Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Therm Biol ; 89: 102572, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32364964

RESUMEN

Rising environmental temperatures have become a global threat for ectotherms, with the increasing risk of overheating promoting population declines. Flexible thermoregulatory behavior might be a plausible mechanism to mitigate the effects of extreme temperatures. We experimentally evaluated thermoregulatory behavior in the bunchgrass lizard, Sceloporus aeneus, at three different environmental temperatures (25, 35 and 45 °C) both with and without a thermal refuge. We recorded themoregulatory behaviors (body posture and movement between hot and cold patches) and compared individual lizards across all experimental temperature and shelter combinations. Behavioral thermoregulation in S. aeneus was characterized by the expression of five body postures, whose frequencies varied based on environmental temperature and microthermal conditions. Behavioral responses allowed lizards to maintain a mean body temperature <40 °C, the critical thermal maximum for temperate species, even at extreme environmental temperatures (45 °C). Although S. aeneus express an array of behavioral postures that provide an effective mechanism to cope with elevating temperatures, the presence of a thermal refuge was important to better achieve this. Together, our study offers a novel method to evaluate microhabitat preference that encompasses both behavioral observations and time-space analysis based on the ambient thermal distribution, a consideration that can aid in the formulation of more accurate predictions on ectotherm vulnerability related to increasing global environmental temperatures.


Asunto(s)
Distribución Animal , Lagartos/fisiología , Termotolerancia , Animales , Temperatura Corporal , Ecosistema , Movimiento , Postura
2.
J Leukoc Biol ; 107(2): 237-250, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31487076

RESUMEN

Macrophage colony-stimulating factor (CSF1 or M-CSF) and interleukin 34 (IL34) are secreted cytokines that control macrophage survival and differentiation. Both act through the CSF1 receptor (CSF1R), a type III transmembrane receptor tyrosine kinase. The functions of CSF1R and both ligands are conserved in birds. We have analyzed protein-coding sequence divergence among avian species. The intracellular tyrosine kinase domain of CSF1R was highly conserved in bird species as in mammals but the extracellular domain of avian CSF1R was more divergent in birds with multiple positively selected amino acids. Based upon crystal structures of the mammalian CSF1/IL34 receptor-ligand interfaces and structure-based alignments, we identified amino acids involved in avian receptor-ligand interactions. The contact amino acids in both CSF1 and CSF1R diverged among avian species. Ligand-binding domain swaps between chicken and zebra finch CSF1 confirmed the function of variants that confer species specificity on the interaction of CSF1 with CSF1R. Based upon genomic sequence analysis, we identified prevalent amino acid changes in the extracellular domain of CSF1R even within the chicken species that distinguished commercial broilers and layers and tropically adapted breeds. The rapid evolution in the extracellular domain of avian CSF1R suggests that at least in birds this ligand-receptor interaction is subjected to pathogen selection. We discuss this finding in the context of expression of CSF1R in antigen-sampling and antigen-presenting cells.


Asunto(s)
Evolución Biológica , Interleucinas/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Polimorfismo Genético , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Pollos , Interleucinas/genética , Ligandos , Factor Estimulante de Colonias de Macrófagos/genética , Filogenia , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Especificidad de la Especie , Pez Cebra
3.
BMC Biol ; 13: 12, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25857347

RESUMEN

BACKGROUND: Macrophages have many functions in development and homeostasis as well as innate immunity. Recent studies in mammals suggest that cells arising in the yolk sac give rise to self-renewing macrophage populations that persist in adult tissues. Macrophage proliferation and differentiation is controlled by macrophage colony-stimulating factor (CSF1) and interleukin 34 (IL34), both agonists of the CSF1 receptor (CSF1R). In the current manuscript we describe the origin, function and regulation of macrophages, and the role of CSF1R signaling during embryonic development, using the chick as a model. RESULTS: Based upon RNA-sequencing comparison to bone marrow-derived macrophages grown in CSF1, we show that embryonic macrophages contribute around 2% of the total embryo RNA in day 7 chick embryos, and have similar gene expression profiles to bone marrow-derived macrophages. To explore the origins of embryonic and adult macrophages, we injected Hamburger-Hamilton stage 16 to 17 chick embryos with either yolk sac-derived blood cells, or bone marrow cells from EGFP+ donors. In both cases, the transferred cells gave rise to large numbers of EGFP+ tissue macrophages in the embryo. In the case of the yolk sac, these cells were not retained in hatched birds. Conversely, bone marrow EGFP+ cells gave rise to tissue macrophages in all organs of adult birds, and regenerated CSF1-responsive marrow macrophage progenitors. Surprisingly, they did not contribute to any other hematopoietic lineage. To explore the role of CSF1 further, we injected embryonic or hatchling CSF1R-reporter transgenic birds with a novel chicken CSF1-Fc conjugate. In both cases, the treatment produced a large increase in macrophage numbers in all tissues examined. There were no apparent adverse effects of chicken CSF1-Fc on embryonic or post-hatch development, but there was an unexpected increase in bone density in the treated hatchlings. CONCLUSIONS: The data indicate that the yolk sac is not the major source of macrophages in adult birds, and that there is a macrophage-restricted, self-renewing progenitor cell in bone marrow. CSF1R is demonstrated to be limiting for macrophage development during development in ovo and post-hatch. The chicken provides a novel and tractable model to study the development of the mononuclear phagocyte system and CSF1R signaling.


Asunto(s)
Pollos/inmunología , Sistema Mononuclear Fagocítico/embriología , Sistema Mononuclear Fagocítico/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal , Animales , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Densidad Ósea/efectos de los fármacos , Células de la Médula Ósea , Diferenciación Celular/efectos de los fármacos , Línea Celular , Embrión de Pollo , Pollos/genética , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/farmacología , Sistema Mononuclear Fagocítico/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Saco Vitelino/citología
4.
Dev Biol ; 402(1): 61-71, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25796573

RESUMEN

Klhl31 is a member of the Kelch-like family in vertebrates, which are characterized by an amino-terminal broad complex tram-track, bric-a-brac/poxvirus and zinc finger (BTB/POZ) domain, carboxy-terminal Kelch repeats and a central linker region (Back domain). In developing somites Klhl31 is highly expressed in the myotome downstream of myogenic regulators (MRF), and it remains expressed in differentiated skeletal muscle. In vivo gain- and loss-of-function approaches in chick embryos reveal a role of Klhl31 in skeletal myogenesis. Targeted mis-expression of Klhl31 led to a reduced size of dermomyotome and myotome as indicated by detection of relevant myogenic markers, Pax3, Myf5, myogenin and myosin heavy chain (MF20). The knock-down of Klhl31 in developing somites, using antisense morpholinos (MO), led to an expansion of Pax3, Myf5, MyoD and myogenin expression domains and an increase in the number of mitotic cells in the dermomyotome and myotome. The mechanism underlying this phenotype was examined using complementary approaches, which show that Klhl31 interferes with ß-catenin dependent Wnt signaling. Klhl31 reduced the Wnt-mediated activation of a luciferase reporter in cultured cells. Furthermore, Klhl31 attenuated secondary axis formation in Xenopus embryos in response to Wnt1 or ß-catenin. Klhl31 mis-expression in the developing neural tube affected its dorso-ventral patterning and led to reduced dermomyotome and myotome size. Co-transfection of a Wnt3a expression vector with Klhl31 in somites or in the neural tube rescued the phenotype and restored the size of dermomyotome and myotome. Thus, Klhl31 is a novel modulator of canonical Wnt signaling, important for vertebrate myogenesis. We propose that Klhl31 acts in the myotome to support cell cycle withdrawal and differentiation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos/fisiología , Músculo Esquelético/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Proliferación Celular , Embrión de Pollo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Hibridación in Situ , Mitosis , Músculos/embriología , Miogenina/biosíntesis , Tubo Neural/metabolismo , Fenotipo , Transducción de Señal , Somitos/metabolismo , Xenopus laevis , beta Catenina/genética
5.
J Immunol ; 194(5): 2338-44, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25637020

RESUMEN

We have identified differences in gene expression in macrophages grown from the bone marrow of male and female chickens in recombinant chicken M-CSF (CSF1). Cells were profiled with or without treatment with bacterial LPS for 24 h. Approximately 600 transcripts were induced by prolonged LPS stimulation to an equal extent in the male and female macrophages. Many transcripts encoded on the Z chromosome were expressed ∼1.6-fold higher in males, reflecting a lack of dosage compensation in the homogametic sex. A smaller set of W chromosome-specific genes was expressed only in females. LPS signaling in mammals is associated with induction of type 1 IFN-responsive genes. Unexpectedly, because IFNs are encoded on the Z chromosome of chickens, unstimulated macrophages from the female birds expressed a set of known IFN-inducible genes at much higher levels than male cells under the same conditions. To confirm that these differences were not the consequence of the actions of gonadal hormones, we induced gonadal sex reversal to alter the hormonal environment of the developing chick and analyzed macrophages cultured from male, female, and female sex-reversed embryos. Gonadal sex reversal did not alter the sexually dimorphic expression of either sex-linked or IFN-responsive genes. We suggest that female birds compensate for the reduced dose of inducible IFN with a higher basal set point of IFN-responsive genes.


Asunto(s)
Proteínas Aviares/inmunología , Pollos/inmunología , Gónadas/inmunología , Macrófagos/inmunología , ARN Mensajero/inmunología , Cromosomas Sexuales/inmunología , Animales , Inhibidores de la Aromatasa/farmacología , Proteínas Aviares/genética , Células Cultivadas , Embrión de Pollo , Pollos/genética , Compensación de Dosificación (Genética) , Fadrozol/farmacología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Gónadas/efectos de los fármacos , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Lipopolisacáridos/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , ARN Mensajero/genética , Caracteres Sexuales
6.
Development ; 141(16): 3255-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25063453

RESUMEN

We have generated the first transgenic chickens in which reporter genes are expressed in a specific immune cell lineage, based upon control elements of the colony stimulating factor 1 receptor (CSF1R) locus. The Fms intronic regulatory element (FIRE) within CSF1R is shown to be highly conserved in amniotes and absolutely required for myeloid-restricted expression of fluorescent reporter genes. As in mammals, CSF1R-reporter genes were specifically expressed at high levels in cells of the macrophage lineage and at a much lower level in granulocytes. The cell lineage specificity of reporter gene expression was confirmed by demonstration of coincident expression with the endogenous CSF1R protein. In transgenic birds, expression of the reporter gene provided a defined marker for macrophage-lineage cells, identifying the earliest stages in the yolk sac, throughout embryonic development and in all adult tissues. The reporter genes permit detailed and dynamic visualisation of embryonic chicken macrophages. Chicken embryonic macrophages are not recruited to incisional wounds, but are able to recognise and phagocytose microbial antigens.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Macrófagos/citología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Aves , Linaje de la Célula , Pollos , Células Dendríticas/citología , Genes Reporteros , Técnicas Genéticas , Sistema Inmunológico , Intrones , Datos de Secuencia Molecular , Fagocitosis , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Transgenes , Saco Vitelino/fisiología
7.
Mol Cell Endocrinol ; 382(1): 150-158, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23994026

RESUMEN

Obesity is characterised by hyperleptinaemia and hypoadiponectinaemia and these metabolic abnormalities may contribute to the progression of several obesity-associated cancers including oesophageal adenocarcinoma (OAC). We have examined the effects of leptin and adiponectin on OE33 OAC cells. Leptin stimulated proliferation, invasion and migration and inhibited apoptosis in a STAT3-dependant manner. Leptin-stimulated MMP-2 secretion in a partly STAT3-dependent manner and MMP-9 secretion via a STAT3-independent pathway. Adiponectin inhibited leptin-induced proliferation, migration, invasion, MMP secretion and reduced the anti-apoptotic effects: these effects of adiponectin were ameliorated by both a non-specific tyrosine phosphatase inhibitor and a specific PTP1B inhibitor. Adiponectin reduced leptin-stimulated JAK2 activation and STAT3 transcriptional activity in a PTP1B-sensitive manner and adiponectin increased both PTP1B protein and activity. We conclude that adiponectin restrains leptin-induced signalling and pro-carcinogenic behaviour by inhibiting the early events in leptin-induced signal transduction by activating PTP1B. Relative adiponectin deficiency in obesity may contribute to the promotion of OAC.


Asunto(s)
Adiponectina/farmacología , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/patología , Leptina/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Janus Quinasa 2/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Modelos Biológicos , Invasividad Neoplásica , Factor de Transcripción STAT3/metabolismo
8.
Dev Comp Immunol ; 42(2): 278-85, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24084378

RESUMEN

Macrophages contribute to innate and acquired immunity as well as many aspects of homeostasis and development. Studies of macrophage biology and function in birds have been hampered by a lack of definitive cell surface markers. As in mammals, avian macrophages proliferate and differentiate in response to CSF1 and IL34, acting through the shared receptor, CSF1R. CSF1R mRNA expression in the chicken is restricted to macrophages and their progenitors. To expedite studies of avian macrophage biology, we produced an avian CSF1R-Fc chimeric protein and generated a monoclonal antibody (designated ROS-AV170) against the chicken CSF1R using the chimeric protein as immunogen. Specific binding of ROS-AV170 to CSF1R was confirmed by FACS, ELISA and immunohistochemistry on tissue sections. CSF1 down-regulated cell surface expression of the CSF1R detected with ROS-AV170, but the antibody did not block CSF1 signalling. Expression of CSF1R was detected on the surface of bone marrow progenitors only after culture in the absence of CSF1, and was induced during macrophage differentiation. Constitutive surface expression of CSF1R distinguished monocytes from other myeloid cells, including heterophils and thrombocytes. This antibody will therefore be of considerable utility for the study of chicken macrophage biology.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Pollos/inmunología , Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Animales , Células de la Médula Ósea/inmunología , Células CHO , Diferenciación Celular/inmunología , Línea Celular , Linaje de la Célula/inmunología , Cricetulus , Femenino , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Monocitos/inmunología , ARN Mensajero/biosíntesis , Receptor de Factor Estimulante de Colonias de Macrófagos/biosíntesis , Receptor de Factor Estimulante de Colonias de Macrófagos/genética
9.
Dev Biol ; 335(1): 143-55, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19716814

RESUMEN

Formation of the vertebrate nervous system requires coordinated cell-cell interactions, intracellular signalling events, gene transcription, and morphogenetic cell movements. Wnt signalling has been involved in regulating a wide variety of biological processes such as embryonic patterning, cell proliferation, cell polarity, motility, and the specification of cell fate. Wnt ligands associate with their receptors, members of the frizzled family (Fz). In Xenopus, five members of the frizzled family are expressed in the early nervous system. We have investigated the role of Xenopus frizzled-10 (Fz10) in neural development. We show that Fz10 is expressed in the dorsal neural ectoderm and neural folds in the region where primary sensory neurons develop. Fz10 mediates canonical Wnt signalling and interacts with Wnt1 and Wnt8 but not Wnt3a as shown in synergy assays. We find that Fz10 is required for the late stages of sensory neuron differentiation. Overexpression of Fz10 in Xenopus leads to an increase in the number of sensory neurons. Loss of Fz10 function using morpholinos inhibits the development of sensory neurons in Xenopus at later stages of neurogenesis and this can be rescued by co-injection of modified Fz10B and beta-catenin. In mouse P19 cells induced by retinoic acid to undergo neural differentiation, overexpression of Xenopus Fz10 leads to an increase in the number of neurons generated while siRNA knockdown of endogenous mouse Fz10 inhibits neurogenesis. Thus we propose Fz10 mediates Wnt1 signalling to determine sensory neural differentiation in Xenopus in vivo and in mouse cell culture.


Asunto(s)
Diferenciación Celular/fisiología , Receptores Frizzled/metabolismo , Neurogénesis/fisiología , Células Receptoras Sensoriales/fisiología , Transducción de Señal/fisiología , Proteínas de Xenopus/metabolismo , Xenopus laevis , Animales , Biomarcadores/metabolismo , Línea Celular , Receptores Frizzled/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Células Receptoras Sensoriales/citología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/anatomía & histología , Xenopus laevis/embriología , Xenopus laevis/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
10.
Chem Biol ; 16(1): 93-104, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19171309

RESUMEN

To dissect the function of matrix metalloproteinases (MMPs) involved in cellular migration in vivo, we undertook both a forward chemical genomic screen and a functional approach to discover modulators of melanophore (pigment cell) migration in Xenopus laevis. We identified the 8-quinolinol derivative NSC 84093 as affecting melanophore migration in the developing embryo and have shown it to act as a MMP inhibitor. Potential targets of NSC 84093 investigated include MMP-14 and MMP-2. MMP-14 is expressed in migrating neural crest cells from which melanophores are derived. MMP-2 is expressed at the relevant time of development and in a pattern that suggests it contributes to melanophore migration. Morpholino-mediated knockdown of both MMPs demonstrates they play a key role in melanophore migration and partially phenocopy the effect of NSC 84093.


Asunto(s)
Compuestos de Anilina/farmacología , Movimiento Celular , Hidroxiquinolinas/farmacología , Metaloproteinasas de la Matriz/metabolismo , Melanóforos/enzimología , Xenopus laevis/embriología , Compuestos de Anilina/química , Animales , Movimiento Celular/genética , Embrión no Mamífero/enzimología , Desarrollo Embrionario , Humanos , Hidroxiquinolinas/química , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Melanóforos/metabolismo , Pigmentación de la Piel , Relación Estructura-Actividad , Xenopus laevis/metabolismo
11.
Mech Dev ; 125(11-12): 1059-70, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18684398

RESUMEN

Macrophages are essential in development, repair and pathology of a variety of tissues via their roles in tissue remodelling, wound healing and inflammation. These biological functions are also associated with a number of human diseases, for example tumour associated macrophages have well defined functions in cancer progression. Xenopus embryonic macrophages arise from a haematopoietic stem cell population by direct differentiation and act as the main mechanism of host defence, before lymphoid cells and a circulatory system have developed. This function is conserved in mouse and human development. Macrophages express a number of matrix metalloproteinases (MMPs), which are central to their function. MMPs are a large family of zinc-dependent endoproteases with multiple roles in extracellular matrix remodelling and the modulation of signalling pathways. We have previously shown MMP-7 to be expressed by Xenopus embryonic macrophages. Here we investigate the role of MMP-7 and two other MMPs (MMP-18 and MMP-9) that are also expressed in the migrating macrophages. Using morpholino (MO) mediated knockdown of each of the MMPs we demonstrate that they are necessary for normal macrophage migration in vivo. The loss-of-function effect can be rescued using the specific MMPs, altered to be resistant to morpholinos but not by overexpression of the other MMPs. Double and triple morpholino knockdowns further suggest that these MMPs act combinatorily to promote embryonic macrophage migration. Thus, our results imply that these three MMPs have distinct functions, which together are crucial to mediate macrophage migration in the developing embryo. This demonstrates conclusively that MMPs are required for normal macrophage cell migration in the whole organism.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Macrófagos/citología , Metaloproteinasas de la Matriz/fisiología , Animales , Movimiento Celular , Matriz Extracelular/metabolismo , Técnicas Genéticas , Corazón/embriología , Macrófagos/enzimología , Macrófagos/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz Secretadas/metabolismo , Modelos Genéticos , Sistemas de Lectura Abierta , Temperatura , Xenopus laevis
12.
Dev Biol ; 298(1): 285-98, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16928367

RESUMEN

The neural crest is a multipotent cell population that migrates from the dorsal edge of the neural tube to various parts of the embryo where it differentiates into a remarkable variety of different cell types. Initial induction of neural crest is mediated by a combination of BMP, Wnt, FGF, Retinoic acid and Notch/Delta signaling. The two-signal model for neural crest induction suggests that BMP signaling induces the competence to become neural crest. The second signal involves Wnt acting through the canonical pathway and leads to expression of neural crest markers such as slug. Wnt signals from the neural plate, non-neural ectoderm and paraxial mesoderm have all been suggested to play a role in neural crest induction. We show that Xenopus frizzled7 (Xfz7) is expressed in the dorsal ectoderm including early neural crest progenitors and is a key mediator of the Wnt inductive signal. We demonstrate that Xfz7 expression is induced in response to a BMP antagonist, noggin, and that Xfz7 can induce neural crest specific genes in noggin-treated ectodermal explants (animal caps). Morpholino-mediated or dominant negative inhibition of Xfz7 inhibits Wnt induced Xslug expression in the animal cap assay and in the whole embryo leading to a loss of neural crest derived pigment cells. Full-length Xfz7 rescues the morpholino-induced phenotype, as does activated beta-catenin, suggesting that Xfz7 is signaling through the canonical pathway. We therefore demonstrate that Xfz7 is regulated by BMP antagonism and is required for neural crest induction by Wnt in the developing vertebrate embryo.


Asunto(s)
Tipificación del Cuerpo , Cresta Neural/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Proteínas Wnt/metabolismo , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Animales , Secuencia de Bases , Embrión no Mamífero , Datos de Secuencia Molecular , Cresta Neural/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA