Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 2797, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493999

RESUMEN

Fat distribution is an independent cardiometabolic risk factor. However, its molecular and cellular underpinnings remain obscure. Here we demonstrate that two independent GWAS signals at RSPO3, which are associated with increased body mass index-adjusted waist-to-hip ratio, act to specifically increase RSPO3 expression in subcutaneous adipocytes. These variants are also associated with reduced lower-body fat, enlarged gluteal adipocytes and insulin resistance. Based on human cellular studies RSPO3 may limit gluteofemoral adipose tissue (AT) expansion by suppressing adipogenesis and increasing gluteal adipocyte susceptibility to apoptosis. RSPO3 may also promote upper-body fat distribution by stimulating abdominal adipose progenitor (AP) proliferation. The distinct biological responses elicited by RSPO3 in abdominal versus gluteal APs in vitro are associated with differential changes in WNT signalling. Zebrafish carrying a nonsense rspo3 mutation display altered fat distribution. Our study identifies RSPO3 as an important determinant of peripheral AT storage capacity.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Distribución de la Grasa Corporal , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trombospondinas/metabolismo , Proteínas de Pez Cebra/metabolismo , Adipocitos/efectos de los fármacos , Tejido Adiposo/metabolismo , Adiposidad/genética , Adulto , Alelos , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Doxiciclina/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Caracteres Sexuales , Células Madre/metabolismo , Trombospondinas/genética , Relación Cintura-Cadera , Vía de Señalización Wnt/efectos de los fármacos , Pez Cebra/genética , Proteínas de Pez Cebra/genética
3.
Adipocyte ; 8(1): 330-338, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31411107

RESUMEN

Adipose tissues often exhibit subtle, quantitative differences between individuals, leading to a graded series of adiposity phenotypes at the population level. Robust, quantitative analyses are vital for studying these differences. In this Commentary we highlight two articles from our lab that employ sensitive new methods in zebrafish capable of delineating complex and quantitative adiposity phenotypes. In the first article, we utilized in vivo imaging to systematically quantify zebrafish adipose tissues. We identified 34 regionally distinct zebrafish adipose tissues and developed statistical models to predict the size and variance of each adipose tissue over the course of zebrafish growth. We then employed these models to identify effects of strain and diet on adipose tissue growth. In the second article, we employed deep phenotyping to study complex disease-related adiposity traits. Using this methodology, we identified that adipose tissues have unique capacities to re-deposit lipid following food restriction and re-feeding. These distinct re-deposition potentials led to widespread fat distribution changes following re-feeding. We discuss how these novel findings may provide relevance to health conditions such as anorexia nervosa. Together, the strategies described in these two articles can be used as unbiased and quantitative methods to uncover new relationships between genotype, diet and adiposity.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad/fisiología , Pez Cebra/metabolismo , Animales , Biomarcadores/metabolismo , Dieta , Modelos Biológicos , Imagen Molecular , Tamaño de los Órganos , Distribución Tisular
4.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661757

RESUMEN

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Asunto(s)
Metabolismo Energético/genética , Melanocortinas/metabolismo , Semaforinas/genética , Adolescente , Adulto , Animales , Peso Corporal , Línea Celular , Niño , Preescolar , Modelos Animales de Enfermedad , Ingestión de Alimentos , Femenino , Variación Genética/genética , Homeostasis , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Obesidad/genética , Obesidad/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Adulto Joven , Pez Cebra
5.
J Lipid Res ; 59(8): 1536-1545, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29794036

RESUMEN

The regional distribution of adipose tissues is implicated in a wide range of diseases. For example, proportional increases in visceral adipose tissue increase the risk for insulin resistance, diabetes, and CVD. Zebrafish offer a tractable model system by which to obtain unbiased and quantitative phenotypic information on regional adiposity, and deep phenotyping can explore complex disease-related adiposity traits. To facilitate deep phenotyping of zebrafish adiposity traits, we used pairwise correlations between 67 adiposity traits to generate stage-specific adiposity profiles that describe changing adiposity patterns and relationships during growth. Linear discriminant analysis classified individual fish according to an adiposity profile with 87.5% accuracy. Deep phenotyping of eight previously uncharacterized zebrafish mutants identified neuropilin 2b as a novel gene that alters adipose distribution. When we applied deep phenotyping to identify changes in adiposity during diet manipulations, zebrafish that underwent food restriction and refeeding had widespread adiposity changes when compared with continuously fed, equivalently sized control animals. In particular, internal adipose tissues (e.g., visceral adipose) exhibited a reduced capacity to replenish lipid following food restriction. Together, these results in zebrafish establish a new deep phenotyping technique as an unbiased and quantitative method to help uncover new relationships between genotype, diet, and adiposity.


Asunto(s)
Adiposidad/efectos de los fármacos , Adiposidad/genética , Dieta/efectos adversos , Predisposición Genética a la Enfermedad/genética , Fenotipo , Pez Cebra/genética , Pez Cebra/metabolismo , Animales , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Pez Cebra/crecimiento & desarrollo
6.
J Exp Biol ; 221(Pt Suppl 1)2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29514883

RESUMEN

Adipose morphology is defined as the number and size distribution of adipocytes (fat cells) within adipose tissue. Adipose tissue with fewer but larger adipocytes is said to have a 'hypertrophic' morphology, whereas adipose with many adipocytes of a smaller size is said to have a 'hyperplastic' morphology. Hypertrophic adipose morphology is positively associated with insulin resistance, diabetes and cardiovascular disease. By contrast, hyperplastic morphology is associated with improved metabolic parameters. These phenotypic associations suggest that adipose morphology influences risk of cardiometabolic disease. Intriguingly, monozygotic twin studies have determined that adipose morphology is in part determined genetically. Therefore, identifying the genetic regulation of adipose morphology may help us to predict, prevent and ameliorate insulin resistance and associated metabolic diseases. Here, we review the current literature regarding adipose morphology in relation to: (1) metabolic and medical implications; (2) the methods used to assess adipose morphology; and (3) transcriptional differences between morphologies. We further highlight three mechanisms that have been hypothesized to promote adipocyte hypertrophy and thus to regulate adipose morphology.


Asunto(s)
Tejido Adiposo/fisiología , Enfermedades Metabólicas/fisiopatología , Tejido Adiposo/fisiopatología , Animales , Humanos , Enfermedades Metabólicas/etiología , Ratones , Ratas
7.
Adipocyte ; 6(4): 277-283, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28792859

RESUMEN

Non-communicable diseases (NCDs) such as cardiovascular disease, diabetes and cancer were responsible for 68% of all deaths worldwide in 2012. The regional distribution of lipid deposited within adipose tissue (AT) - so called body fat distribution (BFD) - is a strong risk factor for NCDs. BFD is highly heritable; however, the genetic basis of BFD is almost entirely unknown. Genome-wide association studies have identified several loci associated with BFD, including at Plexin D1 (PLXND1) - a gene known to modulate angiogenesis. We recently demonstrated that zebrafish homozygous for a null mutation in plxnd1 had a reduced capacity to store lipid in visceral AT (VAT) leading to altered BFD. Moreover, we found that type V collagens were upregulated in plxnd1 mutants, and mediated the inhibitory effect of Plxnd1 on VAT growth. These results strengthen evidence that Plxnd1 influences BFD in human populations, and validate zebrafish as a model to study BFD. However, many pertinent questions remain unanswered. Here we outline potential Plxnd1 mechanisms of action in AT, and describe the genetic architecture at human PLXND1 that is associated with BFD and NCD susceptibility.


Asunto(s)
Adiposidad/genética , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Receptores de Superficie Celular/genética , Proteínas de Pez Cebra/genética , Tejido Adiposo/metabolismo , Animales , Distribución de la Grasa Corporal , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Matriz Extracelular , Estudio de Asociación del Genoma Completo , Receptores de Superficie Celular/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-28588550

RESUMEN

The tropical freshwater zebrafish has recently emerged as a valuable model organism for the study of adipose tissue biology and obesity-related disease. The strengths of the zebrafish model system are its wealth of genetic mutants, transgenic tools, and amenability to high-resolution imaging of cell dynamics within live animals. However, zebrafish adipose research is at a nascent stage and many gaps exist in our understanding of zebrafish adipose physiology and metabolism. By contrast, adipose research within other, closely related, teleost species has a rich and extensive history, owing to the economic importance of these fish as a food source. Here, we compare and contrast knowledge on peroxisome proliferator-activated receptor gamma (PPARG)-mediated adipogenesis derived from both biomedical and aquaculture literatures. We first concentrate on the biomedical literature to (i) briefly review PPARG-mediated adipogenesis in mammals, before (ii) reviewing Pparg-mediated adipogenesis in zebrafish. Finally, we (iii) mine the aquaculture literature to compare and contrast Pparg-mediated adipogenesis in aquaculturally relevant teleosts. Our goal is to highlight evolutionary similarities and differences in adipose biology that will inform our understanding of the role of adipose tissue in obesity and related disease.

9.
Dis Model Mech ; 10(6): 797-809, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28348140

RESUMEN

The zebrafish model system offers significant utility for in vivo imaging of adipose tissue (AT) dynamics and for screening to identify chemical and genetic modifiers of adiposity. In particular, AT can be quantified accurately in live zebrafish using fluorescent lipophilic dyes. Although this methodology offers considerable promise, the comprehensive identification and classification of zebrafish ATs has not been performed. Here, we use fluorescent lipophilic dyes and in vivo imaging systematically to identify, classify and quantify the zebrafish AT pool. We identify 34 regionally distinct zebrafish ATs, including five visceral ATs and 22 subcutaneous ATs. For each of these ATs, we describe detailed morphological characteristics to aid their identification in future studies. Furthermore, we quantify the areas for each AT and construct regression models to allow prediction of expected AT size and variation across a range of developmental stages. Finally, we demonstrate the utility of this resource for identifying effects of strain variation and high-fat diet on AT growth. Altogether, this resource provides foundational information on the identity, dynamics and expected quantities of zebrafish ATs for use as a reference for future studies.


Asunto(s)
Tejido Adiposo/anatomía & histología , Tejido Adiposo/metabolismo , Pez Cebra/metabolismo , Adiposidad , Aletas de Animales/fisiología , Animales , Dieta Alta en Grasa , Embrión no Mamífero/metabolismo , Tamaño de los Órganos , Análisis de Regresión , Pez Cebra/embriología
10.
Proc Natl Acad Sci U S A ; 112(14): 4363-8, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831505

RESUMEN

Genome-wide association studies have implicated PLEXIN D1 (PLXND1) in body fat distribution and type 2 diabetes. However, a role for PLXND1 in regional adiposity and insulin resistance is unknown. Here we use in vivo imaging and genetic analysis in zebrafish to show that Plxnd1 regulates body fat distribution and insulin sensitivity. Plxnd1 deficiency in zebrafish induced hyperplastic morphology in visceral adipose tissue (VAT) and reduced lipid storage. In contrast, subcutaneous adipose tissue (SAT) growth and morphology were unaffected, resulting in altered body fat distribution and a reduced VAT:SAT ratio in zebrafish. A VAT-specific role for Plxnd1 appeared conserved in humans, as PLXND1 mRNA was positively associated with hypertrophic morphology in VAT, but not SAT. In zebrafish plxnd1 mutants, the effect on VAT morphology and body fat distribution was dependent on induction of the extracellular matrix protein collagen type V alpha 1 (col5a1). Furthermore, after high-fat feeding, zebrafish plxnd1 mutant VAT was resistant to expansion, and excess lipid was disproportionately deposited in SAT, leading to an even greater exacerbation of altered body fat distribution. Plxnd1-deficient zebrafish were protected from high-fat-diet-induced insulin resistance, and human VAT PLXND1 mRNA was positively associated with type 2 diabetes, suggesting a conserved role for PLXND1 in insulin sensitivity. Together, our findings identify Plxnd1 as a novel regulator of VAT growth, body fat distribution, and insulin sensitivity in both zebrafish and humans.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Colágeno Tipo V/biosíntesis , Insulina/metabolismo , Grasa Intraabdominal/patología , Glicoproteínas de Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Composición Corporal , Proliferación Celular , Células Endoteliales/citología , Matriz Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lípidos/química , Ratones , Mutación , Obesidad , ARN Mensajero/metabolismo , Transducción de Señal , Pez Cebra
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.
Endocrinology ; 154(4): 1476-87, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23456361

RESUMEN

Somatic growth and adipogenesis are closely associated with the development of obesity in humans. In this study, we identify a zebrafish mutant, vizzini, that exhibits both a severe defect in somatic growth and increased accumulation of adipose tissue. Positional cloning of vizzini revealed a premature stop codon in gh1. Although the effects of GH are largely through igfs in mammals, we found no decrease in the expression of igf transcripts in gh1 mutants during larval development. As development progressed, however, we found overall growth to be progressively retarded and the attainment of specific developmental stages to occur at abnormally small body sizes relative to wild type. Moreover, both subcutaneous (sc) and visceral adipose tissues underwent precocious development in vizzini mutants, and at maturity, the sizes of different fat deposits were greatly expanded relative to wild type. In vivo confocal imaging of sc adipose tissue (SAT) expansion revealed that vizzini mutants exhibit extreme enlargement of adipocyte lipid droplets without a corresponding increase in lipid droplet number. These findings suggest that GH1 signaling restricts SAT hypertrophy in zebrafish. Finally, nutrient deprivation of vizzini mutants revealed that SAT mobilization was greatly diminished during caloric restriction, further implicating GH1 signaling in adipose tissue homeostasis. Overall, the zebrafish gh1 mutant, vizzini, exhibits decreased somatic growth, increased adipose tissue accumulation, and disrupted adipose plasticity after nutrient deprivation and represents a novel model to investigate the in vivo dynamics of vertebrate obesity.


Asunto(s)
Adipocitos/patología , Adipogénesis/genética , Adiposidad/genética , Enanismo/genética , Hormona del Crecimiento/genética , Obesidad/genética , Pez Cebra/genética , Animales , Codón sin Sentido , Hipertrofia
13.
Methods Cell Biol ; 105: 63-86, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21951526

RESUMEN

White adipose tissue (WAT) is the major site of energy storage in bony vertebrates, and also serves central roles in the endocrine regulation of energy balance. The cellular and molecular mechanisms underlying WAT development and physiology are not well understood. This is due in part to difficulties associated with imaging adipose tissues in mammalian model systems, especially during early life stages. The zebrafish (Danio rerio) has recently emerged as a new model system for adipose tissue research, in which WAT can be imaged in a transparent living vertebrate at all life stages. Here we present detailed methods for labeling adipocytes in live zebrafish using fluorescent lipophilic dyes, and for in vivo microscopy of zebrafish WAT.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo Blanco/metabolismo , Biología Evolutiva/métodos , Embrión no Mamífero/metabolismo , Larva/metabolismo , Imagen Molecular/métodos , Coloración y Etiquetado/métodos , Pez Cebra/embriología , Adipocitos/citología , Tejido Adiposo Blanco/química , Tejido Adiposo Blanco/embriología , Animales , Embrión no Mamífero/anatomía & histología , Metabolismo Energético , Femenino , Fijadores/química , Colorantes Fluorescentes/análisis , Larva/anatomía & histología , Larva/crecimiento & desarrollo , Lípidos/análisis , Masculino , Microscopía Fluorescente , Pez Cebra/fisiología
14.
Dev Biol ; 317(2): 508-22, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18417109

RESUMEN

The Pax3/7 gene family has a fundamental and conserved role during neural crest formation. In people, PAX3 mutation causes Waardenburg syndrome, and murine Pax3 is essential for pigment formation. However, it is unclear exactly how Pax3 functions within the neural crest. Here we show that pax3 is expressed before other pax3/7 members, including duplicated pax3b, pax7 and pax7b genes, early in zebrafish neural crest development. Knockdown of Pax3 protein by antisense morpholino oligonucleotides results in defective fate specification of xanthophores, with complete ablation in the trunk. Other pigment lineages are specified and differentiate. As a consequence of xanthophore loss, expression of pax7, a marker of the xanthophore lineage, is reduced in neural crest. Morpholino knockdown of Pax7 protein shows that Pax7 itself is dispensable for xanthophore fate specification, although yellow pigmentation is reduced. Loss of xanthophores after reduction of Pax3 correlates with a delay in melanoblast differentiation followed by significant increase in melanophores, suggestive of a Pax3-driven fate switch within a chromatophore precursor or stem cell. Analysis of other neural crest derivatives reveals that, in the absence of Pax3, the enteric nervous system is ablated from its inception. Therefore, Pax3 in zebrafish is required for specification of two specific lineages of neural crest, xanthophores and enteric neurons.


Asunto(s)
Sistema Nervioso Entérico/embriología , Cresta Neural/embriología , Factor de Transcripción PAX7/metabolismo , Factores de Transcripción Paired Box/metabolismo , Pigmentos Biológicos/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Teorema de Bayes , Western Blotting , Inmunohistoquímica , Hibridación in Situ , Oligonucleótidos/genética , Factor de Transcripción PAX3 , Filogenia
15.
Dev Biol ; 302(2): 504-21, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17094960

RESUMEN

Pax3/7 paired homeodomain transcription factors are important markers of muscle stem cells. Pax3 is required upstream of myod for lateral dermomyotomal cells in the amniote somite to form particular muscle cells. Later Pax3/7-dependent cells generate satellite cells and most body muscle. Here we analyse early myogenesis from, and regulation of, a population of Pax3-expressing dermomyotome-like cells in the zebrafish. Zebrafish pax3 is widely expressed in the lateral somite and, along with pax7, becomes restricted anteriorly and then to the external cells on the lateral somite surface. Midline-derived Hedgehog signals appear to act directly on lateral somite cells to repress Pax3/7. Both Hedgehog and Fgf8, signals that induce muscle formation within the somite, suppress Pax3/7 and promote expression of myogenic regulatory factors (MRFs) myf5 and myod in specific muscle precursor cell populations. Loss of MRF function leads to loss of myogenesis by specific populations of muscle fibres, with parallel up-regulation of Pax3/7. Myod is required for lateral fast muscle differentiation from pax3-expressing cells. In contrast, either Myf5 or Myod is sufficient to promote slow muscle formation from adaxial cells. Thus, myogenic signals act to drive somite cells to a myogenic fate through up-regulation of distinct combinations of MRFs. Our data show that the relationship between Pax3/7 genes and myogenesis is evolutionarily ancient, but that changes in the MRF targets for particular signals contribute to myogenic differences between species.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Hedgehog/fisiología , Proteína MioD/fisiología , Factor 5 Regulador Miogénico/fisiología , Factor de Transcripción PAX7/biosíntesis , Factores de Transcripción Paired Box/biosíntesis , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Proliferación Celular , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos , Fibras Musculares Esqueléticas/fisiología , Mutación , Cresta Neural/embriología , Cresta Neural/metabolismo , Factor de Transcripción PAX3 , Factor de Transcripción PAX7/genética , Factores de Transcripción Paired Box/genética , Transducción de Señal , Somitos/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...