Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
FASEB J ; 34(5): 6791-6807, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32223017

RESUMO

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.


Assuntos
Osso e Ossos/patologia , Cartilagem/patologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/fisiologia , Má Oclusão/patologia , Crista Neural/patologia , Proteína Wnt1/fisiologia , Animais , Osso e Ossos/metabolismo , Cartilagem/metabolismo , Condrogênese , Anormalidades Craniofaciais , Feminino , Masculino , Má Oclusão/genética , Má Oclusão/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Crista Neural/metabolismo , Fenótipo
2.
Brain Res ; 1735: 146726, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32057808

RESUMO

Olfactory ensheathing cells (OECs) are special glial cells localized in olfactory system which secrete large number of neurotrophic factors and promote the migration and survival of neurons. Canonical Wnt/ß-catenin signaling is activated in OECs during development and facilitates the growth and regeneration of axons. In the present study, we investigated the effects of Wnt-activated OECs on the proliferation and differentiation of neural stem cells (NSCs) in vitro. Primary neonatal mouse OECs were cultured and identified by immunostaining of P75 and GFAP. Wnt activation was achieved by lentivirus transfection of dominant-active-ß-catenin (EbC) and examined by immunostaining of PY489. The conditioned medium (CM) of Wnt-activated OECs (wOECs-CM) and control OECs (cOECs-CM) was used to stimulate NSCs. In proliferation assay, wOECs-CM could increase the percentage of Ki67/Sox2 double positive cells. Under differentiation condition, wOECs-CM could maintain the expression of Nestin and promote the differentiation of NSCs into Tuj1-positive neurons. Taken together, our data revealed that wOECs stimulates the proliferation and differentiation of NSCs in a secretory manner, indicating that combined transplantation of wOECs and NSCs may be beneficial for regeneration.


Assuntos
Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Animais , Axônios/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/química , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/metabolismo , Células-Tronco Neurais/fisiologia , Neuroglia/metabolismo , Olfato/fisiologia , Via de Sinalização Wnt/fisiologia , Proteína Wnt1/metabolismo , Proteína Wnt1/fisiologia
3.
PLoS Genet ; 15(9): e1008351, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31527874

RESUMO

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.


Assuntos
Proteínas de Drosophila/metabolismo , Via Secretória/fisiologia , Via de Sinalização Wnt/fisiologia , Proteína Wnt1/metabolismo , Animais , Padronização Corporal/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Células Epiteliais/metabolismo , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Ligantes , Transdução de Sinais , Asas de Animais/embriologia , Asas de Animais/metabolismo , Proteínas Wnt/genética , Via de Sinalização Wnt/genética , Proteína Wnt1/genética , Proteína Wnt1/fisiologia
4.
Arterioscler Thromb Vasc Biol ; 39(8): 1629-1644, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31189430

RESUMO

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.


Assuntos
Adipócitos Marrons/citologia , Adipogenia , Tecido Adiposo/fisiologia , Crista Neural/citologia , Tecido Adiposo/citologia , Angiotensina II/farmacologia , Animais , Aorta Torácica/citologia , Artéria Carótida Primitiva/citologia , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/fisiologia , Proteína Wnt1/fisiologia
5.
Dis Model Mech ; 12(7)2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31213486

RESUMO

The Wnt gene family encodes an evolutionarily conserved group of proteins that regulate cell growth, differentiation and stem cell self-renewal. Aberrant Wnt signaling in human breast tumors has been proposed as a driver of tumorigenesis, especially in the basal-like tumor subtype where canonical Wnt signaling is both enriched and predictive of poor clinical outcomes. The development of effective Wnt-based therapeutics, however, has been slowed in part by a limited understanding of the context-dependent nature with which these aberrations influence breast tumorigenesis. We previously reported that MMTV-Wnt1 mice, an established model for studying Wnt signaling in breast tumors, develop two subtypes of tumors by gene expression classification: Wnt1-EarlyEx and Wnt1-LateEx Here, we extend this initial observation and show that Wnt1-EarlyEx tumors exhibit high expression of canonical Wnt, non-canonical Wnt, and EGFR signaling pathway signatures. Therapeutically, Wnt1-EarlyEx tumors showed a dynamic reduction in tumor volume when treated with an EGFR inhibitor. Wnt1-EarlyEx tumors had primarily Cd49fpos/Epcamneg FACS profiles, but it was not possible to serially transplant these tumors into wild-type FVB female mice. Conversely, Wnt1-LateEx tumors had a bloody gross pathology, which was highlighted by the presence of 'blood lakes' identified by H&E staining. These tumors had primarily Cd49fpos/Epcampos FACS profiles, but also contained a secondary Cd49fpos/Epcamneg subpopulation. Wnt1-LateEx tumors were enriched for activating Hras1 mutations and were capable of reproducing tumors when serially transplanted into wild-type FVB female mice. This study definitively shows that the MMTV-Wnt1 mouse model produces two phenotypically distinct subtypes of mammary tumors that differ in multiple biological aspects including sensitivity to an EGFR inhibitor.


Assuntos
Antineoplásicos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Proteína Wnt1/fisiologia , Animais , Antineoplásicos/farmacologia , Carcinogênese , Separação Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Genes ras , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/virologia , Camundongos , Fenótipo , Via de Sinalização Wnt , Proteína Wnt1/metabolismo
6.
Biochem Pharmacol ; 162: 3-13, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30879494

RESUMO

Autophagy-dependent cell death is a distinct mode of regulated cell death required in a context specific manner. One of the best validated genetic models of autophagy-dependent cell death is the removal of the Drosophila larval midgut during larval-pupal transition. We have previously shown that down-regulation of growth signaling is essential for autophagy induction and larval midgut degradation. Sustained growth signaling through Ras and PI3K blocks autophagy and consequently inhibits midgut degradation. In addition, the morphogen Dpp plays an important role in regulating the correct timing of midgut degradation. Here we explore the potential roles of Hh and Wg signaling in autophagy-dependent midgut cell death. We demonstrate that Hh and Wg signaling are not involved in the regulation of autophagy-dependent cell death. However, surprisingly we found that one key component of these pathways, the Drosophila Glycogen Synthase Kinase 3, Shaggy (Sgg), may regulate midgut cell size independent of Hh and Wg signaling.


Assuntos
Morte Celular Autofágica/fisiologia , Proteínas de Drosophila/fisiologia , Proteínas Hedgehog/fisiologia , Transdução de Sinais/fisiologia , Proteína Wnt1/fisiologia , Animais , Animais Geneticamente Modificados , Drosophila
7.
PLoS Genet ; 14(8): e1007568, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30142157

RESUMO

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.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Elementos Facilitadores Genéticos , Receptores ErbB/fisiologia , Extremidades/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Peptídeos de Invertebrados/fisiologia , Alelos , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/crescimento & desenvolvimento , Receptores ErbB/genética , Deleção de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Discos Imaginais/fisiologia , Neurregulinas/genética , Neurregulinas/fisiologia , Organizadores Embrionários , Receptores de Peptídeos de Invertebrados/genética , Transdução de Sinais , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/fisiologia , Proteína Wnt1/genética , Proteína Wnt1/fisiologia
8.
Dev Biol ; 439(2): 53-64, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29715461

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica/métodos , Imagem Óptica/métodos , Proteína Wnt1/metabolismo , Aciltransferases/metabolismo , Animais , Embrião de Galinha , Galinhas/metabolismo , Ectoderma/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipoilação , Proteínas de Membrana/metabolismo , Camundongos , Crista Neural/metabolismo , Processamento de Proteína Pós-Traducional , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia , Proteína Wnt1/fisiologia , Proteína Wnt3A/metabolismo , Peixe-Zebra/metabolismo
9.
Oncotarget ; 7(44): 71960-71973, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27713126

RESUMO

18ß-glycyrrhetinic acid (GRA) exerts anti-tumor effects on various types of cancer. In the present study, we found that GRA attenuated the severity of gastritis and suppressed gastric tumorigenesis in transgenic mice. We also discovered that miR-149-3p was downregulated in gastric cancer tissues and cell lines as compared to normal gastric tissues and epithelial cells, but was upregulated by GRA. miR-149-3p expression also correlated negatively with lymphnode metastasis. Our functional assays showed that miR-149-3p overexpression inhibited cell proliferation and cell cycle progression while inducing apoptosis, while inhibition of miR-149-3p had the opposite effects. In addition, we identified Wnt-1 as a direct target of miR-149-3p. These data suggest that GRA inhibits the initiation and progression of gastric tumors by ameliorating the inflammatory microenvironment through downregulation of COX-2 expression and by inhibiting Wnt-1 expression through the upregulation of tumor suppressor miR-149-3p. GRA may thus have the potential to serve as a useful therapeutic agent for the prevention and treatment of gastric cancer.


Assuntos
Ácido Glicirretínico/análogos & derivados , MicroRNAs/fisiologia , Neoplasias Gástricas/prevenção & controle , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt1/fisiologia , Adulto , Idoso , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/análise , Feminino , Ácido Glicirretínico/farmacologia , Humanos , Metástase Linfática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neoplasias Gástricas/patologia , Proteína Wnt1/genética
10.
PLoS Biol ; 14(9): e1002536, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27584613

RESUMO

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.


Assuntos
Apoptose/efeitos da radiação , Proteínas de Drosophila/fisiologia , Células Epiteliais/efeitos da radiação , Discos Imaginais/fisiologia , Fatores de Transcrição STAT/fisiologia , Proteína Wnt1/fisiologia , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/efeitos da radiação , Células Epiteliais/fisiologia , Expressão Gênica , Inativação Gênica/efeitos da radiação , Discos Imaginais/citologia , Larva/citologia , Larva/fisiologia , Larva/efeitos da radiação , Lesões Experimentais por Radiação , Regeneração
11.
Dev Biol ; 415(2): 228-241, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988119

RESUMO

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.


Assuntos
Nervos Cranianos/citologia , Células Receptoras Sensoriais/citologia , Animais , Apoptose , Axônios/fisiologia , Diferenciação Celular , Linhagem da Célula , Nervos Cranianos/embriologia , Ectoderma/citologia , Fatores de Crescimento de Fibroblastos/fisiologia , Gânglios Sensitivos/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/citologia , Neurogênese , Fatores de Transcrição/genética , Tretinoína/fisiologia , Proteína Wnt1/fisiologia
12.
J Neurosci ; 35(39): 13385-401, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26424886

RESUMO

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.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Mesencéfalo/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco Pluripotentes/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Contagem de Células , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Sobrevivência Celular/genética , Células Cultivadas , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transcriptoma , Proteína Wnt1/genética , Proteína Wnt1/fisiologia
13.
Cancer Prev Res (Phila) ; 8(6): 492-501, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25813526

RESUMO

Loss of parietal cells initiates the development of spasmolytic polypeptide-expressing metaplasia (SPEM), a precancerous lesion in stomach. CD44 variant (CD44v) that enhances the ability to defend against reactive oxygen species (ROS) in epithelial cells is expressed de novo in SPEM of K19-Wnt1/C2mE mice, a transgenic model of gastric tumorigenesis, and is required for the efficient development of SPEM and gastric tumor in these animals. The role of ROS and its downstream signaling in CD44-dependent gastric tumorigenesis has remained unknown, however. With the use of the K19-Wnt1/C2mE mouse, we now show that parietal cells in the inflamed stomach are highly sensitive to oxidative stress and manifest activation of p38(MAPK) signaling by ROS. Oral treatment with the antioxidant ascorbic acid or genetic ablation of the Ink4a/Arf locus, a major downstream target of ROS-p38(MAPK) signaling, inhibited parietal cell loss and the subsequent gastric tumorigenesis. Our results indicate that signaling activated by oxidative stress in parietal cells plays a key role in CD44-dependent gastric tumorigenesis. .


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Transformação Celular Neoplásica/patologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Receptores de Hialuronatos/metabolismo , Estresse Oxidativo , Células Parietais Gástricas/patologia , Neoplasias Gástricas/patologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Técnicas Imunoenzimáticas , Metaplasia/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Knockout , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células Parietais Gástricas/metabolismo , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Proteína Wnt1/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
PLoS One ; 10(3): e0119467, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25799576

RESUMO

OBJECTIVES: Enteric neural stem cells provide hope of curative treatment for enteric neuropathies. Current protocols for their harvesting from humans focus on the generation of 'neurospheres' from cultures of dissociated gut tissue. The study aims to better understand the derivation, generation and composition of enteric neurospheres. DESIGN: Gut tissue was obtained from Wnt1-Cre;Rosa26Yfp/Yfp transgenic mice (constitutively labeled neural crest cells) and paediatric patients. Gut cells were cultured either unsorted (mixed neural crest/non-neural crest), or following FACS selection into neural crest (murine-YFP+ve/human-p75+ve) or non-neural crest (YFP-ve/p75-ve) populations. Cultures and resultant neurospheres were characterized using immunolabelling in vitro and following transplantation in vivo. RESULTS: Cultures of (i) unsorted, (ii) neural crest, and (iii) non-neural crest cell populations generated neurospheres similar in numbers, size and morphology. Unsorted neurospheres were highly heterogeneous for neural crest content. Neural crest-derived (YFP+ve/p75+ve) neurospheres contained only neural derivatives (neurons and glia) and were devoid of non-neural cells (i.e. negative for SMA, c-Kit), with the converse true for non-neural crest-derived (YFP-ve/p75-ve) 'neurospheres'. Under differentiation conditions only YFP+ve cells gave rise to neural derivatives. Both YFP+ve and YFP-ve cells displayed proliferation and spread upon transplantation in vivo, but YFP-ve cells did not locate or integrate within the host ENS. CONCLUSIONS: Spherical accumulations of cells, so-called 'neurospheres' forming in cultures of dissociated gut contain variable proportions of neural crest-derived cells. If they are to be used for ENS cell replacement therapy then improved protocols for their generation, including cell selection, should be sought in order to avoid inadvertent transplantation of non-therapeutic, non-ENS cells.


Assuntos
Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Sistema Nervoso Entérico/citologia , Trato Gastrointestinal/citologia , Crista Neural/citologia , Células-Tronco Neurais/citologia , Animais , Proteínas de Bactérias/metabolismo , Células Cultivadas , Sistema Nervoso Entérico/metabolismo , Feminino , Trato Gastrointestinal/metabolismo , Humanos , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Proteína Wnt1/fisiologia
15.
Mol Cell Biochem ; 402(1-2): 213-23, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25618247

RESUMO

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.


Assuntos
Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Fibroblastos/fisiologia , Osteoblastos/fisiologia , Ligamento Periodontal/citologia , Proteína Wnt1/fisiologia , Adulto , Fosfatase Alcalina/metabolismo , Calcificação Fisiológica , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Via de Sinalização Wnt , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
16.
Oncogene ; 34(34): 4429-38, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25381821

RESUMO

Understanding the factors contributing to tumor initiation, progression and evolution is of paramount significance. Among them, wild-type p53-induced phosphatase 1 (Wip1) is emerging as an important oncogene by virtue of its negative control on several key tumor suppressor pathways. Originally discovered as a p53-regulated gene, Wip1 has been subsequently found amplified and more recently mutated in a significant fraction of human cancers including breast tumors. Recent development in the field further uncovered the utility of anti-Wip1-directed therapies in delaying tumor onset or in reducing the tumor burden. Furthermore, Wip1 could be an important factor that contributes to tumor heterogeneity, suggesting that its inhibition may decrease the rate of cancer evolution. These effects depend on several signaling pathways modulated by Wip1 phosphatase in a spatial and temporal manner. In this review we discuss the recent development in understanding how Wip1 contributes to tumorigenesis with its relevance to breast cancer.


Assuntos
Neoplasias da Mama/etiologia , Fosfoproteínas Fosfatases/fisiologia , Mama/crescimento & desenvolvimento , Citidina Desaminase/genética , Dano ao DNA , Feminino , Genes BRCA2 , Genes p53 , Heterocromatina , Humanos , Antígenos de Histocompatibilidade Menor , Mutação , Oncogenes , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 2C , Receptor ErbB-2/fisiologia , Proteína Wnt1/fisiologia
17.
PLoS One ; 9(11): e113247, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25401739

RESUMO

Wnt5a is a non-canonical signaling Wnt that has been implicated in tumor suppression. We previously showed that loss of Wnt5a in MMTV-PyVmT tumors resulted in a switch in tumor phenotype resulting in tumors with increased basal phenotype and high Wnt/ß-catenin signaling. The object of this study was to test the hypothesis that Wnt5a can act to inhibit tumors formed by activation of Wnt/ß-catenin signaling. To this end, we characterized tumor and non-tumor mammary tissue from MMTV-Wnt1 and double transgenic MMTV-Wnt1;MMTV-Wnt5a mice. Wnt5a containing mice demonstrated fewer tumors with increased latency when compared to MMTV-Wnt1 controls. Expression of markers for basal-like tumors was down-regulated in the tumors that formed in the presence of Wnt5a indicating a phenotypic switch. Reduced canonical Wnt signaling was detected in double transgenic tumors as a decrease in active ß-catenin protein and a decrease in Axin2 mRNA transcript levels. In non-tumor tissues, over-expression of Wnt5a in MMTV-Wnt1 mammary glands resulted in attenuation of phenotypes normally observed in MMTV-Wnt1 glands including hyperbranching and increased progenitor and basal cell populations. Even though Wnt5a could antagonize Wnt/ß-catenin signaling in primary mammary epithelial cells in culture, reduced Wnt/ß-catenin signaling was not detected in non-tumor MMTV-Wnt1;Wnt5a tissue in vivo. The data demonstrate that Wnt5a suppresses tumor formation and promotes a phenotypic shift in MMTV-Wnt1 tumors.


Assuntos
Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Proteínas Wnt/metabolismo , Proteína Wnt1/fisiologia , Animais , Western Blotting , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Imunofluorescência , Técnicas Imunoenzimáticas , Masculino , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Wnt/genética , Proteína Wnt-5a , beta Catenina
18.
Development ; 141(12): 2483-93, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24917503

RESUMO

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.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteína Wnt1/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Receptores Frizzled/genética , Ligantes , Modelos Biológicos , Fenótipo , Regiões Promotoras Genéticas , Transdução de Sinais , Transcrição Gênica , Regulação para Cima , Asas de Animais/embriologia , Proteína Wnt1/genética
19.
Carcinogenesis ; 35(9): 2102-12, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24832086

RESUMO

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.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Neoplasias Mamárias Experimentais/etiologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Proteína Wnt1/fisiologia , Animais , Animais Lactentes , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Lactação , Células MCF-7 , Masculino , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estresse Oxidativo , Fenótipo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Fatores de Risco , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
20.
J Am Soc Nephrol ; 25(8): 1760-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24578135

RESUMO

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.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Fosfatos/metabolismo , Insuficiência Renal Crônica/metabolismo , Proteína Wnt1/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Animais , Doenças Ósseas Metabólicas/tratamento farmacológico , Doenças Ósseas Metabólicas/etiologia , Proteínas Morfogenéticas Ósseas/metabolismo , Modelos Animais de Doenças , Método Duplo-Cego , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Glicoproteínas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Klotho , Lantânio/uso terapêutico , Masculino , Camundongos Endogâmicos C57BL , Fósforo na Dieta , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...