Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cachexia Sarcopenia Muscle ; 11(4): 929-946, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32159297

RESUMEN

BACKGROUND: Cachexia, a multifactorial syndrome affecting more than 50% of patients with advanced cancer and responsible for ~20% of cancer-associated deaths, is still a poorly understood process without a standard cure available. Skeletal muscle atrophy caused by systemic inflammation is a major clinical feature of cachexia, leading to weight loss, dampening patients' quality of life, and reducing patients' response to anticancer therapy. RAGE (receptor for advanced glycation end-products) is a multiligand receptor of the immunoglobulin superfamily and a mediator of muscle regeneration, inflammation, and cancer. METHODS: By using murine models consisting in the injection of colon 26 murine adenocarcinoma (C26-ADK) or Lewis lung carcinoma (LLC) cells in BALB/c and C57BL/6 or Ager-/- (RAGE-null) mice, respectively, we investigated the involvement of RAGE signalling in the main features of cancer cachexia, including the inflammatory state. In vitro experiments were performed using myotubes derived from C2C12 myoblasts or primary myoblasts isolated from C57BL/6 wild type and Ager-/- mice treated with the RAGE ligand, S100B (S100 calcium-binding protein B), TNF (tumor necrosis factor)α±IFN (interferon) γ, and tumour cell- or masses-conditioned media to analyse hallmarks of muscle atrophy. Finally, muscles of wild type and Ager-/- mice were injected with TNFα/IFNγ or S100B in a tumour-free environment. RESULTS: We demonstrate that RAGE is determinant to activate signalling pathways leading to muscle protein degradation in the presence of proinflammatory cytokines and/or tumour-derived cachexia-inducing factors. We identify the RAGE ligand, S100B, as a novel factor able to induce muscle atrophy per se via a p38 MAPK (p38 mitogen-activated protein kinase)/myogenin axis and STAT3 (signal transducer and activator of transcription 3)-dependent MyoD (myoblast determination protein 1) degradation. Lastly, we found that in cancer conditions, an increase in serum levels of tumour-derived S100B and HMGB1 (high mobility group box 1) occurs leading to chronic activation/overexpression of RAGE, which induces hallmarks of cancer cachexia (i.e. muscle wasting, systemic inflammation, and release of tumour-derived pro-cachectic factors). Absence of RAGE in mice translates into reduced serum levels of cachexia-inducing factors, delayed loss of muscle mass and strength, reduced tumour progression, and increased survival. CONCLUSIONS: RAGE is a molecular determinant in inducing the hallmarks of cancer cachexia, and molecular targeting of RAGE might represent a therapeutic strategy to prevent or counteract the cachectic syndrome.


Asunto(s)
Caquexia/prevención & control , Neoplasias/complicaciones , Receptor para Productos Finales de Glicación Avanzada/fisiología , Animales , Humanos , Ratones , Neoplasias/fisiopatología
2.
J Cachexia Sarcopenia Muscle ; 9(7): 1213-1234, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30334619

RESUMEN

Emerging evidence suggests that the signalling of the Receptor for Advanced Glycation End products (RAGE) is critical for skeletal muscle physiology controlling both the activity of muscle precursors during skeletal muscle development and the correct time of muscle regeneration after acute injury. On the other hand, the aberrant re-expression/activity of RAGE in adult skeletal muscle is a hallmark of muscle wasting that occurs in response to ageing, genetic disorders, inflammatory conditions, cancer, and metabolic alterations. In this review, we discuss the mechanisms of action and the ligands of RAGE involved in myoblast differentiation, muscle regeneration, and muscle pathological conditions. We highlight potential therapeutic strategies for targeting RAGE to improve skeletal muscle function.


Asunto(s)
Músculo Esquelético/metabolismo , Enfermedades Musculares/etiología , Enfermedades Musculares/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Ligandos , Músculo Esquelético/patología , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Enfermedades Musculares/patología , Isoformas de Proteínas , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/química , Receptor para Productos Finales de Glicación Avanzada/genética , Rabdomiosarcoma/etiología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Transducción de Señal
3.
Hum Mol Genet ; 27(21): 3734-3746, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085099

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal X-linked disease affecting striated muscles, which undergo progressive degeneration and chronic inflammation. Receptor for advanced glycation end-products (RAGE), a multiligand receptor involved in myogenesis and inflammation, is absent in healthy adult muscles but is re-expressed in myoblasts, regenerating myofibers and activated immune cells upon acute muscle injury, and in certain myopathies. We show here that RAGE is expressed and chronically stimulated in muscles of mdx mice, an experimental model of DMD, which also release high amounts of the RAGE ligands, HMGB1 and S100B. We generated a double mutant, mdx/Ager-/- mouse lacking dystrophin and RAGE. Compared to mdx mice, muscles of mdx/Ager-/- mice show restrained inflammation, unaffected fibrosis and higher muscle strength. Mdx/Ager-/- macrophages are less responsive to proinflammatory stimuli and express lower levels of Ccr2, Ccl2 and Ccl7, which are involved in monocyte/macrophage chemotaxis and migration. In vivo treatment of dystrophic muscles with a RAGE blocking antibody results in reduced necrosis and inflammatory infiltrate. Our results suggest that RAGE sustains muscle inflammation and necrosis in DMD muscles and that reducing RAGE activity might represent a potential therapeutic tool to counteract muscle inflammation and rescue muscle morphology in DMD conditions.


Asunto(s)
Inflamación , Fuerza Muscular , Distrofia Muscular de Duchenne/metabolismo , Receptor para Productos Finales de Glicación Avanzada/fisiología , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Fibrosis , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular de Duchenne/fisiopatología , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo
4.
Sci Rep ; 7(1): 12537, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970581

RESUMEN

Regeneration of injured skeletal muscles relies on a tightly controlled chain of cellular and molecular events. We show that appropriate levels of S100B protein are required for timely muscle regeneration after acute injury. S100B released from damaged myofibers and infiltrating macrophages expands the myoblast population, attracts macrophages and promotes their polarization into M2 (pro-regenerative) phenotype, and modulates collagen deposition, by interacting with RAGE (receptor for advanced glycation end-products) or FGFR1 (fibroblast growth factor receptor 1) depending on the muscle repair phase and local conditions. However, persistence of high S100B levels compromises the regeneration process prolonging myoblast proliferation and macrophage infiltration, delaying M1/M2 macrophage transition, and promoting deposition of fibrotic tissue via RAGE engagement. Interestingly, S100B is released in high abundance from degenerating muscles of mdx mice, an animal model of Duchenne muscular dystrophy (DMD), and blocking S100B ameliorates histopathology. Thus, levels of S100B differentially affect skeletal muscle repair upon acute injury and in the context of muscular dystrophy, and S100B might be regarded as a potential molecular target in DMD.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Duchenne/genética , Regeneración/genética , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Animales , Modelos Animales de Enfermedad , Humanos , Activación de Macrófagos/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos mdx , Fuerza Muscular/genética , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
5.
J Cell Sci ; 127(Pt 8): 1699-711, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24554430

RESUMEN

Embryonal rhabdomyosarcomas (ERMSs) show elevated levels of PAX7, a transcription factor that marks quiescent adult muscle stem (satellite) cells and is important for proliferation and survival of activated satellite cells and whose timely repression is required for myogenic differentiation. However, the mechanism of PAX7 accumulation in ERMSs and whether high PAX7 causes uncontrolled proliferation in ERMS remains to be elucidated. The receptor for advanced glycation end-products (RAGE, encoded by AGER) transduces a myogenic and anti-proliferative signal in myoblasts, and stable transfection of the ERMS cell line TE671, which does not express RAGE, with AGER results in reduced proliferation and formation of tumor masses in vivo, and enhanced apoptosis and myogenic differentiation. Herein, we show that RAGE expression is low or absent in human ERMSs. We also show that in ERMS cells (1) PAX7 accumulates owing to absent or low RAGE signaling; (2) elevated PAX7 levels reduce RAGE expression and levels of MyoD and myogenin, muscle-specific transcription factors required for myoblast proliferation arrest and differentiation, respectively; (3) PAX7 supports myoblast proliferation by reducing the levels of MyoD, primarily by promoting its degradation; and (4), when ectopically expressed in ERMS cells, that RAGE upregulates myogenin which upregulates MyoD and downregulates PAX7, with consequent inhibition of proliferation and stimulation of differentiation. Thus, failure to express RAGE and, hence, MyoD and myogenin above a critical level in ERMS cells might result in deregulated PAX7 expression leading to uncontrolled proliferation and, potentially, to rhabdomyosarcomagenesis.


Asunto(s)
Proliferación Celular , Factor de Transcripción PAX7/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Rabdomiosarcoma Embrionario/metabolismo , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteína MioD/metabolismo , Mioblastos/metabolismo , Miogenina/metabolismo , Rabdomiosarcoma Embrionario/patología , Transducción de Señal , Regulación hacia Arriba
6.
J Cell Sci ; 125(Pt 6): 1440-54, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22328527

RESUMEN

Expression of the paired-box 7 (PAX7) transcription factor is regulated during both myoblast proliferation and differentiation: high levels of PAX7 compromise myogenic differentiation because of excess and prolonged proliferation, whereas low levels of PAX7 result in precocious differentiation. We showed that myogenin repressed Pax7 transcription in differentiating myoblasts by binding to specific recognition sites in the Pax7 promoter, and that high-mobility group box 1 (HMGB1)-receptor for advanced glycation end-products (RAGE) signaling was required for myogenin induction and myogenin-dependent repression of Pax7 transcription. In addition, PAX7 negatively and myogenin positively regulated RAGE expression. RAGE, a multiligand receptor of the immunoglobulin superfamily, was not expressed in adult skeletal muscles, and was transiently expressed in activated, proliferating and differentiating satellite cells (SCs) in injured muscles. Compared with wild-type muscles, Rage(-/-) muscles exhibited increased numbers of basal SCs that were further increased in injured Rage(-/-) muscles following elevated myoblast asymmetric division; complete regeneration of injured Rage(-/-) muscles was found to be delayed by ~1 week. Thus, RAGE signaling physiologically repressed Pax7 transcription in SCs by upregulating myogenin, thereby accelerating muscle regeneration and limiting SC self-renewal.


Asunto(s)
Proteína HMGB1/fisiología , Homeostasis/fisiología , Miogenina/fisiología , Factor de Transcripción PAX7/genética , Receptores Inmunológicos/genética , Células Satélite del Músculo Esquelético/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mioblastos/citología , Mioblastos/metabolismo , Factor de Transcripción PAX7/biosíntesis , Cultivo Primario de Células , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/deficiencia , Elementos Reguladores de la Transcripción/fisiología , Proteínas Represoras/fisiología , Células Satélite del Músculo Esquelético/citología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...