties and underline the importance of testing platelet mitochondria quality prior to their clinical use.


Asunto(s)
Plaquetas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cicatrización de Heridas
8.
RMD Open ; 5(1): e000811, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30886734

RESUMEN

Objective: The role of interferons (IFN) in the pathophysiology of primary inflammatory and dysimmune myopathies (IDM) is increasingly investigated, notably because specific neutralisation approaches may constitute promising therapeutic tracks. In present work we analysed the muscular expression of specific IFNα/ß and IFNγ-stimulated genes in patients with various types of IDM. Methods: 39 patients with IDM with inclusion body myositis (IBM, n=9), dermatomyositis (DM, n=10), necrotising autoimmune myopathies (NAM, n=10) and antisynthetase myositis (ASM, n=10), and 10 controls were included. Quantification of expression levels of IFNγ, ISG15, an IFNα/ß-inducible gene and of six IFNγ-inducible genes (GBP2, HLA-DOB, HLA-DPB, CIITA, HLA-DRB and HLA-DMB) was performed on muscle biopsy samples. Results: DM usually associated with strong type I IFNα/ß signature, IBM and ASM with prominent type II IFNγ signature and NAM with neither type I nor type II IFN signature. Immunofluorescence study in ASM and IBM showed myofibre expression of major histocompatibility class 2 (MHC-2) and CIITA, confirming the induction of the IFNγ pathway. Furthermore, MHC-2-positive myofibres were observed in close proximity to CD8+ T cells which produce high levels of IFNγ. Conclusion: Distinct IFN signatures allow a more distinct segregation of IDMs and myofibre MHC-2 expression is a reliable biomarker of type II IFN signature.


Asunto(s)
Susceptibilidad a Enfermedades , Interferones/metabolismo , Enfermedades Musculares/etiología , Enfermedades Musculares/metabolismo , Miositis/etiología , Miositis/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Biopsia , Diagnóstico Diferencial , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/diagnóstico , Miositis/diagnóstico , Transducción de Señal
9.
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
10.
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
11.
Proc Natl Acad Sci U S A ; 109(47): E3231-40, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23129614

RESUMEN

Skeletal muscle regeneration mainly depends on satellite cells, a population of resident muscle stem cells. However, our understanding of the molecular mechanisms underlying satellite cell activation is still largely undefined. Here, we show that Cripto, a regulator of early embryogenesis, is a novel regulator of muscle regeneration and satellite cell progression toward the myogenic lineage. Conditional inactivation of cripto in adult satellite cells compromises skeletal muscle regeneration, whereas gain of function of Cripto accelerates regeneration, leading to muscle hypertrophy. Moreover, we provide evidence that Cripto modulates myogenic cell determination and promotes proliferation by antagonizing the TGF-ß ligand myostatin. Our data provide unique insights into the molecular and cellular basis of Cripto activity in skeletal muscle regeneration and raise previously undescribed implications for stem cell biology and regenerative medicine.


Asunto(s)
Linaje de la Célula , Factor de Crecimiento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/fisiología , Miostatina/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Envejecimiento/metabolismo , Animales , Proliferación Celular , Eliminación de Gen , Marcación de Gen , Hipertrofia , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mioblastos/metabolismo , Mioblastos/patología , Miostatina/metabolismo , Transducción de Señal
12.
Cell Stem Cell ; 5(3): 298-309, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19733541

RESUMEN

Mechanisms governing muscle satellite cell withdrawal from cell cycle to enter into quiescence remain poorly understood. We studied the role of angiopoietin 1 (Ang1) and its receptor Tie-2 in the regulation of myogenic precursor cell (mpc) fate. In human and mouse, Tie-2 was preferentially expressed by quiescent satellite cells in vivo and reserve cells (RCs) in vitro. Ang1/Tie-2 signaling, through ERK1/2 pathway, decreased mpc proliferation and differentiation, increased the number of cells in G0, increased expression of RC-associated markers (p130, Pax7, Myf-5, M-cadherin), and downregulated expression of differentiation-associated markers. Silencing Tie-2 had opposite effects. Cells located in the satellite cell neighborhood (smooth muscle cells, fibroblasts) upregulated RC-associated markers by secreting Ang1 in vitro. In vivo, Tie-2 blockade and Ang1 overexpression increased the number of cycling and quiescent satellite cells, respectively. We propose that Ang1/Tie-2 signaling regulates mpc self-renewal by controlling the return to quiescence of a subset of satellite cells.


Asunto(s)
Angiopoyetina 1/metabolismo , Comunicación Autocrina , Comunicación Paracrina , Receptor TIE-2/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Angiopoyetina 1/farmacología , Animales , Apoptosis/efectos de los fármacos , Comunicación Autocrina/efectos de los fármacos , Recuento de Células , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Factor de Transcripción PAX7/metabolismo , Comunicación Paracrina/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/enzimología , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
13.
Exerc Sport Sci Rev ; 37(1): 18-22, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19098520

RESUMEN

Macrophages are necessary for skeletal muscle regeneration after injury. Muscle recruits inflammatory monocytes/macrophages that switch toward an anti-inflammatory profile upon phagocytosis of debris. In vitro, proinflammatory macrophages stimulate myoblast proliferation, whereas anti-inflammatory macrophages stimulate their differentiation. Thus, macrophages are involved in both phases of skeletal muscle regeneration: first, inflammation and cleansing of necrosis, and then myogenic differentiation and tissue repair.


Asunto(s)
Macrófagos/fisiología , Músculo Esquelético/fisiología , Regeneración , Animales , Diferenciación Celular , Humanos , Inflamación/fisiopatología , Monocitos/fisiología , Músculo Esquelético/lesiones , Mioblastos/citología , Fenotipo
14.
Genes Dev ; 22(13): 1747-52, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18593877

RESUMEN

In the fatal degenerative Duchenne muscular dystrophy (DMD), skeletal muscle is progressively replaced by fibrotic tissue. Here, we show that fibrinogen accumulates in dystrophic muscles of DMD patients and mdx mice. Genetic loss or pharmacological depletion of fibrinogen in these mice reduced fibrosis and dystrophy progression. Our results demonstrate that fibrinogen-Mac-1 receptor binding, through induction of IL-1beta, drives the synthesis of transforming growth factor-beta (TGFbeta) by mdx macrophages, which in turn induces collagen production in mdx fibroblasts. Fibrinogen-produced TGFbeta further amplifies collagen accumulation through activation of profibrotic alternatively activated macrophages. Fibrinogen, by engaging its alphavbeta3 receptor on fibroblasts, also directly promotes collagen synthesis. These data unveil a profibrotic role of fibrinogen deposition in muscle dystrophy.


Asunto(s)
Fibrinógeno/fisiología , Activación de Macrófagos/fisiología , Distrofia Muscular de Duchenne/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Niño , Preescolar , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrosis , Humanos , Integrina alfaVbeta3/metabolismo , Interleucina-1beta/metabolismo , Antígeno de Macrófago-1/metabolismo , Macrófagos/fisiología , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/patología , Unión Proteica
15.
Nat Genet ; 40(2): 170-80, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18176562

RESUMEN

HIF prolyl hydroxylases (PHD1-3) are oxygen sensors that regulate the stability of the hypoxia-inducible factors (HIFs) in an oxygen-dependent manner. Here, we show that loss of Phd1 lowers oxygen consumption in skeletal muscle by reprogramming glucose metabolism from oxidative to more anaerobic ATP production through activation of a Pparalpha pathway. This metabolic adaptation to oxygen conservation impairs oxidative muscle performance in healthy conditions, but it provides acute protection of myofibers against lethal ischemia. Hypoxia tolerance is not due to HIF-dependent angiogenesis, erythropoiesis or vasodilation, but rather to reduced generation of oxidative stress, which allows Phd1-deficient myofibers to preserve mitochondrial respiration. Hypoxia tolerance relies primarily on Hif-2alpha and was not observed in heterozygous Phd2-deficient or homozygous Phd3-deficient mice. Of medical importance, conditional knockdown of Phd1 also rapidly induces hypoxia tolerance. These findings delineate a new role of Phd1 in hypoxia tolerance and offer new treatment perspectives for disorders characterized by oxidative stress.


Asunto(s)
Metabolismo Basal , Glucosa/metabolismo , Hipoxia/metabolismo , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/deficiencia , Animales , Dióxido de Carbono/metabolismo , Isótopos de Carbono/metabolismo , Radioisótopos de Carbono/metabolismo , Embrión de Mamíferos , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Fibroblastos/metabolismo , Glutamatos/metabolismo , Homocigoto , Inmunohistoquímica , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Biológicos , Músculo Esquelético/metabolismo , Resonancia Magnética Nuclear Biomolecular , Oxidación-Reducción , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología , Procolágeno-Prolina Dioxigenasa/genética , Tomografía Computarizada por Rayos X
16.
Trends Mol Med ; 12(8): 345-7, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16843725

RESUMEN

A new study by Greenway and colleagues links mutations in the angiogenin gene to patients with amyotrophic lateral sclerosis (ALS)--a progressive and fatal motoneuron disease. This is an unexpected finding because angiogenin was originally identified as a molecule involved in the formation of blood vessels (angiogenesis). Angiogenin bears striking similarity to vascular endothelial growth factor (VEGF), which is the prototypic angiogenic factor that has recently emerged as a molecule with important neuroprotective activities. Besides VEGF, angiogenin is the second so-called angiogenic factor implicated in ALS, raising the question of whether additional angiogenic factors might have a role in ALS. Overall, these findings identify angiogenin as a novel candidate gene in the pathogenesis of ALS--a discovery that ultimately might lead to the development of new therapeutic strategies.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Predisposición Genética a la Enfermedad/genética , Ribonucleasa Pancreática/genética , Esclerosis Amiotrófica Lateral/metabolismo , Humanos , Modelos Biológicos , Ribonucleasa Pancreática/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
J Cell Sci ; 119(Pt 12): 2497-507, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16720640

RESUMEN

The mechanisms underlying stromal cell supportive functions are incompletely understood but probably implicate a mixture of cytokines, matrix components and cell adhesion molecules. Skeletal muscle uses recruited macrophages to support post-injury regeneration. We and others have previously shown that macrophages secrete mitogenic factors for myogenic cells. Here, we focused on macrophage-elicited survival signals. We demonstrated that: (1) macrophage influx is temporally correlated with the disappearance of TUNEL-positive apoptotic myogenic cells during post-injury muscle regeneration in mice; (2) direct cell-cell contacts between human macrophages and myogenic cells rescue myogenic cells from apoptosis, as assessed by decreased annexin V labelling and caspase-3 activity, and by increased DIOC-6 staining, Bcl-2 expression and phosphorylation of Akt and ERK1/2 survival pathways; (3) four pro-survival cell-cell adhesion molecular systems detected by DNA macroarray are expressed by macrophages and myogenic cells in vitro and in vivo - VCAM-1-VLA-4, ICAM-1-LFA-1, PECAM-1-PECAM-1 and CX3CL1-CX3CR1; (4) macrophages deliver anti-apoptotic signals through all four adhesion systems, as assessed by functional analyses with blocking antibodies; and (5) macrophages more strongly rescue differentiated myotubes, which must achieve adhesion-induced stabilisation of their structure to survive. Macrophages could secure these cells until they establish final association with the matrix.


Asunto(s)
Apoptosis/fisiología , Moléculas de Adhesión Celular/fisiología , Macrófagos/fisiología , Fibras Musculares Esqueléticas/fisiología , Mioblastos/fisiología , Animales , Adhesión Celular/fisiología , Membrana Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Venenos Elapídicos/administración & dosificación , Citometría de Flujo , Humanos , Técnicas In Vitro , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Células Musculares/citología , Células Musculares/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/lesiones , Transducción de Señal/fisiología
18.
Mol Biol Cell ; 16(2): 861-70, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15574885

RESUMEN

Knowledge on molecular systems involved in myogenic precursor cell (mpc) fusion into myotubes is fragmentary. Previous studies have implicated the a disintegrin and metalloproteinase (ADAM) family in most mammalian cell fusion processes. ADAM12 is likely involved in fusion of murine mpc and human rhabdomyosarcoma cells, but it requires yet unknown molecular partners to launch myogenic cell fusion. ADAM12 was shown able to mediate cell-to-cell attachment through binding alpha9beta1 integrin. We report that normal human mpc express both ADAM12 and alpha9beta1 integrin during their differentiation. Expression of alpha9 parallels that of ADAM12 and culminates at time of fusion. alpha9 and ADAM12 coimmunoprecipitate and participate to mpc adhesion. Inhibition of ADAM12/alpha9beta1 integrin interplay, by either ADAM12 antisense oligonucleotides or blocking antibody to alpha9beta1, inhibited overall mpc fusion by 47-48%, with combination of both strategies increasing inhibition up to 62%. By contrast with blockade of vascular cell adhesion molecule-1/alpha4beta1, which also reduced fusion, exposure to ADAM12 antisense oligonucleotides or anti-alpha9beta1 antibody did not induce detachment of mpc from extracellular matrix, suggesting specific involvement of ADAM12-alpha9beta1 interaction in the fusion process. Evaluation of the fusion rate with regard to the size of myotubes showed that both ADAM12 antisense oligonucleotides and alpha9beta1 blockade inhibited more importantly formation of large (> or =5 nuclei) myotubes than that of small (2-4 nuclei) myotubes. We conclude that both ADAM12 and alpha9beta1 integrin are expressed during postnatal human myogenic differentiation and that their interaction is mainly operative in nascent myotube growth.


Asunto(s)
Diferenciación Celular , Integrinas/metabolismo , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Desarrollo de Músculos , Músculo Esquelético/embriología , Proteínas ADAM , Proteína ADAM12 , Anticuerpos Bloqueadores/farmacología , Adhesión Celular , Proliferación Celular , Células Cultivadas , Interacciones Farmacológicas , Electroforesis en Gel de Poliacrilamida , Fluoresceína-5-Isotiocianato , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes , Humanos , Immunoblotting , Indoles , Integrinas/antagonistas & inhibidores , Integrinas/efectos de los fármacos , Integrinas/genética , Cinética , Fusión de Membrana/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/efectos de los fármacos , Metaloendopeptidasas/genética , Microscopía Confocal , Fibras Musculares Esqueléticas/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Pruebas de Precipitina , Propidio , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodaminas
19.
J Cell Biol ; 163(5): 1133-43, 2003 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-14662751

RESUMEN

Once escaped from the quiescence niche, precursor cells interact with stromal components that support their survival, proliferation, and differentiation. We examined interplays between human myogenic precursor cells (mpc) and monocyte/macrophages (MP), the main stromal cell type observed at site of muscle regeneration. mpc selectively and specifically attracted monocytes in vitro after their release from quiescence, chemotaxis declining with differentiation. A DNA macroarray-based strategy identified five chemotactic factors accounting for 77% of chemotaxis: MP-derived chemokine, monocyte chemoattractant protein-1, fractalkine, VEGF, and the urokinase system. MP showed lower constitutive chemotactic activity than mpc, but attracted monocytes much strongly than mpc upon cross-stimulation, suggesting mpc-induced and predominantly MP-supported amplification of monocyte recruitment. Determination of [3H]thymidine incorporation, oligosomal DNA levels and annexin-V binding showed that MP stimulate mpc proliferation by soluble factors, and rescue mpc from apoptosis by direct contacts. We conclude that once activated, mpc, which are located close by capillaries, initiate monocyte recruitment and interplay with MP to amplify chemotaxis and enhance muscle growth.


Asunto(s)
Apoptosis , Macrófagos/fisiología , Monocitos/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Células Satélite del Músculo Esquelético/fisiología , Células Cultivadas , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Quimiotaxis , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Humanos , Macrófagos/citología , Datos de Secuencia Molecular , Monocitos/citología , Músculo Esquelético/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Satélite del Músculo Esquelético/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...