Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropathol Appl Neurobiol ; 47(2): 218-235, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32772401

RESUMEN

AIMS: Transforming growth factor-ß (TGF-ß) signalling is thought to contribute to the remodelling of extracellular matrix (ECM) of skeletal muscle and to functional decline in patients with muscular dystrophies. We wanted to determine the role of TGF-ß-induced ECM remodelling in dystrophic muscle. METHODS: We experimentally induced the pathological hallmarks of severe muscular dystrophy by mechanically overloading the plantaris muscle in mice. Furthermore, we determined the role of TGF-ß signalling on dystrophic tissue modulation and on muscle function by (i) overloading myostatin knockout (Mstn-/- ) mice and (ii) by additional pharmacological TGF-ß inhibition via halofuginone. RESULTS: Transcriptome analysis of overloaded muscles revealed upregulation predominantly of genes associated with ECM, inflammation and metalloproteinase activity. Histology revealed in wild-type mice signs of severe muscular dystrophy including myofibres with large variation in size and internalized myonuclei, as well as increased ECM deposition. At the same time, muscle weight had increased by 208% and muscle force by 234%. Myostatin deficiency blunted the effect of overload on muscle mass (59% increase) and force (76% increase), while having no effect on ECM deposition. Concomitant treatment with halofuginone blunted overload-induced muscle hypertrophy and muscle force increase, while reducing ECM deposition and increasing myofibre size. CONCLUSIONS: ECM remodelling is associated with an increase in muscle mass and force in overload-modelled dystrophic muscle. Lack of myostatin is not advantageous and inhibition of ECM deposition by halofuginone is disadvantageous for muscle plasticity in response to stimuli that induce dystrophic muscle.


Asunto(s)
Matriz Extracelular/patología , Matriz Extracelular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
2.
Neuropathol Appl Neurobiol ; 46(6): 602-614, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32573804

RESUMEN

BACKGROUND: Dmdmdx , harbouring the c.2983C>T nonsense mutation in Dmd exon 23, is a mouse model for Duchenne muscular dystrophy (DMD), frequently used to test therapies aimed at dystrophin restoration. Current translational research is methodologically hampered by the lack of a reporter mouse model, which would allow direct visualization of dystrophin expression as well as longitudinal in vivo studies. METHODS: We generated a DmdEGFP-mdx reporter allele carrying in cis the mdx-23 mutation and a C-terminal EGFP-tag. This mouse model allows direct visualization of spontaneously and therapeutically restored dystrophin-EGFP fusion protein either after natural fibre reversion, or for example, after splice modulation using tricyclo-DNA to skip Dmd exon 23, or after gene editing using AAV-encoded CRISPR/Cas9 for Dmd exon 23 excision. RESULTS: Intravital microscopy in anaesthetized mice allowed live-imaging of sarcolemmal dystrophin-EGFP fusion protein of revertant fibres as well as following therapeutic restoration. Dystrophin-EGFP-fluorescence persisted ex vivo, allowing live-imaging of revertant and therapeutically restored dystrophin in isolated fibres ex vivo. Expression of the shorter dystrophin-EGFP isoforms Dp71 in the brain, Dp260 in the retina, and Dp116 in the peripheral nerve remained unabated by the mdx-23 mutation. CONCLUSION: Intravital imaging of DmdEGFP-mdx muscle permits novel experimental approaches such as the study of revertant and therapeutically restored dystrophin in vivo and ex vivo.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Alelos , Animales , Sistemas CRISPR-Cas , Exones , Edición Génica , Terapia Genética , Humanos , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Distrofia Muscular de Duchenne/terapia , Retina/metabolismo , Sarcolema/metabolismo , Sarcolema/ultraestructura
3.
Arch Pediatr ; 22(12 Suppl 1): 12S63-8, 2015 Dec.
Artículo en Francés | MEDLINE | ID: mdl-26773589

RESUMEN

Duchenne muscular dystrophy (DMD) is the most frequent hereditary neuromuscular disorder in childhood. Over the past 30 years, increasingly better standards of care have considerably improved the quality of life as well as the life expectancy of DMD patients. Despite such progress in disease management, DMD remains a devastating disorder with continuous decline of motor and cardiac function. Until recently, corticosteroids were the only treatment available to slow down, however modestly, disease progression. Importantly, novel innovative therapeutic approaches are currently being developed. This review discusses the rational and underlying molecular mechanism of these novel strategies as well as the progress made by recent clinical trials. Importantly, these new therapeutic advances bear the potential to profoundly modify the disease course of DMD.


Asunto(s)
Distrofia Muscular de Duchenne/terapia , Niño , Humanos , Distrofia Muscular de Duchenne/genética
4.
Exp Gerontol ; 48(2): 183-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23201547

RESUMEN

The prolonged effect of myostatin deficiency on muscle performance in knockout mice has as yet been only poorly investigated. We have demonstrated that absolute maximal force is increased in 6-month old female and male knockout mice and 2-year old female knockout mice as compared to age- and sex-matched wildtype mice. Similarly, absolute maximal power is increased by myostatin deficiency in 6-month old female and male knockout mice but not in 2-year old female knockout mice. The increases we observed were greater in 6-month old female than in male knockout mice and can primarily result from muscle hypertrophy. In contrast, fatigue resistance was decreased in 6-month old knockout mice of both sexes as compared to age- and sex-matched wildtype mice. Moreover, in contrast to 2-year old female wildtype mice, aging in 2-year old knockout mice reduced absolute maximal force and power of both sexes as compared to their younger counterparts, although muscle weight did not change. These age-related decreases were lower in 2-year old female than in 2-year old male knockout mice. Together these results suggest that the beneficial effect of myostatin deficiency on absolute maximal force and power is greater in young (versus old) mice and female (versus male) mice. Most of these effects of myostatin deficiency are related neither to changes in the concentration of myofibrillar proteins nor to the slow to fast fiber type transition.


Asunto(s)
Envejecimiento/metabolismo , Contracción Muscular , Fuerza Muscular , Músculo Esquelético/metabolismo , Miostatina/deficiencia , Factores de Edad , Envejecimiento/genética , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fatiga Muscular , Miostatina/genética , Factores Sexuales
5.
Cell Death Differ ; 18(2): 222-34, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20689554

RESUMEN

Satellite cells are the resident stem cells of adult skeletal muscle, supplying myonuclei for homoeostasis, hypertrophy and repair. In this study, we have examined the role of bone morphogenetic protein (BMP) signalling in regulating satellite cell function. Activated satellite cells expressed BMP receptor type 1A (BMPR-1A/Alk-3) and contained phosphorylated Smad proteins, indicating that BMP signalling is operating during proliferation. Indeed, exogenous BMP4 stimulated satellite cell division and inhibited myogenic differentiation. Conversely, interfering with the interactions between BMPs and their receptors by the addition of either the BMP antagonist Noggin or soluble BMPR-1A fragments, induced precocious differentiation. Similarly, blockade of BMP signalling by siRNA-mediated knockdown of BMPR-1A, disruption of the intracellular pathway by either Smad5 or Smad4 knockdown or inhibition of Smad1/5/8 phosphorylation with Dorsomorphin, also caused premature myogenic differentiation. BMP signalling acted to inhibit the upregulation of genes associated with differentiation, in part, through regulating Id1. As satellite cells differentiated, Noggin levels increased to antagonise BMP signalling, since Noggin knockdown enhanced proliferation and impeded myoblast fusion into large multinucleated myotubes. Finally, interference of normal BMP signalling after muscle damage in vivo perturbed the regenerative process, and resulted in smaller regenerated myofibres. In conclusion, BMP signalling operates during routine satellite cell function to help coordinate the balance between proliferation and differentiation, before Noggin is activated to antagonise BMPs and facilitate terminal differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Células Satélite del Músculo Esquelético/citología , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Diferenciación Celular , Proliferación Celular , Ratones , Fosforilación , Pirazoles/farmacología , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo
6.
Acta Myol ; 24(3): 230-41, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16629058

RESUMEN

Over the past decade, signalling cascades have been characterised that control key features of muscle growth, including the proliferation, differentiation of muscle precursors, the control cell size (hypertrophy) and cell death. In this review we highlight how two differing signalling molecules, Insulin-like Growth Factor-1 (IGF-1) and myostatin, regulate key steps during muscle development. We discuss how IGF-1 and myostatin signalling cascades can be manipulated to stimulate muscle growth. We summarise experimental data from mdx mouse, the animal model for Duchenne muscular dystrophy, that suggest a therapeutic value of these strategies for patients suffering from muscular dystrophy without redressing the primary cause of the lesion.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/fisiología , Desarrollo de Músculos/fisiología , Hipertonía Muscular/etiología , Distrofia Muscular de Duchenne/terapia , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Humanos , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/fisiopatología , Miostatina
7.
Development ; 126(5): 1041-53, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9927604

RESUMEN

Embryonic muscle growth requires a fine balance between proliferation and differentiation. In this study we have investigated how this balance is achieved during chick development. Removal of ectoderm from trunk somites results in the down-regulation of Pax-3 expression and cell division of myogenic precursors is halted. This initially leads to an up-regulation of MyoD expression and to a burst in terminal differentiation but further muscle growth is arrested. Locally applied bone morphogenetic protein-4 (BMP-4) to somites mimics the effect of the ectoderm and stimulates Pax-3 expression which eventually results in excessive muscle growth in somites. Surprisingly, BMP-4 up-regulates expression of noggin which encodes a BMP-4 antagonist. This suggests that the proliferation enhancing activity of BMP-4 can be limited via up-regulation of noggin and that myogenic cells differentiate, as an intrinsic property, when deprived of BMP-4 influence. In contrast to BMP-4, Sonic hedgehog (Shh) locally applied to somites arrests muscle growth by down-regulation of Pax-3 and immediate up-regulation of MyoD expression. Such premature muscle differentiation in somites at tongue and limb levels prevents myogenic migration and thus tongue and limb muscle are not formed. Therefore, precise limitation of differentiation, executed by proliferative and Pax-3 promoting signals, is indispensable for continuous embryonic muscle growth.


Asunto(s)
Músculos/embriología , Transactivadores , Factores de Transcripción , Animales , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras , Diferenciación Celular , División Celular , Embrión de Pollo , Proteínas de Unión al ADN/metabolismo , Ectodermo , Extremidades , Proteínas Hedgehog , Músculos/metabolismo , Proteína MioD/metabolismo , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box , Proteínas/metabolismo , Transducción de Señal , Lengua
8.
Curr Biol ; 8(11): 642-52, 1998 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-9635191

RESUMEN

BACKGROUND: Skeletal muscle of trunk, limbs and tongue develops from a small population of cells that originates from somites. Although promoters and inhibitors of muscle differentiation have been isolated, nothing is known about how the amplification of the muscle precursor pool is regulated; this amplification provides muscle mass during development. Furthermore, little is known about how cells accumulate in the pre-muscle masses in the limbs. We investigated the role of bone morphogenetic protein (BMPs) and Sonic hedgehog (Shh) during proliferation, differentiation and positioning of muscle. RESULTS: The proliferation of muscle precursors in limbs was linked to Pax-3 expression. Ectoderm removal downregulated Pax-3 expression, arrested proliferation and prematurely initiated muscle differentiation which exhausted the muscle precursor pool and prevented further muscle growth. BMP-2, BMP-4 and BMP-7 had a dose-dependent effect on pre-myogenic cells: low concentrations maintained a Pax-3-expressing proliferative population, substituting for ectoderm-derived proliferative signals and delaying differentiation, whereas high concentrations prevented muscle development, probably by inducing apoptosis. In the limb, Shh upregulated Bmp-2 and Bmp-7 expression which delayed muscle differentiation, upregulated Pax-3, amplified the muscle precursor population and stimulated excessive muscle growth. CONCLUSIONS: These data indicate that embryonic muscle growth requires muscle differentiation to be delayed. Muscle differentiation may occur through a default pathway after cells escape proliferative signals. Positioning of muscle is regulated by high concentrations of BMPs, thus a single type of signalling molecule can determine crucial steps in muscle development: when and where to proliferate, and when and where to differentiate.


Asunto(s)
Extremidades/embriología , Músculo Esquelético/embriología , Transactivadores , Factores de Transcripción , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular , División Celular , Embrión de Pollo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Inducción Embrionaria , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog , Modelos Biológicos , Músculo Esquelético/citología , Proteína MioD/genética , Proteína MioD/fisiología , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box , Proteínas/genética , Proteínas/fisiología , Transducción de Señal , Factores de Tiempo
9.
Dev Biol ; 178(2): 327-42, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8812133

RESUMEN

We have cloned and sequenced a chick gene, Flik (follistatin-like) that appears to be the homolog of the mammalian TSC36. The ORF encodes a secreted protein of approx 38 kDa, containing a single cysteine-rich domain that shows a strong relationship with the second of the four from which Follistatin is constructed. The remainder of the Flik protein shows no strong family affinities. We describe here the normal expression pattern of the gene during primitive streak and neurula stages. We also give revised data for early neurectodermal expression of chick follistatin (Connolly et al., 1995, Developmental Genetics 17(1), 65-77) and follow the new ectopic expression of both genes during the induction of a second neural axis, after grafting of Hensen's node into a peripheral position in a host blastoderm. Both genes mark the organizer (node and/or mesodermal head process) and early neural plate, and could thus be involved in intercellular signaling during mesodermal dorsalization and neural induction. Flik expression appears earlier than that of follistatin however, and unlike that of follistatin, it is maintained strongly in the dorsal midline with intensity smoothly declining into presumptive lateral regions. We show that both genes are upregulated in host tissue in the neighborhood of node grafts, but whereas follistatin is transcribed after 8-10 hr in host epiblast that has formed new neural plate, Flik is expressed within 4 hr in this region, sometimes detectable before the first structural changes (columnarization) of neuralization. Thus, although ectodermal Flik expression is later confined within neural plate, and mesodermal expression concentrated in dorsal axial tissue, its early distribution is consistent with the idea that the encoded protein may first be involved in generating a graded system that positions the boundaries of both neural and dorsal axial mesodermal territories. The results are discussed in relation to this hypothesis and to other recent findings regarding control of vertebrate dorsoventral patterning.


Asunto(s)
Inducción Embrionaria/genética , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Embrión de Pollo , Clonación Molecular , Proteínas Relacionadas con la Folistatina , Glicoproteínas/metabolismo , Datos de Secuencia Molecular
10.
Dev Biol ; 178(2): 343-62, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8812134

RESUMEN

We report on the normal and experimentally altered expression of two structurally related genes, Follistatin and Follistatin-like (Flik), in the somites of avian embryos. In normal chick embryos, Follistatin expression can first be seen in the cells of the dorsolateral somite quarter. During somite maturation, the cells of the dorsomedial quarter also express this gene. Within the dermomyotome it seems that only the muscle precursors are Follistatin-positive. The migrating precursors of limb and tongue muscle as well as the myotome cells show Follistatin expression. The manipulation experiments reveal that the expression of Follistatin in the somites can be inhibited by notochord signals. This effect can be mimicked by sonic hedgehog protein. Flik is expressed in the dorsomedial compartment of the somite and later on in the myotome. Unlike Follistatin, Flik expression requires signals emanating from the neural tube. Notochordal influences do not alter Flik expression. The expression of both genes does not depend on signals of intermediate or lateral mesoderm. Since the products of both genes are proposed to antagonize TGF-beta superfamily proteins during gastrulation and neuralization, we postulate that during myogenesis follistatin and flik counteract inhibiting effects of related molecules on muscle differentiation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Músculo Esquelético/embriología , Somitos , Animales , Compartimento Celular , Diferenciación Celular , Embrión de Pollo , Folistatina , Proteínas Relacionadas con la Folistatina , Músculo Esquelético/metabolismo
11.
Dev Biol ; 174(2): 448-52, 1996 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-8631514

RESUMEN

Bone morphogenetic proteins (BMPs) induce ectopic cartilage and bone when implanted intramuscularly in adult rats. Expression data suggest that BMPs signal skeletal development in embryos. An important question is which cells are targets of BMP signaling in adult and embryonic tissues. Here, we examined the effect of BMP-2 on micromass cultures of chick limb bud mesenchyme. We report that BMP-2 promotes formation of cartilage and simultaneously inhibits development of muscle cells. To follow the fate of presumptive muscle cells, we replaced chick somites with quail somites at the wing level and made micromass cultures from the chimeric wings. In untreated cultures, quail cells formed muscle but not cartilage. In BMP-2-treated cultures, quail cells disappeared altogether. This suggests that BMP-2 may simultaneously promote cartilage differentiation and reduce the presumptive myogenic cell populations in regions of skeletal development.


Asunto(s)
Cartílago/embriología , Extremidades/embriología , Sustancias de Crecimiento/fisiología , Músculos/embriología , Proteínas/fisiología , Animales , Proteínas Morfogenéticas Óseas , Embrión de Pollo , Sustancias de Crecimiento/genética , Humanos , Plásmidos , Proteínas/genética , Codorniz , Retroviridae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...