Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Res Sq ; 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38746309

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive scarring disease arising from the maladaptive differentiation of lung stem cells into bronchial epithelial cells rather than into alveolar type 1 (AT1) cells, which are responsible for gas exchange. Here, we report that healthy lungs maintain their stem cells through tonic Hippo and ß-catenin signaling, which promote Yap/Taz degradation and allow for low level expression of the Wnt target gene Myc. Inactivation of upstream activators of the Hippo pathway in lung stem cells inhibits this tonic ß-catenin signaling and Myc expression and promotes their Taz mediated differentiation into AT1 cells. Vice versa, increased Myc in collaboration with Yap promotes the differentiation of lung stem cells along the basal and myoepithelial like lineages allowing them to invade and bronchiolize the lung parenchyma in a process reminiscent of submucosal gland development. Our findings indicate that stem cells exhibiting the highest Myc levels become supercompetitors that drive remodeling, whereas loser cells with lower Myc levels terminally differentiate into AT1 cells.

2.
Elife ; 122023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37166104

RESUMO

Idiopathic pulmonary fibrosis (IPF) consists of fibrotic alveolar remodeling and progressive loss of pulmonary function. Genetic and experimental evidence indicates that chronic alveolar injury and failure to properly repair the respiratory epithelium are intrinsic to IPF pathogenesis. Loss of alveolar type 2 (AT2) stem cells or mutations that either impair their self-renewal and/or impair their differentiation into AT1 cells can serve as a trigger of pulmonary fibrosis. Recent reports indicate increased YAP activity in respiratory epithelial cells in IPF lungs. Individual IPF epithelial cells with aberrant YAP activation in bronchiolized regions frequently co-express AT1, AT2, conducting airway selective markers and even mesenchymal or EMT markers, demonstrating 'indeterminate' states of differentiation and suggesting that aberrant YAP signaling might promote pulmonary fibrosis. Yet, Yap and Taz have recently also been shown to be important for AT1 cell maintenance and alveolar epithelial regeneration after Streptococcus pneumoniae-induced injury. To investigate how epithelial Yap/Taz might promote pulmonary fibrosis or drive alveolar epithelial regeneration, we inactivated the Hippo pathway in AT2 stem cells resulting in increased nuclear Yap/Taz, and found that this promotes their alveolar regenerative capacity and reduces pulmonary fibrosis following bleomycin injury by pushing them along the AT1 cell lineage. Vice versa, inactivation of both Yap1 and Wwtr1 (encoding Taz) or Wwtr1 alone in AT2 cell stem cells impaired alveolar epithelial regeneration and resulted in increased pulmonary fibrosis upon bleomycin injury. Interestingly, the inactivation of only Yap1 in AT2 stem cells promoted alveolar epithelial regeneration and reduced pulmonary fibrosis. Together, these data suggest that epithelial Yap promotes, and epithelial Taz reduces pulmonary fibrosis suggesting that targeting Yap but not Taz-mediated transcription might help promote AT1 cell regeneration and treat pulmonary fibrosis.


Assuntos
Via de Sinalização Hippo , Fibrose Pulmonar Idiopática , Humanos , Pulmão/patologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Fatores de Transcrição/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Bleomicina/toxicidade , Bleomicina/metabolismo
3.
Cell Rep ; 41(12): 111863, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36543133

RESUMO

In injured airways of the adult lung, epithelial progenitors are called upon to repair by nearby mesenchymal cells via signals transmitted through the niche. Currently, it is unclear whether repair is coordinated by the mesenchymal cells that maintain the niche or by the airway epithelial cells that occupy it. Here, we show that the spatiotemporal expression of Fgf10 by the niche is primarily orchestrated by the niche's epithelial occupants-both those that reside prior to, and following, injury. During homeostasis, differentiated airway epithelial cells secrete Sonic hedgehog (Shh) to inhibit Fgf10 expression by Gli1+ peribronchial mesenchymal cells in the niche. After injury, remaining epithelial cells produce Wnt7b to induce Fgf10 expression in airway smooth muscle cells in the niche. We find that this reliance on a common activator of airway epithelial stem cells also allows for the recruitment of remote stem cell populations when local populations have been exhausted.


Assuntos
Proteínas Hedgehog , Células-Tronco Mesenquimais , Proteínas Hedgehog/metabolismo , Pulmão/metabolismo , Diferenciação Celular , Células Epiteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo
4.
Front Pharmacol ; 11: 120, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194398

RESUMO

Fgfr1 (Fibroblast growth factor receptor 1) and Fgfr2 are dynamically expressed during lung development, homeostasis, and regeneration. Our current analysis indicates that Fgfr2 is expressed in distal epithelial progenitors AT2, AT1, club, and basal cells but not in ciliated or neuroendocrine cells during lung development and homeostasis. However, after injury, Fgfr2 becomes upregulated in neuroendocrine cells and distal club cells. Epithelial Fgfr1 expression is minimal throughout lung development, homeostasis, and regeneration. We further find both Fgfr1 and Fgfr2 strongly expressed in cartilage progenitors and airway smooth muscle cells during lung development, whereas Fgfr1 but not Fgfr2 was expressed in lipofibroblasts and vascular smooth muscle cells. In the adult lung, Fgfr1 and Fgfr2 were mostly downregulated in smooth muscle cells but became upregulated after injury. Fgfr1 remained expressed in mesenchymal alveolar niche cells or lipofibroblasts with lower levels of expression in their descendant (alveolar) myofibroblasts during alveologenesis.

5.
Stem Cell Reports ; 12(5): 1041-1055, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31056475

RESUMO

Idiopathic pulmonary fibrosis is a common form of interstitial lung disease resulting in alveolar remodeling and progressive loss of pulmonary function because of chronic alveolar injury and failure to regenerate the respiratory epithelium. Histologically, fibrotic lesions and honeycomb structures expressing atypical proximal airway epithelial markers replace alveolar structures, the latter normally lined by alveolar type 1 (AT1) and AT2 cells. Bronchial epithelial stem cells (BESCs) can give rise to AT2 and AT1 cells or honeycomb cysts following bleomycin-mediated lung injury. However, little is known about what controls this binary decision or whether this decision can be reversed. Here we report that inactivation of Fgfr2b in BESCs impairs their contribution to both alveolar epithelial regeneration and honeycomb cysts after bleomycin injury. By contrast overexpression of Fgf10 in BESCs enhances fibrosis resolution by favoring the more desirable outcome of alveolar epithelial regeneration over the development of pathologic honeycomb cysts.


Assuntos
Células Epiteliais Alveolares/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Lesão Pulmonar/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Mucosa Respiratória/metabolismo , Células-Tronco/metabolismo , Células Epiteliais Alveolares/citologia , Animais , Bleomicina , Linhagem Celular , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Humanos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/genética , Masculino , Camundongos Knockout , Camundongos Transgênicos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Regeneração/genética , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Transdução de Sinais/genética , Células-Tronco/citologia
6.
Development ; 146(2)2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651296

RESUMO

Organ growth and tissue homeostasis rely on the proliferation and differentiation of progenitor cell populations. In the developing lung, localized Fgf10 expression maintains distal Sox9-expressing epithelial progenitors and promotes basal cell differentiation in the cartilaginous airways. Mesenchymal Fgf10 expression is induced by Wnt signaling but inhibited by Shh signaling, and epithelial Fgf10 signaling activates ß-catenin signaling. The Hippo pathway is a well-conserved signaling cascade that regulates organ size and stem/progenitor cell behavior. Here, we show that Hippo signaling promotes lineage commitment of lung epithelial progenitors by curbing Fgf10 and ß-catenin signaling. Our findings show that both inactivation of the Hippo pathway (nuclear Yap) or ablation of Yap result in increased ß-catenin and Fgf10 signaling, suggesting a cytoplasmic role for Yap in epithelial lineage commitment. We further demonstrate redundant and non-redundant functions for the two nuclear effectors of the Hippo pathway, Yap and Taz, during lung development.


Assuntos
Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Pulmão/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Citoplasma/metabolismo , Feminino , Via de Sinalização Hippo , Pulmão/embriologia , Masculino , Camundongos , Modelos Biológicos , Organogênese , Fenótipo , Fosfoproteínas/metabolismo , Alvéolos Pulmonares/embriologia , Transativadores , Proteínas de Sinalização YAP
7.
Mol Aspects Med ; 65: 56-69, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30130563

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive and terminal lung disease with no known cure. IPF is a disease of aging, with median age of diagnosis over 65 years. Median survival is between 3 and 5 years after diagnosis. IPF is characterized primarily by excessive deposition of extracellular matrix (ECM) proteins by activated lung fibroblasts and myofibroblasts, resulting in reduced gas exchange and impaired pulmonary function. Growing evidence supports the concept of a pro-fibrotic environment orchestrated by underlying factors such as genetic predisposition, chronic injury and aging, oxidative stress, and impaired regenerative responses may account for disease development and persistence. Currently, two FDA approved drugs have limited efficacy in the treatment of IPF. Many of the genes and gene networks associated with lung development are induced or activated in IPF. In this review, we analyze current knowledge in the field, gained from both basic and clinical research, to provide new insights into the disease process, and potential approaches to treatment of pulmonary fibrosis.


Assuntos
Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Animais , Biomarcadores , Microambiente Celular , Suscetibilidade a Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Miofibroblastos/metabolismo , Fibrose Pulmonar/patologia , Transdução de Sinais , Células Estromais , Transativadores , Fator de Crescimento Transformador beta/metabolismo
8.
Front Genet ; 9: 418, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319693

RESUMO

The lung is morphologically structured into a complex tree-like network with branched airways ending distally in a large number of alveoli for efficient oxygen exchange. At the cellular level, the adult lung consists of at least 40-60 different cell types which can be broadly classified into epithelial, endothelial, mesenchymal, and immune cells. Fibroblast growth factor 10 (Fgf10) located in the lung mesenchyme is essential to regulate epithelial proliferation and lineage commitment during embryonic development and post-natal life, and to drive epithelial regeneration after injury. The cells that express Fgf10 in the mesenchyme are progenitors for mesenchymal cell lineages during embryonic development. During adult lung homeostasis, Fgf10 is expressed in mesenchymal stromal niches, between cartilage rings in the upper conducting airways where basal cells normally reside, and in the lipofibroblasts adjacent to alveolar type 2 cells. Fgf10 protects and promotes lung epithelial regeneration after different types of lung injuries. An Fgf10-Hippo epithelial-mesenchymal crosstalk ensures maintenance of stemness and quiescence during homeostasis and basal stem cell (BSC) recruitment to further promote regeneration in response to injury. Fgf10 signaling is dysregulated in different human lung diseases including bronchopulmonary dysplasia (BPD), idiopathic pulmonary fibrosis (IPF), and chronic obstructive pulmonary disease (COPD), suggesting that dysregulation of the FGF10 pathway is critical to the pathogenesis of several human lung diseases.

9.
Dev Cell ; 43(1): 48-59.e5, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-29017029

RESUMO

The lung harbors its basal stem/progenitor cells (BSCs) in the protected environment of the cartilaginous airways. After major lung injuries, BSCs are activated and recruited to sites of injury. Here, we show that during homeostasis, BSCs in cartilaginous airways maintain their stem cell state by downregulating the Hippo pathway (resulting in increased nuclear Yap), which generates a localized Fgf10-expressing stromal niche; in contrast, differentiated epithelial cells in non-cartilaginous airways maintain quiescence by activating the Hippo pathway and inhibiting Fgf10 expression in airway smooth muscle cells (ASMCs). However, upon injury, surviving differentiated epithelial cells spread to maintain barrier function and recruit integrin-linked kinase to adhesion sites, which leads to Merlin degradation, downregulation of the Hippo pathway, nuclear Yap translocation, and expression and secretion of Wnt7b. Epithelial-derived Wnt7b, then in turn, induces Fgf10 expression in ASMCs, which extends the BSC niche to promote regeneration.


Assuntos
Diferenciação Celular/fisiologia , Fator 10 de Crescimento de Fibroblastos/metabolismo , Pulmão/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Epiteliais/citologia , Via de Sinalização Hippo , Camundongos Transgênicos , Miócitos de Músculo Liso/citologia , Fosfoproteínas/metabolismo
10.
Dev Dyn ; 244(3): 342-66, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25470458

RESUMO

BACKGROUND: The adaptation to terrestrial life required the development of an organ capable of efficient air-blood gas exchange. To meet the metabolic load of cellular respiration, the mammalian respiratory system has evolved from a relatively simple structure, similar to the two-tube amphibian lung, to a highly complex tree-like system of branched epithelial airways connected to a vast network of gas exchanging units called alveoli. The development of such an elaborate organ in a relatively short time window is therefore an extraordinary feat and involves an intimate crosstalk between mesodermal and endodermal cell lineages. RESULTS: This review describes the molecular processes governing lung development with an emphasis on the current knowledge on the role of Wnt and FGF signaling in lung epithelial differentiation. CONCLUSIONS: The Wnt and FGF signaling pathways are crucial for the dynamic and reciprocal communication between epithelium and mesenchyme during lung development. In addition, some of this developmental crosstalk is reemployed in the adult lung after injury to drive regeneration, and may, when aberrantly or chronically activated, result in chronic lung diseases. Novel insights into how the Wnt and FGF pathways interact and are integrated into a complex gene regulatory network will not only provide us with essential information about how the lung regenerates itself, but also enhance our understanding of the pathogenesis of chronic lung diseases, as well as improve the controlled differentiation of lung epithelium from pluripotent stem cells.


Assuntos
Linhagem da Célula/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Pulmão/embriologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Humanos , Pulmão/citologia
11.
PLoS One ; 7(6): e38452, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22719891

RESUMO

Fibroblast growth factor 10 (Fgf10) is a key regulator of diverse organogenetic programs during mouse development, particularly branching morphogenesis. Fgf10-null mice suffer from lung and limb agenesis as well as cecal and colonic atresia and are thus not viable. To date, the Mlcv1v-nLacZ-24 transgenic mouse strain (referred to as Fgf10(LacZ)), which carries a LacZ insertion 114 kb upstream of exon 1 of Fgf10 gene, has been the only strain to allow transient lineage tracing of Fgf10-positive cells. Here, we describe a novel Fgf10(Cre-ERT2) knock-in line (Fgf10(iCre)) in which a Cre-ERT2-IRES-YFP cassette has been introduced in frame with the ATG of exon 1 of Fgf10 gene. Our studies show that Cre-ERT2 insertion disrupts Fgf10 function. However, administration of tamoxifen to Fgf10(iCre); Tomato(flox) double transgenic embryos or adult mice results in specific labeling of Fgf10-positive cells, which can be lineage-traced temporally and spatially. Moreover, we show that the Fgf10(iCre) line can be used for conditional gene inactivation in an inducible fashion during early developmental stages. We also provide evidence that transcription factors located in the first intron of Fgf10 gene are critical for maintaining Fgf10 expression over time. Thus, the Fgf10(iCre) line should serve as a powerful tool to explore the functions of Fgf10 in a controlled and stage-specific manner.


Assuntos
Desenvolvimento Embrionário , Fator 10 de Crescimento de Fibroblastos/genética , Células-Tronco Mesenquimais/metabolismo , Animais , Sequência de Bases , Primers do DNA , Éxons , Inativação Gênica , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Tamoxifeno/administração & dosagem
12.
J Clin Invest ; 121(11): 4409-19, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21985786

RESUMO

During lung development, parabronchial SMC (PSMC) progenitors in the distal mesenchyme secrete fibroblast growth factor 10 (Fgf10), which acts on distal epithelial progenitors to promote their proliferation. ß-catenin signaling within PSMC progenitors is essential for their maintenance, proliferation, and expression of Fgf10. Here, we report that this Wnt/Fgf10 embryonic signaling cascade is reactivated in mature PSMCs after naphthalene-induced injury to airway epithelium. Furthermore, we found that this paracrine Fgf10 action was essential for activating surviving variant Clara cells (the cells in the airway epithelium from which replacement epithelial cells originate) located at the bronchoalveolar duct junctions and adjacent to neuroendocrine bodies. After naphthalene injury, PSMCs secreted Fgf10 to activate Notch signaling and induce Snai1 expression in surviving variant Clara cells, which subsequently underwent a transient epithelial to mesenchymal transition to initiate the repair process. Epithelial Snai1 expression was important for regeneration after injury. We have therefore identified PSMCs as a stem cell niche for the variant Clara cells in the lung and established that paracrine Fgf10 signaling from the niche is critical for epithelial repair after naphthalene injury. These findings also have implications for understanding the misregulation of lung repair in asthma and cancer.


Assuntos
Lesão Pulmonar/patologia , Miócitos de Músculo Liso/patologia , Nicho de Células-Tronco , Células-Tronco/patologia , Animais , Diferenciação Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/genética , Lesão Pulmonar/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Miócitos de Músculo Liso/metabolismo , Naftalenos/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regeneração/genética , Regeneração/fisiologia , Transdução de Sinais , Células-Tronco/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
13.
Dev Biol ; 333(2): 238-50, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19559694

RESUMO

The miR-17 family of microRNAs has recently been recognized for its importance during lung development. The transgenic overexpression of the entire miR-17-92 cluster in the lung epithelium led to elevated cellular proliferation and inhibition of differentiation, while targeted deletion of miR-17-92 and miR-106b-25 clusters showed embryonic or early post-natal lethality. Herein we demonstrate that miR-17 and its paralogs, miR-20a, and miR-106b, are highly expressed during the pseudoglandular stage and identify their critical functional role during embryonic lung development. Simultaneous downregulation of these three miRNAs in explants of isolated lung epithelium altered FGF10 induced budding morphogenesis, an effect that was rescued by synthetic miR-17. E-Cadherin levels were reduced, and its distribution was altered by miR-17, miR-20a and miR-106b downregulation, while conversely, beta-catenin activity was augmented, and expression of its downstream targets, including Bmp4 as well as Fgfr2b, increased. Finally, we identified Stat3 and Mapk14 as key direct targets of miR-17, miR-20a, and miR-106b and showed that simultaneous overexpression of Stat3 and Mapk14 mimics the alteration of E-Cadherin distribution observed after miR-17, miR-20a, and miR-106b downregulation. We conclude that the mir-17 family of miRNA modulates FGF10-FGFR2b downstream signaling by specifically targeting Stat3 and Mapk14, hence regulating E-Cadherin expression, which in turn modulates epithelial bud morphogenesis in response to FGF10 signaling.


Assuntos
Caderinas/metabolismo , Células Epiteliais/citologia , Fator 10 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/embriologia , MicroRNAs/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Pulmão/citologia , Camundongos , Modelos Biológicos
14.
Stem Cells ; 26(11): 2902-11, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18719226

RESUMO

A new source of stem cells has recently been isolated from amniotic fluid; these amniotic fluid stem cells have significant potential for regenerative medicine. These cells are multipotent, showing the ability to differentiate into cell types from each embryonic germ layer. We investigated the ability of human amniotic fluid stem cells (hAFSC) to integrate into murine lung and to differentiate into pulmonary lineages after injury. Using microinjection into cultured mouse embryonic lungs, hAFSC can integrate into the epithelium and express the early human differentiation marker thyroid transcription factor 1 (TTF1). In adult nude mice, following hyperoxia injury, tail vein-injected hAFSC localized in the distal lung and expressed both TTF1 and the type II pneumocyte marker surfactant protein C. Specific damage of Clara cells through naphthalene injury produced integration and differentiation of hAFSC at the bronchioalveolar and bronchial positions with expression of the specific Clara cell 10-kDa protein. These results illustrate the plasticity of hAFSC to respond in different ways to different types of lung damage by expressing specific alveolar versus bronchiolar epithelial cell lineage markers, depending on the type of injury to recipient lung. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Líquido Amniótico/citologia , Células Epiteliais/citologia , Pulmão/citologia , Mucosa Respiratória/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Quimiocina CXCL12/metabolismo , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Feminino , Humanos , Pulmão/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/patologia , Lesão Pulmonar/terapia , Masculino , Mesoderma/citologia , Camundongos , Camundongos Nus , Microinjeções , Naftalenos , Surfactantes Pulmonares/metabolismo , Receptores CXCR4/metabolismo , Transplante de Células-Tronco , Fatores de Transcrição
15.
PLoS One ; 3(1): e1516, 2008 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-18231602

RESUMO

BACKGROUND: The role of ss-catenin signaling in mesodermal lineage formation and differentiation has been elusive. METHODOLOGY: To define the role of ss-catenin signaling in these processes, we used a Dermo1(Twist2)(Cre/+) line to target a floxed beta-catenin allele, throughout the embryonic mesenchyme. Strikingly, the Dermo1(Cre/+); beta-catenin(f/-) conditional Knock Out embryos largely phenocopy Pitx1(-/-)/Pitx2(-/-) double knockout embryos, suggesting that ss-catenin signaling in the mesenchyme depends mostly on the PITX family of transcription factors. We have dissected this relationship further in the developing lungs and find that mesenchymal deletion of beta-catenin differentially affects two major mesenchymal lineages. The amplification but not differentiation of Fgf10-expressing parabronchial smooth muscle progenitor cells is drastically reduced. In the angioblast-endothelial lineage, however, only differentiation into mature endothelial cells is impaired. CONCLUSION: Taken together these findings reveal a hierarchy of gene activity involving ss-catenin and PITX, as important regulators of mesenchymal cell proliferation and differentiation.


Assuntos
Linhagem da Célula , Pulmão/embriologia , Mesoderma/citologia , Transdução de Sinais , beta Catenina/metabolismo , Animais , Western Blotting , Proliferação de Células , Fatores de Crescimento de Fibroblastos/metabolismo , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Pulmão/citologia , Pulmão/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Interferente Pequeno , beta Catenina/genética
16.
Organogenesis ; 4(2): 100-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19279721

RESUMO

Reporter transgene, knockout, and misexpression studies support the notion that Wnt/beta-catenin signaling regulates aspects of branching morphogenesis, regional specialization of the epithelium and mesenchyme, and establishment of progenitor cell pools. As demonstrated for other foregut endoderm-derived organs, beta-catenin and the Wnt/beta-catenin signaling pathway contribute to control of cellular proliferation, differentiation and migration. However, the contribution of Wnt/beta-catenin signaling to these processes is shaped by other signals impinging on target tissues. In this review, we will concentrate on roles for Wnt/beta-catenin in respiratory system development, including segregation of the conducting airway and alveolar compartments, specialization of the mesenchyme, and establishment of tracheal asymmetries and tracheal glands.

17.
Dev Biol ; 299(1): 52-62, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16989802

RESUMO

Fibroblast growth factor (FGF) signaling has been shown to regulate lung epithelial development but its influence on mesenchymal differentiation has been poorly investigated. To study the role of mesenchymal FGF signaling in the differentiation of the mesenchyme and its impact on epithelial morphogenesis, we took advantage of Fgfr2c(+/Delta) mice, which due to a splicing switch express Fgfr2b in mesenchymal tissues and manifest Apert syndrome-like phenotypes. Using a set of in vivo and in vitro studies, we show that an autocrine FGF10-FGFR2b signaling loop is established in the mutant lung mesenchyme, which has several consequences. It prevents the entry of the smooth muscle progenitors into the smooth muscle cell (SMC) lineage and results in reduced fibronectin and elastin deposition. Levels of Fgf10 expression are raised within the mutant mesenchyme itself. Epithelial branching as well as epithelial levels of FGF and canonical Wnt signaling is dramatically reduced. These defects result in arrested development of terminal airways and an "emphysema like" phenotype in postnatal lungs. Our work unravels part of the complex interactions that govern normal lung development and may be pertinent to understanding the basis of respiratory defects in Apert syndrome.


Assuntos
Diferenciação Celular , Pulmão/citologia , Mesoderma/metabolismo , Miócitos de Músculo Liso/citologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Comunicação Autócrina/fisiologia , Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator 10 de Crescimento de Fibroblastos/deficiência , Fator 10 de Crescimento de Fibroblastos/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Pulmão/embriologia , Camundongos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Proteínas Wnt/metabolismo
18.
Dev Biol ; 293(1): 77-89, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16494859

RESUMO

Mesothelial Fibroblast Growth Factor 9 (Fgf9) has been demonstrated by inactivation studies in mouse to be critical for the proliferation of the mesenchyme. We now show that Fgf9 is also expressed at significant levels in the distal epithelium from the mid-pseudoglandular stages. Using mesenchymal-free lung endoderm culture, we show that FGF9 triggers the proliferation of the distal epithelium leading to the formation of a cyst-like structure. On embryonic Fgfr2b-/- lungs, FGF9 induces proliferation of the mesenchyme but fails to trigger a similar effect on the epithelium, therefore involving the FGFR2b receptor in the proliferative response of the epithelium to FGF9. While FGF9 inhibits the differentiation of the mesenchyme, the epithelium appears to differentiate normally. At the molecular level, FGF9 up-regulates Fgf10 expression in the mesenchyme likely via increased expression of Tbx4 and 5 and controls the transcription of Hedgehog targets Ptc and Gli-1 in a Hedgehog-independent manner. We also show that FGF9 inhibits the activation of the canonical Wnt pathway in the epithelium by increasing Dkk1 expression, a canonical Wnt antagonist. Our work shows for the first time that FGF9 acts on the epithelium involving FGFR2b to control its proliferation but not its differentiation and contributes to the regulation of canonical Wnt signaling in the epithelium.


Assuntos
Epitélio/metabolismo , Fator 9 de Crescimento de Fibroblastos/fisiologia , Pulmão/embriologia , Mesoderma/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Epitélio/embriologia , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Pulmão/citologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia , Proteínas com Domínio T/metabolismo , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/fisiologia
19.
Dev Biol ; 282(2): 422-31, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15950607

RESUMO

Branching morphogenesis of many organs, including the embryonic lung, is a dynamic process in which growth factor mediated tyrosine kinase receptor activation is required, but must be tightly regulated to direct ramifications of the terminal branches. However, the specific regulators that modulate growth factor signaling downstream of the tyrosine kinase receptor remain to be determined. Herein, we demonstrate for the first time an important function for the intracellular protein tyrosine phosphatase Shp2 in directing embryonic lung epithelial morphogenesis. We show that Shp2 is specifically expressed in embryonic lung epithelial buds, and that loss of function by the suppression of Shp2 mRNA expression results in a 53% reduction in branching morphogenesis. Furthermore, by intra-tracheal microinjection of a catalytically inactive adenoviral Shp2 construct, we provide direct evidence that the catalytic activity of Shp2 is required for proper embryonic lung branch formation. We demonstrate that Shp2 activity is required for FGF10 induced endodermal budding. Furthermore, a loss of Shp2 catalytic activity in the embryonic lung was associated with a reduction in ERK phosphorylation and epithelial cell proliferation. However, epithelial cell differentiation was not affected. Our results show that the protein tyrosine phosphatase Shp2 plays an essential role in modulating growth factor mediated tyrosine kinase receptor activation in early embryonic lung branching morphogenesis.


Assuntos
Brônquios/embriologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Morfogênese/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais/fisiologia , Adenoviridae , Análise de Variância , Animais , Western Blotting , Catálise , Primers do DNA , DNA Complementar/genética , Epitélio/fisiologia , Fator 10 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/metabolismo , Vetores Genéticos , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligonucleotídeos Antissenso , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Development ; 132(9): 2157-66, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15800000

RESUMO

Lineage formation in the lung mesenchyme is poorly understood. Using a transgenic mouse line expressing LacZ under the control of Fgf10 regulatory sequences, we show that the pool of Fgf10-positive cells in the distal lung mesenchyme contains progenitors of the parabronchial smooth muscle cells. Fgf10 gene expression is slightly repressed in this transgenic line. This allowed us to create a hypomorphic Fgf10 phenotype by expressing the LacZ transgene in a heterozygous Fgf10 background. Hypomorphic Fgf10 mutant lungs display a decrease in beta-galactosidase-positive cells around the bronchial epithelium associated with an accumulation of beta-galactosidase-expressing cells in the distal mesenchyme. This correlates with a marked reduction of alpha smooth muscle actin expression, thereby demonstrating that FGF10 is mostly required for the entry of mesenchymal cells into the parabronchial smooth muscle cell lineage. The failure of exogenous FGF10 to phosphorylate its known downstream targets ERK and AKT in lung mesenchymal cultures strongly suggests that FGF10 acts indirectly on the progenitor population via an epithelial intermediate. We provide support for a role of epithelial BMP4 in mediating the formation of parabronchial smooth muscle cells.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Fatores de Crescimento de Fibroblastos/genética , Mioblastos de Músculo Liso/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/metabolismo , Brônquios/fisiologia , Movimento Celular/fisiologia , Epitélio/fisiologia , Fator 10 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/metabolismo , Pulmão/citologia , Pulmão/fisiologia , Camundongos , Camundongos Transgênicos , Mioblastos de Músculo Liso/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...