Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 36(3): 458-466, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29230914

RESUMEN

Notch signaling is essential to maintain skeletal muscle stem cells in quiescence. However, the precise roles of different Notch receptors are incompletely defined. Here, we demonstrate a role for Notch3 (N3) in the self-renewal of muscle stem cells. We found that N3 is active in quiescent C2C12 reserve cells (RCs), and N3 over-expression and knockdown studies in C2C12 and primary satellite cells reveal a role in self-renewal. The Notch ligand Delta-like 4 (Dll4) is expressed by newly formed myotubes and interaction with this ligand is sufficient to maintain N3 activity in quiescent C2C12 RCs to prevent activation and progression into the cell cycle. Thus, our data suggest a model whereby during regeneration, expression of Dll4 by nascent muscle fibers triggers N3 signaling in associated muscle stem cells to recruit them to quiescence, thereby renewing the stem cell pool. Stem Cells 2018;36:458-466.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Mioblastos Esqueléticos/metabolismo , Receptor Notch3/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular , Células Cultivadas , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/citología , Receptor Notch3/genética , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo
2.
Exp Cell Res ; 313(2): 341-56, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17123512

RESUMEN

To be effective for tissue repair, satellite cells (the stem cells of adult muscle) must survive the initial activation from quiescence. Using an in vitro model of satellite cell activation, we show that erbB1, erbB2 and erbB3, members of the EGF receptor tyrosine kinase family, appear on satellite cells within 6 h of activation. We show that signalling via erbB2 provides an anti-apoptotic survival mechanism for satellite cells during the first 24 h, as they progress to a proliferative state. Inhibition of erbB2 signalling with AG825 reduced satellite cell numbers, concomitant with elevated caspase-8 activation and TUNEL labelling of apoptotic satellite cells. In serum-free conditions, satellite cell apoptosis could be largely prevented by a mixture of erbB1, erbB3 and erbB4 ligand growth factors, but not by neuregulin alone (erbB3/erbB4 ligand). Furthermore, using inhibitors specific to discrete intracellular signalling pathways, we identify MEK as a pro-apoptotic mediator, and the erbB-regulated factor STAT3 as an anti-apoptotic mediator during satellite cell activation. These results implicate erbB2 signalling in the preservation of a full compliment of satellite cells as they activate in the context of a damaged muscle.


Asunto(s)
Células Musculares/citología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-2/metabolismo , Células Satélite del Músculo Esquelético/enzimología , Animales , Apoptosis/efectos de los fármacos , Benzotiazoles/farmacología , Supervivencia Celular , Humanos , Ligandos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Células Musculares/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas , Proteínas Tirosina Quinasas Receptoras/análisis , Receptor ErbB-2/análisis , Receptor ErbB-2/antagonistas & inhibidores , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/fisiología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/enzimología , Tirfostinos/farmacología
3.
J Cell Sci ; 119(Pt 9): 1824-32, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16608873

RESUMEN

Skeletal muscle growth and regeneration are attributed to satellite cells - muscle stem cells resident beneath the basal lamina that surrounds each myofibre. Quiescent satellite cells express the transcription factor Pax7 and when activated, coexpress Pax7 with MyoD. Most then proliferate, downregulate Pax7 and differentiate. By contrast, others maintain Pax7 but lose MyoD and return to a state resembling quiescence. Here we show that Pax7 is able to drive transcription in quiescent and activated satellite cells, and continues to do so in those cells that subsequently cease proliferation and withdraw from immediate differentiation. We found that constitutive expression of Pax7 in satellite-cell-derived myoblasts did not affect MyoD expression or proliferation. Although maintained expression of Pax7 delayed the onset of myogenin expression it did not prevent, and was compatible with, myogenic differentiation. Constitutive Pax7 expression in a Pax7-null C2C12 subclone increased the proportion of cells expressing MyoD, showing that Pax7 can act genetically upstream of MyoD. However these Pax7-null cells were unable to differentiate into normal myotubes in the presence of Pax7. Therefore Pax7 may be involved in maintaining proliferation and preventing precocious differentiation, but does not promote quiescence.


Asunto(s)
Músculo Esquelético/citología , Proteína MioD/metabolismo , Factor de Transcripción PAX7/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Transcripción Genética , Animales , Diferenciación Celular/fisiología , Fusión Celular , Proliferación Celular , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Proteína MioD/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Satélite del Músculo Esquelético/citología
4.
Gene Expr Patterns ; 5(1): 3-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15533812

RESUMEN

Heparin-binding EGF-like growth factor (HB-EGF) is a potent mitogen and chemoattractant for diverse cell types including, keratinocytes, fibroblasts and vascular smooth muscle cells. In adult mice, skeletal muscle and endothelial cells prominently express HB-EGF, although analysis of embryonic expression has been limited to studies of heart and kidney development. Here we survey HB-EGF mRNA expression in E7.5-E15 mouse embryos and show that HB-EGF is expressed in branchial arches, limb buds and, transiently, in mature somites between E9.25 and E11. This somitic expression is restricted to the myotomal compartment. Intriguingly, within myotome pairs, the expression of HB-EGF is stronger on the left side of the body, whilst cognate receptors, ErbB1 and ErbB4, are symmetrically expressed in left and right somite pairs. In iv/iv mutant embryos, with inverted left-right body axis, the expression of HB-EGF was also inverted, now being stronger in myotomes on the right side of the body. Thus, the expression of HB-EGF in myotome pairs is regulated by global cues that define the left-right body axis.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Ratones/embriología , Animales , Región Branquial/metabolismo , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular , Esbozos de los Miembros , Ratones/genética , Ratones/metabolismo , Ratones Endogámicos C57BL , Somitos/metabolismo
5.
Dev Dyn ; 231(4): 795-800, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15499557

RESUMEN

Most muscle originates from the myotomal compartment of the somites, paired structures flanking the neural tube. Whereas vertebrate embryos show molecular and morphological asymmetry about the left-right body axis, somitic myogenesis is thought to occur symmetrically. Here, we provide the first evidence that myotome pairs are transiently left-right asymmetric, with higher expression of alpha-skeletal actin and myosin light chain 3F (MLC3F) on the left side between embryonic day 9.5-10.25. In iv mutants with situs inversus, the asymmetric expression of alpha-skeletal actin and MLC3F was inverted, showing that this process is regulated by global left-right axis cues, initiated before gastrulation. However, although left-sided identity is later maintained by Pitx2 genes, we found that Pitx2c null embryos have normal left-biased expression of alpha-skeletal actin and MLC3F. Myotome asymmetry, therefore, is downstream of the iv mutation but upstream of, or unrelated to, the Pitx2c pathway.


Asunto(s)
Actinas/genética , Desarrollo Embrionario/fisiología , Músculo Esquelético/embriología , Cadenas Ligeras de Miosina/genética , Situs Inversus/fisiopatología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Esquelético/fisiología , Proteínas Nucleares/genética , Embarazo , Situs Inversus/genética , Factores de Transcripción , Proteína del Homeodomínio PITX2
6.
Circulation ; 110(11 Suppl 1): II219-24, 2004 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-15364866

RESUMEN

BACKGROUND: Poor survival of grafted cells is a major factor hindering the therapeutic effect of cell transplantation; however, the causes of cell death remain unclear. We hypothesized that interleukin-1beta (IL-1beta) might play a role in the acute inflammatory response and graft death after cell transplantation and that inhibition of IL-1beta might improve graft survival. METHODS AND RESULTS: 14C-labeled male skeletal muscle precursor cells were implanted into female mouse hearts by direct intramuscular injection. The amount of 14C-label provides an estimate of the surviving cell number, whereas the amount of male-specific Smcy gene measured by polymerase chain reaction indicates the total (surviving+proliferated) number of donor-derived cells. At 10 minutes after implantation, 44.8+/-2.4% of the grafted cells survived and this steadily decreased to 14.6+/-1.1% by 24 hours, and to 7.9+/-0.6% by 72 hours (n=6 in each point). Proliferation of the surviving cells, which began after 24 hours, resulted in an increase in the total cell number from 15.5+/-0.8% at 24 hours to 24.4+/-1.6% at 72 hours. Acute inflammation was prominent at 24 hours and was reduced by 72 hours, in parallel with IL-1beta expression. Administration of anti-IL-1beta antibody improved graft survival at both 24 (25.6+/-1.6%) and 72 hours (14.8+/-1.1%) and resulted in a 2-fold increase in the total cell number at 72 hours (45.8+/-2.4%). The effects of IL-1beta inhibition corresponded with a reduced inflammatory response. CONCLUSIONS: IL-1beta is involved in acute inflammation and graft death after direct intramyocardial cell transplantation. Targeted inhibition of IL-1beta may be a useful strategy to improve graft survival.


Asunto(s)
Interleucina-1/fisiología , Mioblastos/trasplante , Miocarditis/etiología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Biomarcadores , Diferenciación Celular , División Celular , Línea Celular Transformada/trasplante , Supervivencia Celular/efectos de los fármacos , Trasplante de Células/efectos adversos , Femenino , Supervivencia de Injerto/efectos de los fármacos , Histona Demetilasas , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Interleucina-1/antagonistas & inhibidores , Interleucina-1/biosíntesis , Interleucina-1/genética , Masculino , Ratones , Mioblastos/patología , Miocarditis/tratamiento farmacológico , Miocarditis/prevención & control , Miocardio/metabolismo , Peroxidasa/análisis , Proteínas/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Dev Biol ; 273(2): 454-65, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15328025

RESUMEN

The myogenic regulatory factor Myf5 is integral to the initiation and control of skeletal muscle formation. In adult muscle, Myf5 is expressed in satellite cells, stem cells of mature muscle, but not in the myonuclei that sustain the myofibre. Using the Myf5(nlacZ/+) mouse, we now show that Myf5 is also constitutively expressed in muscle spindles-stretch-sensitive mechanoreceptors, while muscle denervation induces extensive reactivation of the Myf5 gene in myonuclei. To identify the elements involved in the regulation of Myf5 in adult muscle, we analysed reporter gene expression in a transgenic bacterial artificial chromosome (BAC) deletion series of the Mrf4/Myf5 locus. A BAC carrying 140 kb upstream of the Myf5 transcription start site was sufficient to drive all aspects of Myf5 expression in adult muscle. In contrast, BACs carrying 88 and 59 kb upstream were unable to drive consistent expression in satellite cells, although expression in muscle spindles and reactivation of the locus in myonuclei were retained. Therefore, as during development, multiple enhancers are required to generate the full expression pattern of Myf5 in the adult. Together, these observations show that elements controlling adult Myf5 expression are genetically separable and possibly distinct from those that control Myf5 during development. These studies are a first step towards identifying cognate transcription factors involved in muscle stem cell regulation.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Musculares/genética , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Transactivadores/genética , Animales , Cromosomas Artificiales Bacterianos/genética , Técnicas de Cultivo , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Mecanorreceptores/crecimiento & desarrollo , Mecanorreceptores/metabolismo , Ratones , Ratones Transgénicos , Desnervación Muscular , Husos Musculares/crecimiento & desarrollo , Husos Musculares/metabolismo , Músculo Esquelético/citología , Factor 5 Regulador Miogénico , Células Satélite del Músculo Esquelético/metabolismo
8.
J Cell Biol ; 166(3): 347-57, 2004 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-15277541

RESUMEN

Growth, repair, and regeneration of adult skeletal muscle depends on the persistence of satellite cells: muscle stem cells resident beneath the basal lamina that surrounds each myofiber. However, how the satellite cell compartment is maintained is unclear. Here, we use cultured myofibers to model muscle regeneration and show that satellite cells adopt divergent fates. Quiescent satellite cells are synchronously activated to coexpress the transcription factors Pax7 and MyoD. Most then proliferate, down-regulate Pax7, and differentiate. In contrast, other proliferating cells maintain Pax7 but lose MyoD and withdraw from immediate differentiation. These cells are typically located in clusters, together with Pax7-ve progeny destined for differentiation. Some of the Pax7+ve/MyoD-ve cells then leave the cell cycle, thus regaining the quiescent satellite cell phenotype. Significantly, noncycling cells contained within a cluster can be stimulated to proliferate again. These observations suggest that satellite cells either differentiate or switch from terminal myogenesis to maintain the satellite cell pool.


Asunto(s)
Diferenciación Celular/fisiología , Células Satélite del Músculo Esquelético/fisiología , Animales , Regulación hacia Abajo , Genes Reporteros , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Proteína MioD/metabolismo , Factor de Transcripción PAX7
9.
FASEB J ; 18(10): 1153-5, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15155562

RESUMEN

Survival and proliferation of skeletal myoblasts within the cardiac environment are crucial to the therapeutic efficacy of myoblast transplantation to the heart. We have analyzed the early dynamics of myoblasts implanted into the myocardium and investigated the mechanisms underlying graft attrition. At 10 min after implantation of [14C]thymidine-labeled male myoblasts into female mice hearts, 14C measurement showed that 39.2 +/- 3.0% of the grafted cells survived, and this steadily decreased to 16.0 +/- 1.7% by 24 h and to 7.4 +/- 0.9% by 72 h. PCR of male-specific Smcy gene calculated that the total (surviving plus proliferated) number of donor-derived cells was 18.3 +/- 1.6 and 23.3 +/- 1.3% at 24 and 72 h, respectively, indicating that proliferation of the surviving cells began after 24 h. Acute inflammation became prominent by 24 h and was reduced by 72 h as indicated by myeloperoxidase activity and histological findings. Multiplex RT-PCR revealed corresponding changes in IL-1beta, TGF-beta, IL-6, and TNF-alpha expression. Treatment with CuZn-superoxide dismutase attenuated the initial rapid death and resulted in enhanced cell numbers afterward, giving a twofold increased total number at 72 h compared with the nontreatment. This effect was associated with reduced inflammatory response, suggesting a causative role for superoxide in the initial rapid graft death and subsequent inflammation. These data describe the early dynamics of myoblasts implanted into the myocardium and suggest that initial oxidative stress and following inflammatory response may be important mechanisms contributing to acute graft attrition, both of which could be potential therapeutic targets to improve the efficiency of cell transplantation to the heart.


Asunto(s)
Rechazo de Injerto/patología , Mioblastos Esqueléticos/trasplante , Miocardio/patología , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Recuento de Células/instrumentación , Muerte Celular , Línea Celular/trasplante , Citocinas/análisis , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mioblastos Esqueléticos/patología , Miocarditis/etiología , Miocarditis/patología , Estrés Oxidativo , Peroxidasa/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/farmacología , Superóxido Dismutasa/uso terapéutico
10.
Proc Natl Acad Sci U S A ; 101(12): 4216-21, 2004 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-15020774

RESUMEN

After myocardial infarction (MI), adverse remodeling with left ventricular (LV) dilatation is a major determinant of poor outcome. Skeletal myoblast (SkM) implantation improves cardiac function post-MI, although the mechanism is unclear. IL-1 influences post-MI hypertrophy and collagen turnover and is implicated in SkM death after grafting. We hypothesized that SkM expressing secretory IL-1 receptor antagonist (sIL-1ra) at MI border zones would specifically attenuate adverse remodeling and exhibit improved graft cell number. Stable murine male SkM lines (5 x 10(5) cells), expressing or nonexpressing (cont) for sIL-1ra, were implanted into infarct border zones of female nude mice immediately after left coronary artery occlusion. LV ejection fraction (LVEF), end-diastolic diameter, and transmitral peak early/late (E/A) flow velocity ratio were determined by echocardiography. Cardiac myocyte hypertrophy and fibrosis were assessed by morphometry, picrosirius red staining, and hydroxyproline assay. At 3 weeks, cont-SkM-engrafted hearts showed reduced hypertrophy, improved LVEF (55.7 +/- 1.2% vs. MI-only: 40.3 +/- 2.9%), and preserved E/A ratios. sIL-1ra-SkM implantation enhanced these effects (LVEF, 67.0 +/- 2.3%) and significantly attenuated LV dilatation (LV end-diastolic diameter, 4.0 +/- 1.1 mm vs. cont-SkM, 4.5 +/- 1.2 mm vs. MI-only, 4.8 +/- 1.8 mm); this was associated with greater graft numbers, as shown by PCR for male-specific smcy gene. Enzyme zymography showed attenuated matrix metalloproteinase-2 and -9 up-regulation post-MI by either donor SkM type, although infarct-remote zone collagen was reduced only with sIL-1ra-SkM. These results suggest that SkM implantation improves cardiac function post-MI by modulation of adverse remodeling, and that this effect can be significantly enhanced by targeting IL-1 as a key upstream regulator of both adverse remodeling and graft cell death.


Asunto(s)
Interleucina-1/antagonistas & inhibidores , Mioblastos Esqueléticos/trasplante , Miocardio/metabolismo , Sialoglicoproteínas/genética , Animales , Colágeno/biosíntesis , Colágeno/genética , Matriz Extracelular/metabolismo , Proteína Antagonista del Receptor de Interleucina 1 , Metaloproteinasas de la Matriz/biosíntesis , Metaloproteinasas de la Matriz/genética , Ratones , Mioblastos Esqueléticos/metabolismo , Infarto del Miocardio , Miocitos Cardíacos/metabolismo , Receptores de Interleucina-1/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sialoglicoproteínas/metabolismo
11.
Cell Tissue Res ; 315(2): 233-42, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14648195

RESUMEN

Demonstration of the importance of the paired box transcription factor Pax7 for the murine myosatellite cell population, with persistent expression in mature skeletal muscle, prompted us to investigate the distribution of Pax7 protein in biopsy samples of normal and pathological human skeletal limb muscle. Immunostaining for M-cadherin, an adhesion molecule present at the interface between myofibre and satellite cell, and the characteristic position adjacent to the muscle fibre and beneath the fibre's basement membrane were used to identify satellite cells. Anti-Pax7 reactivity was found in the majority of satellite cells but a small population was Pax7 negative. Neither could we identify Pax7-positive nuclei in freshly regenerating myotubes or in presumed myoblasts in these biopsies. Similarly, in myogenic cell cultures derived from the explantation of human foetal muscle Pax7 expression was low or undetectable at the proliferative myoblast stage but it became prominent in an increasing proportion of mononucleate cells after the induction of differentiation. This expression was, however, restricted to mononucleate cells; it did not persist into the differentiation stage of newly formed multinucleate myotubes. Despite this, in the biopsy samples, we occasionally found Pax7-positive nuclei in muscle fibres that seemed to be undergoing degenerative changes. Most of these were found to be the nuclei of cells engaged in focal regenerative processes, but Pax7 re-expression by myonuclei "in distress" cannot be ruled out entirely.


Asunto(s)
Cadherinas/metabolismo , Núcleo Celular/metabolismo , Proteínas de Homeodominio/metabolismo , Músculo Esquelético/patología , Células Satélite del Músculo Esquelético/citología , Biopsia , Células Cultivadas , Humanos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Miofibrillas/metabolismo , Factor de Transcripción PAX7 , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo
12.
Exp Cell Res ; 281(1): 39-49, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12441128

RESUMEN

The satellite cell compartment provides skeletal muscle with a remarkable capacity for regeneration. Here, we have used isolated myofibers to investigate the activation and proliferative potential of satellite cells. We have previously shown that satellite cells are heterogeneous: the majority express Myf5 and M-cadherin protein, presumably reflecting commitment to myogenesis, while a minority is negative for both. Although MyoD is rarely detected in quiescent satellite cells, over 98% of satellite cells contain MyoD within 24 h of stimulation. Significantly, MyoD is only observed in cells that are already expressing Myf5. In contrast, a minority population does not activate by the criteria of Myf5 or MyoD expression. Following the synchronous activation of the myogenic regulatory factor+ve satellite cells, their daughter myoblasts proliferate with a doubling time of approximately 17 h, irrespective of the fiber type (type I, IIa, or IIb) from which they originate. Although fast myofibers have fewer associated satellite cells than slow, and accordingly produce fewer myoblasts, each myofiber phenotype is associated with a complement of satellite cells that has sufficient proliferative potential to fully regenerate the parent myofiber within 4 days. This time course is similar to that observed in vivo following acute injury and indicates that cells other than satellite cells are not required for complete myofiber regeneration.


Asunto(s)
Proteínas de Unión al ADN , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Proteína MioD/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/citología , Transactivadores , Animales , División Celular , Linaje de la Célula , Núcleo Celular/metabolismo , Células Cultivadas , Cinética , Ratones , Ratones Mutantes , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteína MioD/genética , Factor 5 Regulador Miogénico , Células Satélite del Músculo Esquelético/metabolismo
13.
J Cell Biol ; 157(4): 693-702, 2002 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-12011114

RESUMEN

Environmental influences have profound yet reversible effects on the behavior of resident cells. Earlier data have indicated that the amount of muscle formed from implanted myogenic cells is greatly augmented by prior irradiation (18 Gy) of the host mouse muscle. Here we confirm this phenomenon, showing that it varies between host mouse strains. However, it is unclear whether it is due to secretion of proliferative factors or reduction of antiproliferative agents. To investigate this further, we have exploited the observation that the immortal myogenic C2 C12 cell line forms tumors far more rapidly in irradiated than in nonirradiated host muscle. We show that the effect of preirradiation on tumor formation is persistent and dose dependent. However, C2 C12 cells are not irreversibly compelled to form undifferentiated tumor cells by the irradiated muscle environment and are still capable of forming large amounts of muscle when reimplanted into a nonirradiated muscle. In a clonal analysis of this effect, we discovered that C2 C12 cells have a bimodal propensity to form tumors; some clones form no tumors even after extensive periods in irradiated graft sites, whereas others rapidly form extensive tumors. This illustrates the subtle interplay between the phenotype of implanted cells and the factors in the muscle environment.


Asunto(s)
Diferenciación Celular/efectos de la radiación , División Celular/efectos de la radiación , Transformación Celular Neoplásica/efectos de la radiación , Supervivencia de Injerto/efectos de la radiación , Músculo Esquelético/efectos de la radiación , Neoplasias Inducidas por Radiación/metabolismo , Regeneración/efectos de la radiación , Trasplante de Células Madre , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Línea Celular Transformada , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Clonales/citología , Células Clonales/metabolismo , Células Clonales/efectos de la radiación , Distrofina/deficiencia , Distrofina/genética , Supervivencia de Injerto/fisiología , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Neoplasias Inducidas por Radiación/patología , Neoplasias Inducidas por Radiación/fisiopatología , Fenotipo , Regeneración/fisiología , Células Madre/citología , Células Madre/metabolismo , Trasplante de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...