Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 151(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39092607

RESUMEN

Branching morphogenesis is a characteristic feature of many essential organs, such as the lung and kidney, and most glands, and is the net result of two tissue behaviors: branch point initiation and elongation. Each branched organ has a distinct architecture customized to its physiological function, but how patterning occurs in these ramified tubular structures is a fundamental problem of development. Here, we use quantitative 3D morphometrics, time-lapse imaging, manipulation of ex vivo cultured mouse embryonic organs and mice deficient in the planar cell polarity component Vangl2 to address this question in the developing mammary gland. Our results show that the embryonic epithelial trees are highly complex in topology owing to the flexible use of two distinct modes of branch point initiation: lateral branching and tip bifurcation. This non-stereotypy was contrasted by the remarkably constant average branch frequency, indicating a ductal growth invariant, yet stochastic, propensity to branch. The probability of branching was malleable and could be tuned by manipulating the Fgf10 and Tgfß1 pathways. Finally, our in vivo data and ex vivo time-lapse imaging suggest the involvement of tissue rearrangements in mammary branch elongation.


Asunto(s)
Glándulas Mamarias Animales , Morfogénesis , Animales , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Femenino , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Factor de Crecimiento Transformador beta1/metabolismo , Imagen de Lapso de Tiempo , Polaridad Celular , Embrión de Mamíferos/metabolismo , Transducción de Señal
2.
Development ; 150(22)2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37982496

RESUMEN

Tissue interactions are essential for guiding organ development and regeneration. Hair follicle formation relies on inductive signalling between two tissues, the embryonic surface epithelium and the adjacent mesenchyme. Although previous research has highlighted the hair-inducing potential of the mesenchymal component of the hair follicle - the dermal papilla and its precursor, the dermal condensate - the source and nature of the primary inductive signal before dermal condensate formation have remained elusive. Here, we performed epithelial-mesenchymal tissue recombination experiments using hair-forming back skin and glabrous plantar skin from mouse embryos to unveil that the back skin mesenchyme is inductive even before dermal condensate formation. Moreover, the naïve, unpatterned mesenchyme was sufficient to trigger hair follicle formation even in the oral epithelium. Building on previous knowledge, we explored the hair-inductive ability of the Wnt agonist R-spondin 1 and a Bmp receptor inhibitor in embryonic skin explants. Although R-spondin 1 instigated precocious placode-specific transcriptional responses, it was insufficient for hair follicle induction, either alone or in combination with Bmp receptor inhibition. Our findings pave the way for identifying the hair follicle-inducing cue.


Asunto(s)
Folículo Piloso , Cabello , Ratones , Animales , Folículo Piloso/fisiología , Piel , Mesodermo/fisiología , Receptores de Proteínas Morfogenéticas Óseas
3.
J Mammary Gland Biol Neoplasia ; 28(1): 17, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37450065

RESUMEN

On 8 December 2022 the organizing committee of the European Network for Breast Development and Cancer labs (ENBDC) held its fifth annual Think Tank meeting in Amsterdam, the Netherlands. Here, we embraced the opportunity to look back to identify the most prominent breakthroughs of the past ten years and to reflect on the main challenges that lie ahead for our field in the years to come. The outcomes of these discussions are presented in this position paper, in the hope that it will serve as a summary of the current state of affairs in mammary gland biology and breast cancer research for early career researchers and other newcomers in the field, and as inspiration for scientists and clinicians to move the field forward.


Asunto(s)
Neoplasias de la Mama , Glándulas Mamarias Humanas , Humanos , Femenino , Mama , Biología
4.
Evol Dev ; 25(6): 410-417, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37070415

RESUMEN

The development of an individual must be capable of resisting the harmful effects of internal and external perturbations. This capacity, called robustness, can make the difference between normal variation and disease. Some systems and organs are more resilient in their capacity to correct the effects of internal disturbances such as mutations. Similarly, organs and organisms differ in their capacity to be resilient against external disturbances, such as changes in temperature. Furthermore, all developmental systems must be somewhat flexible to permit evolutionary change, and understanding robustness requires a comparative framework. Over the last decades, most research on developmental robustness has been focusing on specific model systems and organs. Hence, we lack tools that would allow cross-species and cross-organ comparisons. Here, we emphasize the need for a uniform framework to experimentally test and quantify robustness across study systems and suggest that the analysis of fluctuating asymmetry might be a powerful proxy to do so. Such a comparative framework will ultimately help to resolve why and how organs of the same and different species differ in their sensitivity to internal (e.g., mutations) and external (e.g., temperature) perturbations and at what level of biological organization buffering capacities exist and therefore create robustness of the developmental system.


Asunto(s)
Evolución Biológica , Modelos Biológicos , Animales , Temperatura
5.
Genes Dev ; 27(4): 450-8, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23431057

RESUMEN

In hair follicle development, a placode-derived signal is believed to induce formation of the dermal condensation, an essential component of ectodermal organs. However, the identity of this signal is unknown. Furthermore, although induction and patterning of hair follicles are intimately linked, it is not known whether the mesenchymal condensation is necessary for inducing the initial epithelial pattern. Here, we show that fibroblast growth factor 20 (Fgf20) is expressed in hair placodes and is induced by and functions downstream from epithelial ectodysplasin (Eda)/Edar and Wnt/ß-Catenin signaling to initiate formation of the underlying dermal condensation. Fgf20 governs formation of primary and secondary dermal condensations in developing hair follicles and subsequent formation of guard, awl, and auchene hairs. Although primary dermal condensations are absent in Fgf20 mutant mice, a regular array of hair placodes is formed, demonstrating that the epithelial patterning process is independent of known histological and molecular markers of underlying mesenchymal patterns during the initial stages of hair follicle development.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Folículo Piloso/embriología , Animales , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Ratones , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
6.
J Mammary Gland Biol Neoplasia ; 25(4): 409-416, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33009602

RESUMEN

Branching morphogenesis of the murine mammary gland starts during late embryogenesis. It is regulated by the signals emanating both from the epithelium and the mesenchyme, yet the molecular mechanisms regulating this process remain poorly understood. We have previously developed a unique whole organ culture technique for embryonic mammary glands, which provides a powerful tool to monitor and manipulate branching morphogenesis ex vivo. Nowadays, RNA sequencing and other transcriptional profiling techniques provide robust methods to identify components of gene regulatory networks driving branching morphogenesis. However, validation of the candidate genes still mainly depends on the use of the transgenic mouse models, especially in mammary gland studies. By comparing different serotypes of recombinant adeno-associated virus (rAAVs), we found out that rAAVs provide sufficient efficiency for gene transfer with different tissue preferences depending on the serotypes of the virus. AAV-2 and AAV-8 preferentially target epithelial and mesenchymal compartments, respectively, while AAV-9 infects both tissues. Here, we describe a protocol for AAV-mediated gene transfer in ex vivo cultured murine embryonic mammary gland facilitating gene function studies on mammary gland branching morphogenesis.


Asunto(s)
Técnicas de Transferencia de Gen , Glándulas Mamarias Animales/crecimiento & desarrollo , Morfogénesis/genética , Técnicas de Cultivo de Órganos/métodos , Animales , Dependovirus/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos/genética , Ratones , Serogrupo
7.
Nature ; 512(7512): 44-8, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25079326

RESUMEN

The evolutionary relationships of extinct species are ascertained primarily through the analysis of morphological characters. Character inter-dependencies can have a substantial effect on evolutionary interpretations, but the developmental underpinnings of character inter-dependence remain obscure because experiments frequently do not provide detailed resolution of morphological characters. Here we show experimentally and computationally how gradual modification of development differentially affects characters in the mouse dentition. We found that intermediate phenotypes could be produced by gradually adding ectodysplasin A (EDA) protein in culture to tooth explants carrying a null mutation in the tooth-patterning gene Eda. By identifying development-based character inter-dependencies, we show how to predict morphological patterns of teeth among mammalian species. Finally, in vivo inhibition of sonic hedgehog signalling in Eda null teeth enabled us to reproduce characters deep in the rodent ancestry. Taken together, evolutionarily informative transitions can be experimentally reproduced, thereby providing development-based expectations for character-state transitions used in evolutionary studies.


Asunto(s)
Evolución Biológica , Fósiles , Diente/anatomía & histología , Diente/crecimiento & desarrollo , Animales , Simulación por Computador , Ectodisplasinas/deficiencia , Ectodisplasinas/genética , Ectodisplasinas/farmacología , Femenino , Eliminación de Gen , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Técnicas In Vitro , Masculino , Ratones , Diente Molar/anatomía & histología , Diente Molar/efectos de los fármacos , Diente Molar/crecimiento & desarrollo , Fenotipo , Transducción de Señal/efectos de los fármacos , Diente/efectos de los fármacos
8.
J Mammary Gland Biol Neoplasia ; 24(3): 201-206, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31494779

RESUMEN

The eleventh annual workshop of the European Network for Breast Development and Cancer, Methods in mammary gland biology and breast cancer, took place on the 16th to 18th of May 2019 in Weggis, Switzerland. The main topics of the meeting were high resolution genomics and proteomics for the study of mammary gland development and cancer, breast cancer signaling, tumor microenvironment, preclinical models of breast cancer, and tissue morphogenesis. Exciting novel findings in, or highly relevant to, mammary gland biology and breast cancer field were presented, with insights into the methods used to obtain them. Among others, the discussed methods included single-cell RNA sequencing, genetic barcoding, lineage tracing, spatial transcriptomics, optogenetics, genetic mouse models and organoids.


Asunto(s)
Neoplasias de la Mama/patología , Mama/patología , Carcinogénesis/patología , Microambiente Tumoral , Animales , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Femenino , Genómica , Humanos , Proteómica , Transducción de Señal , Sociedades Científicas
10.
Development ; 142(22): 3954-63, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26450968

RESUMEN

Epithelial morphogenesis generates the shape of the tooth crown. This is driven by patterned differentiation of cells into enamel knots, root-forming cervical loops and enamel-forming ameloblasts. Enamel knots are signaling centers that define the positions of cusp tips in a tooth by instructing the adjacent epithelium to fold and proliferate. Here, we show that the forkhead-box transcription factor Foxi3 inhibits formation of enamel knots and cervical loops and thus the differentiation of dental epithelium in mice. Conditional deletion of Foxi3 (Foxi3 cKO) led to fusion of molars with abnormally patterned shallow cusps. Foxi3 was expressed in the epithelium, and its expression was reduced in the enamel knots and cervical loops and in ameloblasts. Bmp4, a known inducer of enamel knots and dental epithelial differentiation, downregulated Foxi3 in wild-type teeth. Using genome-wide gene expression profiling, we showed that in Foxi3 cKO there was an early upregulation of differentiation markers, such as p21, Fgf15 and Sfrp5. Different signaling pathway components that are normally restricted to the enamel knots were expanded in the epithelium, and Sostdc1, a marker of the intercuspal epithelium, was missing. These findings indicated that the activator-inhibitor balance regulating cusp patterning was disrupted in Foxi3 cKO. In addition, early molar bud morphogenesis and, in particular, formation of the suprabasal epithelial cell layer were impaired. We identified keratin 10 as a marker of suprabasal epithelial cells in teeth. Our results suggest that Foxi3 maintains dental epithelial cells in an undifferentiated state and thereby regulates multiple stages of tooth morphogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Epitelio/fisiología , Factores de Transcripción Forkhead/metabolismo , Diente Molar/embriología , Morfogénesis/fisiología , Transducción de Señal/fisiología , Corona del Diente/embriología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Epitelio/metabolismo , Técnica del Anticuerpo Fluorescente , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Análisis por Matrices de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Nature ; 483(7389): 324-7, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22398444

RESUMEN

One of the fascinating aspects of the history of life is the apparent increase in morphological complexity through time, a well known example being mammalian cheek tooth evolution. In contrast, experimental studies of development more readily show a decrease in complexity, again well exemplified by mammalian teeth, in which tooth crown features called cusps are frequently lost in mutant and transgenic mice. Here we report that mouse tooth complexity can be increased substantially by adjusting multiple signalling pathways simultaneously. We cultured teeth in vitro and adjusted ectodysplasin (EDA), activin A and sonic hedgehog (SHH) pathways, all of which are individually required for normal tooth development. We quantified tooth complexity using the number of cusps and a topographic measure of surface complexity. The results show that whereas activation of EDA and activin A signalling, and inhibition of SHH signalling, individually cause subtle to moderate increases in complexity, cusp number is doubled when all three pathways are adjusted in unison. Furthermore, the increase in cusp number does not result from an increase in tooth size, but from an altered primary patterning phase of development. The combination of a lack of complex mutants, the paucity of natural variants with complex phenotypes, and our results of greatly increased dental complexity using multiple pathways, suggests that an increase may be inherently different from a decrease in phenotypic complexity.


Asunto(s)
Evolución Biológica , Diente Molar/anatomía & histología , Diente Molar/metabolismo , Transducción de Señal , Activinas/metabolismo , Activinas/farmacología , Animales , Biología Evolutiva , Ectodisplasinas/metabolismo , Ectodisplasinas/farmacología , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/farmacología , Ratones , Diente Molar/efectos de los fármacos , Diente Molar/embriología , Mutación , Técnicas de Cultivo de Órganos , Fenotipo , Transducción de Señal/efectos de los fármacos
12.
Semin Immunol ; 26(3): 220-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24928340

RESUMEN

Ectodysplasin (Eda) is the most studied tumor necrosis ligand in the field of developmental biology. In all vertebrates studied so far, inactivating germline mutations in Eda lead to the genetic disease called hypohidrotic ectodermal dysplasia (HED). In humans, HED is a life-threatening condition in particular in infants due to absent or severely reduced sweating leading to hyperthermia. HED is also characterized by sparse hair, and oligo- or anodontia. Research of the Eda pathway has not only increased our knowledge on ectodermal appendage development and etiology of developmental disorders, but also on evolution of several vertebrate species including humankind. Studies on mouse and dog models of HED has led to one of the most stunning breakthroughs in applied developmental biology research by showing that a short-term treatment of neonates with a synthetic ligand corrects many of the HED-associated traits. Eighteen years after the identification of EDA as the causative gene in HED, a phase II trial aiming at permanent correction of the disease is now ongoing. This review summarizes the latest discoveries in the Eda field and points to areas that need further investigation such as the possible involvement of Eda in cell migration, stem cell maintenance, or cancer.


Asunto(s)
Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Animales , Displasia Ectodermal Anhidrótica Tipo 1/genética , Displasia Ectodermal Anhidrótica Tipo 1/metabolismo , Ectodisplasinas/genética , Humanos , Transducción de Señal
13.
PLoS Genet ; 11(11): e1005676, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26581094

RESUMEN

Mammary gland development commences during embryogenesis with the establishment of a species typical number of mammary primordia on each flank of the embryo. It is thought that mammary cell fate can only be induced along the mammary line, a narrow region of the ventro-lateral skin running from the axilla to the groin. Ectodysplasin (Eda) is a tumor necrosis factor family ligand that regulates morphogenesis of several ectodermal appendages. We have previously shown that transgenic overexpression of Eda (K14-Eda mice) induces formation of supernumerary mammary placodes along the mammary line. Here, we investigate in more detail the role of Eda and its downstream mediator transcription factor NF-κB in mammary cell fate specification. We report that K14-Eda mice harbor accessory mammary glands also in the neck region indicating wider epidermal cell plasticity that previously appreciated. We show that even though NF-κB is not required for formation of endogenous mammary placodes, it is indispensable for the ability of Eda to induce supernumerary placodes. A genome-wide profiling of Eda-induced genes in mammary buds identified several Wnt pathway components as potential transcriptional targets of Eda. Using an ex vivo culture system, we show that suppression of canonical Wnt signalling leads to a dose-dependent inhibition of supernumerary placodes in K14-Eda tissue explants.


Asunto(s)
Ectodisplasinas/genética , Glándulas Mamarias Humanas/crecimiento & desarrollo , Morfogénesis/genética , Vía de Señalización Wnt/genética , Animales , Diferenciación Celular/genética , Ectodisplasinas/biosíntesis , Ectodisplasinas/metabolismo , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/crecimiento & desarrollo , Humanos , Glándulas Mamarias Humanas/citología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo
14.
Stem Cells ; 34(7): 1896-908, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26992132

RESUMEN

The hair follicle is an ideal system to study stem cell specification and homeostasis due to its well characterized morphogenesis and stereotypic cycles of stem cell activation upon each hair cycle to produce a new hair shaft. The adult hair follicle stem cell niche consists of two distinct populations, the bulge and the more activation-prone secondary hair germ (HG). Hair follicle stem cells are set aside during early stages of morphogenesis. This process is known to depend on the Sox9 transcription factor, but otherwise the establishment of the hair follicle stem cell niche is poorly understood. Here, we show that that mutation of Foxi3, a Forkhead family transcription factor mutated in several hairless dog breeds, compromises stem cell specification. Further, loss of Foxi3 impedes hair follicle downgrowth and progression of the hair cycle. Genome-wide profiling revealed a number of downstream effectors of Foxi3 including transcription factors with a recognized function in hair follicle stem cells such as Lhx2, Runx1, and Nfatc1, suggesting that the Foxi3 mutant phenotype results from simultaneous downregulation of several stem cell signature genes. We show that Foxi3 displays a highly dynamic expression pattern during hair morphogenesis and cycling, and identify Foxi3 as a novel secondary HG marker. Absence of Foxi3 results in poor hair regeneration upon hair plucking, and a sparse fur phenotype in unperturbed mice that exacerbates with age, caused by impaired secondary HG activation leading to progressive depletion of stem cells. Thus, Foxi3 regulates multiple aspects of hair follicle development and homeostasis. Stem Cells 2016;34:1896-1908.


Asunto(s)
Factores de Transcripción Forkhead/deficiencia , Folículo Piloso/citología , Células Madre/citología , Células Madre/metabolismo , Animales , Biomarcadores/metabolismo , Compartimento Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Embrión de Mamíferos/metabolismo , Retroalimentación Fisiológica/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Regeneración/efectos de los fármacos , Transducción de Señal , Células Madre/efectos de los fármacos , Proteínas Wnt/farmacología
15.
Semin Cell Dev Biol ; 25-26: 11-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24487243

RESUMEN

The embryonic surface ectoderm gives rise to the epidermis and ectodermal appendages including hair follicles, teeth, scales, feathers, and mammary, sweat, and salivary glands. Their early development proceeds largely the same through the induction, placode, and bud stages prior to diversification of epithelial morphogenesis which ultimately produces the wide array of mature organs. In this review we summarize the current knowledge on the molecular and cellular processes driving the shared stages of skin appendage development revealed by analysis of mouse mutants. We focus on three mammalian organs: hair follicle, tooth, and mammary gland. We reevaluate the information gained from classic epithelial-mesenchymal tissue recombination experiments in light of current molecular knowledge. We place special emphasis on the signaling pathways that mediate tissue interactions, and attempt to link the signaling outputs to changes in cellular behavior that ultimately shape the developing organ.


Asunto(s)
Ectodermo/crecimiento & desarrollo , Piel/crecimiento & desarrollo , Animales , Ectodermo/citología , Ectodermo/embriología , Transición Epitelial-Mesenquimal , Ratones , Morfogénesis , Piel/citología , Piel/enzimología
16.
N Engl J Med ; 378(17): 1637-1638, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29694818
17.
Development ; 139(17): 3189-99, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22833125

RESUMEN

Uncovering the origin and nature of phenotypic variation within species is the first step in understanding variation between species. Mouse models with altered activities of crucial signal pathways have highlighted many important genes and signal networks regulating the morphogenesis of complex structures, such as teeth. The detailed analyses of these models have indicated that the balanced actions of a few pathways regulating cell behavior modulate the shape and number of teeth. Currently, however, most mouse models studied have had gross alteration of morphology, whereas analyses of more subtle modification of morphology are required to link developmental studies to evolutionary change. Here, we have analyzed a signaling network involving ectodysplasin (Eda) and fibroblast growth factor 20 (Fgf20) that subtly affects tooth morphogenesis. We found that Fgf20 is a major downstream effector of Eda and affects Eda-regulated characteristics of tooth morphogenesis, including the number, size and shape of teeth. Fgf20 function is compensated for by other Fgfs, in particular Fgf9 and Fgf4, and is part of an Fgf signaling loop between epithelium and mesenchyme. We showed that removal of Fgf20 in an Eda gain-of-function mouse model results in an Eda loss-of-function phenotype in terms of reduced tooth complexity and third molar appearance. However, the extra anterior molar, a structure lost during rodent evolution 50 million years ago, was stabilized in these mice.


Asunto(s)
Ectodisplasinas/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Morfogénesis/fisiología , Transducción de Señal/fisiología , Diente/embriología , Animales , Evolución Biológica , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Hibridación in Situ , Indoles , Luciferasas , Ratones , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética
18.
Proc Natl Acad Sci U S A ; 109(15): 5744-9, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22451941

RESUMEN

Ductal growth of the mammary gland occurs in two distinct stages. The first round of branching morphogenesis occurs during embryogenesis, and the second round commences at the onset of puberty. Currently, relatively little is known about the genetic networks that control the initial phases of ductal expansion, which, unlike pubertal development, proceeds independent of hormonal input in female mice. Here we identify NF-κB downstream of the TNF-like ligand ectodysplasin (Eda) as a unique regulator of embryonic and prepubertal ductal morphogenesis. Loss of Eda, or inhibition of NF-κB, led to smaller ductal trees with fewer branches. On the other hand, overexpression of Eda caused a dramatic NF-κB-dependent phenotype in both female and male mice characterized by precocious and highly increased ductal growth and branching that correlated with enhanced cell proliferation. We have identified several putative transcriptional target genes of Eda/NF-κB, including PTHrP, Wnt10a, and Wnt10b, as well as Egf family ligands amphiregulin and epigen. We developed a mammary bud culture system that allowed us to manipulate mammary development ex vivo and found that recombinant PTHrP, Wnt3A, and Egf family ligands stimulate embryonic branching morphogenesis, suggesting that these pathways may cooperatively mediate the effects of Eda.


Asunto(s)
Ectodisplasinas/metabolismo , Hormonas/farmacología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Morfogénesis/efectos de los fármacos , FN-kappa B/metabolismo , Anfirregulina , Andrógenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Familia de Proteínas EGF , Desarrollo Embrionario/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Epigen , Epitelio/efectos de los fármacos , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Femenino , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ligandos , Masculino , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Ratones , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas Wnt/metabolismo
19.
Am J Hum Genet ; 89(1): 67-81, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21741611

RESUMEN

Craniosynostosis and supernumerary teeth most often occur as isolated developmental anomalies, but they are also separately manifested in several malformation syndromes. Here, we describe a human syndrome featuring craniosynostosis, maxillary hypoplasia, delayed tooth eruption, and supernumerary teeth. We performed homozygosity mapping in three unrelated consanguineous Pakistani families and localized the syndrome to a region in chromosome 9. Mutational analysis of candidate genes in the region revealed that all affected children harbored homozygous missense mutations (c.662C>G [p.Pro221Arg], c.734C>G [p.Ser245Cys], or c.886C>T [p.Arg296Trp]) in IL11RA (encoding interleukin 11 receptor, alpha) on chromosome 9p13.3. In addition, a homozygous nonsense mutation, c.475C>T (p.Gln159X), and a homozygous duplication, c.916_924dup (p.Thr306_Ser308dup), were observed in two north European families. In cell-transfection experiments, the p.Arg296Trp mutation rendered the receptor unable to mediate the IL11 signal, indicating that the mutation causes loss of IL11RA function. We also observed disturbed cranial growth and suture activity in the Il11ra null mutant mice, in which reduced size and remodeling of limb bones has been previously described. We conclude that IL11 signaling is essential for the normal development of craniofacial bones and teeth and that its function is to restrict suture fusion and tooth number. The results open up the possibility of modulation of IL11 signaling for the treatment of craniosynostosis.


Asunto(s)
Craneosinostosis/genética , Interleucina-11/metabolismo , Transducción de Señal , Erupción Dental/genética , Diente Supernumerario/genética , Animales , Línea Celular , Niño , Preescolar , Mapeo Cromosómico , Codón sin Sentido , Biología Computacional , Análisis Mutacional de ADN , Regulación hacia Abajo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Interleucina-11/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Linaje , Diente Supernumerario/patología , Factores de Transcripción/genética
20.
Development ; 138(13): 2681-91, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21652647

RESUMEN

The developing submandibular salivary gland (SMG) is a well-studied model for tissue interactions and branching morphogenesis. Its development shares similar features with other ectodermal appendages such as hair and tooth. The ectodysplasin (Eda) pathway is essential for the formation and function of several ectodermal organs. Mutations in the signaling components of the Eda pathway lead to a human syndrome known as hypohidrotic ectodermal dysplasia (HED), which is characterized by missing and malformed teeth, sparse hair and reduced sweating. Individuals with HED suffer also from dry mouth because of reduced saliva flow. In order to understand the underlying mechanism, we analyzed salivary gland development in mouse models with altered Eda pathway activities. We have found that Eda regulates growth and branching of the SMG via transcription factor NF-κB in the epithelium, and that the hedgehog pathway is an important mediator of Eda/NF-κB. We also sought to determine whether a similar reciprocal interplay between the Eda and Wnt/ß-catenin pathways, which are known to operate in other skin appendages, functions in developing SMG. Surprisingly and unlike in developing hair follicles and teeth, canonical Wnt signaling activity did not colocalize with Edar/NF-κB in salivary gland epithelium. Instead, we observed high mesenchymal Wnt activity and show that ablation of mesenchymal Wnt signaling either in vitro or in vivo compromised branching morphogenesis. We also provide evidence suggesting that the effects of mesenchymal Wnt/ß-catenin signaling are mediated, at least in part, through regulation of Eda expression.


Asunto(s)
Ectodisplasinas/metabolismo , Glándulas Salivales/embriología , Glándulas Salivales/metabolismo , Proteínas Wnt/metabolismo , Animales , Ectodisplasinas/genética , Femenino , Hibridación in Situ , Masculino , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Técnicas de Cultivo de Órganos , Reacción en Cadena de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Wnt/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA