Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Coral Reefs ; 41(4): 1147-1159, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37334145

RESUMEN

Reported divergent responses of coral growth and skeletal microstructure to the nutrient environment complicate knowledge-based management of water quality in coral reefs. By re-evaluating published results considering the taxonomy of the studied corals and the N:P stoichiometry of their nutrient environment, we could resolve some of the major apparent contradictions. Our analysis suggests that Acroporids behave differently to several other common genera and show distinct responses to specific nutrient treatments. We hypothesised that both the concentrations of dissolved inorganic N and P in the water and their stoichiometry shape skeletal growth and microstructure. We tested this hypothesis by exposing Acropora polystoma fragments to four nutrient treatments for > 10 weeks: high nitrate/high phosphate (HNHP), high nitrate/low phosphate (HNLP), low nitrate/high phosphate (LNHP) and low nitrate/low phosphate (LNLP). HNHP corals retained high zooxanthellae densities and their linear extension and calcification rates were up to ten times higher than in the other treatments. HNLP and LNLP corals bleached through loss of symbionts. The photochemical efficiency (Fv/Fm) of residual symbionts in HNLP corals was significantly reduced, indicating P-starvation. Micro-computed tomography (µCT) of the skeletal microstructure revealed that reduced linear extension in nutrient limited or nutrient starved conditions (HNLP, LNHP, LNLP) was associated with significant thickening of skeletal elements and reduced porosity. These changes can be explained by the strongly reduced linear extension rate in combination with a smaller reduction in the calcification rate. Studies using increased skeletal density as a proxy for past thermal bleaching events should consider that such an increase in density may also be associated with temperature-independent response to the nutrient environment. Furthermore, the taxonomy of corals and seawater N:P stoichiometry should be considered when analysing and managing the impacts of nutrient pollution. Supplementary Information: The online version contains supplementary material available at 10.1007/s00338-022-02223-0.

2.
Dev Cell ; 1(3): 435-40, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11702954

RESUMEN

Development of the arterial pole of the heart is a critical step in cardiogenesis, yet its embryological origin remains obscure. We have analyzed a transgenic mouse line in which beta-galactosidase activity is observed in the embryonic right ventricle and outflow tract of the heart and in contiguous splanchnic and pharyngeal mesoderm. The nlacZ transgene has integrated upstream of the fibroblast growth factor 10 (Fgf10) gene and comparison with the expression pattern of Fgf10 in pharyngeal mesoderm indicates transgene control by Fgf10 regulatory sequences. Dil labeling shows a progressive movement of cells from the pharyngeal arch region into the growing heart tube between embryonic days 8.25 and 10.5. These data suggest that arterial pole myocardium originates outside the classical heart field.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Corazón/embriología , Mesodermo/metabolismo , Faringe/embriología , Animales , Carbocianinas/metabolismo , Factor 10 de Crecimiento de Fibroblastos , Colorantes Fluorescentes/metabolismo , Corazón/fisiología , Hibridación in Situ , Mesodermo/citología , Ratones , Ratones Transgénicos , Miocardio/citología , Miocardio/metabolismo , Faringe/metabolismo , Transgenes , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
3.
J Cell Biol ; 129(2): 383-96, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7721942

RESUMEN

The myosin light chain IF/3F locus contains two independent promoters, MLC1F and MLC3F, which are differentially activated during skeletal muscle development. Transcription at this locus is regulated by a 3' skeletal muscle enhancer element, which directs correct temporal and tissue-specific expression from the MLC1F promoter in transgenic mice. To investigate the role of this enhancer in regulation of the MLC3F promoter in vivo, we have analyzed reporter gene expression in transgenic mice containing lacZ under transcriptional control of the mouse MLC3F promoter and 3' enhancer element. Our results show that these regulatory elements direct strong expression of lacZ in skeletal muscle; the transgene, however, is activated 4-5 d before the endogenous MLC3F promoter, at the time of initiation of MLC1F transcription. In adult mice, transgene activity is downregulated in muscles that have reduced contributions of type IIB fibers (soleus and diaphragm). The rostrocaudal positional gradient of transgene expression documented for MLC1F transgenic mice (Donoghue, M., J. P. Merlie, N. Rosenthal, and J. R. Sanes. 1991. Proc. Natl. Acad. Sci. USA. 88:5847-5851) is not seen in MLC3F transgenic mice. Although MLC3F was previously thought to be restricted to skeletal striated muscle, the MLC3F-lacZ transgene is expressed in cardiac muscle from 7.5 d of development in a spatially restricted manner in the atria and left ventricular compartments, suggesting that transcriptional differences exist between cardiomyocytes in left and right compartments of the heart. We show here that transgene-directed expression of the MLC3F promoter reflects low level expression of endogenous MLC3F transcripts in the mouse heart.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Corazón/fisiología , Músculo Esquelético/fisiología , Miosinas/genética , Regiones Promotoras Genéticas/genética , Animales , Secuencia de Bases , Regulación hacia Abajo , Desarrollo Embrionario y Fetal , Femenino , Corazón Fetal/fisiología , Genes Reporteros/genética , Corazón/embriología , Corazón/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Desarrollo de Músculos , Músculo Esquelético/embriología , Músculo Esquelético/crecimiento & desarrollo , Miocardio/citología , ARN Mensajero/análisis , Activación Transcripcional , beta-Galactosidasa/genética
4.
J Cell Biol ; 119(4): 811-21, 1992 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1429837

RESUMEN

The spatial and temporal expression of the dystrophin gene has been examined during mouse embryogenesis, using in situ hybridization on tissue sections with a probe from the 5' end of the dystrophin coding sequence. In striated muscle, dystrophin transcripts are detectable from about 9 d in the heart and slightly later in skeletal muscle. However, there is an important difference between the two types of muscle: the heart is already functional as a contractile organ before the appearance of dystrophin transcripts, whereas this is not the case in skeletal muscle, where dystrophin and myosin heavy chain transcripts are first detectable at the same time. In the heart, dystrophin transcripts accumulate initially in the outflow tract and, at later stages, in both the atria and ventricles. In skeletal muscle, the gene is expressed in all myocytes irrespective of fiber type. In smooth muscle dystrophin transcripts are first detectable from 11 d post coitum in blood vessels, and subsequently in lung bronchi and in the digestive tract. The other major tissue where the dystrophin gene is expressed is the brain, where transcripts are clearly detectable in the cerebellum from 13 d. High-level expression of the gene is also seen in particular regions of the forebrain involved in the regulation of circadian rhythms, the endocrine system, and olfactory function, not previously identified in this context. The findings are discussed in the context of the pathology of Duchenne muscular dystrophy.


Asunto(s)
Encéfalo/embriología , Distrofina/genética , Corazón Fetal/metabolismo , Expresión Génica , Músculos/embriología , Animales , Encéfalo/metabolismo , Distrofina/biosíntesis , Desarrollo Embrionario y Fetal , Hibridación in Situ , Ratones , Músculo Liso/embriología , Músculo Liso/metabolismo , Músculos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
J Cell Biol ; 123(4): 823-35, 1993 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8227143

RESUMEN

We have previously reported the identification of a distinct myosin heavy chain (MyHC) isoform in a major subpopulation of rat skeletal muscle fibers, referred to as 2X fibers (Schiaffino, S., L. Gorza, S. Sartore, L. Saggin, M. Vianello, K. Gundersen, and T. Lømo. 1989. J. Muscle Res. Cell Motil. 10:197-205). However, it was not known whether 2X-MyHC is the product of posttranslational modification of other MyHCs or is coded by a distinct mRNA. We report here the isolation and characterization of cDNAs coding a MyHC isoform that is expressed in type 2X skeletal muscle fibers. 2X-MyHC transcripts differ from other MyHC transcripts in their restriction map and 3' end sequence and are thus derived from a distinct gene. In situ hybridization analyses show that 2X-MyHC transcripts are expressed at high levels in the diaphragm and fast hindlimb muscles and can be coexpressed either with 2B- or 2A-MyHC transcripts in a number of fibers. At the single fiber level the distribution of each MyHC mRNA closely matches that of the corresponding protein, determined by specific antibodies on serial sections. In hindlimb muscles 2X-, 2A-, and 2B-MyHC transcripts are first detected by postnatal day 2-5 and display from the earliest stages a distinct pattern of distribution in different muscles and different fibers. The emergence of type 2 MyHC isoforms thus defines a distinct neonatal phase of fiber type differentiation during muscle development. The functional significance of MyHC isoforms is discussed with particular reference to the velocity of shortening of skeletal muscle fibers.


Asunto(s)
Regulación de la Expresión Génica , Músculos/metabolismo , Miosinas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Diferenciación Celular , ADN , Estimulación Eléctrica , Hibridación in Situ , Datos de Secuencia Molecular , Desarrollo de Músculos , Miosinas/metabolismo , Ratas , Mapeo Restrictivo , Hormonas Tiroideas/fisiología , Transcripción Genética
6.
J Cell Biol ; 113(6): 1255-65, 1991 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2045411

RESUMEN

The spatial and temporal expression pattern of the muscle regulatory gene Myf-6 (MRF4/herculin) has been investigated by in situ hybridization during embryonic and fetal mouse development. Here, we report that the Myf-6 gene shows a biphasic pattern of expression. Myf-6 transcripts are first detected in the most rostral somites of the mouse embryo at 9 d of gestation and accumulate progressively in myotomal cells along the rostro-caudal axis. This expression is transient and Myf-6 mRNA can no longer be detected in myotomal cells after day 12 post coitum (p.c.). In contrast to other muscle determination genes (MyoD1, myogenin, Myf-5), Myf-6 mRNA is not detected in limb buds or visceral arches and skeletal muscle of the mouse embryo (day 8-15 p.c.). In fetal mice, Myf-6 transcripts appear at day 16 p.c. in all skeletal muscles, and the gene continues to be expressed at a high level after birth. These results suggest that early Myf-6 expression may be restricted to a population of myogenic cells that does not contribute to the embryonic muscle masses in limb buds and visceral arches. The reappearance of Myf-6 mRNA in fetal skeletal muscle coincides approximately with secondary muscle fiber formation and the onset of innervation.


Asunto(s)
Genes Reguladores , Proteínas Musculares/genética , Músculos/metabolismo , Factores Reguladores Miogénicos , Secuencia de Bases , Northern Blotting , ADN , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Desarrollo de Músculos , Músculos/embriología , Miogenina , Hibridación de Ácido Nucleico
7.
J Cell Biol ; 111(4): 1465-76, 1990 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2211821

RESUMEN

Using in situ hybridization, we have investigated the temporal sequence of myosin gene expression in the developing skeletal muscle masses of mouse embryos. The probes used were isoform-specific, 35S-labeled antisense cRNAs to the known sarcomeric myosin heavy chain and myosin alkali light chain gene transcripts. Results showed that both cardiac and skeletal myosin heavy chain and myosin light chain mRNAs were first detected between 9 and 10 d post coitum (p.c.) in the myotomes of the most rostral somites. Myosin transcripts appeared in more caudal somites at later stages in a developmental gradient. The earliest myosin heavy chain transcripts detected code for the embryonic skeletal (MHCemb) and beta-cardiac (MHC beta) isoforms. Perinatal myosin heavy chain (MHCpn) transcripts begin to accumulate at 10.5 d p.c., which is much earlier than previously reported. At this stage, MHCemb is the major MHC transcript. By 12.5 d p.c., MHCpn and MHCemb mRNAs are present to an equal extent, and by 15.5 d p.c. the MHCpn transcript is the major MHC mRNA detected. Cardiac MHC beta transcripts are always present as a minor component. In contrast, the cardiac MLC1A mRNA is initially more abundant than that encoding the skeletal MLC1F isoform. By 12.5 d p.c. the two MLC mRNAs are present at similar levels, and by 15.5 d p.c., MLC1F is the predominant MLC transcript detected. Transcripts for the ventricular/slow (MLC1V) and another fast skeletal myosin light chain (MLC3F) are not detected in skeletal muscle before 15 d p.c., which marks the beginning of the fetal stage of muscle development. This is the first stage at which we can detect differences in expression of myosin genes between developing muscle fibers. We conclude that, during the development of the myotome and body wall muscles, different myosin genes follow independent patterns of activation and accumulation. The data presented are the first detailed study of myosin gene expression at these early stages of skeletal muscle development.


Asunto(s)
Músculos/embriología , Miosinas/genética , Animales , Secuencia de Bases , Diferenciación Celular , Regulación de la Expresión Génica , Edad Gestacional , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Músculos/citología , Músculos/metabolismo , Miosinas/biosíntesis , Hibridación de Ácido Nucleico , Fenotipo , Sondas ARN , ARN Mensajero/metabolismo , Radioisótopos de Azufre
8.
J Cell Biol ; 111(6 Pt 1): 2427-36, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2277065

RESUMEN

Expression of the two isoforms of cardiac myosin heavy chain (MHC), MHC alpha and MHC beta, in mammals is regulated postnatally by a variety of stimuli, including serum hormone levels. Less is known about the factors that regulate myosin gene expression in rapidly growing cardiac muscle in embryos. Using isoform-specific 35S-labeled cRNA probes corresponding to the two MHC genes and the two myosin alkali light chain (MLC) genes expressed in cardiac muscle, we have investigated the temporal and spatial pattern of expression of these different genes in the developing mouse heart by in situ hybridization. Between 7.5 and 8 d post coitum (p.c.), the newly formed cardiac tube begins to express MHC alpha, MHC beta, MLC1 atrial (MLC1A), and MLC1 ventricular (MLC1V) gene transcripts at high levels throughout the myocardium. As a distinct ventricular chamber forms between 8 and 9 d p.c., MHC beta mRNAs begin to be restricted to ventricular myocytes. This process is complete by 10.5 d p.c. During this time, MHC alpha mRNA levels decrease in ventricular muscle cells but continue to be expressed at high levels in atrial muscle cells. MHC alpha transcripts continue to decrease in ventricular myocytes until 16 d p.c., when they are detectable at low levels, but then increase, and finally replace MHC beta mRNAs in ventricular muscle by 7 d after birth. Like MHC beta, MLC1V transcripts become restricted to ventricular myocytes, but at a slower rate. MLC1V mRNAs continue to be detected at low levels in atrial cells until 15.5 d p.c. MLC1A mRNA levels gradually decrease but are still detectable in ventricular cells until a few days after birth. This dynamic pattern of changes in the myosin phenotype in the prenatal mouse heart suggests that there are different regulatory mechanisms for cell-specific expression of myosin isoforms during cardiac development.


Asunto(s)
Regulación de la Expresión Génica , Genes , Corazón/embriología , Miosinas/genética , Envejecimiento , Animales , Secuencia de Bases , Edad Gestacional , Corazón/crecimiento & desarrollo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Transcripción Genética
9.
J Cell Biol ; 151(6): 1221-34, 2000 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-11121437

RESUMEN

Skeletal muscle is one of a several adult post-mitotic tissues that retain the capacity to regenerate. This relies on a population of quiescent precursors, termed satellite cells. Here we describe two novel markers of quiescent satellite cells: CD34, an established marker of hematopoietic stem cells, and Myf5, the earliest marker of myogenic commitment. CD34(+ve) myoblasts can be detected in proliferating C2C12 cultures. In differentiating cultures, CD34(+ve) cells do not fuse into myotubes, nor express MyoD. Using isolated myofibers as a model of synchronous precursor cell activation, we show that quiescent satellite cells express CD34. An early feature of their activation is alternate splicing followed by complete transcriptional shutdown of CD34. This data implicates CD34 in the maintenance of satellite cell quiescence. In heterozygous Myf5(nlacZ/+) mice, all CD34(+ve) satellite cells also express beta-galactosidase, a marker of activation of Myf5, showing that quiescent satellite cells are committed to myogenesis. All such cells are positive for the accepted satellite cell marker, M-cadherin. We also show that satellite cells can be identified on isolated myofibers of the myosin light chain 3F-nlacZ-2E mouse as those that do not express the transgene. The numbers of satellite cells detected in this way are significantly greater than those identified by the other three markers. We conclude that the expression of CD34, Myf5, and M-cadherin defines quiescent, committed precursors and speculate that the CD34(-ve), Myf5(-ve) minority may be involved in maintaining the lineage-committed majority.


Asunto(s)
Antígenos CD34/aislamiento & purificación , Proteínas de Unión al ADN , Proteínas Musculares/aislamiento & purificación , Músculo Esquelético/citología , Células Madre/citología , Transactivadores , Animales , Diferenciación Celular , Linaje de la Célula , Ratones , Ratones Transgénicos , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/embriología , Factor 5 Regulador Miogénico , Fragmentos de Péptidos/aislamiento & purificación , ARN Mensajero/aislamiento & purificación , Regeneración
10.
J Cell Biol ; 116(5): 1243-55, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1310995

RESUMEN

The accumulation of two myogenic regulatory proteins, MyoD and myogenin, was investigated by double-immunocytochemistry and correlated with myosin heavy chain expression in different classes of myoblasts in culture and during early myogenesis in vivo. During in vitro differentiation of fetal myoblasts, MyoD-positive cells were detected first, followed by the appearance of cells positive for both MyoD and myogenin and finally by the appearance of differentiated myocytes and myotubes expressing myosin heavy chain (MHC). A similar pattern of expression was observed in cultures of embryonic and satellite cells. In contrast, most myogenic cells isolated from newly formed somites, expressed MHC in the absence of detectable levels of myogenin or MyoD. In vivo, the appearance of both myogenin and MyoD proteins was only detected at 10.5 d postcoitum (d.p.c.), when terminally differentiated muscle cells could already be identified in the myotome. Parasagittal sections of the caudal myotomes of 10.5-d-old embryos showed that expression of contractile proteins preceded the expression of myogenin or MyoD and, when coexpressed, MHC and myogenin did not co-localize within all the cells of the myotome. In the limb bud, however, many myogenin (or MyoD) positive/MHC negative cells could be observed in the proximal region at day 11. During further embryonic development the expression of these proteins remained constant in all the muscle anlagens examined, decreasing to a low level during the late fetal period. Western and Northern analysis confirmed that the myogenin protein could only be detected after 10.5 d.p.c. while the corresponding message was clearly present at 9.5 d.p.c., strongly suggesting a posttranscriptional regulation of myogenin during this stage of embryonic development. These data show that the first myogenic cells which appear in the mouse myotome, and can be cultured from it, accumulate muscle structural proteins in their cytoplasm without expressing detectable levels of myogenin protein (although the message is clearly accumulated). Neither MyoD message or protein are detectable in these cells, which may represent a distinct myogenic population whose role in development remains to be established.


Asunto(s)
Proteínas Musculares/análisis , Músculos/embriología , Animales , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Músculos/citología , Proteína MioD , Miogenina , Miosinas/análisis , ARN Mensajero/análisis
11.
Neuron ; 13(4): 813-21, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7946330

RESUMEN

Embryonic mouse neural tubes produce a variety of terminally differentiated cells in vitro, mostly neurons and glia. We report here that some of these cells differentiate into skeletal muscle cells. The possibility of mesoderm contamination was ruled out as follows. First, Dil+ muscle cells were present in cultures from a Dil-labeled neuroepithelium. Second, a small fraction of cultured neural tube cells coexpressed muscle myosin and neuronal beta III tubulin within the same cell. Third, embryos generated from embryonic stem cells in which nlacZ was targeted into the myogenic gene myf-5 expressed nlacZ in a localized region of the neural tube. These myf-5+ cells coexpress neuronal and muscle markers in culture. The developmental significance of this phenomenon is discussed in the context of overlapping regulatory networks between myogenesis and neurogenesis.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Proteínas de Unión al ADN , Músculo Esquelético/citología , Transactivadores , Animales , Carbocianinas , Diferenciación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes , Marcación de Gen , Inmunohistoquímica , Ratones , Proteínas Musculares/análisis , Proteínas Musculares/genética , Factor 5 Regulador Miogénico , Sarcómeros/inmunología , Tubulina (Proteína)/análisis , beta-Galactosidasa/análisis
12.
Curr Opin Genet Dev ; 11(4): 440-8, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11448631

RESUMEN

Research in the past year has added to our understanding of the signalling systems that specify myogenic identity in the embryo and of the regulation and roles of MyoD family members. New insights into the movement of muscle precursor cells include the demonstration that Lbx1 is essential for their migration from the somite to some but not all sites of muscle formation elsewhere. Later in development, ras as well as calcineurin signalling is now implicated in the definition of slow versus fast fibre types. The myogenic identity of precursor cells in the adult depends on Pax7, the orthologue of Pax3 which is required for early myogenesis; this finding is of major importance for muscle regeneration and the active field of stem cell research.


Asunto(s)
Proteínas de Unión al ADN , Músculo Esquelético/embriología , Transducción de Señal/fisiología , Transactivadores , Animales , Movimiento Celular/fisiología , Regulación de la Expresión Génica , Proteínas Musculares/genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Factor 5 Regulador Miogénico , Vertebrados
13.
Curr Opin Genet Dev ; 4(5): 745-51, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7849514

RESUMEN

The study of myogenesis in the embryo is a rapidly expanding field. In this context, the consequences of mutating different members of the MyoD family, together with an increasing number of observations that point to the importance of the MEF2 or RSRF family as myogenic regulators, and the identification of Pax-3 as a marker of early myogenic cells, have advanced our understanding of the molecular embryology of skeletal muscle. Novel cardiac regulatory factors such as Nkx-2.5 and GATA-4, in addition to MEF2 isoforms, are also beginning to be identified. At the molecular level, crystallographic studies have led to a structural model of the actinomyosin complex and also to information about how MyoD contacts DNA.


Asunto(s)
Músculos/embriología , Animales , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Humanos , Factores de Transcripción MEF2 , Ratones , Músculos/fisiología , Proteína MioD/genética , Factores Reguladores Miogénicos , Transducción de Señal , Factores de Transcripción/genética
14.
Anaesthesia ; 68(5): 546, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23573866
15.
Curr Biol ; 4(1): 61-3, 1994 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-7922315

RESUMEN

Mice lacking myogenin have little skeletal muscle as fibres fail to differentiate. Lack of both MyoD and myf-5 results in no skeletal muscle and apparently no myoblasts, suggesting that these factors act earlier in muscle development.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Musculares/fisiología , Músculos/fisiología , Proteína MioD/fisiología , Miogenina/fisiología , Transactivadores , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Desarrollo Embrionario y Fetal , Expresión Génica , Ratones , Ratones Mutantes , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/biosíntesis , Músculos/citología , Músculos/embriología , Proteína MioD/biosíntesis , Factor 5 Regulador Miogénico
16.
Trends Genet ; 8(4): 144-8, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1321521

RESUMEN

Classical embryology has provided a conceptual basis for our understanding of where muscle comes from. Histological and morphological studies of muscle fibre formation in the foetus and neonate have provided information on how muscle matures. More recent advances in molecular genetics have led to the characterization of muscle structural genes, and to the striking discovery of the MyoD family of myogenic regulatory factors. The question of how myogenesis takes place can now be formulated in terms of gene regulation, and molecular tools can be used to describe this process in the embryo and foetus.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas Musculares/fisiología , Músculos/embriología , Músculos/fisiología , Animales , Diferenciación Celular/fisiología , Proteína MioD , Factores de Transcripción/fisiología
17.
Trends Genet ; 12(6): 218-23, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8928226

RESUMEN

Skeletal myoblasts are derived from paraxial mesoderm, but how myoblasts acquire their identity is still a matter of speculation. The characterization of molecular markers and, in some cases, the analysis of mutations in the corresponding genes, has now made it possible to ask specific questions about this process. Specification of somite cell fate depends on epigenetic factors. Adjacent tissues, such as the neural tube, notochord, dorsal ectoderm and lateral mesoderm, act either positively or negatively on the different myogenic precursor populations in the somite. Candidate molecules for this complex signalling activity include sonic hedgehog and the Wnt proteins as positive signals, and BMP4 as a possible inhibitor. Although it is generally assumed that induction is required, some observations suggest that embryonic cells might have a tendency to undergo myogenesis as a 'default' pathway. By analogy with Drosophila, where the neurogenic genes affect myogenesis, the vertebrate homologues of notch and its ligands could be candidate molecules for a repression or derepression mechanism. Similar studies with cultured muscle cells also implicate other HLH factors as potential inhibitors of the MyoD family and, hence, of inappropriate myogenesis.


Asunto(s)
Músculo Esquelético/inmunología , Animales , Biomarcadores , Drosophila , Inducción Embrionaria , Regulación del Desarrollo de la Expresión Génica , Vías Nerviosas/embriología
18.
Mol Cell Biol ; 14(5): 3504-13, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-8164695

RESUMEN

A DNase I-hypersensitive site analysis of the 5'-flanking region of the mouse alpha-cardiac actin gene with muscle cell lines derived from C3H mice shows the presence of two such sites, at about -5 and -7 kb. When tested for activity in cultured cells with homologous and heterologous promoters, both sequences act as muscle-specific enhancers. Transcription from the proximal promoter of the alpha-cardiac actin gene is increased 100-fold with either enhancer. The activity of the distal enhancer in C2/7 myotubes is confined to an 800-bp fragment, which contains multiple E boxes. In transfection assays, this sequence does not give detectable transactivation by any of the myogenic factors even though one of the E boxes is functionally important. Bandshift assays showed that MyoD and myogenin can bind to this E box. However, additional sequences are also required for activity. We conclude that in the case of this muscle enhancer, myogenic factors alone are not sufficient to activate transcription either directly via an E box or indirectly through activation of genes encoding other muscle factors. In BALB/c mice, in which cardiac actin mRNA levels are 8- to 10-fold lower, the alpha-cardiac actin locus is perturbed by a 9.5-kb insertion (I. Garner, A. J. Minty, S. Alonso, P. J. Barton, and M. E. Buckingham, EMBO J. 5:2559-2567, 1986). This is located at -6.5 kb, between the two enhancers. The insertion therefore distances the distal enhancer from the promoter and from the proximal enhancer of the bona fide cardiac actin gene, probably thus perturbing transcriptional activity.


Asunto(s)
Actinas/genética , ADN/genética , Elementos de Facilitación Genéticos , Músculos/metabolismo , Miocardio/metabolismo , ARN Mensajero/biosíntesis , Animales , Secuencia de Bases , Línea Celular , Cloranfenicol O-Acetiltransferasa/biosíntesis , Cloranfenicol O-Acetiltransferasa/metabolismo , Clonación Molecular , ADN/aislamiento & purificación , ADN/metabolismo , Sondas de ADN , Desoxirribonucleasa I , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Mapeo Restrictivo , Transfección
19.
Mol Cell Biol ; 24(7): 2944-57, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15024082

RESUMEN

Expression of the mouse cardiac actin gene depends on a distal enhancer (-7 kbp) which has been shown, in transgenic mice, to direct expression to embryonic skeletal muscle. The presence of this distal sequence is also associated with reproducible expression of cardiac actin transgenes. In differentiated skeletal muscle cells, activity of the enhancer is driven by an E box, binding MyoD family members, and by a 3' AT-rich sequence which is in the location of a DNase I-hypersensitive site. This sequence does not bind MEF2 proteins, or other known muscle transcription factors, directly. Oct1 and Emb, a class VI POU domain protein, bind to consensus sites on the DNA, and it is the binding of Emb which is important for activity. Emb binds as a major complex with MEF2D and the histone transacetylase p300. The form of Emb present in this complex and as a major form in muscle cell extracts is longer (80 kDa) than that previously described. These results demonstrate the importance of this novel complex in the transcriptional regulation of the cardiac actin gene and suggest a potential role in chromatin remodeling associated with muscle gene activation.


Asunto(s)
Acetiltransferasas/metabolismo , Actinas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Histonas/metabolismo , Miocardio/metabolismo , Factores de Transcripción/metabolismo , Actinas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Huella de ADN , Histona Acetiltransferasas , Factores de Transcripción MEF2 , Sustancias Macromoleculares , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Factores Reguladores Miogénicos , Alineación de Secuencia , Activación Transcripcional , Factores de Transcripción p300-CBP
20.
Mol Cell Biol ; 15(8): 4585-96, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7623850

RESUMEN

The mouse myosin light-chain 1A (MLC1A) gene, expressed in the atria of the adult heart, is one of the first muscle genes to be activated when skeletal as well as cardiac muscles form in the embryo. It is also transcribed in skeletal muscle cell lines at the onset of differentiation. Transient transfection assays of mouse skeletal muscle cell lines with DNA constructs containing MLC1A promoter fragments fused to the chloramphenicol acetyltransferase (CAT) gene show that the first 630 bp of the promoter is sufficient to direct expression of the reporter gene during myotube formation. Two E boxes located at bp -76 and -519 are necessary for this regulation. MyoD and myogenin proteins bind to them as heterodimers with E12 protein and, moreover, transactivate them in cotransfection experiments with the MLC1A promoter in nonmuscle cells. Interestingly, the effect of mutating each E box is less striking in primary cultures than in the C2 or Sol8 muscle cell line. A DNA fragment from bp -36 to -597 confers tissue- and stage-specific activity to the herpes simplex virus thymidine kinase promoter in both orientations, showing that the skeletal muscle-specific regulation of the MLC1A gene is under the control of a muscle-specific enhancer which extends into the proximal promoter region. At bp -89 is a diverged CArG box, CC(A/T)6AG, which binds the serum response factor (SRF) in myotube nuclear extracts, as does the wild-type sequence, CC(A/T)6GG. Both types of CArG box also bind a novel myotube-enriched complex which has contact points with the AT-rich part of the CArG box and adjacent 3' nucleotides. Mutations within the CArG box distinguish between the binding of this complex and binding of SRF; only SRF binding is directly involved in the specific regulation of the MLC1A gene in skeletal muscle cell lines.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Desarrollo de Músculos , Músculo Esquelético/crecimiento & desarrollo , Factores Reguladores Miogénicos/metabolismo , Miosinas/genética , Regiones Promotoras Genéticas/genética , Factores de Transcripción , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Análisis Mutacional de ADN , Proteínas de Unión al ADN , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Ratones , Datos de Secuencia Molecular , Proteína MioD/metabolismo , Miogenina/metabolismo , Proteínas Nucleares/metabolismo , Eliminación de Secuencia , Factores de Transcripción TCF , Proteína 1 Similar al Factor de Transcripción 7 , Activación Transcripcional , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA