Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Physiol ; 215(1): 265-75, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18064665

RESUMEN

Prolonged limb immobilization, which is often the outcome of injury and illness, results in the atrophy of skeletal muscles. The basis of muscle atrophy needs to be better understood in order to allow development of effective countermeasures. The present study focused on determining whether skeletal muscle stem cells, satellite cells, are directly affected by long-term immobilization as well as on investigating the potential of pharmacological and physiological avenues to counterbalance atrophy-induced muscle deterioration. We used external fixation (EF), as a clinically relevant model, to gain insights into the relationships between muscle degenerative and regenerative conditions to the myogenic properties and abundance of bona fide satellite cells. Rats were treated with tetracycline (Tet) through the EF period, or exercise trained on a treadmill for 2 weeks after the cessation of the atrophic stimulus. EF induced muscle mass loss; declined expression of the muscle specific regulatory factors (MRFs) Myf5, MyoD, myogenin, and also of satellite cell numbers and myogenic differentiation aptitude. Tet enhanced the expression of MRFs, but did not prevent the decline of the satellite cell pool. After exercise running, however, muscle mass, satellite cell numbers (enumerated through the entire length of myofibers), and myogenic differentiation aptitude (determined by the lineal identity of clonal cultures of satellite cells) were re-gained to levels prior to EF. Together, our results point to Tet and exercise running as promising and relevant approaches for enhancing muscle recovery after atrophy.


Asunto(s)
Fijación de Fractura/métodos , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Condicionamiento Físico Animal , Células Madre/citología , Tetraciclina/farmacología , Animales , Separación Celular , Células Clonales , Regulación de la Expresión Génica/efectos de los fármacos , Miembro Posterior/diagnóstico por imagen , Miembro Posterior/efectos de los fármacos , Miembro Posterior/patología , Masculino , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/metabolismo , Tamaño de los Órganos , Radiografía , Ratas , Ratas Long-Evans , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Madre/efectos de los fármacos
2.
J Anim Sci ; 86(14 Suppl): E207-16, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17878281

RESUMEN

Satellite cells, the main source of myoblasts in postnatal muscle, are located beneath the myofiber basal lamina. The myogenic potential of satellite cells was initially documented based on their capacity to produce progeny that fused into myotubes. More recently, molecular markers of resident satellite cells were identified, further contributing to defining these cells as myogenic stem cells that produce differentiating progeny and self-renew. Herein, we discuss aspects of the satellite cell transcriptional milieu that have been intensively investigated in our research. We elaborate on the expression patterns of the paired box (Pax) transcription factors Pax3 and Pax7, and on the myogenic regulatory factors myogenic factor 5 (Myf5), myogenic determination factor 1 (MyoD), and myogenin. We also introduce original data on MyoD upregulation in newly activated satellite cells, which precedes the first round of cell proliferation. Such MyoD upregulation occurred even when parent myofibers with their associated satellite cells were exposed to pharmacological inhibitors of hepatocyte growth factor and fibroblast growth factor receptors, which are typically involved in promoting satellite cell proliferation. These observations support the hypothesis that most satellite cells in adult muscle are committed to rapidly entering myogenesis. We also detected expression of serum response factor in resident satellite cells prior to MyoD expression, which may facilitate the rapid upregulation of MyoD. Aspects of satellite cell self-renewal based on the reemergence of cells expressing Pax7, but not MyoD, in myogenic cultures are discussed further herein. We conclude by describing our recent studies using transgenic mice in which satellite cells are traced and isolated based on their expression of green fluorescence protein driven by regulatory elements of the nestin promoter (nestin-green fluorescence protein). This feature provides us with a novel means of studying satellite cell transcriptional signatures, heterogeneity among muscle groups, and the role of the myogenic niche in directing satellite cell self-renewal.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Músculo Esquelético/crecimiento & desarrollo , Células Satélite del Músculo Esquelético/fisiología , Transcripción Genética , Animales , Animales Modificados Genéticamente , Ratones , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Transactivadores , Factores de Transcripción , Regulación hacia Arriba
3.
J Histochem Cytochem ; 49(4): 455-62, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11259448

RESUMEN

Isolated chicken myoblasts had previously been utilized in many studies aiming at understanding the emergence and regulation of the adult myogenic precursors (satellite cells). However, in recent years only a small number of chicken satellite cell studies have been published compared to the increasing number of studies with rodent satellite cells. In large part this is due to the lack of markers for tracing avian myogenic cells before they become terminally differentiated and express muscle-specific structural proteins. We previously demonstrated that myoblasts isolated from fetal and adult chicken muscle display distinct schedules of myosin heavy-chain isoform expression in culture. We further showed that myoblasts isolated from newly hatched and young chickens already possess the adult myoblast phenotype. In this article, we report on the use of polyclonal antibodies against the chicken myogenic regulatory factor proteins MyoD and myogenin for monitoring fetal and adult chicken myoblasts as they progress from proliferation to differentiation in culture. Fetal-type myoblasts were isolated from 11-day-old embryos and adult-type myoblasts were isolated from 3-week-old chickens. We conclude that fetal myoblasts express both MyoD and myogenin within the first day in culture and rapidly transit into the differentiated myosin-expressing state. In contrast, adult myoblasts are essentially negative for MyoD and myogenin by culture Day 1 and subsequently express first MyoD and then myogenin before expressing sarcomeric myosin. The delayed MyoD-to-myogenin transition in adult myoblasts is accompanied by a lag in the fusion into myotubes, compared to fetal myoblasts. We also report on the use of a commercial antibody against the myocyte enhancer factor 2A (MEF2A) to detect terminally differentiated chicken myoblasts by their MEF2+ nuclei. Collectively, the results support the hypothesis that fetal and adult myoblasts represent different phenotypic populations. The fetal myoblasts may already be destined for terminal differentiation at the time of their isolation, and the adult myoblasts may represent progenitors that reside in an earlier compartment of the myogenic lineage.


Asunto(s)
Músculo Esquelético/metabolismo , Proteína MioD/metabolismo , Miogenina/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos , Células Cultivadas , Pollos , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Técnica del Anticuerpo Fluorescente , Factores de Transcripción MEF2 , Desarrollo de Músculos , Músculo Esquelético/crecimiento & desarrollo , Proteína MioD/inmunología , Factores Reguladores Miogénicos , Miogenina/inmunología , Miosinas/inmunología , Miosinas/metabolismo , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
4.
Histochem J ; 32(8): 467-74, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11095071

RESUMEN

Immunoflorescence microscopy of sections of the voodoo lily Sauromatum guttatum appendix stained with monoclonal antibodies against alpha-smooth muscle actin and cytoplasmic actin revealed different staining intensity of different parts of the cell. The anti-cytoplasmic-actin recognized antigens present mainly in the cytoplasm, and the anti-alpha-smooth muscle-actin recognized more intensively antigens present in the nuclei. A positive staining of the nucleus was also obtained with FITC-phalloidin confirming the presence of actin in its filamenous form in the nucleus. The presence of a nuclear alpha-smooth muscle-actin-like protein was further confirmed by confocal laser microscopy. On Western blots, the two anti-actins labelled a protein band that comigrated with standard actin at the approximate molecular weight of 43 kDa. Several other proteins interacted with the two antibodies to a different degree. The monoclonal antibodies against beta-tubulin subunit stained only the periphery of the cytoplasm and anti-pan cytoplasmic myosin stained the cytoplasm weakly. On a Western blot, anti-beta-tubulin subunit primarily recognized a protein band at the appropriate molecular weight of 50 kDa. This is the first cytochemical evidence for the presence of alpha-smooth muscle-actin-like protein in the plant nucleus.


Asunto(s)
Actinas/análisis , Núcleo Celular/ultraestructura , Liliaceae/citología , Núcleo Celular/metabolismo , Técnica del Anticuerpo Fluorescente , Liliaceae/metabolismo
5.
J Histochem Cytochem ; 48(9): 1173-93, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10950875

RESUMEN

Smooth and skeletal muscle tissues are composed of distinct cell types that express related but distinct isoforms of the structural genes used for contraction. These two muscle cell types are also believed to have distinct embryological origins. Nevertheless, the phenomenon of a phenotypic switch from smooth to skeletal muscle has been demonstrated in several in vivo studies. This switch has been minimally analyzed at the cellular level, and the mechanism driving it is unknown. We used immunofluorescence and RT-PCR to demonstrate the expression of the skeletal muscle-specific regulatory genes MyoD and myogenin, and of several skeletal muscle-specific structural genes in cultures of the established rat smooth muscle cell lines PAC1, A10, and A7r5. The skeletal muscle regulatory gene Myf5 was not detected in these three cell lines. We further isolated clonal sublines from PAC1 cultures that homogeneously express smooth muscle characteristics at low density and undergo a coordinated increase in skeletal muscle-specific gene expression at high density. In some of these PAC1 sublines, this process culminates in the high-frequency formation of myotubes. As in the PAC1 parental line, Myf5 was not expressed in the PAC1 sublines. We show that the PAC1 sublines that undergo a more robust transition into the skeletal muscle phenotype also express significantly higher levels of the insulin-like growth factor (IGF1 and IGF2) genes and of FGF receptor 4 (FGFR4) gene. Our results suggest that MyoD expression in itself is not a sufficient condition to promote a coordinated program of skeletal myogenesis in the smooth muscle cells. Insulin administered at a high concentration to PAC1 cell populations with a poor capacity to undergo skeletal muscle differentiation enhances the number of cells displaying the skeletal muscle differentiated phenotype. The findings raise the possibility that the IGF signaling system is involved in the phenotypic switch from smooth to skeletal muscle. The gene expression program described here can now be used to investigate the mechanisms that may underlie the propensity of certain smooth muscle cells to adopt a skeletal muscle identity.(J Histochem Cytochem 48:1173-1193, 2000)


Asunto(s)
Proteínas de Unión al ADN , Proteínas Musculares/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteína MioD/metabolismo , Transactivadores , Animales , Diferenciación Celular , Línea Celular , Expresión Génica , Inmunohistoquímica , Proteínas Musculares/genética , Proteína MioD/genética , Factor 5 Regulador Miogénico , Penetrancia , Fenotipo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
J Histochem Cytochem ; 48(8): 1079-96, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10898801

RESUMEN

Satellite cells are the myogenic precursors in postnatal muscle and are situated beneath the myofiber basement membrane. We previously showed that fibroblast growth factor 2 (FGF2, basic FGF) stimulates a greater number of satellite cells to enter the cell cycle but does not modify the overall schedule of a short proliferative phase and a rapid transition to the differentiated state as the satellite cells undergo myogenesis in isolated myofibers. In this study we investigated whether other members of the FGF family can maintain the proliferative state of the satellite cells in rat myofiber cultures. We show that FGF1, FGF4, and FGF6 (as well as hepatocyte growth factor, HGF) enhance satellite cell proliferation to a similar degree as that seen with FGF2, whereas FGF5 and FGF7 are ineffective. None of the growth factors prolongs the proliferative phase or delays the transition of the satellite cells to the differentiating, myogenin(+) state. However, FGF6 retards the rapid exit of the cells from the myogenin(+) state that routinely occurs in myofiber cultures. To determine which of the above growth factors might be involved in regulating satellite cells in vivo, we examined their mRNA expression patterns in cultured rat myofibers using RT-PCR. The expression of all growth factors, excluding FGF4, was confirmed. Only FGF6 was expressed at a higher level in the isolated myofibers and not in the connective tissue cells surrounding the myofibers or in satellite cells dissociated away from the muscle. By Western blot analysis, we also demonstrated the presence of FGF6 protein in the skeletal musle tissue. Our studies therefore suggest that the myofibers serve as the main source for the muscle FGF6 in vivo. We also used RT-PCR to analyze the expression patterns of the four tyrosine kinase FGF receptors (FGFR1-FGFR4) and of the HGF receptor (c-met) in the myofiber cultures. Depending on the time in culture, expression of all receptors was detected, with FGFR2 and FGFR3 expressed only at a low level. Only FGFR4 was expressed at a higher level in the myofibers but not the connective tissue cell cultures. FGFR4 was also expressed at a higher level in satellite cells compared to the nonmyogenic cells when the two cell populations were released from the muscle tissue and fractionated by Percoll density centrifugation. The unique localization patterns of FGF6 and FGFR4 may reflect specific roles for these members of the FGF signaling complex during myogenesis in adult skeletal muscle.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Expresión Génica , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Diferenciación Celular , División Celular , Células Cultivadas , Células del Tejido Conectivo/metabolismo , Factor 6 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente , Factor de Crecimiento de Hepatocito/farmacología , Masculino , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Neuropathol Exp Neurol ; 59(4): 323-32, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10759188

RESUMEN

Secretion of regulatory peptides by macrophages in injured skeletal muscle constitutes a pivotal determinator of tissue homeostasis. We analyzed expression of a novel Ca2+- binding peptide expressed by activated macrophages, the allograft inflammatory factor-1 (AIF-1), in rat devascularized skeletal muscle. AIF-1 expression was observed in 94% of all macrophages at the site of the injury 48 hours postdevascularization. The physiological function of AIF-1 in injured skeletal muscle was analyzed using a rat in-vitro model of satellite cell proliferation and differentiation. Addition of AIF-1 to the culture medium resulted in a concentration-dependent and reversible reduction of the total number of cells expressing M-cadherin (p < or = 0.0001), a mediator of the differentiation process of skeletal muscle cells, the proliferation associated PCNA (p < or = 0.0001), and the initiator of muscle differentiation myogenin (p < or = 0.0001). These results provide convincing evidence that activated AIF-1 expressing macrophages constitute the predominant cell type in skeletal muscle 48 hours postinjury, and that AIF-1 regulates reduced proliferation, differentiation, and activation of satellite cells.


Asunto(s)
Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Cadherinas/biosíntesis , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/farmacología , Recuento de Células/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inmunohistoquímica , Isquemia/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/citología , Masculino , Proteínas de Microfilamentos , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Miogenina/biosíntesis , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología
8.
J Histochem Cytochem ; 47(11): 1375-84, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10544211

RESUMEN

Satellite cells (SCs) in normal adult muscle are quiescent. They can enter the mitotic program when stimulated with growth factors such as basic FGF. Short-term denervation stimulates SC to enter the mitotic cycle in vivo, whereas long-term denervation depletes the SC pool. The molecular basis for the neural influence on SCs has not been established. We studied the phenotype and the proliferative capacity of SCs from muscle that had been denervated before being cultured in vitro. The expression of PCNA, myogenin, and muscle (M)-cadherin in SCs of normal and denervated muscle fibers was examined at the single-cell level by immunolabeling in a culture system of isolated rat muscle fibers with attached SCs. Immediately after plating (Day 0), neither PCNA nor myogenin was present on normal muscle fibers, but we detected an average of 0.5 M-cadherin(+) SCs per muscle fiber. The number of these M-cadherin(+) cells (which are negative for PCNA and myogenin) increased over the time course examined. A larger fraction of cells negative for M-cadherin underwent mitosis and expressed PCNA, followed by myogenin. The kinetics of SCs from muscle fibers denervated for 4 days before culturing were similar to those of normal controls. Denervation from 1 to 32 weeks before plating, however, suppressed PCNA and myogenin expression almost completely. The fraction of M-cadherin(+) (PCNA(-)/myogenin(-)) SCs was decreased after 1 week of denervation, increased above normal after denervation for 4 or 8 weeks, and decreased again after denervation for 16 or 32 weeks. We suggest that the M-cadherin(+) cells are nondividing SCs because they co-express neither PCNA or myogenin, whereas the cells positive for PCNA or myogenin (and negative for M-cadherin) have entered the mitotic cycle. SCs from denervated muscle were different from normal controls when denervated for 1 week or longer. The effect of denervation on the phenotypic modulation of SCs includes resistance to recruitment into the mitotic cycle under the conditions studied here and a robust extension of the nonproliferative compartment. These characteristics of SCs deprived of neural influence may account for the failure of denervated muscle to fully regenerate. (J Histochem Cytochem 47:1375-1383, 1999)


Asunto(s)
Desnervación Muscular , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Músculo Esquelético/inervación , Animales , Biomarcadores , Cadherinas/análisis , Ciclo Celular , División Celular , Células Cultivadas , Inmunohistoquímica , Masculino , Mitosis , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Miogenina/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , Ratas , Ratas Wistar , Valores de Referencia , Nervio Ciático/fisiología , Factores de Tiempo
9.
Dev Biol ; 210(2): 440-55, 1999 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10357902

RESUMEN

Satellite cells from adult rat muscle coexpress proliferating cell nuclear antigen and MyoD upon entry into the cell cycle, suggesting that MyoD plays a role during the recruitment of satellite cells. Moreover, the finding that muscle regeneration is compromised in MyoD-/- mice, has provided evidence for the role of MyoD during myogenesis in adult muscle. In order to gain further insight into the role of MyoD during myogenesis in the adult, we compared satellite cells from MyoD-/- and wildtype mice as they progress through myogenesis in single-myofiber cultures and in tissue-dissociated cell cultures (primary cultures). Satellite cells undergoing proliferation and differentiation were traced immunohistochemically using antibodies against various regulatory proteins. In addition, an antibody against the mitogen-activated protein kinases ERK1 and ERK2 was used to localize the cytoplasm of the fiber-associated satellite cells regardless of their ability to express specific myogenic regulatory factor proteins. We show that during the initial days in culture the myofibers isolated from both the MyoD-/- and the wildtype mice contain the same number of proliferating, ERK+ satellite cells. However, the MyoD-/- satellite cells continue to proliferate and only a very small number of cells transit into the myogenin+ state, whereas the wildtype cells exit the proliferative compartment and enter the myogenin+ stage. Analyzing tissue-dissociated cultures of MyoD-/- satellite cells, we identified numerous cells whose nuclei were positive for the Myf5 protein. In contrast, quantification of Myf5+ cells in the wildtype cultures was difficult due to the low level of Myf5 protein present. The Myf5+ cells in the MyoD-/- cultures were often positive for desmin, similar to the MyoD+ cells in the wildtype cultures. Myogenin+ cells were identified in the MyoD-/- primary cultures, but their appearance was delayed compared to the wildtype cells. These "delayed" myogenin+ cells can express other differentiation markers such as MEF2A and cyclin D3 and fuse into myotubes. Taken together, our studies suggest that the presence of MyoD is critical for the normal progression of satellite cells into the myogenin+, differentiative state. It is further proposed that the Myf5+/MyoD- phenotype may represent the myogenic stem cell compartment which is capable of maintaining the myogenic precursor pool in the adult muscle.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos , Músculo Esquelético/citología , Proteína MioD/fisiología , Transactivadores , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/análisis , Ciclo Celular , Diferenciación Celular , División Celular , Células Cultivadas , Ciclina D3 , Ciclinas/análisis , Proteínas de Unión al ADN/análisis , Desmina/análisis , Diafragma/citología , Factores de Transcripción MEF2 , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/análisis , Músculo Esquelético/fisiología , Proteína MioD/genética , Factor 5 Regulador Miogénico , Factores Reguladores Miogénicos , Miogenina/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , Ratas , Factores de Transcripción/análisis
10.
J Histochem Cytochem ; 47(1): 23-42, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9857210

RESUMEN

Although the role of satellite cells in muscle growth and repair is well recognized, understanding of the molecular events that accompany their activation and proliferation is limited. In this study, we used the single myofiber culture model for comparing the proliferative dynamics of satellite cells from growing (3-week-old), young adult (8- to 10-week-old), and old (9- to 11-month-old) rats. In these fiber cultures, the satellite cells are maintained in their in situ position underneath the fiber basement membrane. We first demonstrate that the cytoplasm of fiber-associated satellite cells can be monitored with an antibody against the extracellular signal regulated kinases 1 and 2 (ERK1 and ERK2), which belong to the mitogen-activated protein kinase (MAPK) superfamily. With this immunocytological marker, we show that the satellite cells from all three age groups first proliferate and express PCNA and MyoD, and subsequently, about 24 hr later, exit the PCNA+/MyoD+ state and become positive for myogenin. For all three age groups, fibroblast growth factor 2 (FGF2) enhances by about twofold the number of satellite cells that are capable of proliferation, as determined by monitoring the number of cells that transit from the MAPK+ phenotype to the PCNA+/MAPK+ or MyoD+/MAPK+ phenotype. Furthermore, contrary to the commonly accepted convention, we show that in the fiber cultures FGF2 does not suppress the subsequent transition of the proliferating cells into the myogenin+ compartment. Although myogenesis of satellite cells from growing, young adult, and old rats follows a similar program, two distinctive features were identified for satellite cells in fiber cultures from the old rats. First, a large number of MAPK+ cells do not appear to enter the MyoD-myogenin expression program. Second, the maximal number of proliferating satellite cells is attained a day later than in cultures from the young adults. This apparent "lag" in proliferation was not affected by hepatocyte growth factor (HGF), which has been implicated in accelerating the first round of satellite cell proliferation. HGF and FGF2 were equally efficient in promoting proliferation of satellite cells in fibers from old rats. Collectively, the investigation suggests that FGF plays a critical role in the recruitment of satellite cells into proliferation.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Músculo Esquelético/citología , Envejecimiento , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Citarabina/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Immunoblotting , Inmunohistoquímica , Masculino , Desarrollo de Músculos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Proteína MioD/metabolismo , Miogenina/metabolismo , Miosinas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Isoformas de Proteínas , Ratas , Ratas Sprague-Dawley
11.
Anat Embryol (Berl) ; 197(6): 421-37, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9682974

RESUMEN

Whereas the understanding of the mechanisms underlying skeletal and cardiac muscle development has been increased dramatically in recent years, the understanding of smooth muscle development is still in its infancy. This paper summarizes studies on the ontogeny of chicken smooth muscle cells in the wall of the aorta and aortic arch-derived arteries. Employing immunocytochemistry with antibodies against smooth muscle contractile and extracellular matrix proteins we trace smooth muscle cell patterning from early development throughout adulthood. Comparing late stage embryos to young and adult chickens we demonstrate, for all the stages analyzed, that the cells in the media of aortic arch-derived arteries and of the thoracic aorta are organized in alternating lamellae. The lamellar cells, but not the interlamellar cells, express smooth muscle specific contractile proteins and are surrounded by basement membrane proteins. This smooth muscle cell organization of lamellar and interlamellar cells is fully acquired by embryonic day 11 (ED 11). We further show that, during earlier stages of embryogenesis (ED3 through ED7), cells expressing smooth muscle proteins appear only in the peri-endothelial region of the aortic and aortic arch wall and are organized as a narrow band of cells that does not demonstrate the lamellar-interlamellar pattern. On ED9, infrequent cells organized in lamellar-interlamellar organization can be detected and their frequency increases by ED10. In addition to changes in cell organization, we show that there is a characteristic sequence of contractile and extracellular matrix protein expression during development of the aortic wall. At ED3 the peri-endothelial band of differentiated smooth muscle cells is already positive for smooth muscle alpha actin (alphaSM-actin) and fibronectin. By the next embryonic day the peri-endothelial cell layer is also positive for smooth muscle myosin light chain kinase (SM-MLCK). Subsequently, by ED5 this peri-endothelial band of differentiated smooth muscle cells is positive for alphaSM-actin, SM-MLCK, SM-calponin, fibronectin, and collagen type IV. However, laminin and desmin (characteristic basement membrane and contractile proteins of smooth muscle) are first seen only at the onset of the lamellar-interlamellar cell organization (ED9 to ED10). We conclude that the development of chicken aortic smooth muscle involves transitions in cell organization and in expression of smooth muscle proteins until the adult-like phenotype is achieved by mid-embryogenesis. This detailed analysis of the ontogeny of chick aortic smooth muscle should provide a sound basis for future studies on the regulatory mechanisms underlying vascular smooth muscle development.


Asunto(s)
Aorta Torácica/crecimiento & desarrollo , Diferenciación Celular , Proteínas Contráctiles/análisis , Proteínas de la Matriz Extracelular/análisis , Desarrollo de Músculos , Músculo Liso Vascular/crecimiento & desarrollo , Factores de Edad , Animales , Aorta Torácica/citología , Aorta Torácica/metabolismo , Membrana Basal/crecimiento & desarrollo , Embrión de Pollo , Pollos/crecimiento & desarrollo , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Músculo Liso Vascular/anatomía & histología , Músculo Liso Vascular/embriología , Músculo Liso Vascular/metabolismo , Conejos
12.
J Histochem Cytochem ; 46(1): 59-67, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9407021

RESUMEN

Thyroid hormone (T3) affects muscle development and muscle regeneration. It also interacts with the muscle regulatory gene MyoD in culture and affects myoblast proliferation. We studied the localization of MyoD protein using a well-characterized polyclonal antibody for immunohistochemistry. Relative numbers of myogenic precursor cells per field were identified by their MyoD expression during muscle regeneration in normal and mdx dystrophic mice, with particular reference to the expression in mononuclear cells and myotubes at various T3 levels. In regeneration by normal muscles, relatively few MyoD+ nuclei per field were present in mononuclear cells of euthyroid and hypothyroid mice. MyoD staining of mononuclear cell nuclei was approximately doubled in fields of regenerating muscles of normal hyperthyroid compared to euthyroid mice, and was observed in precursors that appeared to be aligned before fusion into myotubes. In mdx regenerating muscle, twofold more mononuclear cells positive for MyoD were present in all three treatment groups compared to normal muscles regenerating under the same conditions. Localization was similar to the pattern in normal euthyroid mice. However, in muscles regenerating in hyperthyroid mdx mice, both mononuclear cell nuclei and centrally located nuclei in a subpopulation (about 15%) of new myotubes formed after the crush injury were intensely stained for MyoD protein. The changes observed are consistent with reports on T3-induced alteration of muscle repair, and propose a link between MyoD regulation and the accelerated differentiation during regeneration under high T3 conditions. (J Histochem Cytochem 46:59-67, 1998)


Asunto(s)
Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Proteína MioD/biosíntesis , Triyodotironina/metabolismo , Cicatrización de Heridas/fisiología , Heridas no Penetrantes/metabolismo , Animales , Hipotiroidismo/inducido químicamente , Hipotiroidismo/tratamiento farmacológico , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Microscopía Fluorescente , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Miofibrillas/efectos de los fármacos , Miofibrillas/metabolismo , Propiltiouracilo , Especificidad de la Especie , Triyodotironina/farmacología , Cicatrización de Heridas/efectos de los fármacos , Heridas no Penetrantes/patología
13.
Growth Factors ; 15(1): 1-27, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9401815

RESUMEN

We have previously demonstrated that PDGF-BB enhances proliferation of C2 myoblasts. This has led us to examine whether the mitogenic influence of PDGF-BB in the C2 model correlates with modulation of specific steps associated with myogenic differentiation. C2 myoblasts transiting through these differentiation specific steps were monitored via immunocytochemistry. We show that the influence of PDGF on enhancing cell proliferation correlates with a delay in the emergence of cells positive for sarcomeric myosin. We further monitored the influence of PDGF-BB on differentiation steps preceding the emergence of myosin+ cells. We demonstrate that mononucleated C2 cells first express MyoD (MyoD+/myogenin- cells) and subsequently, myogenin. Cells negative for both MyoD and myogenin (the phenotype preceding the MyoD+ state) were present at all times in culture and comprised the majority, if not all, of the cells which responded mitogenically to PDGF. Additionally, the frequency of the MyoD+/myogenin+ cell phenotype was reduced in cultures receiving PDGF, suggesting that PDGF can modulate the transition of the cells into the myogenin+ state. We determined that many of the myogenin+ cells subsequently become MEF2A+ and this phenomenon is not influenced by PDGF-BB. FGF-2 also enhanced the proliferation of C2 myoblasts and suppressed the appearance of the myogenin+ cells, but did not influence the subsequent transition into the MEF2A+ state. The study raises the possibility that PDGF-BB and FGF-2 might delay the transition of the C2 cells into the MyoD+/myogenin+ state by depressing a paracrine signal that enhances differentiation.


Asunto(s)
Proteínas de Unión al ADN/análisis , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Proteína MioD/análisis , Miogenina/análisis , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factores de Transcripción/análisis , Animales , Becaplermina , Bromodesoxiuridina/metabolismo , Recuento de Células/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Células Clonales/citología , Células Clonales/metabolismo , Ciclina A/análisis , Desmina/análisis , Factor 2 de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Factores de Transcripción MEF2 , Ratones , Músculo Esquelético/química , Factores Reguladores Miogénicos , Miosinas/análisis , Fenotipo , Proteínas Proto-Oncogénicas c-sis
14.
Microsc Res Tech ; 30(5): 366-80, 1995 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-7787236

RESUMEN

The myogenic precursor cells of postnatal and adult skeletal muscle are situated underneath the basement membrane of the myofibers. It is because of their unique positions that these precursor cells are often referred to as satellite cells. Such defined satellite cells can first be detected following the formation of a distinct basement membrane around the fiber, which takes place in late stages of embryogenesis. Like myoblasts found during development, satellite cells can proliferate, differentiate, and fuse into myofibers. However, in the normal, uninjured adult muscle, satellite cells are mitotically quiescent. In recent years several important questions concerning the biology of satellite cells have been asked. One aspect has been the relationship between satellite cells and myoblasts found in the developing muscle: are these myogenic populations identical or different? Another aspect has been the physiological cues that control the quiescent, proliferative, and differentiative states of these myogenic precursors: what are the growth regulators and how do they function? These issues are discussed, referring to previous work by others and further emphasizing our own studies on avian and rodent satellite cells. Collectively, the studies presented indicate that satellite cells represent a distinct myogenic population that becomes dominant in late stages of embryogenesis. Moreover, although satellite cells are already destined to be myogenic precursors, they do not express any of the four known myogenic regulatory genes unless their activation is induced in the animal or in culture. Furthermore, multiple growth factors are important regulators of satellite cell proliferation and differentiation. Our work on the role of one of these growth factors [platelet-derived growth factor (PDGF)] during proliferation of adult myoblasts is further discussed with greater detail and the possibility that PDGF is involved in the transition from fetal to adult myoblasts in late embryogenesis is brought forward.


Asunto(s)
Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Animales , Diferenciación Celular , División Celular , Pollos , Sustancias de Crecimiento/fisiología , Morfogénesis , Proteínas Musculares/fisiología , Músculo Esquelético/ultraestructura , Factores Reguladores Miogénicos/fisiología , Ratas
15.
Cell Mol Biol Res ; 41(4): 241-9, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-8775982

RESUMEN

We describe our studies on the characterization of the cell phenotypes in the wall of the aortic-arch-derived arteries from "late" chicken embryos. Using immunocytochemistry with antibodies against smooth muscle cytoskeletal and basement membrane proteins, we show that the smooth muscle of the aortic-arch-derived arteries from 13- to 19-d-old embryos contains two cell phenotypes organized in interchanging lamellae. One cell phenotype (lamellar cells), but not the other cell phenotype (interlamellar cells), expresses the cytoskeletal proteins desmin and alpha smooth muscle actin (alpha SMactin). Both cell phenotypes express the cytoskeletal protein vimentin. Furthermore, the lamellar cells but not the interlamellar cells are surrounded by the basement membrane proteins laminin and collagen type IV. Performing quail-chick transplantation experiments and using a quail specific antibody, we show that both lamellar and interlamellar cells in the "aortic arches" of a 15-d-old chimeric embryo are derived from neural crest cells. We conclude that the aortic smooth muscle cells from "late" chicken embryos consist of two distinct cell phenotypes which are derived from a common lineage.


Asunto(s)
Aorta/embriología , Membrana Basal/metabolismo , Proteínas del Citoesqueleto/metabolismo , Músculo Liso Vascular/embriología , Animales , Aorta Torácica/citología , Diferenciación Celular , Linaje de la Célula , Embrión de Pollo , Quimera , Colágeno/metabolismo , Inmunohistoquímica , Laminina/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Cresta Neural/citología , Codorniz , Vimentina/metabolismo
16.
J Cell Physiol ; 160(3): 563-72, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8077294

RESUMEN

Extracts from gently crushed adult mouse skeletal muscles (CMEs) contain potent myoblast mitogens, and may be used as a model system to investigate myotrophic factors released by adult muscles following injury. CME was separated into four peaks of mitogenic activity by heparin affinity chromatography. The fraction of CME that did not bind to heparin contained transferrin (Tf). Three peaks of mitogenic activity were eluted from the heparin-agarose columns at NaCl concentrations of 0.4 M, 0.9 M, and 2.0 M. A 46 kDa protein that shared antigenicity with the BB isoform of platelet-derived growth factor (PDGF-BB) was present in the 0.4 M NaCl eluant. Mitogenic activity in the 2.0 M NaCl peak eluted identically to purified basic fibroblast growth factor (bFGF), did not act additively to saturating amounts of purified bFGF, and was neutralized by anti-bFGF antibodies. The 0.9 M NaCl eluant acted additively to the combination of three known growth factors for myoblasts, bFGF, insulin-like growth factor 1, and epidermal growth factor, to stimulate C2 myoblast proliferation, suggesting this fraction contains a mitogenic activity which does not utilize (and hence compete for) receptors for the known mitogens for myoblasts. Additionally, the 0.9 M NaCl eluant did not stimulate proliferation of fibroblast-like cells derived from muscle tissue. The unbound, 0.4 M NaCl, 0.9 M NaCl, and 2.0 M NaCl eluants from the heparin-agarose column acted additively to one another to stimulate myoblast proliferation. Our data suggest that Tf, PDGF-BB-like molecules, bFGF-like activity, and an uncharacterized heparin-binding myoblast mitogen could be released after muscle injury and act to stimulate satellite cell proliferation.


Asunto(s)
Cromatografía de Afinidad/métodos , Heparina , Mitógenos/fisiología , Músculos/lesiones , Músculos/metabolismo , Extractos de Tejidos/fisiología , Animales , Antígenos/inmunología , Becaplermina , Cromatografía en Agarosa , Factor 2 de Crecimiento de Fibroblastos/fisiología , Heparina/metabolismo , Ratones , Proteínas Musculares/fisiología , Concentración Osmolar , Péptidos/fisiología , Factor de Crecimiento Derivado de Plaquetas/inmunología , Proteínas Proto-Oncogénicas c-sis , Cloruro de Sodio/farmacología , Extractos de Tejidos/inmunología , Extractos de Tejidos/metabolismo , Transferrina/metabolismo
17.
Dev Biol ; 164(2): 588-603, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7913900

RESUMEN

Myogenic precursors in adult skeletal muscle (satellite cells) are mitotically quiescent but can proliferate in response to a variety of stresses including muscle injury. To gain further understanding of adult myoblasts, we analyzed myogenesis of satellite cells on intact fibers isolated from adult rat muscle. In this culture model, satellite cells are maintained in their in situ position underneath the fiber basement membrane. In the present study patterns of satellite cell proliferation, expression of myogenic regulatory factor proteins, and expression of differentiation-specific, cytoskeletal proteins were determined, via immunohistochemistry of cultured fibers. The temporal appearance and the numbers of cells positive for proliferating cell nuclear antigen (PCNA) or for MyoD were similar, suggesting that MyoD is present in detectable amounts in proliferating but not quiescent satellite cells. Satellite cells positive for myogenin, alpha-smooth muscle actin (alpha SMactin), or developmental sarcomeric myosin (DEVmyosin) appeared following the decline in PCNA and MyoD expression. However, expression of myogenin and alpha SMactin was transient, while DEV-myosin expression was continuously maintained. Moreover, the number of DEVmyosin + cells was only half of the number of myogenin + or alpha SMactin + cells--indicating, perhaps, that only 50% of the satellite cell descendants entered the phase of terminal differentiation. We further determined that the number of proliferating satellite cells can be modulated by basic FGF but the overall schedule of cell cycle entry, proliferation, differentiation, and temporal expression of regulatory and structural proteins was unaffected. We thus conclude that satellite cells conform to a highly coordinated program when undergoing myogenesis at their native position along the muscle fiber.


Asunto(s)
Proteínas Musculares/biosíntesis , Músculos/metabolismo , Animales , Diferenciación Celular , División Celular , Medios de Cultivo , Técnicas de Cultivo , Factor 2 de Crecimiento de Fibroblastos/fisiología , Inmunohistoquímica , Masculino , Músculos/citología , Proteína MioD/metabolismo , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación , Ratas , Ratas Sprague-Dawley
18.
Dev Biol ; 156(2): 307-18, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8462733

RESUMEN

In the present study we measured the level of PDGF receptor expression by chicken myoblasts and the effect of the three different PDGF isoforms (AA, AB, BB) on DNA synthesis by myoblasts. We examined PDGF receptor expression and function on clonally derived myoblasts in order to eliminate contaminating fibroblasts which are present in myogenic cultures and which bind PDGF. Furthermore, since we have previously shown that fetal myoblasts are replaced with adult myoblasts during late chicken embryogenesis, we compared PDGF receptor expression and function on myoblasts from Embryonic Day 10 (E10, mid development) and from Embryonic Day 19 (E19, late development). We found that all myogenic clones from late embryos (E19) express many receptors for PDGF-BB, far fewer receptors for PDGF-AB, and even fewer, if any, receptors for PDGF-AA. Myoblast clones derived from E10 were more heterogeneous in their PDGF binding pattern ranging from clones similar to E19 clones to clones having very few PDGF binding sites. We also found that both PDGF-AB and PDGF-BB can promote DNA synthesis by clonally derived chicken myoblasts maintained in 2.5% fetal bovine serum whereas PDGF-AA has no detectable effect. Finally, we observed that primary myogenic cultures from E10 and E19 differ strikingly in levels of PDGF binding; E19 cultures bind much more PDGF than do E10 cultures. We conclude that PDGF can enhance the proliferation of chicken myoblasts and that myoblasts responsive to PDGF are more frequent in late than in mid stages of development. We propose that PDGF may be a modulator of myogenesis of adult but not fetal myoblasts.


Asunto(s)
Músculos/embriología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Embrión de Pollo , Edad Gestacional , Músculos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/química
19.
Prog Clin Biol Res ; 383B: 575-85, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8115374

RESUMEN

In here we summarize some of our recent studies on adult chicken myoblasts. We discuss a) myosin isoform expression by the differentiated myoblasts, and b) the role of PDGF during proliferation of the cells. These studies demonstrate that adult myoblasts represent a unique myogenic population (or lineage) which emerges in late development. Furthermore, we discuss possible lineal relationship between adult and somitic/limb bud cells. Unless otherwise noted, the source of myoblasts was chicken pectoralis muscle, and all procedures and antibodies were as described in previous publications (Yablonka-Reuveni, et al., 1987; Yablonka-Reuveni and Seifert, submitted; Hartley, et al., 1991; 1992).


Asunto(s)
Músculos/citología , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Pollos , Extremidades/embriología , Desarrollo de Músculos , Músculos/embriología , Miosinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Codorniz/embriología , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...