Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 143(5): 872-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26839363

RESUMEN

Pax3 and Foxc2 have been shown genetically to mutually repress each other in the mouse somite. Perturbation of this balance in multipotent cells of the dermomyotome influences cell fate; upregulation of Foxc2 favours a vascular fate, whereas higher levels of Pax3 lead to myogenesis. Foxc1 has overlapping functions with Foxc2. In Foxc1/2 double-mutant embryos, somitogenesis is severely affected, precluding analysis of somite derivatives. We have adopted a conditional approach whereby mutations in Foxc1 and Foxc2 genes were targeted to Pax3-expressing cells. Inclusion of a conditional reporter allele in the crosses made it possible to follow cells that had expressed Pax3. At the forelimb level, endothelial and myogenic cells migrate from adjacent somites into the limb bud. This population of endothelial cells is compromised in the double mutant, whereas excessive production of myogenic cells is observed in the trunk. However, strikingly, myogenic progenitors fail to enter the limbs, leading to the absence of skeletal muscle. Pax3-positive migratory myogenic progenitors, marked by expression of Lbx1, are specified in the somite at forelimb level, but endothelial progenitors are absent. The myogenic progenitors do not die, but differentiate prematurely adjacent to the somite. We conclude that the small proportion of somite-derived endothelial cells in the limb is required for the migration of myogenic limb progenitors.


Asunto(s)
Células Endoteliales/metabolismo , Miembro Anterior/embriología , Factores de Transcripción Forkhead/genética , Desarrollo de Músculos/fisiología , Factores de Transcripción Paired Box/metabolismo , Somitos/metabolismo , Animales , Movimiento Celular , Separación Celular , Cruzamientos Genéticos , Femenino , Citometría de Flujo , Miembro Anterior/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Hibridación in Situ , Esbozos de los Miembros/embriología , Masculino , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Mutación , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Fenotipo
2.
Stem Cells ; 35(3): 725-738, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27612437

RESUMEN

The transcription factor MEF2C (Myocyte Enhancer Factor 2C) plays an established role in the early steps of myogenic differentiation. However, the involvement of MEF2C in adult myogenesis and in muscle regeneration has not yet been systematically investigated. Alternative splicing of mammalian MEF2C transcripts gives rise to two mutually exclusive protein variants: MEF2Cα2 which exerts a positive control of myogenic differentiation, and MEF2Cα1, in which the α1 domain acts as trans-repressor of the MEF2C pro-differentiation activity itself. However, MEF2Cα1 variants are persistently expressed in differentiating cultured myocytes, suggesting a role in adult myogenesis. We found that overexpression of both MEF2Cα1/α2 proteins in a mouse model of muscle injury promotes muscle regeneration and hypertrophy, with each isoform promoting different stages of myogenesis. Besides the ability of MEF2Cα2 to increase differentiation, we found that overexpressed MEF2Cα1 enhances both proliferation and differentiation of primary myoblasts, and activates the AKT/mTOR/S6K anabolic signaling pathway in newly formed myofibers. The multiple activities of MEF2Cα1 are modulated by phosphorylation of Ser98 and Ser110, two amino acid residues located in the α1 domain of MEF2Cα1. These specific phosphorylations allow the interaction of MEF2Cα1 with the peptidyl-prolyl isomerase PIN1, a regulator of MEF2C functions. Overall, in this study we established a novel regulatory mechanism in which the expression and the phosphorylation of MEF2Cα1 are critically required to sustain the adult myogenesis. The described molecular mechanism will represent a new potential target for the development of therapeutical strategies to treat muscle-wasting diseases. Stem Cells 2017;35:725-738.


Asunto(s)
Empalme Alternativo/genética , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Regeneración , Envejecimiento/metabolismo , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Hipertrofia , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Endogámicos C57BL , Mioblastos/metabolismo , Células 3T3 NIH , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Fosforilación , Unión Proteica , Dominios Proteicos , Células Satélite del Músculo Esquelético/metabolismo , Serina/metabolismo
3.
Development ; 140(6): 1231-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23406902

RESUMEN

The myogenic regulatory factor Myod and insulin-like growth factor 2 (Igf2) have been shown to interact in vitro during myogenic differentiation. In order to understand how they interact in vivo, we produced double-mutant mice lacking both the Myod and Igf2 genes. Surprisingly, these mice display neonatal lethality due to severe diaphragm atrophy. Alteration of diaphragm muscle development occurs as early as 15.5 days post-coitum in the double-mutant embryos and leads to a defect in the terminal differentiation of muscle progenitor cells. A negative-feedback loop was detected between Myod and Igf2 in embryonic muscles. Igf2 belongs to the imprinted H19-Igf2 locus. Molecular analyses show binding of Myod on a mesodermal enhancer (CS9) of the H19 gene. Chromatin conformation capture experiments reveal direct interaction of CS9 with the H19 promoter, leading to increased H19 expression in the presence of Myod. In turn, the non-coding H19 RNA represses Igf2 expression in trans. In addition, Igf2 also negatively regulates Myod expression, possibly by reducing the expression of the Srf transcription factor, a known Myod activator. In conclusion, Igf2 and Myod are tightly co-regulated in skeletal muscles and act in parallel pathways in the diaphragm, where they affect the progression of myogenic differentiation. Igf2 is therefore an essential player in the formation of a functional diaphragm in the absence of Myod.


Asunto(s)
Diafragma/embriología , Epistasis Genética/fisiología , Factor II del Crecimiento Similar a la Insulina/genética , Proteína MioD/genética , ARN Largo no Codificante/genética , Animales , Animales Recién Nacidos , Diafragma/crecimiento & desarrollo , Diafragma/metabolismo , Embrión de Mamíferos , Femenino , Sitios Genéticos , Factor II del Crecimiento Similar a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Desarrollo de Músculos/genética , Proteína MioD/fisiología , Organogénesis/genética , Embarazo , ARN Largo no Codificante/fisiología
4.
J Cell Sci ; 124(Pt 23): 3980-8, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22159413

RESUMEN

We report expression of Pax3, an important regulator of skeletal muscle stem cell behaviour, in the brachial and femoral arteries of adult mice. In these contractile arteries of the limb, but not in the elastic arteries of the trunk, bands of GFP-positive cells were observed in Pax3(GFP/+) mice. Histological and biochemical examination of the vessels, together with clonal analysis after purification of Pax3-GFP-positive cells by flow cytometry, established their vascular smooth muscle identity. These blood-vessel-derived cells do not respond to inducers of other mesodermal cell types, such as bone, however, they can contribute to muscle fibre formation when co-cultured with skeletal muscle cells. This myogenic conversion depends on the expression of Pax3, but is rare and non-cell autonomous as it requires cell fusion. Myocardin, which promotes acquisition of a mature smooth muscle phenotype in these Pax3-GFP-positive cells, antagonises their potential for skeletal muscle differentiation. Genetic manipulation shows that myocardin is, however, positively regulated by Pax3, unlike genes for other myocardin-related factors, MRTFA, MRTFB or SRF. Expression of Pax3 overlaps with that reported for Msx2, which is required for smooth muscle differentiation of blood vessel-derived multipotent mesoangioblasts. These observations are discussed with respect to the origin and function of Pax3-expressing cells in blood vessels, and more general questions of cell fate determination and adult cell plasticity and reprogramming.


Asunto(s)
Arteria Braquial/metabolismo , Arteria Femoral/metabolismo , Miocitos del Músculo Liso/metabolismo , Factores de Transcripción Paired Box/metabolismo , Animales , Arteria Braquial/citología , Diferenciación Celular , Técnicas de Cocultivo , Arteria Femoral/citología , Citometría de Flujo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Transactivadores/genética , Transactivadores/metabolismo , Transfección
5.
FASEB J ; 26(11): 4584-91, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22859371

RESUMEN

Brown fat or brown adipose tissue (BAT), found in newborn mammals as small depots localized in the interscapular region, plays a prominent role in regulating thermogenesis perinatally. The physiological importance of functional BAT has been recently reasserted in human adults. Because myoblasts and adipoblasts emerge from a common mesodermal precursor, we investigated developmental determination and the reciprocal relationship between muscle and adipocyte commitment. Here we show that a mutant mouse defective for both Igf2 and Myod genes exhibits massive BAT hypertrophy compared with wild-type and single-mutant newborns. The increased adipocyte proliferation in BAT of double-mutant newborns was associated with overexpression of the brown fat-specific marker Ucp1. More strikingly, expression of the master key gene Prdm16 involved in the switch between myogenic and brown adipogenic lineages was drastically enhanced. We further demonstrate that concomitant Myod and Igf2 inactivation accelerates differentiation of a brown preadipocyte cell line and induces lipid accumulation and increased Ucp1 and Prdm16 expression. This in vitro approach brings additional support for the implication of both Myod and Igf2 in BAT development. These results provide the first in vivo evidence that a myogenic regulator together with a growth factor act simultaneously but through independent pathways to repress Prdm16, which opens potential therapeutic perspectives for human metabolic disorders.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Proteína MioD/metabolismo , Factores de Transcripción/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/fisiología , Animales , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Heterocigoto , Homocigoto , Factor II del Crecimiento Similar a la Insulina/genética , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteína MioD/genética , ARN Interferente Pequeño , Factores de Transcripción/genética , Proteína Desacopladora 1
6.
Neurodegener Dis ; 11(3): 153-64, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22797053

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is the most common adult-onset neurodegenerative disease characterized by ascending muscle weakness, atrophy and paralysis. Early muscle abnormalities that precede motor neuron loss in ALS may destabilize neuromuscular junctions, and we have previously demonstrated alterations in myogenic regulatory factor (MRF) expression in vivo and in the activation of myofiber-associated skeletal muscle satellite cells (SMSCs) in the mouse model of ALS (SOD1-G93A). METHODS: To elucidate niche dependence versus cell-autonomous mutant SOD1 (mSOD1) toxicity in this model, we measured in vitro proliferation potential and MRF and cyclin gene expression in SMSC cultures derived from fast-twitch extensor digitorum longus and slow-twitch soleus muscles of SOD1-G93A mice. RESULTS: SMSCs from early presymptomatic (p40) to terminal, semi-paralytic (p120) SOD1-G93A mice demonstrated generally lower proliferation potential compared with age-matched controls. However, induced proliferation was observed in surgically denervated wild-type animals and SOD1-G93A animals at p90, when critical denervation arises. SMSCs from fast and slow muscles were similarly affected by mSOD1 expression. Lowered proliferation rate was generally corroborated with decreased relative MRF expression levels, although this was most prominent in early age and was modulated by muscle type origin. Cyclins controlling cell proliferation did not show modifications in their mRNA levels; however, the expression of cyclin-dependent kinase inhibitor 1A (Cdkn1a), which is known to promote myoblast differentiation, was decreased in SOD1-G93A cultures. CONCLUSIONS: Our data suggest that the function of SMSCs is impaired in SOD1-G93A satellite cells from the earliest stages of the disease when no critical motor neuron loss has been described.


Asunto(s)
Proliferación Celular , Células Satélite del Músculo Esquelético/enzimología , Células Satélite del Músculo Esquelético/patología , Superóxido Dismutasa/fisiología , Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/patología , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Transgénicos
7.
Curr Opin Genet Dev ; 18(4): 330-6, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18625314

RESUMEN

In this review we shall discuss recent publications on the heterogeneity of muscle stem cells, signaling pathways that affect their behaviour and regulatory mechanisms that underlie their myogenic fate, with reference to insights provided by work on skeletal muscle formation in the embryo as well as the adult, with the mouse as a model of reference.


Asunto(s)
Músculo Esquelético/embriología , Mioblastos Esqueléticos/fisiología , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Modelos Biológicos , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/citología , Células Satélite del Músculo Esquelético/fisiología , Transducción de Señal/genética
8.
Proc Natl Acad Sci U S A ; 106(32): 13383-7, 2009 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-19666532

RESUMEN

Skeletal muscle stem cells are regulated by Pax3/7. During development, Pax3 is required for the maintenance of these cells in the somite and their migration to sites of myogenesis; high levels of Pax3 interfere with muscle cell differentiation, both in the embryo and in the adult. Quantitative fine-tuning of Pax3 is critical, and microRNAs provide a potential mechanism. We identify microRNA-27b (miR-27b), which directly targets the 3'-UTR of Pax3 mRNA, as such a regulator. miR-27b is expressed in the differentiating skeletal muscle of the embryonic myotome and in activated satellite cells of adult muscle. In vivo overexpression of a miR-27b transgene in Pax3-positive cells in the embryo leads to down-regulation of Pax3, resulting in interference with progenitor cell migration and in premature differentiation. In a complementary experiment, miR-27b inhibitors were transfected into cultures of adult muscle satellite cells that normally express miR-27b at the onset of differentiation, when Pax3 protein levels undergo rapid down-regulation. Interference with miR-27b function results in continuing Pax3 expression leading to more proliferation and a delay in the onset of differentiation. Pax7 levels are not affected. Introduction of miR-27b antagomirs at a site of muscle injury in vivo also affects Pax3 expression and regeneration in vivo. We therefore conclude that miR-27b regulates Pax3 protein levels and this down-regulation ensures rapid and robust entry into the myogenic differentiation program.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Músculo Esquelético/citología , Factores de Transcripción Paired Box/genética , Células Madre/citología , Células Madre/metabolismo , Regiones no Traducidas 3'/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Humanos , Ratones , Ratones Transgénicos , MicroARNs/genética , Datos de Secuencia Molecular , Músculo Esquelético/fisiología , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Somitos/embriología , Somitos/metabolismo
9.
J Cell Biochem ; 112(10): 2825-36, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21608019

RESUMEN

During postnatal growth and after muscle injury, satellite cells proliferate and differentiate into myotubes to form and repair musculature. Comparison of studies on satellite cell proliferation and differentiation characteristics is confounded by the heterogeneity of the experimental conditions used. To examine the influence of sex, age, and fiber-type origin on in vitro properties of satellite cells derived from postnatal muscles, fast extensor digitorum longus (EDL) and slow soleus (SOL) muscles were extracted from male and female mice of 1 week to 3 months of age. Myoblast proliferation and myogenic regulatory factor (MRF) expression was measured from cultures of freshly isolated satellite cells. Higher proliferation rate and elevated Myod1 expression was found in male EDL and SOL derived cells compared with females at age of 40, 60, and 120 days, whereas inverse tendency for cell proliferation was apparent in EDL of juvenile (7-day-old) pups. Myogenin and Mrf4 transcripts were generally elevated in males of 40 and 60 days of age and in female EDL of juveniles. However, these differentiation markers did not significantly correlate with proliferation rate at all ages. Pax7, whose overexpression can block myogenesis, was up-regulated especially in 40-day-old females where MRF expression was low. These results indicate that gender, postnatal age, and muscle fiber origin affect proliferation and muscle transcription factor expression in vitro. The results also support the view that satellite cells originating from slow and fast muscles are intrinsically different and warrant further studies on the effect of cell origin for therapeutic approaches.


Asunto(s)
Células Satélite del Músculo Esquelético/citología , Factores de Edad , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Femenino , Inmunohistoquímica , Masculino , Ratones , Proteína MioD/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Factores Sexuales
10.
J Cell Biol ; 172(1): 91-102, 2006 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-16380438

RESUMEN

The growth and repair of skeletal muscle after birth depends on satellite cells that are characterized by the expression of Pax7. We show that Pax3, the paralogue of Pax7, is also present in both quiescent and activated satellite cells in many skeletal muscles. Dominant-negative forms of both Pax3 and -7 repress MyoD, but do not interfere with the expression of the other myogenic determination factor, Myf5, which, together with Pax3/7, regulates the myogenic differentiation of these cells. In Pax7 mutants, satellite cells are progressively lost in both Pax3-expressing and -nonexpressing muscles. We show that this is caused by satellite cell death, with effects on the cell cycle. Manipulation of the dominant-negative forms of these factors in satellite cell cultures demonstrates that Pax3 cannot replace the antiapoptotic function of Pax7. These findings underline the importance of cell survival in controlling the stem cell populations of adult tissues and demonstrate a role for upstream factors in this context.


Asunto(s)
Músculo Esquelético/citología , Proteína MioD/metabolismo , Factor de Transcripción PAX7/fisiología , Factores de Transcripción Paired Box/fisiología , Células Satélite del Músculo Esquelético/fisiología , Animales , Apoptosis , Ciclo Celular , Diferenciación Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Mutación , Factor de Transcripción PAX3 , Factor de Transcripción PAX7/genética , Factores de Transcripción Paired Box/genética , Células Satélite del Músculo Esquelético/citología
11.
Stem Cells ; 27(5): 1098-108, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19415780

RESUMEN

Cell replacement therapy using stem cell transplantation holds much promise in the field of regenerative medicine. In the area of hematopoietic stem cell transplantation, O(6)-methylguanine-DNA methyltransferase MGMT (P140K) gene-mediated drug resistance-based in vivo enrichment strategy of donor stem cells has been shown to achieve up to 75%-100% donor cell engraftment in the host's hematopoietic stem cell compartment following repeated rounds of selection. This strategy, however, has not been applied in any other organ system. We tested the feasibility of using this MGMT (P140K)-mediated enrichment strategy for cell transplantation in skeletal muscles of mice. We demonstrate that muscle cells expressing an MGMT (P140K) drug resistance gene can be protected and selectively enriched in response to alkylating chemotherapy both in vitro and in vivo. Upon transplantation of MGMT (P140K)-expressing male CD34(+ve) donor stem cells isolated from regenerating skeletal muscle into injured female muscle treated with alkylating chemotherapy, donor cells showed enhanced engraftment in the recipient muscle 7 days following transplantation as examined by quantitative-polymerase chain reaction using Y-chromosome specific primers. Fluorescent in situ hybridization analysis using a Y-chromosome paint probe revealed donor-derived de novo muscle fiber formation in the recipient muscle 14 days following transplantation, with approximately 12.5% of total nuclei within the regenerated recipient muscle being of donor origin. Following engraftment, the chemo-protected donor CD34(+ve) cells induced substantial endogenous regeneration of the chemo-ablated host muscle that is otherwise unable to self-regenerate. We conclude that the MGMT (P140K)-mediated enrichment strategy can be successfully implemented in muscle.


Asunto(s)
Resistencia a Medicamentos , Músculo Esquelético/enzimología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Trasplante de Células Madre , Animales , Animales Modificados Genéticamente , Antígenos CD34/metabolismo , Carmustina/farmacología , Diferenciación Celular/efectos de los fármacos , Separación Celular , Supervivencia Celular/efectos de los fármacos , Guanina/análogos & derivados , Guanina/farmacología , Humanos , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/enzimología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Mioblastos/citología , Mioblastos/efectos de los fármacos , Mioblastos/enzimología , Regeneración/efectos de los fármacos , Retroviridae/genética , Transducción Genética
13.
Elife ; 72018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30106373

RESUMEN

Skeletal muscle regeneration depends on satellite cells. After injury these muscle stem cells exit quiescence, proliferate and differentiate to regenerate damaged fibres. We show that this progression is accompanied by metabolic changes leading to increased production of reactive oxygen species (ROS). Using Pitx2/3 single and double mutant mice that provide genetic models of deregulated redox states, we demonstrate that moderate overproduction of ROS results in premature differentiation of satellite cells while high levels lead to their senescence and regenerative failure. Using the ROS scavenger, N-Acetyl-Cysteine (NAC), in primary cultures we show that a physiological increase in ROS is required for satellite cells to exit the cell cycle and initiate differentiation through the redox activation of p38α MAP kinase. Subjecting cultured satellite cells to transient inhibition of P38α MAP kinase in conjunction with NAC treatment leads to their rapid expansion, with striking improvement of their regenerative potential in grafting experiments.


Asunto(s)
Proteínas de Homeodominio/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Regeneración/genética , Factores de Transcripción/genética , Acetilcisteína/administración & dosificación , Animales , Diferenciación Celular/genética , Senescencia Celular/genética , Ratones , Músculo Esquelético/citología , Mutación , Oxidación-Reducción , Especies Reactivas de Oxígeno , Células Satélite del Músculo Esquelético , Células Madre/citología , Proteína del Homeodomínio PITX2
14.
Eur J Cardiothorac Surg ; 31(3): 444-51, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17223566

RESUMEN

OBJECTIVE: Cell therapy may be a promising alternative or adjunct to current treatment modalities for ischemic heart failure. But little is known on the impact of myogenic cell transplantation in large animal models of non-ischemic cardiomyopathy. The aim of the present study was to explore whether an ovine model of toxin-induced heart disease could benefit from non-cultured skeletal muscle cell transplantation. METHODS: Sequential intracoronary injections of doxorubicin (0.75 mg/kg) were carried out every 2 weeks until echocardiographic detection of myocardial dysfunction. Sheep were then randomly assigned to either non-cultured cell transplantation (n=8) or placebo injection (n=5). For the cell therapy group, a skeletal muscle biopsy (about 10 g) was explanted from each animal approximately 3h before grafting. After thoracotomy, 20 epicardial injections were carried out. The animals were assessed one last time before sacrifice, 2 months after the thoracotomy. Cells were tracked with cmDiI (red fluorescence) and characterized with immunohistochemistry with monoclonal antibodies to a fast skeletal isoform of myosin heavy chain. RESULTS: Two months after intramyocardial grafting, tissue Doppler imaging and conventional echocardiographic assessment of the groups showed a marked improvement in the non-cultured cell therapy group. Ejection fraction (EF) (p<0.05) as well as systolic endocardial velocities (p<0.01) improved versus the placebo group. CmDiI and skeletal myosin heavy chain expression was detected in all animals at 2 months after implantation confirming engraftment of skeletal muscle cells. CONCLUSIONS: In conclusion, our data indicate that non-cultured muscle cell transplantation is feasible and may translate into a functional benefit in an ovine model of dilated heart failure.


Asunto(s)
Insuficiencia Cardíaca/terapia , Fibras Musculares Esqueléticas/trasplante , Animales , Velocidad del Flujo Sanguíneo , Supervivencia Celular , Células Cultivadas , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Doxorrubicina , Ecocardiografía Doppler , Estudios de Factibilidad , Supervivencia de Injerto , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/fisiopatología , Miocardio/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Oveja Doméstica , Volumen Sistólico
15.
Circulation ; 107(24): 3088-92, 2003 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-12810607

RESUMEN

BACKGROUND: There is compelling evidence showing that cellular cardiomyoplasty can improve cardiac function. Considering the potential benefit of using noncultured muscle cells (little time, lower cost, reduced risk of contamination), we investigated the feasibility of grafting cells obtained directly after enzymatic dissociation of skeletal muscle biopsies into ovine myocardium. We hypothesized that those noncultured muscle cells would engraft massively. METHODS AND RESULTS: Autologous, intramyocardial skeletal muscle cell implantation was performed in 8 sheep. A skeletal muscle biopsy sample ( approximately 10 g) was explanted from each animal. The sheep were left to recover for approximately 3 hours and reanesthetized when the cells were ready for implantation. A left fifth intercostal thoracotomy was performed, and 10 epicardial injections of the muscle preparation (between 10 and 20 million cells) were carried out. All sheep were euthanized 3 weeks after myocardial implantation. Immunohistochemistry was performed with monoclonal antibodies to a fast skeletal isoform of myosin heavy chain. Skeletal myosin heavy-chain expression was detected in all slides at 3 weeks after implantation in 8 of 8 animals, confirming engraftment of skeletal muscle cells. Massive areas of engraftment (from 2 to 9 mm in diameter) or discrete loci were noted within the myocardial wall. CONCLUSIONS: Our results indicate that noncultured skeletal muscle cells can successfully and massively engraft in ovine myocardium. Thus, avoiding the cell culture expansion phase is feasible and could become a promising option for cellular cardiomyoplasty.


Asunto(s)
Supervivencia de Injerto/fisiología , Músculo Esquelético/citología , Músculo Esquelético/trasplante , Miocardio/citología , Animales , Separación Celular , Células Cultivadas , Estudios de Factibilidad , Inmunohistoquímica , Músculo Esquelético/metabolismo , Proteína MioD/biosíntesis , Cadenas Pesadas de Miosina/biosíntesis , Ovinos , Trasplante Autólogo
16.
J Thorac Cardiovasc Surg ; 129(5): 1119-27, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15867789

RESUMEN

OBJECTIVE: Beyond the first 2 months of life, pulmonary artery banding is warranted before two-stage arterial switch operation. The aim of this study was to explore whether myogenic cell transplantation could contribute to right ventricular function during pulmonary artery constriction in an ovine model. METHODS: Sixteen rams were assigned to one of the following groups: group 1, simple pulmonary artery banding (n = 5); group 2, pulmonary artery banding and cell implantation in the right ventricle (n = 7); and group 3, pulmonary artery banding and placebo injection in the right ventricle (n = 4). Hemodynamic assessment with pressure-volume loops was performed on days 0 and 60. The pulmonary artery banding and the injections were achieved through a left fourth intercostal thoracotomy. Autologous myogenic cell implantation was carried out with a noncultured cell preparation, as previously described by our group. Implanted sites were processed with monoclonal antibodies to a fast skeletal-specific isoform of myosin heavy chain (MY32). RESULTS: Skeletal myosin heavy chain expression was detected at 2 months after noncultured cell implantation in all grafted animals. Right ventricular training resulted in statistically significant increased signs of contractility in all three groups. There was no observed difference, however, between the cell therapy group and the other two groups with respect to signs of cardiac function. CONCLUSION: Successful engraftment of noncultured cells into right ventricular myocardium did not translate into a functional benefit that we could demonstrate in our ovine model. Cellular therapy thus is probably not useful to strengthen a left ventricle being retrained through pulmonary artery banding before arterial switch operation. However, cell transplantation may affect the outcome of right ventricular failure long term after atrial switch operation. Although preliminary, this investigation paves the way for further research into cellular cardiomyoplasty, right ventricular failure, and congenital heart disease.


Asunto(s)
Trasplante de Células/métodos , Modelos Animales de Enfermedad , Arteria Pulmonar/cirugía , Células Satélite del Músculo Esquelético/trasplante , Transposición de los Grandes Vasos/cirugía , Factores de Edad , Animales , Biopsia , Cateterismo Cardíaco , Gasto Cardíaco , Supervivencia Celular , Terapia Combinada , Constricción , Ventrículos Cardíacos , Inyecciones Intramusculares , Masculino , Contracción Miocárdica , Miocardio , Presión Esfenoidal Pulmonar , Distribución Aleatoria , Ovinos , Volumen Sistólico , Trasplante Autólogo , Transposición de los Grandes Vasos/fisiopatología , Resultado del Tratamiento , Función Ventricular Derecha
18.
Neurosci Lett ; 352(1): 70-2, 2003 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-14615053

RESUMEN

The transplantation of progenitor muscle cells in striated skeletal muscle of mdx mice, a model of dystrophin deficiency, is well known to induce the formation of mosaic fibres expressing dystrophin near the site of injection. We tried to determine if the number of injected cells is related to the number of dystrophin-positive fibres. The grafted cells provided by 5 day-old C57Bl10 mice are syngenic to mdx mice and were cultured to select undifferentiated progenitors. Dystrophin-positive fibres distinct to 'revertant' fibres were detectable 10 days following the graft of as few as 10(3) cells. The number of dystrophin-positive fibres increases logarithmically with the number of grafted cells. The data indicate that the number of dystrophin-positive fibres plateaus above 5x10(5)-10(6) grafted cells and that a greater number of progenitor cells is not required to obtain a better result.


Asunto(s)
Distrofina/biosíntesis , Células Musculares/trasplante , Músculo Esquelético/trasplante , Células Madre/citología , Células Madre/metabolismo , Trasplantes , Animales , Distrofina/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/trasplante , Músculo Esquelético/química , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Células Madre/química
19.
Free Radic Biol Med ; 75 Suppl 1: S37, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26461356

RESUMEN

Adult tissue homeostasis and regeneration rely on tissue stem cell populations that generate committed precursors and differentiated cells while maintaining a pool of stem cells. In adult skeletal muscle, such cells, called satellite cells, remain quiescent at the periphery of muscle fibers. Upon injury they undergo activation, proliferation and differentiation to replace damaged fibers and also self-renew to reconstitute the muscle stem cell pool. During regeneration, the transition from a quiescent muscle stem cell to a differentiated fiber is accompanied by major metabolic changes. Such changes, and notably the switch from a glycolytic proliferative progenitor state to an oxidative post-mitotic differentiated state, require extensive mitochondrial biogenesis that takes place at the onset of differentiation and leads to increased ROS production. However, it is unclear whether this enhanced ROS production/mitochondrial content reflects an adaptation to the rising energy demand or whether it constitutes an essential regulation element of the differentiation program.To investigate the potential role of this metabolic switch and more specifically of reactive oxygen species during muscle regeneration, we took advantage of mouse mutants for Pitx2 and Pitx3 genes. Both genes are involved in foetal myogenesis where they have been identified as key regulators of the redox state preventing excessive ROS levels and DNA damage as cells undergo differentiation. We have now analyzed adult single and double Pitx2:Pitx3 conditional mutant mouse lines targeted to the muscle stem cell compartment. Double mutant satellite cells undergo senescence with impaired regeneration after injury, whereas in single Pitx3 mutants, premature differentiation occurs. We show that these effects are directly linked to dose-dependent changes in ROS levels and can be reversed by lowering ROS with the N-acetylcystein, supporting the notion that a controlled increase in ROS is required for differentiation of muscle stem cells.

20.
Dev Cell ; 29(4): 392-405, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24871946

RESUMEN

During development, major metabolic changes occur as cells become more specialized within a lineage. In the case of skeletal muscle, differentiation is accompanied by a switch from a glycolytic proliferative progenitor state to an oxidative postmitotic differentiated state. Such changes require extensive mitochondrial biogenesis leading to increased reactive oxygen species (ROS) production that needs to be balanced by an antioxidant system. Our analysis of double conditional Pitx2/3 mouse mutants, both in vivo during fetal myogenesis and ex vivo in primary muscle cell cultures, reveals excessive upregulation of ROS levels leading to DNA damage and apoptosis of differentiating cells. This is a consequence of downregulation of Nrf1 and genes for antioxidant enzymes, direct targets of Pitx2/3, leading to decreased expression of antioxidant enzymes, as well as impairment of mitochondrial function. Our analysis identifies Pitx2 and Pitx3 as key regulators of the intracellular redox state preventing DNA damage as cells undergo differentiation.


Asunto(s)
Proteínas de Homeodominio/genética , Factor Nuclear 1 de Respiración/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética , Animales , Apoptosis/genética , Diferenciación Celular/genética , Células Cultivadas , Daño del ADN/genética , Ratones , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Músculo Esquelético/crecimiento & desarrollo , Mutación , Factor Nuclear 1 de Respiración/biosíntesis , Oxidación-Reducción , Regulación hacia Arriba , Proteína del Homeodomínio PITX2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA