Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
FASEB J ; 35(4): e21246, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33769615

RESUMEN

Osteoporosis is the most common age-related metabolic bone disorder, which is characterized by low bone mass and deterioration in bone architecture, with a propensity to fragility fractures. The best treatment for osteoporosis relies on stimulation of osteoblasts to form new bone and restore bone structure, however, anabolic therapeutics are few and their use is time restricted. Here, we report that Syndecan-3 increases new bone formation through enhancement of WNT signaling in osteoblasts. Young adult Sdc3-/- mice have low bone volume, reduced bone formation, increased bone marrow adipose tissue, increased bone fragility, and a blunted anabolic bone formation response to mechanical loading. This premature osteoporosis-like phenotype of Sdc3-/- mice is due to delayed osteoblast maturation and impaired osteoblast function, with contributing increased osteoclast-mediated bone resorption. Indeed, overexpressing Sdc3 in osteoblasts using the Col1a1 promoter rescues the low bone volume phenotype of the Sdc3-/- mice, and also increases bone volume in WT mice. Mechanistically, SDC3 enhances canonical WNT signaling in osteoblasts through stabilization of Frizzled 1, making SDC3 an attractive target for novel bone anabolic drug development.


Asunto(s)
Desarrollo Óseo/fisiología , Sindecano-3/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Animales Recién Nacidos , Anticuerpos , Proliferación Celular , Desarrollo Fetal , Masculino , Ratones , Ratones Noqueados , Osteoblastos , Osteoclastos , Sindecano-3/genética
2.
Proteomics ; 15(18): 3152-62, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26037908

RESUMEN

The mzQuantML standard has been developed by the Proteomics Standards Initiative for capturing, archiving and exchanging quantitative proteomic data, derived from mass spectrometry. It is a rich XML-based format, capable of representing data about two-dimensional features from LC-MS data, and peptides, proteins or groups of proteins that have been quantified from multiple samples. In this article we report the development of an open source Java-based library of routines for mzQuantML, called the mzqLibrary, and associated software for visualising data called the mzqViewer. The mzqLibrary contains routines for mapping (peptide) identifications on quantified features, inference of protein (group)-level quantification values from peptide-level values, normalisation and basic statistics for differential expression. These routines can be accessed via the command line, via a Java programming interface access or a basic graphical user interface. The mzqLibrary also contains several file format converters, including import converters (to mzQuantML) from OpenMS, Progenesis LC-MS and MaxQuant, and exporters (from mzQuantML) to other standards or useful formats (mzTab, HTML, csv). The mzqViewer contains in-built routines for viewing the tables of data (about features, peptides or proteins), and connects to the R statistical library for more advanced plotting options. The mzqLibrary and mzqViewer packages are available from https://code.google.com/p/mzq-lib/.


Asunto(s)
Sistemas de Administración de Bases de Datos , Bases de Datos de Proteínas/normas , Proteómica/métodos , Proteómica/normas , Programas Informáticos
3.
Front Physiol ; 14: 1301804, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38130476

RESUMEN

Introduction: The skeletal muscle deformity of commercial chickens (Gallus gallus), known as the wooden breast (WB), is associated with fibrotic myopathy of unknown etiology. For future breeding strategies and genetic improvements, it is essential to identify the molecular mechanisms underlying the phenotype. The pathophysiological hallmarks of WB include severe skeletal muscle fibrosis, inflammation, myofiber necrosis, and multifocal degeneration of muscle tissue. The transmembrane proteoglycans syndecans have a wide spectrum of biological functions and are master regulators of tissue homeostasis. They are upregulated and shed (cleaved) as a regulatory mechanism during tissue repair and regeneration. During the last decades, it has become clear that the syndecan family also has critical functions in skeletal muscle growth, however, their potential involvement in WB pathogenesis is unknown. Methods: In this study, we have categorized four groups of WB myopathy in broiler chickens and performed a comprehensive characterization of the molecular and histological profiles of two of them, with a special focus on the role of the syndecans and remodeling of the extracellular matrix (ECM). Results and discussion: Our findings reveal differential expression and shedding of the four syndecan family members and increased matrix metalloproteinase activity. Additionally, we identified alterations in key signaling pathways such as MAPK, AKT, and Wnt. Our work provides novel insights into a deeper understanding of WB pathogenesis and suggests potential therapeutic targets for this condition.

4.
J Cell Mol Med ; 16(5): 1013-25, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21615681

RESUMEN

Post-natal growth and regeneration of skeletal muscle is highly dependent on a population of resident myogenic precursors known as satellite cells. Transcription factors from the Pax gene family, Pax3 and Pax7, are critical for satellite cell biogenesis, survival and potentially self-renewal; however, the underlying molecular mechanisms remain unsolved. This is particularly true in the case of Pax7, which appears to regulate myogenesis at multiple levels. Accordingly, recent data have highlighted the importance of a functional relationship between Pax7 and the MyoD family of muscle regulatory transcription factors during normal muscle formation and disease. Here we will critically review key findings suggesting that Pax7 may play a dual role by promoting resident muscle progenitors to commit to the skeletal muscle lineage while preventing terminal differentiation, thus keeping muscle progenitors poised to differentiate upon environmental cues. In addition, potential regulatory mechanisms for the control of Pax7 activity will be proposed.


Asunto(s)
Desarrollo de Músculos/fisiología , Factor de Transcripción PAX7/fisiología , Células Satélite del Músculo Esquelético/fisiología , Animales , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Ratones , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/fisiología , Mioblastos/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Receptores Notch/fisiología , Sindecanos/fisiología , Proteínas de la Superfamilia TGF-beta/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Vía de Señalización Wnt/fisiología
5.
Matrix Biol ; 113: 61-82, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36152781

RESUMEN

Muscle stem cells (MuSCs) are indispensable for muscle regeneration. A multitude of extracellular stimuli direct MuSC fate decisions from quiescent progenitors to differentiated myocytes. The activity of these signals is modulated by coreceptors such as syndecan-3 (SDC3). We investigated the global landscape of SDC3-mediated regulation of myogenesis using a phosphoproteomics approach which revealed, with the precision level of individual phosphosites, the large-scale extent of SDC3-mediated regulation of signal transduction in MuSCs. We then focused on INSR/AKT/mTOR as a key pathway regulated by SDC3 during myogenesis and mechanistically dissected SDC3-mediated inhibition of insulin receptor signaling in MuSCs. SDC3 interacts with INSR ultimately limiting signal transduction via AKT/mTOR. Both knockdown of INSR and inhibition of AKT restore Sdc3-/- MuSC differentiation to wild type levels. Since SDC3 is rapidly downregulated at the onset of differentiation, our study suggests that SDC3 acts a timekeeper to restrain proliferating MuSC response and prevent premature differentiation.


Asunto(s)
Músculo Esquelético , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sindecano-3/genética , Sindecano-3/metabolismo , Células Cultivadas , Músculo Esquelético/metabolismo , Desarrollo de Músculos/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Diferenciación Celular
6.
J Cell Biol ; 172(2): 233-44, 2006 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-16401724

RESUMEN

The mechanism of skeletal myoblast fusion is not well understood. We show that endogenous nitric oxide (NO) generation is required for myoblast fusion both in embryonic myoblasts and in satellite cells. The effect of NO is concentration and time dependent, being evident only at the onset of differentiation, and direct on the fusion process itself. The action of NO is mediated through a tightly regulated activation of guanylate cyclase and generation of cyclic guanosine monophosphate (cGMP), so much so that deregulation of cGMP signaling leads to a fusion-induced hypertrophy of satellite-derived myotubes and embryonic muscles, and to the acquisition of fusion competence by myogenic precursors in the presomitic mesoderm. NO and cGMP induce expression of follistatin, and this secreted protein mediates their action in myogenesis. These results establish a hitherto unappreciated role of NO and cGMP in regulating myoblast fusion and elucidate their mechanism of action, providing a direct link with follistatin, which is a key player in myogenesis.


Asunto(s)
Fusión Celular , GMP Cíclico/metabolismo , Folistatina/metabolismo , Mioblastos Esqueléticos/fisiología , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Femenino , Folistatina/genética , Ratones , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/fisiología , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos Esqueléticos/citología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Óxido Nítrico Sintasa/metabolismo , Transcripción Genética
7.
Sci Rep ; 10(1): 20487, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33235244

RESUMEN

Rheumatoid arthritis (RA) is a debilitating and painful inflammatory autoimmune disease characterised by the accumulation of leukocytes in the synovium, cartilage destruction and bone erosion. The immunomodulatory effects of bone marrow derived mesenchymal stem cells (MSCs) has been widely studied and the recent observations that syndecan-3 (SDC3) is selectively pro-inflammatory in the joint led us to hypothesise that SDC3 might play an important role in MSC biology. MSCs isolated from bone marrow of wild type and Sdc3-/- mice were used to assess immunophenotype, differentiation, adhesion and migration properties and cell signalling pathways. While both cell types show similar differentiation potential and forward scatter values, the cell complexity in wild type MSCs was significantly higher than in Sdc3-/- cells and was accompanied by lower spread surface area. Moreover, Sdc3-/- MSCs adhered more rapidly to collagen type I and showed a dramatic increase in AKT phosphorylation, accompanied by a decrease in ERK1/2 phosphorylation compared with control cells. In a mouse model of antigen-induced inflammatory arthritis, intraarticular injection of Sdc3-/- MSCs yielded enhanced efficacy compared to injection of wild type MSCs. In conclusion, our data suggest that syndecan-3 regulates MSC adhesion and efficacy in inflammatory arthritis, likely via induction of the AKT pathway.


Asunto(s)
Artritis/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Eliminación de Gen , Inflamación/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Sindecano-3/metabolismo , Animales , Artritis/complicaciones , Artritis/terapia , Células de la Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Colágeno/farmacología , Modelos Animales de Enfermedad , Inflamación/complicaciones , Inflamación/terapia , Masculino , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
8.
Front Cell Dev Biol ; 8: 730, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850844

RESUMEN

BACKGROUND: Extracellular matrix (ECM) remodeling is essential for skeletal muscle development and adaption in response to environmental cues such as exercise and injury. The cell surface proteoglycan syndecan-4 has been reported to be essential for muscle differentiation, but few molecular mechanisms are known. Syndecan-4-/- mice are unable to regenerate damaged muscle, and display deficient satellite cell activation, proliferation, and differentiation. A reduced myofiber basal lamina has also been reported in syndecan-4-/- muscle, indicating possible defects in ECM production. To get a better understanding of the underlying molecular mechanisms, we have here investigated the effects of syndecan-4 genetic ablation on molecules involved in ECM remodeling and muscle growth, both under steady state conditions and in response to exercise. METHODS: Tibialis anterior (TA) muscles from sedentary and exercised syndecan-4-/- and WT mice were analyzed by immunohistochemistry, real-time PCR and western blotting. RESULTS: Compared to WT, we found that syndecan-4-/- mice had reduced body weight, reduced muscle weight, muscle fibers with a smaller cross-sectional area, and reduced expression of myogenic regulatory transcription factors. Sedentary syndecan-4-/- had also increased mRNA levels of syndecan-2, decorin, collagens, fibromodulin, biglycan, and LOX. Some of these latter ECM components were reduced at protein level, suggesting them to be more susceptible to degradation or less efficiently translated when syndecan-4 is absent. At the protein level, TRPC7 was reduced, whereas activation of the Akt/mTOR/S6K1 and Notch/HES-1 pathways were increased. Finally, although exercise induced upregulation of several of these components in WT, a further upregulation of these molecules was not observed in exercised syndecan-4-/- mice. CONCLUSION: Altogether our data suggest an important role of syndecan-4 in muscle development.

9.
J Clin Invest ; 116(10): 2791-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16981010

RESUMEN

Obesity is associated with chronic low-grade inflammation. Thus, at metabolically relevant sites, including adipose tissue and muscle, there is abnormal production of proinflammatory cytokines such as TNF-alpha. Here we demonstrate that eNOS expression was reduced, with a concomitant reduction of mitochondrial biogenesis and function, in white and brown adipose tissue and in the soleus muscle of 3 different animal models of obesity. The genetic deletion of TNF receptor 1 in obese mice restored eNOS expression and mitochondrial biogenesis in fat and muscle; this was associated with less body weight gain than in obese wild-type controls. Furthermore, TNF-alpha downregulated eNOS expression and mitochondrial biogenesis in cultured white and brown adipocytes and muscle satellite cells of mice. The NO donors DETA-NO and SNAP prevented the reduction of mitochondrial biogenesis observed with TNF-alpha. Our findings demonstrate that TNF-alpha impairs mitochondrial biogenesis and function in different tissues of obese rodents by downregulating eNOS expression and suggest a novel pathophysiological process that sustains obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Citocromos c/metabolismo , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Mitocondrias/genética , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/genética , Factor Nuclear 1 de Respiración/genética , Obesidad/genética , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Zucker , Receptores del Factor de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/farmacología
10.
Methods Mol Biol ; 1889: 301-317, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30367422

RESUMEN

Protein phosphorylation is a universal covalent chemical modification of amino acids involved in a large number of biological processes including cell signaling, metabolism, proliferation, differentiation, survival/death, ageing, and many more. Regulation of protein phosphorylation is essential in myogenesis and indeed, when the enzymatic activity of protein kinases is distrupted in myoblasts, myogenesis is affected. In this chapter we describe a method to profile the phosphoproteome of myoblasts using mass spectrometry. Phosphate groups are labile and easily lost during the processing of samples for mass spectrometry. Thus, effective methods to enrich for phosphopeptides from protein extracts have been developed. Here, we discuss and present in detail two such methods that we routinely employ. These methods are based on a sample enrichment step performed on titanium dioxide matrices followed by label-free tandem mass spectrometry and semi-quantitation.


Asunto(s)
Desarrollo de Músculos , Mioblastos/metabolismo , Fosfoproteínas/metabolismo , Proteoma , Proteómica , Transducción de Señal , Cromatografía Liquida , Fosfopéptidos/metabolismo , Fosforilación , Proteómica/métodos , Espectrometría de Masas en Tándem , Flujo de Trabajo
11.
Curr Top Dev Biol ; 126: 23-65, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29305000

RESUMEN

The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.


Asunto(s)
Músculo Esquelético/fisiología , Enfermedades Musculares/fisiopatología , Células Satélite del Músculo Esquelético/fisiología , Nicho de Células Madre/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Músculo Esquelético/citología , Enfermedades Musculares/patología , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Madre/citología
12.
Stem Cell Reports ; 10(3): 970-983, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29429962

RESUMEN

Satellite cells are adult muscle stem cells residing in a specialized niche that regulates their homeostasis. How niche-generated signals integrate to regulate gene expression in satellite cell-derived myoblasts is poorly understood. We undertook an unbiased approach to study the effect of the satellite cell niche on satellite cell-derived myoblast transcriptional regulation and identified the tumor suppressor p53 as a key player in the regulation of myoblast quiescence. After activation and proliferation, a subpopulation of myoblasts cultured in the presence of the niche upregulates p53 and fails to differentiate. When satellite cell self-renewal is modeled ex vivo in a reserve cell assay, myoblasts treated with Nutlin-3, which increases p53 levels in the cell, fail to differentiate and instead become quiescent. Since both these Nutlin-3 effects are rescued by small interfering RNA-mediated p53 knockdown, we conclude that a tight control of p53 levels in myoblasts regulates the balance between differentiation and return to quiescence.


Asunto(s)
Diferenciación Celular/fisiología , Mioblastos/metabolismo , Mioblastos/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Proliferación Celular/fisiología , Imidazoles/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/metabolismo , ARN Interferente Pequeño/metabolismo , Transcripción Genética/fisiología , Regulación hacia Arriba/fisiología
13.
Cell Rep ; 21(6): 1507-1520, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29117557

RESUMEN

Regular endurance training improves muscle oxidative capacity and reduces the risk of age-related disorders. Understanding the molecular networks underlying this phenomenon is crucial. Here, by exploiting the power of computational modeling, we show that endurance training induces profound changes in gene regulatory networks linking signaling and selective control of translation to energy metabolism and tissue remodeling. We discovered that knockdown of the mTOR-independent factor Eif6, which we predicted to be a key regulator of this process, affects mitochondrial respiration efficiency, ROS production, and exercise performance. Our work demonstrates the validity of a data-driven approach to understanding muscle homeostasis.


Asunto(s)
Factores Eucarióticos de Iniciación/metabolismo , Ejercicio Físico , Músculo Esquelético/metabolismo , Acetilación , Animales , Calorimetría , Cromatografía Líquida de Alta Presión , Regulación hacia Abajo , Metabolismo Energético/fisiología , Factores Eucarióticos de Iniciación/deficiencia , Factores Eucarióticos de Iniciación/genética , Redes Reguladoras de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/metabolismo , Condicionamiento Físico Animal , Proteoma/análisis , Especies Reactivas de Oxígeno/metabolismo , Ribosomas/metabolismo , Espectrometría de Masas en Tándem , Transcripción Genética , Regulación hacia Arriba
14.
J Gerontol A Biol Sci Med Sci ; 61(2): 125-35, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16510856

RESUMEN

Interleukin (IL)-12 is the major inducer of T helper cell (Th) 1-type responses. Despite a higher IL-12 production, phytohemagglutinin (PHA)-stimulated peripheral blood mononuclear cells (PBMC), as well as CD4(+) or CD8(+) T cells from elderly donors released interferon (IFN)-gamma amounts similar to those observed in young controls, and underwent only a slight increase in IFN-gamma production after IL-12 costimulation. These findings were not due to an age-related reduction in IL-12 receptor expression. Interestingly, no difference in PHA-triggered signal transducer and activator of transcription 4 (STAT4) phosphorylation between young and elderly donors was found, and a significant IL-12-induced STAT4 activation occurred only in PHA-preactivated cells from the younger group. The age-related defect in IL-12 signaling was STAT4-restricted as it did not involve the p38 mitogen-activated protein kinase (MAPK) pathway. Finally, suppressor of cytokine signaling 3 (SOCS3) expression was significantly higher in unstimulated cells from elderly individuals, and it did not diminish after cell stimulation. These results indicate that a defective STAT4 activation, likely dependent on elevated SOCS3 levels, is involved in the impaired IL-12-dependent T-cell functions with aging.


Asunto(s)
Envejecimiento/fisiología , Interleucina-12/fisiología , Factor de Transcripción STAT4/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Linfocitos T Colaboradores-Inductores/fisiología , Anciano , Anciano de 80 o más Años , Células Cultivadas , Femenino , Humanos , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación , Fitohemaglutininas/farmacología , Proteína 3 Supresora de la Señalización de Citocinas
15.
Cancer Res ; 64(11): 3767-71, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15172982

RESUMEN

Susceptibility of dendritic cells (DCs) to tumor-induced apoptosis reduces their efficacy in cancer therapy. Here we show that delivery within exponentially growing B16 melanomas of DCs treated ex vivo with nitric oxide (NO), released by the NO donor (z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA-NO), significantly reduced tumor growth, with cure of 37% of animals. DETA-NO-treated DCs became resistant to tumor-induced apoptosis because DETA-NO prevented tumor-induced changes in the expression of Bcl-2, Bax, and Bcl-xL; activation of caspase-9; and a reduction in the mitochondrial membrane potential. DETA-NO also increased DC cytotoxic activity against tumor cells and DC ability to trigger T-lymphocyte proliferation. All of the effects of DETA-NO were mediated through cGMP generation. NO and NO-generating drugs may therefore be used to increase the anticancer efficacy of DCs.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Óxido Nítrico/farmacología , Animales , Apoptosis/inmunología , Caspasa 9 , Caspasas/metabolismo , GMP Cíclico/metabolismo , Activación Enzimática , Femenino , Prueba de Cultivo Mixto de Linfocitos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Donantes de Óxido Nítrico/farmacología , Triazenos/farmacología
16.
Skelet Muscle ; 6: 34, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27757223

RESUMEN

BACKGROUND: The skeletal muscle stem cell niche provides an environment that maintains quiescent satellite cells, required for skeletal muscle homeostasis and regeneration. Syndecan-3, a transmembrane proteoglycan expressed in satellite cells, supports communication with the niche, providing cell interactions and signals to maintain quiescent satellite cells. RESULTS: Syndecan-3 ablation unexpectedly improves regeneration in repeatedly injured muscle and in dystrophic mice, accompanied by the persistence of sublaminar and interstitial, proliferating myoblasts. Additionally, muscle aging is improved in syndecan-3 null mice. Since syndecan-3 null myofiber-associated satellite cells downregulate Pax7 and migrate away from the niche more readily than wild type cells, syxndecan-3 appears to regulate satellite cell homeostasis and satellite cell homing to the niche. CONCLUSIONS: Manipulating syndecan-3 provides a promising target for development of therapies to enhance muscle regeneration in muscular dystrophies and in aged muscle.


Asunto(s)
Homeostasis , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/fisiología , Nicho de Células Madre , Sindecano-3/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Células Satélite del Músculo Esquelético/patología , Sindecano-3/genética
17.
Mech Ageing Dev ; 123(10): 1389-402, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12297341

RESUMEN

The age-related impairment of phytohaemagglutinin (PHA)-triggered peripheral blood mononuclear cell (PBMC) proliferation was paralleled by an expansion of CD28 (-) T lymphocytes with a poor capacity to undergo lectin-induced blastogenesis. However, both CD28 (-) and CD28 (+) T cells isolated from aged individuals exhibited a significant reduction of proliferative response to PHA in comparison with young controls, this implies that the CD28-mediated signaling is not the only defective pathway in the elderly. Thus, PBMC or T cell subsets plus monocytes from aged donors were stimulated with PHA and assayed for the production of, or the response to cytokines known to regulate T cell functions. Results can be so summarized: (i). interleukin (IL)-2 as well as IL-10 release was unaffected by age; (ii). in both groups of subjects, IL-15 concentrations were similar to those spontaneously released by PBMC; (iii). surprisingly, IL-12 p70 and IL-12 p40 production by PBMC was markedly increased in the aged group; (iv) in spite of this finding and of the experimental outcome that IFN-gamma synthesis was almost completely dependent on IL-12. PBMC from old individuals did not release higher amounts of IFN-gamma in comparison with young controls; (v). moreover, only a slight increase in IFN-gamma production was observed in PBMC cultures from the aged group as a result of IL-12 and/or IL-15 costimulation; (vi) at the same time, even though IL-12 as well as IL-15 were necessary for an efficient T cell proliferation, the addition of exceeding doses of cytokines proved to be ineffective in enhancing the proliferative outcome of PBMC or of both CD28 (+) and CD28 (-) T cells in the aged group. Taken together, the data outline the role of CD28 and IL-12/IL-15 signaling impairment in T cell proliferative deficiency during senescence.


Asunto(s)
Envejecimiento/inmunología , Células Presentadoras de Antígenos/inmunología , Antígenos CD28/inmunología , Interleucina-12/inmunología , Interleucina-15/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , División Celular , Células Cultivadas , Femenino , Humanos , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-15/biosíntesis , Interleucina-2/biosíntesis , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Fitohemaglutininas/farmacología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/citología
18.
Muscles Ligaments Tendons J ; 2(1): 1-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23738267

RESUMEN

Skeletal muscle is a highly dynamic tissue that can change in size in response to physiological demands and undergo successful regeneration even upon extensive injury. A population of resident stem cells, termed satellite cells, accounts for skeletal muscle plasticity, maintenance and regeneration. Mammalian satellite cells, generated from muscle precursor cells during development, are maintained quiescent in the musculature throughout a lifespan, but ready to activate, proliferate and differentiate into myocytes upon demand. Syndecans are transmembrane heparan sulfate proteoglycans expressed in muscle precursors during embryonic development and in satellite cells during postnatal life. In the last decades a number of crucial functions for syndecans in myogenesis and muscle disease have been described. Here we review the current knowledge of the multiple roles played by syndecans in the skeletal muscle of several animal models and explore future perspectives for human muscle health, with a focus on muscle aging and muscular dystrophy.

19.
J Cell Biol ; 195(1): 147-63, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21949413

RESUMEN

Skeletal muscle contains progenitor cells (satellite cells) that maintain and repair muscle. It also contains muscle side population (SP) cells, which express Abcg2 and may participate in muscle regeneration or may represent a source of satellite cell replenishment. In Abcg2-null mice, the SP fraction is lost in skeletal muscle, although the significance of this loss was previously unknown. We show that cells expressing Abcg2 increased upon injury and that muscle regeneration was impaired in Abcg2-null mice, resulting in fewer centrally nucleated myofibers, reduced myofiber size, and fewer satellite cells. Additionally, using genetic lineage tracing, we demonstrate that the progeny of Abcg2-expressing cells contributed to multiple cell types within the muscle interstitium, primarily endothelial cells. After injury, Abcg2 progeny made a minor contribution to regenerated myofibers. Furthermore, Abcg2-labeled cells increased significantly upon injury and appeared to traffic to muscle from peripheral blood. Together, these data suggest an important role for Abcg2 in positively regulating skeletal muscle regeneration.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Fibras Musculares Esqueléticas/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Células Satélite del Músculo Esquelético/citología
20.
J Cell Biol ; 190(3): 427-41, 2010 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-20696709

RESUMEN

Skeletal muscle postnatal growth and repair depend on satellite cells and are regulated by molecular signals within the satellite cell niche. We investigated the molecular and cellular events that lead to altered myogenesis upon genetic ablation of Syndecan-3, a component of the satellite cell niche. In the absence of Syndecan-3, satellite cells stall in S phase, leading to reduced proliferation, increased cell death, delayed onset of differentiation, and markedly reduced numbers of Pax7(+) satellite cells accompanied by myofiber hypertrophy and an increased number of centrally nucleated myofibers. We show that the aberrant cell cycle and impaired self-renewal of explanted Syndecan-3-null satellite cells are rescued by ectopic expression of the constitutively active Notch intracellular domain. Furthermore, we show that Syndecan-3 interacts with Notch and is required for Notch processing by ADAM17/tumor necrosis factor-alpha-converting enzyme (TACE) and signal transduction. Together, our data support the conclusion that Syndecan-3 and Notch cooperate in regulating homeostasis of the satellite cell population and myofiber size.


Asunto(s)
Desarrollo de Músculos , Receptores Notch/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Sindecano-3/metabolismo , Animales , Ciclo Celular , Diferenciación Celular , Membrana Celular/enzimología , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Ratones , Ratones Endogámicos , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/citología , Transducción de Señal , Sindecano-3/deficiencia , Sindecano-3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA