Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36975217

RESUMEN

Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.


Asunto(s)
Técnicas de Transferencia de Gen , Pez Cebra , Animales , Ratones , Pez Cebra/genética , Pez Cebra/metabolismo , Animales Modificados Genéticamente , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Elementos Transponibles de ADN/genética
2.
PLoS Genet ; 17(5): e1009579, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34033651

RESUMEN

We sought to understand how perturbation of signaling pathways and their targets generates variable phenotypes. In humans, GATA3 associates with highly variable defects, such as HDR syndrome, microsomia and choanal atresia. We previously characterized a zebrafish point mutation in gata3 with highly variable craniofacial defects to the posterior palate. This variability could be due to residual Gata3 function, however, we observe the same phenotypic variability in gata3 null mutants. Using hsp:GATA3-GFP transgenics, we demonstrate that Gata3 function is required between 24 and 30 hpf. At this time maxillary neural crest cells fated to generate the palate express gata3. Transplantation experiments show that neural crest cells require Gata3 function for palatal development. Via a candidate approach, we determined if Bmp signaling was upstream of gata3 and if this pathway explained the mutant's phenotypic variation. Using BRE:d2EGFP transgenics, we demonstrate that maxillary neural crest cells are Bmp responsive by 24 hpf. We find that gata3 expression in maxillary neural crest requires Bmp signaling and that blocking Bmp signaling, in hsp:DN-Bmpr1a-GFP embryos, can phenocopy gata3 mutants. Palatal defects are rescued in hsp:DN-Bmpr1a-GFP;hsp:GATA3-GFP double transgenic embryos, collectively demonstrating that gata3 is downstream of Bmp signaling. However, Bmp attenuation does not alter phenotypic variability in gata3 loss-of-function embryos, implicating a different pathway. Due to phenotypes observed in hypomorphic shha mutants, the Sonic Hedgehog (Shh) pathway was a promising candidate for this pathway. Small molecule activators and inhibitors of the Shh pathway lessen and exacerbate, respectively, the phenotypic severity of gata3 mutants. Importantly, inhibition of Shh can cause gata3 haploinsufficiency, as observed in humans. We find that gata3 mutants in a less expressive genetic background have a compensatory upregulation of Shh signaling. These results demonstrate that the level of Shh signaling can modulate the phenotypes observed in gata3 mutants.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Factor de Transcripción GATA3/genética , Proteínas Hedgehog/metabolismo , Fenotipo , Transducción de Señal , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Factor de Transcripción GATA3/metabolismo , Haploinsuficiencia , Mutación con Pérdida de Función , Mutación , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Organogénesis , Cráneo/citología , Cráneo/embriología , Pez Cebra/embriología
3.
Genesis ; 59(11): e23460, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34739740

RESUMEN

Fetal alcohol spectrum disorder (FASD) describes a wide range of structural deficits and cognitive impairments. FASD impacts up to 5% of children born in the United States each year, making ethanol one of the most common teratogens. Due to limitations and ethical concerns, studies in humans are limited in their ability to study FASD. Animal models have proven critical in identifying and characterizing the mechanisms underlying FASD. In this review, we will focus on the attributes of zebrafish that make it a strong model in which to study ethanol-induced developmental defects. Zebrafish have several attributes that make it an ideal model in which to study FASD. Zebrafish produced large numbers of externally fertilized, translucent embryos. With a high degree of genetic amenability, zebrafish are at the forefront of identifying and characterizing the gene-ethanol interactions that underlie FASD. Work from multiple labs has shown that embryonic ethanol exposures result in defects in craniofacial, cardiac, ocular, and neural development. In addition to structural defects, ethanol-induced cognitive and behavioral impairments have been studied in zebrafish. Building upon these studies, work has identified ethanol-sensitive loci that underlie the developmental defects. However, analyses show there is still much to be learned of these gene-ethanol interactions. The zebrafish is ideally suited to expand our understanding of gene-ethanol interactions and their impact on FASD. Because of the conservation of gene function between zebrafish and humans, these studies will directly translate to studies of candidate genes in human populations and allow for better diagnosis and treatment of FASD.


Asunto(s)
Modelos Animales de Enfermedad , Trastornos del Espectro Alcohólico Fetal/genética , Pez Cebra/genética , Animales , Trastornos del Espectro Alcohólico Fetal/metabolismo , Trastornos del Espectro Alcohólico Fetal/patología , Pez Cebra/metabolismo
4.
Development ; 143(11): 2000-11, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27122171

RESUMEN

The endodermal pouches are a series of reiterated structures that segment the pharyngeal arches and help pattern the vertebrate face. Multiple pathways regulate the complex process of endodermal development, including the Bone morphogenetic protein (Bmp) pathway. However, the role of Bmp signaling in pouch morphogenesis is poorly understood. Using genetic and chemical inhibitor approaches, we show that pouch morphogenesis requires Bmp signaling from 10-18 h post-fertilization, immediately following gastrulation. Blocking Bmp signaling during this window results in morphological defects to the pouches and craniofacial skeleton. Using genetic chimeras we show that Bmp signals directly to the endoderm for proper morphogenesis. Time-lapse imaging and analysis of reporter transgenics show that Bmp signaling is necessary for pouch outpocketing via the Fibroblast growth factor (Fgf) pathway. Double loss-of-function analyses demonstrate that Bmp and Fgf signaling interact synergistically in craniofacial development. Collectively, our analyses shed light on the tissue and signaling interactions that regulate development of the vertebrate face.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Endodermo/embriología , Factores de Crecimiento de Fibroblastos/metabolismo , Morfogénesis , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Recuento de Células , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Cara/embriología , Morfogénesis/efectos de los fármacos , Cresta Neural/efectos de los fármacos , Cresta Neural/metabolismo , Cresta Neural/patología , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/patología , Factores de Tiempo
5.
Dev Dyn ; 245(6): 641-52, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26971580

RESUMEN

BACKGROUND: One of the most prevalent congenital birth defects is cleft palate. The palatal skeleton is derived from the cranial neural crest and platelet-derived growth factors (Pdgf) are critical in palatogenesis. Of the two Pdgf receptors, pdgfra is required for neural crest migration and palatogenesis. However, the role pdgfrb plays in the neural crest, or whether pdgfra and pdgfrb interact during palatogenesis is unclear. RESULTS: We find that pdgfrb is dispensable for craniofacial development in zebrafish. However, the palatal defect in pdgfra;pdgfrb double mutants is significantly more severe than in pdgfra single mutants. Data in mouse suggest this interaction is conserved and that neural crest requires both genes. In zebrafish, pdgfra and pdgfrb are both expressed by neural crest within the pharyngeal arches, and pharmacological analyses demonstrate Pdgf signaling is required at these times. While neither proliferation nor cell death appears affected, time-lapsed confocal analysis of pdgfra;pdgfrb mutants shows a failure of proper neural crest condensation during palatogenesis. CONCLUSIONS: We provide data showing that pdgfra and pdgfrb interact during palatogenesis in both zebrafish and mouse. In zebrafish, this interaction affects proper condensation of maxillary neural crest cells, revealing a previously unknown interaction between Pdgfra and Pdgfrb during palate formation. Developmental Dynamics 245:641-652, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Fisura del Paladar/embriología , Fisura del Paladar/genética , Fisura del Paladar/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Cresta Neural/embriología , Cresta Neural/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Development ; 140(15): 3254-65, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23861062

RESUMEN

Human birth defects are highly variable and this phenotypic variability can be influenced by both the environment and genetics. However, the synergistic interactions between these two variables are not well understood. Fetal alcohol spectrum disorders (FASD) is the umbrella term used to describe the wide range of deleterious outcomes following prenatal alcohol exposure. Although FASD are caused by prenatal ethanol exposure, FASD are thought to be genetically modulated, although the genes regulating sensitivity to ethanol teratogenesis are largely unknown. To identify potential ethanol-sensitive genes, we tested five known craniofacial mutants for ethanol sensitivity: cyp26b1, gata3, pdgfra, smad5 and smoothened. We found that only platelet-derived growth factor receptor alpha (pdgfra) interacted with ethanol during zebrafish craniofacial development. Analysis of the PDGF family in a human FASD genome-wide dataset links PDGFRA to craniofacial phenotypes in FASD, prompting a mechanistic understanding of this interaction. In zebrafish, untreated pdgfra mutants have cleft palate due to defective neural crest cell migration, whereas pdgfra heterozygotes develop normally. Ethanol-exposed pdgfra mutants have profound craniofacial defects that include the loss of the palatal skeleton and hypoplasia of the pharyngeal skeleton. Furthermore, ethanol treatment revealed latent haploinsufficiency, causing palatal defects in ∼62% of pdgfra heterozygotes. Neural crest apoptosis partially underlies these ethanol-induced defects in pdgfra mutants, demonstrating a protective role for Pdgfra. This protective role is mediated by the PI3K/mTOR pathway. Collectively, our results suggest a model where combined genetic and environmental inhibition of PI3K/mTOR signaling leads to variability within FASD.


Asunto(s)
Anomalías Craneofaciales/prevención & control , Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/prevención & control , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/anomalías , Animales , Animales Modificados Genéticamente , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Fisura del Paladar/etiología , Fisura del Paladar/genética , Anomalías Craneofaciales/etiología , Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Trastornos del Espectro Alcohólico Fetal/genética , Interacción Gen-Ambiente , Heterocigoto , Humanos , Mutación , Cresta Neural/anomalías , Cresta Neural/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
7.
Alcohol Clin Exp Res ; 38(8): 2160-3, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25156611

RESUMEN

BACKGROUND: Due to its profound impact on human development, ethanol (EtOH) teratogenicity is a field of intense study. The complexity of variables that influence the outcomes of embryonic or prenatal EtOH exposure compels the use of animal models in which these variables can be isolated. METHODS: Numerous model systems have been used in these studies. The zebrafish is a powerful model system, which has seen a recent increase in usage for EtOH studies. RESULTS: Those using zebrafish for alcohol studies often face 2 questions: (i) How physiologically relevant are the doses of EtOH administered to zebrafish embryos? and (ii) Will the mechanisms of EtOH teratogenesis be conserved to other model systems and human? CONCLUSIONS: The current article by Flentke and colleagues () helps to shed important light on these questions and clearly demonstrates that the zebrafish will be a valuable model system with which to understand EtOH teratogenicity.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal/etiología , Síndromes de Neurotoxicidad/embriología , Animales
8.
bioRxiv ; 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37425959

RESUMEN

Fetal Alcohol Spectrum Disorders (FASD) describe ethanol-induced developmental defects including craniofacial malformations. While ethanol-sensitive genetic mutations contribute to facial malformations, the impacted cellular mechanisms remain unknown. Bmp signaling is a key regulator of epithelial morphogenesis driving facial development, providing a possible ethanol-sensitive mechanism. We found that zebrafish mutants for Bmp signaling components are ethanol-sensitive and affect anterior pharyngeal endoderm shape and gene expression, indicating ethanol-induced malformations of the anterior pharyngeal endoderm cause facial malformations. Integrating FASD patient data, we provide the first evidence that variants in the human Bmp receptor gene BMPR1B associate with ethanol-related differences in jaw volume. Our results show that ethanol exposure disrupts proper morphogenesis of, and tissue interactions between, facial epithelia that mirror overall viscerocranial shape changes and are predictive for Bmp-ethanol associations in human jaw development. Our data provide a mechanistic paradigm linking ethanol to disrupted epithelial cell behaviors that underlie facial defects in FASD.

9.
Eukaryot Cell ; 10(7): 869-83, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21622903

RESUMEN

The phytopathogenic fungus Ustilago maydis undergoes a dimorphic transition in response to mating pheromone, host, and environmental cues. On a solid medium deficient in ammonium (SLAD [0.17% yeast nitrogen base without ammonium sulfate or amino acids, 2% dextrose, 50 µM ammonium sulfate]), U. maydis produces a filamentous colony morphology, while in liquid SLAD, the cells do not form filaments. The p21-activated protein kinases (PAKs) play a substantial role in regulating the dimorphic transition in fungi. The PAK-like Ste20 homologue Smu1 is required for a normal response to pheromone, via upregulation of pheromone expression, and virulence, and its disruption affects both processes. Our experiments suggest that Smu1 also regulates cell length and the filamentous response on solid SLAD medium. Yeast two-hybrid analysis suggested an Hsl7 homologue as a potential interacting partner of Smu1, and a unique open reading frame for such an arginine methyltransferase was detected in the U. maydis genome sequence. Hsl7 regulates cell length and the filamentous response to solid SLAD in a fashion opposite to that of Smu1, but neither overexpression nor disruption of hsl7 attenuates virulence. Simultaneous disruption of hsl7 and overexpression of smu1 lead to a hyperfilamentous response on solid SLAD. Moreover, only this double mutant strain forms filaments in liquid SLAD. The double mutant strain was also significantly reduced in virulence. A similar filamentous response in both solid and liquid SLAD was observed in strains lacking another PAK-like protein kinase involved in cytokinesis and polar growth, Cla4. Our data suggest that Hsl7 may regulate cell cycle progression, while both Smu1 and Cla4 appear to be involved in the filamentous response in U. maydis.


Asunto(s)
Transducción de Señal , Ustilago/enzimología , Quinasas p21 Activadas/metabolismo , Secuencia de Aminoácidos , Medios de Cultivo/química , Citoesqueleto/metabolismo , Eliminación de Gen , Técnicas de Inactivación de Genes , Genes Fúngicos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Feromonas/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Compuestos de Amonio Cuaternario/química , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos , Ustilago/genética , Ustilago/crecimiento & desarrollo , Ustilago/patogenicidad
10.
J Vis Exp ; (189)2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36533820

RESUMEN

Fetal alcohol spectrum disorders (FASD) are characterized by a highly variable set of structural defects and cognitive impairments that arise due to prenatal ethanol exposure. Due to the complex pathology of FASD, animal models have proven critical to our current understanding of ethanol-induced developmental defects. Zebrafish have proven to be a powerful model to examine ethanol-induced developmental defects due to the high degree of conservation of both genetics and development between zebrafish and humans. As a model system, zebrafish possess many attributes that make them ideal for developmental studies, including large numbers of externally fertilized embryos that are genetically tractable and translucent. This allows researchers to precisely control the timing and dosage of ethanol exposure in multiple genetic contexts. One important genetic tool available in zebrafish is transgenesis. However, generating transgenic constructs and establishing transgenic lines can be complex and difficult. To address this issue, zebrafish researchers have established the transposon-based Tol2 transgenesis system. This modular system uses a multisite Gateway cloning approach for the quick assembly of complete Tol2 transposon-based transgenic constructs. Here, we describe the flexible Tol2 system toolbox and a protocol for generating transgenic constructs ready for zebrafish transgenesis and their use in ethanol studies.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Pez Cebra , Animales , Femenino , Humanos , Embarazo , Pez Cebra/genética , Técnicas de Transferencia de Gen , Animales Modificados Genéticamente/genética , Etanol , Elementos Transponibles de ADN
11.
J Vis Exp ; (156)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32116298

RESUMEN

Fetal Alcohol Spectrum Disorders (FASD) describe a highly variable continuum of ethanol-induced developmental defects, including facial dysmorphologies and neurological impairments. With a complex pathology, FASD affects approximately 1 in 100 children born in the United States each year. Due to the highly variable nature of FASD, animal models have proven critical in our current mechanistic understanding of ethanol-induced development defects. An increasing number of laboratories has focused on using zebrafish to examine ethanol-induced developmental defects. Zebrafish produce large numbers of externally fertilized, genetically tractable, translucent embryos. This allows researchers to precisely control timing and dosage of ethanol exposure in multiple genetic contexts and quantify the impact of embryonic ethanol exposure through live imaging techniques. This, combined with the high degree of conservation of both genetics and development with humans, has proven zebrafish to be a powerful model in which to study the mechanistic basis of ethanol teratogenicity. However, ethanol exposure regimens have varied between different zebrafish studies, which has confounded the interpretation of zebrafish data across these studies. Here is a protocol to quantify ethanol concentrations in zebrafish embryos using head space gas chromatography.


Asunto(s)
Cromatografía de Gases/métodos , Embrión no Mamífero/efectos de los fármacos , Etanol/metabolismo , Pez Cebra/embriología , Animales , Modelos Animales de Enfermedad , Femenino , Embarazo
12.
J Vis Exp ; (83): e51190, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24514435

RESUMEN

Time-lapse imaging is a technique that allows for the direct observation of the process of morphogenesis, or the generation of shape. Due to their optical clarity and amenability to genetic manipulation, the zebrafish embryo has become a popular model organism with which to perform time-lapse analysis of morphogenesis in living embryos. Confocal imaging of a live zebrafish embryo requires that a tissue of interest is persistently labeled with a fluorescent marker, such as a transgene or injected dye. The process demands that the embryo is anesthetized and held in place in such a way that healthy development proceeds normally. Parameters for imaging must be set to account for three-dimensional growth and to balance the demands of resolving individual cells while getting quick snapshots of development. Our results demonstrate the ability to perform long-term in vivo imaging of fluorescence-labeled zebrafish embryos and to detect varied tissue behaviors in the cranial neural crest that cause craniofacial abnormalities. Developmental delays caused by anesthesia and mounting are minimal, and embryos are unharmed by the process. Time-lapse imaged embryos can be returned to liquid medium and subsequently imaged or fixed at later points in development. With an increasing abundance of transgenic zebrafish lines and well-characterized fate mapping and transplantation techniques, imaging any desired tissue is possible. As such, time-lapse in vivo imaging combines powerfully with zebrafish genetic methods, including analyses of mutant and microinjected embryos.


Asunto(s)
Huesos Faciales/embriología , Microscopía Confocal/métodos , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Anomalías Craneofaciales/genética , Morfogénesis/genética , Morfogénesis/fisiología , Cresta Neural/embriología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores Acoplados a Proteínas G/genética , Proteína Smad5/genética , Imagen de Lapso de Tiempo/métodos , Pez Cebra/genética , Proteínas de Pez Cebra/genética
13.
Alcohol ; 48(6): 595-602, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25012627

RESUMEN

Fetal Alcohol Spectrum Disorders (FASD) describes a wide range of phenotypic defects affecting facial and neurological development associated with ethanol teratogenicity. It affects approximately 1 in 100 children born in the United States each year. Genetic predisposition along with timing and dosage of ethanol exposure are critical in understanding the prevalence and variability of FASD. The zebrafish attributes of external fertilization, genetic tractability, and high fecundity make it a powerful tool for FASD studies. However, a lack of consensus of ethanol treatment paradigms has limited the interpretation of these various studies. Here we address this concern by examining ethanol tissue concentrations across timing and genetic background. We utilize headspace gas chromatography to determine ethanol concentration in the AB, fli1:EGFP, and Tu backgrounds. In addition, we treated these embryos with ethanol over two different developmental time windows, 6-24 h post fertilization (hpf) and 24-48 hpf. Our analysis demonstrates that embryos rapidly equilibrate to a sub-media level of ethanol. Embryos then maintain this level of ethanol for the duration of exposure. The ethanol tissue concentration level is independent of genetic background, but is timing-dependent. Embryos exposed from 6 to 24 hpf were 2.7-4.2-fold lower than media levels, while embryos were 5.7-6.2-fold lower at 48 hpf. This suggests that embryos strengthen one or more barriers to ethanol as they develop. In addition, both the embryo and, to a lesser extent, the chorion, surrounding the embryo are barriers to ethanol. Overall, this work will help tighten ethanol treatment regimens and strengthen zebrafish as a model of FASD.


Asunto(s)
Etanol/metabolismo , Pez Cebra/metabolismo , Animales , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Trastornos del Espectro Alcohólico Fetal/etiología , Pez Cebra/embriología
14.
PLoS One ; 8(3): e59533, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555697

RESUMEN

In human, mutation of the transcription factor SATB2 causes severe defects to the palate and jaw. The expression and sequence of SATB2 is highly conserved across vertebrate species, including zebrafish. We sought to understand the regulation of satb2 using the zebrafish model system. Due to the normal expression domains of satb2, we analyzed satb2 expression in mutants with disrupted Hh signaling or defective ventral patterning. While satb2 expression appears independent of Edn1 signaling, appropriate expression requires Shha, Smo, Smad5 and Hand2 function. Transplantation experiments show that neural crest cells receive both Bmp and Hh signaling to induce satb2 expression. Dorsomorphin- and cyclopamine-mediated inhibition of Bmp and Hh signaling, respectively, suggests that proper satb2 expression requires a relatively earlier Bmp signal and a later Hh signal. We propose that Bmp signaling establishes competence for the neural crest to respond to Hh signaling, thus inducing satb2 expression.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Región Branquial/citología , Región Branquial/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Región Branquial/embriología , Endodermo/citología , Endodermo/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Pez Cebra/embriología
15.
Commun Integr Biol ; 4(6): 670-3, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22446524

RESUMEN

Ustilago maydis, the fungal pathogen of maize, undergoes a dimorphic transition from budding yeast-like growth to filamentous growth, both as part of its program for pathogenesis and distinctly, in response to environmental cues, such as acid pH or low nitrogen availability. Smu1 is a p21-activated protein kinase (PAK) with roles in both the mating response required for the former function, as well as for the nutrient response. Hsl7 may be a negative regulator of Smu1 and appears to play a role in cell length and cell cycle.  Additional proteins that participate in cell polarity and filamentation pathways include the small G protein, Rac1, and its effector PAK kinase, Cla4. Here we describe further experiments that explore the roles of Cla4 and Rac1 in the response to nitrogen availability. While deletion of rac1severely delays filamentous growth on solid media low in ammonium (SLAD), we found that deletion of cla4 does not abolish filamentous cell morphology on solid SLAD. Unexpectedly, however, the Dcla4 mutants also filament in liquid SLAD. The filamentous cell morphology of the cla4 mutant in liquid SLAD has only been seen previously for one other mutant, a strain deleted for hsl7 that simultaneously over-expresses smu1. 

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA