Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 15(9): e1008351, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31527874

RESUMEN

Wnt proteins are secreted signaling factors that regulate cell fate specification and patterning decisions throughout the animal kingdom. In the Drosophila wing epithelium, Wingless (Wg, the homolog of Wnt1) is secreted from a narrow strip of cells at the dorsal-ventral boundary. However, the route of Wg secretion in polarized epithelial cells remains poorly understood and key proteins involved in this process are still unknown. Here, we performed an in vivo RNAi screen and identified members of the exocyst complex to be required for apical but not basolateral Wg secretion. Specifically blocking the apical Wg secretion leads to reduced downstream signaling. Using an in vivo 'temporal-rescue' assay, our results further indicate that apically secreted Wg activates target genes that require high signaling activity. In conclusion, our results demonstrate that the exocyst is required for an apical route of Wg secretion from polarized wing epithelial cells.


Asunto(s)
Proteínas de Drosophila/metabolismo , Vías Secretoras/fisiología , Vía de Señalización Wnt/fisiología , Proteína Wnt1/metabolismo , Animales , Tipificación del Cuerpo/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Células Epiteliales/metabolismo , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ligandos , Transducción de Señal , Alas de Animales/embriología , Alas de Animales/metabolismo , Proteínas Wnt/genética , Vía de Señalización Wnt/genética , Proteína Wnt1/genética , Proteína Wnt1/fisiología
2.
FASEB J ; 34(5): 6791-6807, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32223017

RESUMEN

The frontal craniofacial skeleton derived from neural crest cells is vital for facial structure and masticatory functions. The exact role of Indian hedgehog (Ihh) in facial and masticatory development has not been fully explored. In this study, we generated craniofacial neural crest cells-specific Ihh deletion mice (Wnt1-Cre;Ihhfl/fl ;Tomatofl/+ ) and found the gradual dwarfism without perinatal lethality. Morphological and histological analyses revealed unambiguous craniofacial phenotypes in mutants, where we observed skeletal malocclusion accompanied by markedly hypoplastic nasomaxillary complex and reversed incisor occlusion. Both the replacement of nasal concha cartilage by turbinate bones and the endochondral ossification of nasal septum ethmoid bone were substantially delayed. We also observed hypoplastic mandibles in mutants where the mandibular ramus was unexpectedly the most affected. Both the condylar process and mandibular angle cartilages were distorted. However, dental examination showed no significant changes in teeth and dentition. Finally, a comprehensive RNA sequence analysis utilizing condylar cartilage identified Ihh-associated gene network including several cell cycle genes and 16 genes related to the extracellular matrix, sulfate transporters, transcription factors, receptors, a ciliogenesis factor, and an adhesion molecule. Our data provide direct in vivo evidence that Ihh plays crucial roles in midface and masticatory system formation, likely by activating key genes.


Asunto(s)
Huesos/patología , Cartílago/patología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/fisiología , Maloclusión/patología , Cresta Neural/patología , Proteína Wnt1/fisiología , Animales , Huesos/metabolismo , Cartílago/metabolismo , Condrogénesis , Anomalías Craneofaciales , Femenino , Masculino , Maloclusión/genética , Maloclusión/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Cresta Neural/metabolismo , Fenotipo
3.
PLoS Genet ; 14(8): e1007568, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30142157

RESUMEN

We characterized the establishment of an Epidermal Growth Factor Receptor (EGFR) organizing center (EOC) during leg development in Drosophila melanogaster. Initial EGFR activation occurs in the center of leg discs by expression of the EGFR ligand Vn and the EGFR ligand-processing protease Rho, each through single enhancers, vnE and rhoE, that integrate inputs from Wg, Dpp, Dll and Sp1. Deletion of vnE and rhoE eliminates vn and rho expression in the center of the leg imaginal discs, respectively. Animals with deletions of both vnE and rhoE (but not individually) show distal but not medial leg truncations, suggesting that the distal source of EGFR ligands acts at short-range to only specify distal-most fates, and that multiple additional 'ring' enhancers are responsible for medial fates. Further, based on the cis-regulatory logic of vnE and rhoE we identified many additional leg enhancers, suggesting that this logic is broadly used by many genes during Drosophila limb development.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Elementos de Facilitación Genéticos , Receptores ErbB/fisiología , Extremidades/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Receptores de Péptidos de Invertebrados/fisiología , Alelos , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/crecimiento & desarrollo , Receptores ErbB/genética , Eliminación de Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Discos Imaginales/fisiología , Neurregulinas/genética , Neurregulinas/fisiología , Organizadores Embrionarios , Receptores de Péptidos de Invertebrados/genética , Transducción de Señal , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/fisiología , Proteína Wnt1/genética , Proteína Wnt1/fisiología
4.
Arterioscler Thromb Vasc Biol ; 39(8): 1629-1644, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189430

RESUMEN

OBJECTIVE: Periaortic arch adipose tissue (PAAT) plays critical roles in regulating vascular homeostasis; however, its anatomic features, developmental processes, and origins remain unclear. Approach and Results: Anatomic analysis and genetic lineage tracing of Wnt1 (wingless-type MMTV [mouse mammary tumor virus] integration site family member 1)-Cre+;Rosa26RFP/+ mice, Myf5 (myogenic factor 5)-Cre+;Rosa26RFP/+ mice, and SM22α-Cre+;Rosa26RFP/+ mice are performed, and the results show that PAAT has unique anatomic features, and the developmental processes of PAAT are independent of the others periaortic adipose tissues. PAAT adipocytes are mainly derived from neural crest cells (NCCs) rather than from Myf5+ progenitors. Most PAAT adipocyte progenitors expressed SM22α+ (smooth muscle protein 22-alpha) during development. Using Wnt1-Cre+;PPARγflox/flox mice, we found that knockout of PPAR (peroxisome proliferator-activated receptor)-γ in NCCs results in PAAT developmental delay and dysplasia, further confirming that NCCs contribute to PAAT formation. And we further indicated PAAT dysplasia aggravates Ang II (angiotensin II)-induced inflammation and remodeling of the common carotid artery close to aorta arch. We also found that NCCs can be differentiated into both brown and white adipocytes in vivo and in vitro. RNA sequencing results suggested NCC-derived adipose tissue displays a distinct transcriptional profile compared with the non-NCC-derived adipose tissue in PAAT. CONCLUSIONS: PAAT has distinctive anatomic features and developmental processes. Most PAAT adipocytes are originated from NCCs which derive from ectoderm. NCCs are progenitors not only of white adipocytes but also of brown adipocytes. This study indicates that the PAAT is derived from multiple cell lineages, the adipocytes derived from different origins have distinct transcriptional profiles, and PAAT plays a critical role in Ang II-induced common carotid artery inflammation and remodeling.Visual OvervieW: An online visual overview is available for this article.


Asunto(s)
Adipocitos Marrones/citología , Adipogénesis , Tejido Adiposo/fisiología , Cresta Neural/citología , Tejido Adiposo/citología , Angiotensina II/farmacología , Animales , Aorta Torácica/citología , Arteria Carótida Común/citología , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/fisiología , Proteína Wnt1/fisiología
5.
Dev Biol ; 439(2): 53-64, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29715461

RESUMEN

Paracrine Wnt signals are critical regulators of cell proliferation, specification, and differentiation during embryogenesis. Consistent with the discovery that Wnt ligands are post-translationally modified with palmitoleate (a 16 carbon mono-unsaturated fatty acid), our studies show that the vast majority of bioavailable chick WNT1 (cWNT1) produced in stably transfected L cells is cell-associated. Thus, it seems unlikely that the WNT1 signal is propagated by diffusion alone. Unfortunately, the production and transport of vertebrate Wnt proteins has been exceedingly difficult to study as few antibodies are able to detect endogenous Wnt proteins and fixation is known to disrupt the architecture of cells and tissues. Furthermore, vertebrate Wnts have been extraordinarily refractory to tagging. To help overcome these obstacles, we have generated a number of tools that permit the detection of WNT1 in palmitoylation assays and the visualization of chick and zebrafish WNT1 in live cells and tissues. Consistent with previous studies in fixed cells, live imaging of cells and tissues with overexpressed cWNT1-moxGFP shows predominant localization of the protein to a reticulated network that is likely to be the endoplasmic reticulum. As PORCN and WLS are important upstream regulators of Wnt gradient formation, we also undertook the generation of mCherry-tagged variants of both proteins. While co-expression of PORCN-mCherry had no discernible effect on the localization of WNT1-moxGFP, co-expression of WLS-mCherry caused a marked redistribution of WNT1-moxGFP to the cell surface and cellular projections in cultured cells as well as in neural crest and surface ectoderm cells in developing chick embryos. Our studies further establish that the levels of WLS, and not PORCN, are rate limiting with respect to WNT1 trafficking.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Imagen Óptica/métodos , Proteína Wnt1/metabolismo , Aciltransferasas/metabolismo , Animales , Embrión de Pollo , Pollos/metabolismo , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipoilación , Proteínas de la Membrana/metabolismo , Ratones , Cresta Neural/metabolismo , Procesamiento Proteico-Postraduccional , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , Proteína Wnt1/fisiología , Proteína Wnt3A/metabolismo , Pez Cebra/metabolismo
6.
PLoS Biol ; 14(9): e1002536, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27584613

RESUMEN

Drosophila melanogaster larvae irradiated with doses of ionizing radiation (IR) that kill about half of the cells in larval imaginal discs still develop into viable adults. How surviving cells compensate for IR-induced cell death to produce organs of normal size and appearance remains an active area of investigation. We have identified a subpopulation of cells within the continuous epithelium of Drosophila larval wing discs that shows intrinsic resistance to IR- and drug-induced apoptosis. These cells reside in domains of high Wingless (Wg, Drosophila Wnt-1) and STAT92E (sole Drosophila signal transducer and activator of transcription [STAT] homolog) activity and would normally form the hinge in the adult fly. Resistance to IR-induced apoptosis requires STAT and Wg and is mediated by transcriptional repression of the pro-apoptotic gene reaper. Lineage tracing experiments show that, following irradiation, apoptosis-resistant cells lose their identity and translocate to areas of the wing disc that suffered abundant cell death. Our findings provide a new paradigm for regeneration in which it is unnecessary to invoke special damage-resistant cell types such as stem cells. Instead, differences in gene expression within a population of genetically identical epithelial cells can create a subpopulation with greater resistance, which, following damage, survive, alter their fate, and help regenerate the tissue.


Asunto(s)
Apoptosis/efectos de la radiación , Proteínas de Drosophila/fisiología , Células Epiteliales/efectos de la radiación , Discos Imaginales/fisiología , Factores de Transcripción STAT/fisiología , Proteína Wnt1/fisiología , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/efectos de la radiación , Células Epiteliales/fisiología , Expresión Génica , Silenciador del Gen/efectos de la radiación , Discos Imaginales/citología , Larva/citología , Larva/fisiología , Larva/efectos de la radiación , Traumatismos Experimentales por Radiación , Regeneración
7.
Dev Biol ; 415(2): 228-241, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26988119

RESUMEN

We compared apparent origins, cellular diversity and regulation of initial axon growth for differentiating cranial sensory neurons. We assessed the molecular and cellular composition of the developing olfactory and otic placodes, and cranial sensory ganglia to evaluate contributions of ectodermal placode versus neural crest at each site. Special sensory neuron populations-the olfactory and otic placodes, as well as those in vestibulo-acoustic ganglion- are entirely populated with cells expressing cranial placode-associated, rather than neural crest-associated markers. The remaining cranial sensory ganglia are a mosaic of cells that express placode-associated as well as neural crest-associated markers. We found two distinct populations of neural crest in the cranial ganglia: the first, as expected, is labeled by Wnt1:Cre mediated recombination. The second is not labeled by Wnt1:Cre recombination, and expresses both Sox10 and FoxD3. These populations-Wnt1:Cre recombined, and Sox10/Foxd3-expressing- are proliferatively distinct from one another. Together, the two neural crest-associated populations are substantially more proliferative than their placode-associated counterparts. Nevertheless, the apparently placode- and neural crest-associated populations are similarly sensitive to altered signaling that compromises cranial morphogenesis and differentiation. Acute disruption of either Fibroblast growth factor (Fgf) or Retinoic acid (RA) signaling alters axon growth and cell death, but does not preferentially target any of the three distinct populations. Apparently, mosaic derivation and diversity of precursors and early differentiating neurons, modulated uniformly by local signals, supports early cranial sensory neuron differentiation and growth.


Asunto(s)
Nervios Craneales/citología , Células Receptoras Sensoriales/citología , Animales , Apoptosis , Axones/fisiología , Diferenciación Celular , Linaje de la Célula , Nervios Craneales/embriología , Ectodermo/citología , Factores de Crecimiento de Fibroblastos/fisiología , Ganglios Sensoriales/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Cresta Neural/citología , Neurogénesis , Factores de Transcripción/genética , Tretinoina/fisiología , Proteína Wnt1/fisiología
8.
Development ; 141(12): 2483-93, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24917503

RESUMEN

The potent activity of Wnt/Wingless (Wg) signals necessitates sophisticated mechanisms that spatially and temporally regulate their distribution and range of action. The two main receptor components for Wg - Arrow (Arr) and Frizzled 2 (Fz2) - are transcriptionally downregulated by Wg signaling, thus forming gradients that oppose that of Wg. Here, we analyze the relevance of this transcriptional regulation for the formation of the Wg gradient in the Drosophila wing disc by combining in vivo receptor overexpression with an in silico model of Wg receptor interactions. Our experiments show that ubiquitous upregulation of Arr and Fz2 has no significant effects on Wg output, whereas clonal overexpression of these receptors leads to signaling discontinuities that have detrimental phenotypic consequences. These findings are supported by our in silico model for Wg diffusion and signal transduction, which suggests that abrupt changes in receptor levels causes discontinuities in Wg signaling. Furthermore, we identify a 200 bp regulatory element in the arr locus that can account for the Arr gradient, and we show that this is indirectly negatively controlled by Wg activity. Finally, we analyze the role of Frizzled 3 (Fz3) in this system and find that its expression, which is induced by Wg, contributes to the establishment of the Arr and Fz2 gradients through counteracting canonical signaling. Taken together, our results provide a model in which the regulatory network of Wg and the three receptor components account for the range and shape of this prototypical morphogen system.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Proteína Wnt1/fisiología , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Receptores Frizzled/genética , Ligandos , Modelos Biológicos , Fenotipo , Regiones Promotoras Genéticas , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba , Alas de Animales/embriología , Proteína Wnt1/genética
9.
J Neurosci ; 35(39): 13385-401, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26424886

RESUMEN

Wingless-related MMTV integration site 1 (WNT1)/ß-catenin signaling plays a crucial role in the generation of mesodiencephalic dopaminergic (mdDA) neurons, including the substantia nigra pars compacta (SNc) subpopulation that preferentially degenerates in Parkinson's disease (PD). However, the precise functions of WNT1/ß-catenin signaling in this context remain unknown. Stem cell-based regenerative (transplantation) therapies for PD have not been implemented widely in the clinical context, among other reasons because of the heterogeneity and incomplete differentiation of the transplanted cells. This might result in tumor formation and poor integration of the transplanted cells into the dopaminergic circuitry of the brain. Dickkopf 3 (DKK3) is a secreted glycoprotein implicated in the modulation of WNT/ß-catenin signaling. Using mutant mice, primary ventral midbrain cells, and pluripotent stem cells, we show that DKK3 is necessary and sufficient for the correct differentiation of a rostrolateral mdDA neuron subset. Dkk3 transcription in the murine ventral midbrain coincides with the onset of mdDA neurogenesis and is required for the activation and/or maintenance of LMX1A (LIM homeobox transcription factor 1α) and PITX3 (paired-like homeodomain transcription factor 3) expression in the corresponding mdDA precursor subset, without affecting the proliferation or specification of their progenitors. Notably, the treatment of differentiating pluripotent stem cells with recombinant DKK3 and WNT1 proteins also increases the proportion of mdDA neurons with molecular SNc DA cell characteristics in these cultures. The specific effects of DKK3 on the differentiation of rostrolateral mdDA neurons in the murine ventral midbrain, together with its known prosurvival and anti-tumorigenic properties, make it a good candidate for the improvement of regenerative and neuroprotective strategies in the treatment of PD. Significance statement: We show here that Dickkopf 3 (DKK3), a secreted modulator of WNT (Wingless-related MMTV integration site)/ß-catenin signaling, is both necessary and sufficient for the proper differentiation and survival of a rostrolateral (parabrachial pigmented nucleus and dorsomedial substantia nigra pars compacta) mesodiencephalic dopaminergic neuron subset, using Dkk3 mutant mice and murine primary ventral midbrain and pluripotent stem cells. The progressive loss of these dopamine-producing mesodiencephalic neurons is a hallmark of human Parkinson's disease, which can up to now not be halted by clinical treatments of this disease. Thus, the soluble DKK3 protein might be a promising new agent for the improvement of current protocols for the directed differentiation of pluripotent and multipotent stem cells into mesodiencephalic dopaminergic neurons and for the promotion of their survival in situ.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Mesencéfalo/fisiología , Células-Madre Neurales/fisiología , Células Madre Pluripotentes/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Recuento de Células , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Supervivencia Celular/genética , Células Cultivadas , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacología , Péptidos y Proteínas de Señalización Intercelular/genética , Mesencéfalo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transcriptoma , Proteína Wnt1/genética , Proteína Wnt1/fisiología
10.
EMBO J ; 31(19): 3901-17, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-22948071

RESUMEN

The ability to regenerate following stress is a hallmark of self-renewing tissues. However, little is known about how regeneration differs from homeostatic tissue maintenance. Here, we study the role and regulation of Wingless (Wg)/Wnt signalling during intestinal regeneration using the Drosophila adult midgut. We show that Wg is produced by the intestinal epithelial compartment upon damage or stress and it is exclusively required for intestinal stem cell (ISC) proliferation during tissue regeneration. Reducing Wg or downstream signalling components from the intestinal epithelium blocked tissue regeneration. Importantly, we demonstrate that Wg from the undifferentiated progenitor cell, the enteroblast, is required for Myc-dependent ISC proliferation during regeneration. Similar to young regenerating tissues, ageing intestines required Wg and Myc for ISC hyperproliferation. Unexpectedly, our results demonstrate that epithelial but not mesenchymal Wg is essential for ISC proliferation in response to damage, while neither source of the ligand is solely responsible for ISC maintenance and tissue self-renewal in unchallenged tissues. Therefore, fine-tuning Wnt results in optimal balance between the ability to respond to stress without negatively affecting organismal viability.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Intestinos/fisiología , Regeneración/fisiología , Células Madre/fisiología , Proteína Wnt1/fisiología , Animales , Proliferación Celular , Femenino , Transducción de Señal/fisiología
11.
EMBO J ; 31(2): 429-42, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22085926

RESUMEN

Wnts are required for cardiogenesis but the role of specific Wnts in cardiac repair remains unknown. In this report, we show that a dynamic Wnt1/ßcatenin injury response activates the epicardium and cardiac fibroblasts to promote cardiac repair. Acute ischaemic cardiac injury upregulates Wnt1 that is initially expressed in the epicardium and subsequently by cardiac fibroblasts in the region of injury. Following cardiac injury, the epicardium is activated organ-wide in a Wnt-dependent manner, expands, undergoes epithelial-mesenchymal transition (EMT) to generate cardiac fibroblasts, which localize in the subepicardial space. The injured regions in the heart are Wnt responsive as well and Wnt1 induces cardiac fibroblasts to proliferate and express pro-fibrotic genes. Disruption of downstream Wnt signalling in epicardial cells decreases epicardial expansion, EMT and leads to impaired cardiac function and ventricular dilatation after cardiac injury. Furthermore, disruption of Wnt/ßcatenin signalling in cardiac fibroblasts impairs wound healing and decreases cardiac performance as well. These findings reveal that a pro-fibrotic Wnt1/ßcatenin injury response is critically required for preserving cardiac function after acute ischaemic cardiac injury.


Asunto(s)
Fibroblastos/metabolismo , Corazón/fisiología , Infarto del Miocardio/patología , Pericardio/metabolismo , Regeneración/fisiología , Transducción de Señal/fisiología , Proteína Wnt1/fisiología , beta Catenina/fisiología , Animales , División Celular , Transición Epitelial-Mesenquimal , Fibrosis , Regulación de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Pericardio/patología , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología , Regulación hacia Arriba , Proteína Wnt1/biosíntesis , Proteína Wnt1/genética , Cicatrización de Heridas/fisiología
12.
Development ; 140(2): 413-22, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23250215

RESUMEN

Wnts and bone morphogenetic proteins (BMPs) are signaling elements that are crucial for a variety of events in animal development. In Drosophila, Wingless (Wg, a Wnt ligand) and Decapentaplegic (Dpp, a BMP homolog) are thought to function through distinct signal transduction pathways and independently direct the patterning of the wing. However, recent studies suggest that Mothers against Dpp (Mad), the key transducer of Dpp signaling, might serve as a node for the crosstalk between these two pathways, and both positive and negative roles of Mad in Wg signaling have been suggested. Here, we describe a novel molecular mechanism by which Dpp signaling suppresses Wg outputs. Brinker (Brk), a transcriptional repressor that is downregulated by Dpp, directly represses naked cuticle (nkd), which encodes a feedback inhibitor of Wg signaling, in vitro and in vivo. Through genetic studies, we demonstrate that Brk is required for Wg target gene expression in fly wing imaginal discs and that loss or gain of brk during wing development mimics loss or gain of Wg signaling, respectively. Finally, we show that Dpp positively regulates the expression of nkd and negatively regulates the Wg target gene Distal-less (Dll). These data support a model in which different signaling pathways interact via a negative-feedback mechanism. Such a mechanism might explain how organs coordinate inputs from multiple signaling cues.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Represoras/fisiología , Proteína Wnt1/fisiología , Animales , Tipificación del Cuerpo , Inmunoprecipitación de Cromatina , Genes Reporteros , Hibridación in Situ , Intrones , Ligandos , Modelos Genéticos , Reacción en Cadena de la Polimerasa/métodos , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Proteína Wnt1/metabolismo
13.
Development ; 140(6): 1342-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23444360

RESUMEN

Wnt1-expressing progenitors generate midbrain dopamine (MbDA) and cerebellum (Cb) neurons in distinct temporal windows and from spatially discrete progenitor domains. It has been shown that Wnt1 and Lmx1a participate in a cross-regulatory loop that is utilized during MbDA neuron development. However, Wnt1 expression dynamically changes over time and precedes that of Lmx1a. The spatial and temporal requirements of Wnt1 in development and specifically its requirement for MbDA neurons remain to be determined. To address these issues, we generated a conditional Wnt1 allele and temporally deleted Wnt1 coupled with genetic lineage analysis. Using this approach, we show that patterning of the midbrain (Mb) and Cb by Wnt1 occurs between the one-somite and the six- to eight-somite stages and is solely dependent on Wnt1 function in the Mb, but not in the Cb. Interestingly, an En1-derived domain persists after the early deletion of Wnt1 and mutant cells express OTX2. However, the En1-derived Wnt1-mutant domain does not contain LMX1a-expressing progenitors, and MbDA neurons are depleted. Thus, we demonstrate an early requirement of Wnt1 for all MbDA neurons. Subsequently, we deleted Wnt1 in the ventral Mb and show a continued late requirement for Wnt1 in MbDA neuron development, but not in LMX1a-expressing progenitors. Specifically, Wnt1 deletion disrupts the birthdating of MbDA neurons and causes a depletion of MbDA neurons positioned medially and a concomitant expansion of MbDA neurons positioned laterally during embryogenesis. Collectively, our analyses resolve the spatial and temporal function of Wnt1 in Mb and Cb patterning and in MbDA neuron development in vivo.


Asunto(s)
Diferenciación Celular/genética , Neuronas Dopaminérgicas/fisiología , Mesencéfalo/embriología , Proteína Wnt1/fisiología , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Diferenciación Celular/fisiología , Cerebelo/embriología , Cerebelo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Cinética , Mesencéfalo/citología , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Especificidad de Órganos/genética , Embarazo , Factores de Tiempo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
14.
Mol Cell Biochem ; 402(1-2): 213-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25618247

RESUMEN

Canonical Wnt signaling is thought to enhance osteogenic differentiation of human periodontal ligament fibroblasts (hPLFs). However, the mechanism of this enhancement has not yet been defined. We investigated the effects of Wnt1 on osteoblast differentiation of hPLFs and explored the mechanisms of the effects. Treating hPLFs with Wnt1 induced cytosolic accumulation and nuclear translocation of ß-catenin with concomitant increases in alkaline phosphatase (ALP) activity and calcium content in a time-dependent and dose-dependent manner. Wnt1-stimulated differentiation of hPLFs was accompanied by augmented phosphorylation of glycogen synthase kinase (GSK)-3ß and expression of the bone-specific factors runt-related transcription factor 2 (Runx2), osterix2 (Osx2), ALP, type I collagen, osteopontin, and osteocalcin. Pretreatment with Dickkopf-1 inhibited Wnt1-stimulated differentiation of hPLFs by suppressing GSK-3ß phosphorylation, nuclear translocation of ß-catenin, and expression of the bone-specific factors. Small interfering (si) RNA-mediated knockdown of ß-catenin, or pretreatment with FH535, markedly prevented Wnt1-stimulated differentiation of cells by blocking Runx2 and its downstream factors at the mRNA and protein levels. siRNA-mediated silencing of Runx2 also inhibited Wnt1-stimulated mineralization of cells, accompanied by a reduction in the levels of Osx2 and other early and late bone-formation regulatory factors. However, Wnt1-mediated nuclear translocation of ß-catenin and GSK-3ß phosphorylation were not inhibited by knockdown of Runx2 or FH535. Collectively, our findings suggested that Wnt1 stimulates osteogenic differentiation and mineralization of hPLFs, mainly by activating the canonical Wnt/ß-catenin pathway, in which Runx2 is a key downstream regulator.


Asunto(s)
Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Fibroblastos/fisiología , Osteoblastos/fisiología , Ligamento Periodontal/citología , Proteína Wnt1/fisiología , Adulto , Fosfatasa Alcalina/metabolismo , Calcificación Fisiológica , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Vía de Señalización Wnt , Adulto Joven , beta Catenina/genética , beta Catenina/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(9): 3389-94, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22331866

RESUMEN

The Drosophila melanogaster larval hematopoietic organ, the lymph gland, is a model to study in vivo the function of the hematopoietic niche. A small cluster of cells in the lymph gland, the posterior signaling center (PSC), maintains the balance between hematopoietic progenitors (prohemocytes) and their differentiation into specialized blood cells (hemocytes). Here, we show that Decapentaplegic/bone morphogenetic protein (Dpp/BMP) signaling activity in PSC cells controls niche size. In the absence of BMP signaling, the number of PSC cells increases. Correlatively, no hemocytes differentiate. Controlling PSC size is, thus, essential for normal blood cell homeostasis. Activation of BMP signaling in the PSC requires expression of the Dally-like heparan-sulfate proteoglycan, under the control of the Collier/early B-cell factor (EBF) transcription factor. A Dpp > dpp autoregulatory loop maintains BMP signaling, which limits PSC cell proliferation by repressing the protooncogene dmyc. Dpp antagonizes activity of wingless (Wg)/Wnt signaling, which positively regulates the number of PSC cells via the control of Dmyc expression. Together, our data show that Collier controls hemocyte homeostasis via coordinate regulation of PSC cell number and PSC signaling to prohemocytes. In mouse, EBF2, BMP, and Wnt signaling in osteoblasts is required for the proper number of niche and hematopoietic stem cells. Our findings bring insights to niche size control and draw parallels between Drosophila and mammalian hematopoiesis.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Hematopoyesis/fisiología , Hemocitos/citología , Nicho de Células Madre , Factores de Transcripción/fisiología , Animales , Recuento de Células , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Genes myc , Hemocitos/metabolismo , Larva , Ratones , Índice Mitótico , Proteoglicanos/genética , Proteoglicanos/fisiología , Transducción de Señal/fisiología , Especificidad de la Especie , Factores de Transcripción/genética , Vertebrados/fisiología , Proteína Wnt1/genética , Proteína Wnt1/fisiología
16.
J Am Soc Nephrol ; 25(8): 1760-73, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24578135

RESUMEN

In chronic kidney disease, vascular calcification, renal osteodystrophy, and phosphate contribute substantially to cardiovascular risk and are components of CKD-mineral and bone disorder (CKD-MBD). The cause of this syndrome is unknown. Additionally, no therapy addresses cardiovascular risk in CKD. In its inception, CKD-MBD is characterized by osteodystrophy, vascular calcification, and stimulation of osteocyte secretion. We tested the hypothesis that increased production of circulating factors by diseased kidneys causes the CKD-MBD in diabetic mice subjected to renal injury to induce stage 2 CKD (CKD-2 mice). Compared with non-CKD diabetic controls, CKD-2 mice showed increased renal production of Wnt inhibitor family members and higher levels of circulating Dickkopf-1 (Dkk1), sclerostin, and secreted klotho. Neutralization of Dkk1 in CKD-2 mice by administration of a monoclonal antibody after renal injury stimulated bone formation rates, corrected the osteodystrophy, and prevented CKD-stimulated vascular calcification. Mechanistically, neutralization of Dkk1 suppressed aortic expression of the osteoblastic transcription factor Runx2, increased expression of vascular smooth muscle protein 22-α, and restored aortic expression of klotho. Neutralization of Dkk1 did not affect the elevated plasma levels of osteocytic fibroblast growth factor 23 but decreased the elevated levels of sclerostin. Phosphate binder therapy restored plasma fibroblast growth factor 23 levels but had no effect on vascular calcification or osteodystrophy. The combination of the Dkk1 antibody and phosphate binder therapy completely treated the CKD-MBD. These results show that circulating Wnt inhibitors are involved in the pathogenesis of CKD-MBD and that the combination of Dkk1 neutralization and phosphate binding may have therapeutic potential for this disorder.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Fosfatos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Proteína Wnt1/fisiología , Proteínas Adaptadoras Transductoras de Señales , Adulto , Animales , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/etiología , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Animales de Enfermedad , Método Doble Ciego , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Klotho , Lantano/uso terapéutico , Masculino , Ratones Endogámicos C57BL , Fósforo Dietético , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/etiología
17.
Carcinogenesis ; 35(9): 2102-12, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24832086

RESUMEN

Many adult chronic diseases are thought to be influenced during early life by maternal nutrition; however, the underlying mechanisms remain largely unknown. Obesity-related diseases may be due partly to high fat consumption. Herein, we evaluated mammary tumor risk in female mouse mammary tumor virus-Wnt-1 transgenic (Tg) offspring exposed to high-fat diet (HFD) or control diet (CD) (45% and 17% kcal from fat, respectively) during gestation and lactation, with CD provided to progeny at weaning. In Tg offspring, maternal HFD exposure increased mammary tumor incidence and decreased tumor latency without affecting tumor volume. Tumor risk was associated with higher tumor necrosis factor-α and insulin and altered oxidative stress biomarkers in sera and with early changes in mammary expression of genes linked to tumor promotion [interleukin 6 (Il6)] or inhibition [phosphatase and tensin homolog deleted on chromosome 10 (Pten), B-cell lymphoma 2 (Bcl2)]. Corresponding wild-type progeny exposed to maternal HFD displayed accelerated mammary development, higher mammary adiposity, increased insulin resistance and early changes in Pten, Bcl2 and Il6, than CD-exposed offspring. Dams-fed HFD showed higher serum glucose and oxidative stress biomarkers but comparable adiposity compared with CD-fed counterparts. In human breast cancer MCF-7 cells, sera from maternal HFD-exposed Tg offspring elicited changes in PTEN, BCL2 and IL6 gene expression, mimicking in vivo exposure; increased cell viability and mammosphere formation and induced measures [insulin receptor substrate-1 (IRS-1), IRS-2] of insulin sensitivity. Serum effects on IRS-1 were recapitulated by exogenous insulin and the PTEN-specific inhibitor SF1670. Hyperinsulinemia and PTEN loss-of-function may thus, couple maternal HFD exposure to enhanced insulin sensitivity via increased mammary IRS-1 expression in progeny, to promote breast cancer risk.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Neoplasias Mamarias Experimentales/etiología , Efectos Tardíos de la Exposición Prenatal/etiología , Proteína Wnt1/fisiología , Animales , Animales Lactantes , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Lactancia , Células MCF-7 , Masculino , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Oxidativo , Fenotipo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Factores de Riesgo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
18.
Development ; 138(22): 4955-67, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22028028

RESUMEN

During metazoan development, the Wnt/Wingless signal transduction pathway is activated repetitively to direct cell proliferation, fate specification, differentiation and apoptosis. Distinct outcomes are elicited by Wnt stimulation in different cellular contexts; however, mechanisms that confer context specificity to Wnt signaling responses remain largely unknown. Starting with an unbiased forward genetic screen in Drosophila, we recently uncovered a novel mechanism by which the cell-specific co-factor Earthbound 1 (Ebd1), and its human homolog jerky, promote interaction between the Wnt pathway transcriptional co-activators ß-catenin/Armadillo and TCF to facilitate context-dependent Wnt signaling responses. Here, through the same genetic screen, we find an unanticipated requirement for Erect Wing (Ewg), the fly homolog of the human sequence-specific DNA-binding transcriptional activator nuclear respiratory factor 1 (NRF1), in promoting contextual regulation of Wingless signaling. Ewg and Ebd1 functionally interact with the Armadillo-TCF complex and mediate the same context-dependent Wingless signaling responses. In addition, Ewg and Ebd1 have similar cell-specific expression profiles, bind to each other directly and also associate with chromatin at shared genomic sites. Furthermore, recruitment of Ebd1 to chromatin is abolished in the absence of Ewg. Our findings provide in vivo evidence that recruitment of a cell-specific co-factor complex to specific chromatin sites, coupled with its ability to facilitate Armadillo-TCF interaction and transcriptional activity, promotes contextual regulation of Wnt/Wingless signaling responses.


Asunto(s)
Proteínas del Dominio Armadillo/metabolismo , Proteína B del Centrómero/metabolismo , Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Neuropéptidos/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Vía de Señalización Wnt/fisiología , Proteína Wnt1/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas del Dominio Armadillo/genética , Proteína B del Centrómero/genética , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/genética , Embrión no Mamífero , Células HEK293 , Humanos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Especificidad de Órganos/genética , Unión Proteica/fisiología , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Factores de Transcripción/genética , Vía de Señalización Wnt/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(27): 11139-44, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690416

RESUMEN

Sexual dimorphism is widespread throughout the metazoa and plays important roles in mate recognition and preference, sex-based niche partitioning, and sex-specific coadaptation. One notable example of sex-specific differences in insect body morphology is presented by the higher diptera, such as Drosophila, in which males develop fewer abdominal segments than females. Because diversity in segment number is a distinguishing feature of major arthropod clades, it is of fundamental interest to understand how different numbers of segments can be generated within the same species. Here we show that sex-specific and segment-specific regulation of the Wingless (Wg) morphogen underlies the development of sexually dimorphic adult segment number in Drosophila. Wg expression is repressed in the developing terminal male abdominal segment by the combination of the Hox protein Abdominal-B (Abd-B) and the sex-determination regulator Doublesex (Dsx). The subsequent loss of the terminal male abdominal segment during pupation occurs through a combination of developmental processes including segment compartmental transformation, apoptosis, and suppression of cell proliferation. Furthermore, we show that ectopic expression of Wg is sufficient to rescue this loss. We propose that dimorphic Wg regulation, in concert with monomorphic segment-specific programmed cell death, are the principal mechanisms of sculpting the sexually dimorphic abdomen of Drosophila.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Proteína Wnt1/genética , Abdomen/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Apoptosis/genética , Apoptosis/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Masculino , Mitosis/genética , Mitosis/fisiología , Morfogénesis/genética , Morfogénesis/fisiología , Caracteres Sexuales , Alas de Animales/crecimiento & desarrollo , Proteína Wnt1/fisiología
20.
Carcinogenesis ; 34(2): 464-74, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23144318

RESUMEN

Despite the well-accepted notion that early maternal influences persist beyond fetal life and may underlie many adult diseases, the risks imposed by the maternal environment on breast cancer development and underlying biological mechanisms remain poorly understood. In this study, we investigated whether early exposure to blueberry (BB) via maternal diet alters oncogene Wnt1-induced mammary tumorigenesis in offspring. Wnt1-transgenic female mice were exposed to maternal Casein (CAS, control) or blueberry-supplemented (CAS + 3%BB) diets throughout pregnancy and lactation. Offspring were weaned to CAS and mammary tumor development was followed until age 8 months. Tumor incidence and latency were similar for both groups; however, tumor weight at killing and tumor volume within 2 weeks of initial detection were lower (by 50 and 60%, respectively) in offspring of BB- versus control-fed dams. Dietary BB exposure beginning at weaning did not alter mammary tumor parameters. Tumors from maternal BB-exposed offspring showed higher tumor suppressor (Pten and Cdh1) and lower proproliferative (Ccnd1), anti-apoptotic (Bcl2) and proangiogenic (Figf, Flt1 and Ephb4) transcript levels, and displayed attenuated microvessel density. Expression of Pten and Cdh1 genes was also higher in mammary tissues of maternal BB-exposed offspring. Mammary tissues and tumors of maternal BB-exposed offspring showed increased chromatin-modifying enzyme Dnmt1 and Ezh2 transcript levels. Body weight, serum insulin and serum leptin/adiponectin ratio were lower for maternal BB-exposed than control tumor-bearing offspring. Tumor weights and serum insulin were positively correlated. Results suggest that dietary influences on the maternal environment contribute to key developmental programs in the mammary gland to modify breast cancer outcome in adult progeny.


Asunto(s)
Arándanos Azules (Planta) , Dieta , Insulina/sangre , Neoplasias Mamarias Animales/prevención & control , Fitoterapia , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteína Wnt1/fisiología , Animales , Western Blotting , Femenino , Humanos , Técnicas para Inmunoenzimas , Lactancia , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Transgénicos , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA