Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 499: 47-58, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37121308

RESUMEN

Slow myosin heavy chain 1 (Smyhc1) is the major sarcomeric myosin driving early contraction by slow skeletal muscle fibres in zebrafish. New mutant alleles lacking a functional smyhc1 gene move poorly, but recover motility as the later-formed fast muscle fibres of the segmental myotomes mature, and are adult viable. By motility analysis and inhibiting fast muscle contraction pharmacologically, we show that a slow muscle motility defect persists in mutants until about 1 month of age. Breeding onto a genetic background marking slow muscle fibres with EGFP revealed that mutant slow fibres undergo terminal differentiation, migration and fibre formation indistinguishable from wild type but fail to generate large myofibrils and maintain cellular orientation and attachments. In mutants, initial myofibrillar structures with 1.67 â€‹µm periodic actin bands fail to mature into the 1.96 â€‹µm sarcomeres observed in wild type, despite the presence of alternative myosin heavy chain molecules. The poorly-contractile mutant slow muscle cells generate numerous cytoplasmic organelles, but fail to grow and bundle myofibrils or to increase in cytoplasmic volume despite passive movements imposed by fast muscle. The data show that both slow myofibril maturation and cellular volume increase depend on the function of a specific myosin isoform and suggest that appropriate force production regulates muscle fibre growth.


Asunto(s)
Miofibrillas , Cadenas Pesadas de Miosina , Animales , Contracción Muscular , Fibras Musculares Esqueléticas , Músculo Esquelético/fisiología , Miofibrillas/química , Cadenas Pesadas de Miosina/genética , Miosinas , Pez Cebra/genética
2.
J Physiol ; 602(3): 427-443, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38160435

RESUMEN

MYH13 is a unique type of sarcomeric myosin heavy chain (MYH) first detected in mammalian extraocular (EO) muscles and later also in vocal muscles, including laryngeal muscles of some mammals and syringeal muscles of songbirds. All these muscles are specialized in generating very fast contractions while producing relatively low force, a design appropriate for muscles acting against a much lower load than most skeletal muscles inserting into the skeleton. The definition of the physiological properties of muscle fibres containing MYH13 has been complicated by the mixed fibre type composition of EO muscles and the coexistence of different MYH types within the same fibre. A major advance in this area came from studies on isolated recombinant myosin motors and the demonstration that the affinity of actin-bound human MYH13 for ADP is much weaker than those of fast-type MYH1 (type 2X) and MYH2 (type 2A). This property is consistent with a very fast detachment of myosin from actin, a major determinant of shortening velocity. The MYH13 gene arose early during vertebrate evolution but was characterized only in mammals and birds and appears to have been lost in some teleost fish. The MYH13 gene is located at the 3' end of the mammalian fast/developmental gene cluster and in a similar position to the orthologous cluster in syntenic regions of the songbird genome. MYH13 gene regulation is controlled by a super-enhancer in the mammalian locus and deletion of the neighbouring fast MYH1 and MYH4 genes leads to abnormal MYH13 expression in mouse leg muscles.


Asunto(s)
Actinas , Cadenas Pesadas de Miosina , Animales , Humanos , Ratones , Actinas/metabolismo , Mamíferos/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosinas/metabolismo , Músculos Oculomotores/metabolismo
3.
Development ; 147(8)2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32345657

RESUMEN

Skeletal muscle derives from dorsal mesoderm formed during vertebrate gastrulation. Fibroblast growth factor (Fgf) signalling cooperates with Tbx transcription factors to promote dorsal mesoderm formation, but their role in myogenesis has been unclear. Using zebrafish, we show that dorsally derived Fgf signals act through Tbx16 and Tbxta to induce slow and fast trunk muscle precursors at distinct dorsoventral positions. Tbx16 binds to and directly activates the myf5 and myod genes, which are required for commitment to myogenesis. Tbx16 activity depends on Fgf signalling from the organiser. In contrast, Tbxta is not required for myf5 expression, but binds a specific site upstream of myod that is not bound by Tbx16 and drives (dependent on Fgf signals) myod expression in adaxial slow precursors, thereby initiating trunk myogenesis. After gastrulation, when similar muscle cell populations in the post-anal tail are generated from tailbud, declining Fgf signalling is less effective at initiating adaxial myogenesis, which is instead initiated by Hedgehog signalling from the notochord. Our findings suggest a hypothesis for ancestral vertebrate trunk myogenic patterning and how it was co-opted during tail evolution to generate similar muscle by new mechanisms.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Desarrollo de Músculos , Proteína MioD/metabolismo , Proteínas de Dominio T Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/genética , Animales , Tipificación del Cuerpo/genética , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Desarrollo de Músculos/genética , Proteína MioD/genética , Transducción de Señal , Proteínas de Dominio T Box/genética , Transcripción Genética , Regulación hacia Arriba/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
4.
Development ; 147(4)2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32054660

RESUMEN

La-related protein 6 (Larp6) is a conserved RNA-binding protein found across eukaryotes that has been suggested to regulate collagen biogenesis, muscle development, ciliogenesis, and various aspects of cell proliferation and migration. Zebrafish have two Larp6 family genes: larp6a and larp6b Viable and fertile single and double homozygous larp6a and larp6b zygotic mutants revealed no defects in muscle structure, and were indistinguishable from heterozygous or wild-type siblings. However, larp6a mutant females produced eggs with chorions that failed to elevate fully and were fragile. Eggs from larp6b single mutant females showed minor chorion defects, but chorions from eggs laid by larp6a;larp6b double mutant females were more defective than those from larp6a single mutants. Electron microscopy revealed defective chorionogenesis during oocyte development. Despite this, maternal zygotic single and double mutants were viable and fertile. Mass spectrometry analysis provided a description of chorion protein composition and revealed significant reductions in a subset of zona pellucida and lectin-type proteins between wild-type and mutant chorions that paralleled the severity of the phenotype. We conclude that Larp6 proteins are required for normal oocyte development, chorion formation and egg activation.


Asunto(s)
Autoantígenos/genética , Autoantígenos/fisiología , Corion/fisiología , Oocitos/fisiología , Ribonucleoproteínas/genética , Ribonucleoproteínas/fisiología , Animales , Movimiento Celular , Proliferación Celular , Colágeno/fisiología , Proteínas del Huevo/fisiología , Femenino , Edición Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma , Genotipo , Heterocigoto , Homocigoto , Lectinas/fisiología , Masculino , Mutación , Oocitos/citología , Oogénesis/fisiología , Fenotipo , Pez Cebra , Zona Pelúcida/fisiología , Antígeno SS-B
5.
Proc Natl Acad Sci U S A ; 117(49): 31208-31218, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229575

RESUMEN

Muscle tissue shows diurnal variations in function, physiology, and metabolism. Whether such variations are dependent on the circadian clock per se or are secondary to circadian differences in physical activity and feeding pattern is unclear. By measuring muscle growth over 12-h periods in live prefeeding larval zebrafish, we show that muscle grows more during day than night. Expression of dominant negative CLOCK (ΔCLK), which inhibits molecular clock function, ablates circadian differences and reduces muscle growth. Inhibition of muscle contraction reduces growth in both day and night, but does not ablate the day/night difference. The circadian clock and physical activity are both required to promote higher muscle protein synthesis during the day compared to night, whereas markers of protein degradation, murf messenger RNAs, are higher at night. Proteasomal inhibitors increase muscle growth at night, irrespective of physical activity, but have no effect during the day. Although physical activity enhances TORC1 activity, and the TORC1 inhibitor rapamycin inhibits clock-driven daytime growth, no effect on muscle growth at night was detected. Importantly, day/night differences in 1) muscle growth, 2) protein synthesis, and 3) murf expression all persist in entrained larvae under free-running constant conditions, indicating circadian drive. Removal of circadian input by exposure to either permanent darkness or light leads to suboptimal muscle growth. We conclude that diurnal variations in muscle growth and metabolism are a circadian property that is independent of, but augmented by, physical activity, at least during development.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Locomoción/genética , Músculo Esquelético/crecimiento & desarrollo , Animales , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Larva/genética , Larva/crecimiento & desarrollo , Luz , Locomoción/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Fotoperiodo , Sirolimus/farmacología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
6.
Dev Biol ; 470: 95-107, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33245870

RESUMEN

During heart formation, the heart grows and undergoes dramatic morphogenesis to achieve efficient embryonic function. Both in fish and amniotes, much of the growth occurring after initial heart tube formation arises from second heart field (SHF)-derived progenitor cell addition to the arterial pole, allowing chamber formation. In zebrafish, this process has been extensively studied during embryonic life, but it is unclear how larval cardiac growth occurs beyond 3 days post-fertilisation (dpf). By quantifying zebrafish myocardial growth using live imaging of GFP-labelled myocardium we show that the heart grows extensively between 3 and 5 dpf. Using methods to assess cell division, cellular development timing assay and Kaede photoconversion, we demonstrate that proliferation, CM addition, and hypertrophy contribute to ventricle growth. Mechanistically, we show that reduction in Mef2c activity (mef2ca+/-;mef2cb-/-), downstream or in parallel with Nkx2.5 and upstream of Ltbp3, prevents some CM addition and differentiation, resulting in a significantly smaller ventricle by 3 dpf. After 3 dpf, however, CM addition in mef2ca+/-;mef2cb-/- mutants recovers to a normal pace, and the heart size gap between mutants and their siblings diminishes into adulthood. Thus, as in mice, there is an early time window when SHF contribution to the myocardium is particularly sensitive to loss of Mef2c activity.


Asunto(s)
Ventrículos Cardíacos/embriología , Corazón/embriología , Factores de Transcripción MEF2/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Factores de Transcripción MEF2/genética , Proteínas Musculares/genética , Mutación , Tamaño de los Órganos , Organogénesis , Pez Cebra , Proteínas de Pez Cebra/genética
7.
FASEB J ; 33(8): 9116-9130, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31100023

RESUMEN

Tendons are an essential part of the musculoskeletal system, connecting muscle and skeletal elements to enable force generation. The transcription factor scleraxis marks vertebrate tendons from early specification. Scleraxis-null mice are viable and have a range of tendon and bone defects in the trunk and limbs but no described cranial phenotype. We report the expression of zebrafish scleraxis orthologs: scleraxis homolog (scx)-a and scxb in cranial and intramuscular tendons and in other skeletal elements. Single mutants for either scxa or scxb, generated by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), are viable and fertile as adult fish. Although scxb mutants show no obvious phenotype, scxa mutant embryos have defects in cranial tendon maturation and muscle misalignment. Mutation of both scleraxis genes results in more severe defects in cranial tendon differentiation, muscle and cartilage dysmorphogenesis and paralysis, and lethality by 2-5 wk, which indicates an essential function of scleraxis for craniofacial development. At juvenile and adult stages, ribs in scxa mutants fail to mineralize and/or are small and heavily fractured. Scxa mutants also have smaller muscle volume, abnormal swim movement, and defects in bone growth and composition. Scleraxis function is therefore essential for normal craniofacial form and function and vital for fish development.-Kague, E., Hughes, S. M., Lawrence, E. A., Cross, S., Martin-Silverstone, E., Hammond, C. L., Hinits, Y. Scleraxis genes are required for normal musculoskeletal development and for rib growth and mineralization in zebrafish.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Desarrollo Musculoesquelético/genética , Proteínas de Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Desarrollo Óseo/genética , Calcificación Fisiológica/genética , Regulación del Desarrollo de la Expresión Génica , Mutación , Costillas/anomalías , Costillas/crecimiento & desarrollo , Costillas/metabolismo , Tendones/anomalías , Tendones/crecimiento & desarrollo , Tendones/metabolismo , Pez Cebra/metabolismo
8.
Genes Dev ; 26(18): 2103-17, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22987640

RESUMEN

How muscle diversity is generated in the vertebrate body is poorly understood. In the limb, dorsal and ventral muscle masses constitute the first myogenic diversification, as each gives rise to distinct muscles. Myogenesis initiates after muscle precursor cells (MPCs) have migrated from the somites to the limb bud and populated the prospective muscle masses. Here, we show that Sonic hedgehog (Shh) from the zone of polarizing activity (ZPA) drives myogenesis specifically within the ventral muscle mass. Shh directly induces ventral MPCs to initiate Myf5 transcription and myogenesis through essential Gli-binding sites located in the Myf5 limb enhancer. In the absence of Shh signaling, myogenesis is delayed, MPCs fail to migrate distally, and ventral paw muscles fail to form. Thus, Shh production in the limb ZPA is essential for the spatiotemporal control of myogenesis and coordinates muscle and skeletal development by acting directly to regulate the formation of specific ventral muscles.


Asunto(s)
Extremidades/embriología , Proteínas Hedgehog/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Mioblastos/citología , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Esbozos de los Miembros/citología , Esbozos de los Miembros/embriología , Ratones , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Transducción de Señal
9.
Semin Cell Dev Biol ; 72: 33-44, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29154822

RESUMEN

Mef2 is a conserved and significant transcription factor in the control of muscle gene expression. In cell culture Mef2 synergises with MyoD-family members in the activation of gene expression and in the conversion of fibroblasts into myoblasts. Amongst its in vivo roles, Mef2 is required for both Drosophila muscle development and mammalian muscle regeneration. Mef2 has functions in other cell-types too, but this review focuses on skeletal muscle and surveys key findings on Mef2 from its discovery, shortly after that of MyoD, up to the present day. In particular, in vivo functions, underpinning mechanisms and areas of uncertainty are highlighted. We describe how Mef2 sits at a nexus in the gene expression network that controls the muscle differentiation program, and how Mef2 activity must be regulated in time and space to orchestrate specific outputs within the different aspects of muscle development. A theme that emerges is that there is much to be learnt about the different Mef2 proteins (from different paralogous genes, spliced transcripts and species) and how the activity of these proteins is controlled.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción MEF2/genética , Músculo Esquelético/metabolismo , Animales , Redes Reguladoras de Genes , Modelos Genéticos , Desarrollo de Músculos/genética , Músculo Esquelético/citología , Músculo Esquelético/embriología , Mioblastos/citología , Mioblastos/metabolismo , Regeneración/genética
10.
Dev Biol ; 431(2): 321-335, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28887016

RESUMEN

Balancing the number of stem cells and their progeny is crucial for tissue development and repair. Here we examine how cell numbers and overall muscle size are tightly regulated during zebrafish somitic muscle development. Muscle stem/precursor cell (MPCs) expressing Pax7 are initially located in the dermomyotome (DM) external cell layer, adopt a highly stereotypical distribution and thereafter a proportion of MPCs migrate into the myotome. Regional variations in the proliferation and terminal differentiation of MPCs contribute to growth of the myotome. To probe the robustness of muscle size control and spatiotemporal regulation of MPCs, we compared the behaviour of wild type (wt) MPCs with those in mutant zebrafish that lack the muscle regulatory factor Myod. Myodfh261 mutants form one third fewer multinucleate fast muscle fibres than wt and show a significant expansion of the Pax7+ MPC population in the DM. Subsequently, myodfh261 mutant fibres generate more cytoplasm per nucleus, leading to recovery of muscle bulk. In addition, relative to wt siblings, there is an increased number of MPCs in myodfh261 mutants and these migrate prematurely into the myotome, differentiate and contribute to the hypertrophy of existing fibres. Thus, homeostatic reduction of the excess MPCs returns their number to normal levels, but fibre numbers remain low. The GSK3 antagonist BIO prevents MPC migration into the deep myotome, suggesting that canonical Wnt pathway activation maintains the DM in zebrafish, as in amniotes. BIO does not, however, block recovery of the myodfh261 mutant myotome, indicating that homeostasis acts on fibre intrinsic growth to maintain muscle bulk. The findings suggest the existence of a critical window for early fast fibre formation followed by a period in which homeostatic mechanisms regulate myotome growth by controlling fibre size. The feedback controls we reveal in muscle help explain the extremely precise grading of myotome size along the body axis irrespective of fish size, nutrition and genetic variation and may form a paradigm for wider matching of organ size.


Asunto(s)
Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Somitos/metabolismo , Pez Cebra/embriología , Animales , Diferenciación Celular , Movimiento Celular , Núcleo Celular/metabolismo , Proliferación Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Larva/metabolismo , Fibras Musculares Esqueléticas/citología , Mutación/genética , Factor de Transcripción PAX7/metabolismo , Somitos/embriología , Proteínas de Pez Cebra/metabolismo
11.
Development ; 140(14): 2972-84, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23760954

RESUMEN

Striated muscles that enable mouth opening and swallowing during feeding are essential for efficient energy acquisition, and are likely to have played a fundamental role in the success of early jawed vertebrates. The developmental origins and genetic requirements of these muscles are uncertain. Here, we determine by indelible lineage tracing in mouse that fibres of sternohyoid muscle (SHM), which is essential for mouth opening during feeding, and oesophageal striated muscle (OSM), which is crucial for voluntary swallowing, arise from Pax3-expressing somite cells. In vivo Kaede lineage tracing in zebrafish reveals the migratory route of cells from the anteriormost somites to OSM and SHM destinations. Expression of pax3b, a zebrafish duplicate of Pax3, is restricted to the hypaxial region of anterior somites that generate migratory muscle precursors (MMPs), suggesting that Pax3b plays a role in generating OSM and SHM. Indeed, loss of pax3b function led to defective MMP migration and OSM formation, disorganised SHM differentiation, and inefficient ingestion and swallowing of microspheres. Together, our data demonstrate Pax3-expressing somite cells as a source of OSM and SHM fibres, and highlight a conserved role of Pax3 genes in the genesis of these feeding muscles of vertebrates.


Asunto(s)
Esófago/embriología , Maxilares/embriología , Desarrollo de Músculos , Músculo Estriado/embriología , Factores de Transcripción Paired Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Movimiento Celular , Deglución , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Esófago/citología , Feto/citología , Feto/metabolismo , Maxilares/citología , Ratones , Músculo Estriado/citología , Músculo Estriado/metabolismo , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Somitos/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
12.
PLoS Biol ; 11(10): e1001679, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24143132

RESUMEN

Muscle fiber size is activity-dependent and clinically important in ageing, bed-rest, and cachexia, where muscle weakening leads to disability, prolonged recovery times, and increased costs. Inactivity causes muscle wasting by triggering protein degradation and may simultaneously prevent protein synthesis. During development, muscle tissue grows by several mechanisms, including hypertrophy of existing fibers. As in other tissues, the TOR pathway plays a key role in promoting muscle protein synthesis by inhibition of eIF4EBPs (eukaryotic Initiation Factor 4E Binding Proteins), regulators of the translational initiation. Here, we tested the role of TOR-eIF4EBP in a novel zebrafish muscle inactivity model. Inactivity triggered up-regulation of eIF4EBP3L (a zebrafish homolog of eIF4EBP3) and diminished myosin and actin content, myofibrilogenesis, and fiber growth. The changes were accompanied by preferential reduction of the muscle transcription factor Mef2c, relative to Myod and Vinculin. Polysomal fractionation showed that Mef2c decrease was due to reduced translation of mef2ca mRNA. Loss of Mef2ca function reduced normal muscle growth and diminished the reduction in growth caused by inactivity. We identify eIF4EBP3L as a key regulator of Mef2c translation and protein level following inactivity; blocking eIF4EBP3L function increased Mef2ca translation. Such blockade also prevented the decline in mef2ca translation and level of Mef2c and slow myosin heavy chain proteins caused by inactivity. Conversely, overexpression of active eIF4EBP3L mimicked inactivity by decreasing the proportion of mef2ca mRNA in polysomes, the levels of Mef2c and slow myosin heavy chain, and myofibril content. Inhibiting the TOR pathway without the increase in eIF4EBP3L had a lesser effect on myofibrilogenesis and muscle size. These findings identify eIF4EBP3L as a key TOR-dependent regulator of muscle fiber size in response to activity. We suggest that by selectively inhibiting translational initiation of mef2ca and other mRNAs, eIF4EBP3L reprograms the translational profile of muscle, enabling it to adjust to new environmental conditions.


Asunto(s)
Proteínas Portadoras/metabolismo , Factores de Transcripción MEF2/genética , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Factores Reguladores Miogénicos/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factores de Transcripción MEF2/metabolismo , Modelos Biológicos , Miofibrillas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Regulación hacia Arriba , Pez Cebra/genética , Proteínas de Pez Cebra/genética
13.
Dev Biol ; 369(2): 199-210, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22750409

RESUMEN

Mef2 transcription factors have been strongly linked with early heart development. D-mef2 is required for heart formation in Drosophila, but whether Mef2 is essential for vertebrate cardiomyocyte (CM) differentiation is unclear. In mice, although Mef2c is expressed in all CMs, targeted deletion of Mef2c causes lethal loss of second heart field (SHF) derivatives and failure of cardiac looping, but first heart field CMs can differentiate. Here we examine Mef2 function in early heart development in zebrafish. Two Mef2c genes exist in zebrafish, mef2ca and mef2cb. Both are expressed similarly in the bilateral heart fields but mef2cb is strongly expressed in the heart poles at the primitive heart tube stage. By using fish mutants for mef2ca and mef2cb and antisense morpholinos to knock down either or both Mef2cs, we show that Mef2ca and Mef2cb have essential but redundant roles in myocardial differentiation. Loss of both Mef2ca and Mef2cb function does not interfere with early cardiogenic markers such as nkx2.5, gata4 and hand2 but results in a dramatic loss of expression of sarcomeric genes and myocardial markers such as bmp4, nppa, smyd1b and late nkx2.5 mRNA. Rare residual CMs observed in mef2ca;mef2cb double mutants are ablated by a morpholino capable of knocking down other Mef2s. Mef2cb over-expression activates bmp4 within the cardiogenic region, but no ectopic CMs are formed. Surprisingly, anterior mesoderm and other tissues become skeletal muscle. Mef2ca single mutants have delayed heart development, but form an apparently normal heart. Mef2cb single mutants have a functional heart and are viable adults. Our results show that the key role of Mef2c in myocardial differentiation is conserved throughout the vertebrate heart.


Asunto(s)
Proteínas Musculares/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Factores Reguladores Miogénicos/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Diferenciación Celular/genética , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Desarrollo de Músculos/genética , Mutación , Miocardio/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Methods Mol Biol ; 2640: 21-43, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995585

RESUMEN

Adult skeletal musculature experiences continuous physical stress, and hence requires maintenance and repair to ensure its continued efficient functioning. The population of resident muscle stem cells (MuSCs), termed satellite cells, resides beneath the basal lamina of adult myofibers, contributing to both muscle hypertrophy and regeneration. Upon exposure to activating stimuli, MuSCs proliferate to generate new myoblasts that differentiate and fuse to regenerate or grow myofibers. Moreover, many teleost fish undergo continuous growth throughout life, requiring continual nuclear recruitment from MuSCs to initiate and grow new fibers, a process that contrasts with the determinate growth observed in most amniotes. In this chapter, we describe a method for the isolation, culture, and immunolabeling of adult zebrafish myofibers that permits examination of both myofiber characteristics ex vivo and the MuSC myogenic program in vitro. Morphometric analysis of isolated myofibers is suitable to assess differences among slow and fast muscles or to investigate cellular features such as sarcomeres and neuromuscular junctions. Immunostaining for Pax7, a canonical stemness marker, identifies MuSCs on isolated myofibers for study. Furthermore, the plating of viable myofibers allows MuSC activation and expansion and downstream analysis of their proliferative and differentiative dynamics, thus providing a suitable, parallel alternative to amniote models for the study of vertebrate myogenesis.


Asunto(s)
Células Satélite del Músculo Esquelético , Pez Cebra , Animales , Músculo Esquelético , Diferenciación Celular , Desarrollo de Músculos , Fibras Musculares Esqueléticas
15.
Methods Mol Biol ; 2640: 227-248, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995599

RESUMEN

Muscle regeneration models have revealed mechanisms of inflammation, wound clearance, and stem cell-directed repair of damage, thereby informing therapy. Whereas studies of muscle repair are most advanced in rodents, the zebrafish is emerging as an additional model organism with genetic and optical advantages. Various muscle wounding protocols (both chemical and physical) have been published. Here we describe simple, cheap, precise, adaptable, and effective wounding protocols and analysis methods for two stages of a larval zebrafish skeletal muscle regeneration model. We show examples of how muscle damage, ingression of muscle stem cells, immune cells, and regeneration of fibers can be monitored over an extended timecourse in individual larvae. Such analyses have the potential to greatly enhance understanding, by reducing the need to average regeneration responses across individuals subjected to an unavoidably variable wound stimulus.


Asunto(s)
Células Satélite del Músculo Esquelético , Pez Cebra , Animales , Pez Cebra/fisiología , Fibras Musculares Esqueléticas , Células Madre , Células Satélite del Músculo Esquelético/fisiología , Proliferación Celular , Músculo Esquelético/fisiología
16.
Dev Biol ; 350(2): 464-75, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21147088

RESUMEN

Differentiation often requires conversion of analogue signals to a stable binary output through positive feedback. Hedgehog (Hh) signalling promotes myogenesis in the vertebrate somite, in part by raising the activity of muscle regulatory factors (MRFs) of the Myod family above a threshold. Hh is known to enhance MRF expression. Here we show that Hh is also essential at a second step that increases Myod protein activity, permitting it to promote Myogenin expression. Hh acts by inducing expression of cdkn1c (p57(Kip2)) in slow muscle precursor cells, but neither Hh nor Cdkn1c is required for their cell cycle exit. Cdkn1c co-operates with Myod to drive differentiation of several early zebrafish muscle fibre types. Myod in turn up-regulates cdkn1c, thereby providing a positive feedback loop that switches myogenic cells to terminal differentiation.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Desarrollo de Músculos , Proteína MioD/fisiología , Animales , Diferenciación Celular , Retroalimentación Fisiológica , Proteínas Hedgehog/fisiología , Factor 5 Regulador Miogénico/fisiología , Miogenina/genética , Transducción de Señal , Pez Cebra
17.
Dev Biol ; 358(1): 102-12, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21798255

RESUMEN

Myogenic regulatory factors of the myod family (MRFs) are transcription factors essential for mammalian skeletal myogenesis. Here we show that a mutation in the zebrafish myod gene delays and reduces early somitic and pectoral fin myogenesis, reduces miR-206 expression, and leads to a persistent reduction in somite size until at least the independent feeding stage. A mutation in myog, encoding a second MRF, has little obvious phenotype at early stages, but exacerbates the loss of somitic muscle caused by lack of Myod. Mutation of both myod and myf5 ablates all skeletal muscle. Haploinsufficiency of myod leads to reduced embryonic somite muscle bulk. Lack of Myod causes a severe reduction in cranial musculature, ablating most muscles including the protractor pectoralis, a putative cucullaris homologue. This phenotype is accompanied by a severe dysmorphology of the cartilaginous skeleton and failure of maturation of several cranial bones, including the opercle. As myod expression is restricted to myogenic cells, the data show that myogenesis is essential for proper skeletogenesis in the head.


Asunto(s)
Huesos/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Haploinsuficiencia/genética , Desarrollo de Músculos/fisiología , Proteína MioD/genética , Cráneo/embriología , Pez Cebra/embriología , Animales , Cartílago/embriología , Haploinsuficiencia/fisiología , Inmunohistoquímica , Hibridación in Situ , Larva/fisiología , Músculo Esquelético/embriología , Mutación/genética , Proteína MioD/metabolismo , Extremidad Superior/embriología , Pez Cebra/genética
18.
FEBS J ; 289(6): 1428-1456, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33755332

RESUMEN

From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.


Asunto(s)
Ejercicio Físico , Contracción Muscular , Adenosina Trifosfato , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas , Músculo Esquelético/fisiología , Miosinas
19.
J Vis Exp ; (184)2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35781279

RESUMEN

A number of methods can be used to visualize individual cells throughout the body of live embryonic, larval or juvenile zebrafish. We show that live fish with fluorescently-marked plasma membranes can be scanned in a confocal laser scanning microscope in order to determine the volume of muscle tissue and the number of muscle fibers present. Efficient approaches for the measurement of cell number and size in live animals over time are described and validated against more arduous segmentation methods. Methods are described that permit the control of muscle electrical, and thus contractile, activity. Loss of skeletal muscle contractile activity greatly reduced muscle growth. In larvae, a protocol is described that allows reintroduction of patterned electrical-evoked contractile activity. The described methods minimize the effect of inter-individual variability and will permit analysis of the effect of electrical, genetic, drug, or environmental stimuli on a variety of cellular and physiological growth parameters in the context of the living organism. Long-term follow-up of the measured effects of a defined early-life intervention on individuals can subsequently be performed.


Asunto(s)
Fenómenos Fisiológicos del Sistema Nervioso , Pez Cebra , Animales , Larva , Contracción Muscular , Fibras Musculares Esqueléticas
20.
Biol Open ; 11(8)2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35972050

RESUMEN

To address questions of stem cell diversity during skeletal myogenesis, a Brainbow-like genetic cell lineage tracing method, dubbed Musclebow2, was derived by enhancer trapping in zebrafish. It is shown that, after initial formation of the primary myotome, at least 15 muscle precursor cells (mpcs) seed each somite, where they proliferate but contribute little to muscle growth prior to hatching. Thereafter, dermomyotome-derived mpc clones rapidly expand while some progeny undergo terminal differentiation, leading to stochastic clonal drift within the mpc pool. No evidence of cell-lineage-based clonal fate diversity was obtained. Neither fibre nor mpc death was observed in uninjured animals. Individual marked muscle fibres persist across much of the lifespan indicating low rates of nuclear turnover. In adulthood, early-marked mpc clones label stable blocks of tissue comprising a significant fraction of either epaxial or hypaxial somite. Fusion of cells from separate early-marked clones occurs in regions of clone overlap. Wounds are regenerated from several local mpcs; no evidence for specialised stem mpcs was obtained. In conclusion, our data indicate that most mpcs in muscle tissue contribute to local growth and repair and suggest that cellular turnover is low in the absence of trauma.


Asunto(s)
Longevidad , Pez Cebra , Animales , Desarrollo de Músculos , Músculo Esquelético , Somitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA