Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Lab Invest ; 99(9): 1363-1375, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31028279

RESUMEN

Proper lung development depends on the precise temporal and spatial expression of several morphogenic factors, including Fgf10, Fgf9, Shh, Bmp4, and Tgf-ß. Over- or under-expression of these molecules often leads to aberrant embryonic or postnatal lung development. Herein, we deleted the Tgf-ß1 gene specifically within the lung embryonic mesenchymal compartment at specific gestational stages to determine the contribution of this cytokine to lung development. Mutant embryos developed severe lung hypoplasia and died at birth due to the inability to breathe. Despite the markedly reduced lung size, proliferation and differentiation of the lung epithelium was not affected by the lack of mesenchymal expression of the Tgf-ß1 gene, while apoptosis was significantly increased in the mutant lung parenchyma. Lack of mesenchymal expression of the Tgf-ß1 gene was also associated with reduced lung branching morphogenesis, with accompanying inhibition of the local FGF10 signaling pathway as well as abnormal development of the vascular system. To shed light on the mechanism of lung hypoplasia, we quantified the phosphorylation of 226 proteins in the mutant E12.5 lung compared with control. We identified five proteins, Hrs, Vav2, c-Kit, the regulatory subunit of Pi3k (P85), and Fgfr1, that were over- or under-phosphorylated in the mutant lung, suggesting that they could be indispensable effectors of the TGF-ß signaling program during embryonic lung development. In conclusion, we have uncovered novel roles of the mesenchyme-specific Tgf-ß1 ligand in embryonic mouse lung development and generated a mouse model that may prove helpful to identify some of the key pathogenic mechanisms underlying lung hypoplasia in humans.


Asunto(s)
Técnicas de Inactivación de Genes/métodos , Pulmón/embriología , Mesodermo/embriología , Morfogénesis/genética , Factor de Crecimiento Transformador beta1 , Animales , Animales Recién Nacidos , Apoptosis , Técnicas de Cultivo de Célula , Femenino , Pulmón/patología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L196-L207, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27941074

RESUMEN

The signaling cross talk between the tracheal mesenchyme and epithelium has not been researched extensively, leaving a substantial gap of knowledge in the mechanisms dictating embryonic development of the proximal airways by the adjacent mesenchyme. Recently, we reported that embryos lacking mesenchymal expression of Sox9 did not develop tracheal cartilage rings and showed aberrant differentiation of the tracheal epithelium. Here, we propose that tracheal cartilage provides local inductive signals responsible for the proper differentiation, metabolism, and inflammatory status regulation of the tracheal epithelium. The tracheal epithelium of mesenchyme-specific Sox9Δ/Δ mutant embryos showed altered mRNA expression of various epithelial markers such as Pb1fa1, surfactant protein B (Sftpb), secretoglobulin, family 1A, member 1 (Scgb1a1), and trefoil factor 1 (Tff1). In vitro tracheal epithelial cell cultures confirmed that tracheal chondrocytes secrete factors that inhibit club cell differentiation. Whole gene expression profiling and ingenuity pathway analysis showed that the tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and transforming growth factor-ß (TGF-ß) signaling pathways were significantly altered in the Sox9 mutant trachea. TNF-α and IFN-γ interfered with the differentiation of tracheal epithelial progenitor cells into mature epithelial cell types in vitro. Mesenchymal knockout of Tgf-ß1 in vivo resulted in altered differentiation of the tracheal epithelium. Finally, mitochondrial enzymes involved in fat and glycogen metabolism, cytochrome c oxidase subunit VIIIb (Cox8b) and cytochrome c oxidase subunit VIIa polypeptide 1 (Cox7a1), were strongly upregulated in the Sox9 mutant trachea, resulting in increases in the number and size of glycogen storage vacuoles. Our results support a role for tracheal cartilage in modulation of the differentiation and metabolism and the expression of inflammatory-related genes in the tracheal epithelium by feeding into the TNF-α, IFN-γ, and TGF-ß signaling pathways.


Asunto(s)
Cartílago/embriología , Embrión de Mamíferos/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inflamación/genética , Tráquea/citología , Tráquea/embriología , Animales , Biomarcadores/metabolismo , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Diferenciación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Medios de Cultivo Condicionados/farmacología , Embrión de Mamíferos/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/embriología , Epitelio/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glucógeno/metabolismo , Interferón gamma/metabolismo , Masculino , Mesodermo/efectos de los fármacos , Mesodermo/embriología , Ratones Noqueados , Modelos Biológicos , Mutación/genética , Oxidación-Reducción/efectos de los fármacos , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/efectos de los fármacos , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
BMC Biol ; 13: 42, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-26092405

RESUMEN

BACKGROUND: Adenomatous polyposis coli (Apc) is a tumor suppressor that inhibits Wnt/Ctnnb1. Mutations of Apc will not only lead to familial adenomatous polyposis with associated epithelial lesions, but will also cause aggressive fibromatosis in mesenchymal cells. However, the roles of Apc in regulating mesenchymal cell biology and organogenesis during development are unknown. RESULTS: We have specifically deleted the Apc gene in lung mesenchymal cells during early lung development in mice. Loss of Apc function resulted in immediate mesenchymal cell hyperproliferation through abnormal activation of Wnt/Ctnnb1, followed by a subsequent inhibition of cell proliferation due to cell cycle arrest at G0/G1, which was caused by a mechanism independent of Wnt/Ctnnb1. Meanwhile, abrogation of Apc also disrupted lung mesenchymal cell differentiation, including decreased airway and vascular smooth muscle cells, the presence of Sox9-positive mesenchymal cells in the peripheral lung, and excessive versican production. Moreover, lung epithelial branching morphogenesis was drastically inhibited due to disrupted Bmp4-Fgf10 morphogen production and regulation in surrounding lung mesenchyme. Lastly, lung mesenchyme-specific Apc conditional knockout also resulted in altered lung vasculogenesis and disrupted pulmonary vascular continuity through a paracrine mechanism, leading to massive pulmonary hemorrhage and lethality at mid-gestation when the pulmonary circulation should have started. CONCLUSIONS: Our study suggests that Apc in lung mesenchyme plays central roles in coordinating the proper development of several quite different cellular compartments including lung epithelial branching and pulmonary vascular circulation during lung organogenesis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Pulmón/anomalías , Pulmón/crecimiento & desarrollo , Mesodermo/citología , Animales , Diferenciación Celular , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Mesodermo/anomalías , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
5.
J Cell Sci ; 125(Pt 17): 4036-48, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22685326

RESUMEN

Little is known about the regulatory mechanisms underlying lung epithelial tight junction (TJ) assembly, which is inextricably linked to the preservation of epithelial polarity, and is highly coordinated by proteins that regulate epithelial cell polarity, such as aPKCζ. We recently reported that Eya1 phosphatase functions through aPKCζ-Notch1 signaling to control cell polarity in the lung epithelium. Here, we have extended these observations to TJ formation to demonstrate that Eya1 is crucial for the maintenance of TJ protein assembly in the lung epithelium, probably by controlling aPKCζ phosphorylation levels, aPKCζ-mediated TJ protein phosphorylation and Notch1-Cdc42 activity. Thus, TJs are disassembled after interfering with Eya1 function in vivo or during calcium-induced TJ assembly in vitro. These effects are reversed by reintroduction of wild-type Eya1 or partially inhibiting aPKCζ in Eya1siRNA cells. Moreover, genetic activation of Notch1 rescues Eya1(-/-) lung epithelial TJ defects. These findings uncover novel functions for the Eya1-aPKCζ-Notch1-Cdc42 pathway as a crucial regulatory mechanism of TJ assembly and polarity of the lung epithelium, providing a conceptual framework for future mechanistic and translational studies in this area.


Asunto(s)
Epitelio/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/citología , Pulmón/enzimología , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Uniones Estrechas/metabolismo , Animales , Calcio/metabolismo , Membrana Celular/metabolismo , Activación Enzimática , Células Epiteliales/citología , Células Epiteliales/enzimología , Epitelio/embriología , Femenino , Eliminación de Gen , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Pulmón/embriología , Ratones , Proteínas Nucleares/deficiencia , Fosforilación , Proteína Quinasa C/metabolismo , Transporte de Proteínas , Proteínas Tirosina Fosfatasas/deficiencia , Receptor Notch1/metabolismo , Transducción de Señal , Activación Transcripcional , Proteína de Unión al GTP cdc42/metabolismo
7.
Development ; 138(7): 1395-407, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21385765

RESUMEN

Cell polarity, mitotic spindle orientation and asymmetric division play a crucial role in the self-renewal/differentiation of epithelial cells, yet little is known about these processes and the molecular programs that control them in embryonic lung distal epithelium. Herein, we provide the first evidence that embryonic lung distal epithelium is polarized with characteristic perpendicular cell divisions. Consistent with these findings, spindle orientation-regulatory proteins Insc, LGN (Gpsm2) and NuMA, and the cell fate determinant Numb are asymmetrically localized in embryonic lung distal epithelium. Interfering with the function of these proteins in vitro randomizes spindle orientation and changes cell fate. We further show that Eya1 protein regulates cell polarity, spindle orientation and the localization of Numb, which inhibits Notch signaling. Hence, Eya1 promotes both perpendicular division as well as Numb asymmetric segregation to one daughter in mitotic distal lung epithelium, probably by controlling aPKCζ phosphorylation. Thus, epithelial cell polarity and mitotic spindle orientation are defective after interfering with Eya1 function in vivo or in vitro. In addition, in Eya1(-/-) lungs, perpendicular division is not maintained and Numb is segregated to both daughter cells in mitotic epithelial cells, leading to inactivation of Notch signaling. As Notch signaling promotes progenitor cell identity at the expense of differentiated cell phenotypes, we test whether genetic activation of Notch could rescue the Eya1(-/-) lung phenotype, which is characterized by loss of epithelial progenitors, increased epithelial differentiation but reduced branching. Indeed, genetic activation of Notch partially rescues Eya1(-/-) lung epithelial defects. These findings uncover novel functions for Eya1 as a crucial regulator of the complex behavior of distal embryonic lung epithelium.


Asunto(s)
Diferenciación Celular/fisiología , Polaridad Celular/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/embriología , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Huso Acromático/metabolismo , Animales , Western Blotting , Proteínas de Ciclo Celular , Epitelio/embriología , Epitelio/metabolismo , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Pulmón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Mitosis/fisiología , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Fosforilación/fisiología , Proteínas Tirosina Fosfatasas/genética , Receptores Notch/genética , Huso Acromático/genética
8.
Cell Microbiol ; 15(1): 63-81, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22963587

RESUMEN

The interaction of outer membrane protein A (OmpA) with its receptor, Ecgp96 (a homologue of Hsp90ß), is critical for the pathogenesis of Escherichia coli K1 meningitis. Since Hsp90 chaperones Toll-like receptors (TLRs), we examined the role of TLRs in E. coli K1 infection. Herein, we show that newborn TLR2(-/-) mice are resistant to E. coli K1 meningitis, while TLR4(-/-) mice succumb to infection sooner. In vitro, OmpA+ E. coli infection selectively upregulates Ecgp96 and TLR2 in human brain microvascular endothelial cells (HBMEC), whereas OmpA- E. coli upregulates TLR4 in these cells. Furthermore, infection with OmpA+ E. coli causes Ecgp96 and TLR2 translocate to the plasma membrane of HBMEC as a complex. Immunoprecipitation studies of the plasma membrane fractions from infected HBMEC reveal that the C termini of Ecgp96 and TLR2 are critical for OmpA+ E. coli invasion. Knockdown of TLR2 using siRNA results in inefficient membrane translocation of Ecgp96 and significantly reduces invasion. In addition, the interaction of Ecgp96 andTLR2 induces a bipartite signal, one from Ecgp96 through PKC-α while the other from TLR2 through MyD88, ERK1/2 and NF-κB. This bipartite signal ultimately culminates in the efficient production of NO, which in turn promotes E. coli K1 invasion of HBMEC.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Endocitosis , Células Endoteliales/microbiología , Escherichia coli/patogenicidad , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Línea Celular , Análisis Mutacional de ADN , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Noqueados , Mapeo de Interacción de Proteínas , Receptor Toll-Like 2/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
9.
BMC Biol ; 11: 117, 2013 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-24274029

RESUMEN

BACKGROUND: Embryonic lung development is instructed by crosstalk between mesenchyme and epithelia, which results in activation of transcriptional factors, such as Sox9, in a temporospatial manner. Sox9 is expressed in both distal lung epithelium and proximal lung mesenchyme. Here, we investigated the effect of lung mesenchyme-specific inducible deletion of Sox9 during murine lung development. RESULTS: Transgenic mice lacking Sox9 expression were unable to breathe and died at birth, with noticeable tracheal defects. Cartilage rings were missing, and the tracheal lumen was collapsed in the mutant trachea. In situ hybridization showed an altered expression pattern of Tbx4, Tbx5 and Fgf10 genes and marked reduction of Collagen2 expression in the tracheal mesenchyme. The tracheal phenotype was increasingly severe, with longer duration of deletion. Lymphatic vasculature was underdeveloped in the mutant trachea: Prox1, Lyve1, and Vegfr3 were decreased after Sox9 knockout. We also found that compared with normal tracheal epithelium, the mutant tracheal epithelium had an altered morphology with fewer P63-positive cells and more CC10-positive cells, fewer goblet cells, and downregulation of surfactant proteins A and C. CONCLUSION: The appropriate temporospatial expression of Sox9 in lung mesenchyme is necessary for appropriate tracheal cartilage formation, lymphatic vasculature system development, and epithelial differentiation. We uncovered a novel mechanism of lung epithelium differentiation: tracheal cartilage rings instruct the tracheal epithelium to differentiate properly during embryonic development. Thus, besides having a mechanical function, tracheal cartilage also appears to be a local signaling structure in the embryonic lung.


Asunto(s)
Pulmón/embriología , Mesodermo/embriología , Factor de Transcripción SOX9/metabolismo , Tráquea/embriología , Animales , Cartílago/embriología , Doxiciclina , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Pulmón/citología , Pulmón/metabolismo , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Factor de Transcripción SOX9/genética , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
BMC Biol ; 11: 111, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24225400

RESUMEN

BACKGROUND: Reciprocal interactions between lung mesenchymal and epithelial cells play essential roles in lung organogenesis and homeostasis. Although the molecular markers and related animal models that target lung epithelial cells are relatively well studied, molecular markers of lung mesenchymal cells and the genetic tools to target and/or manipulate gene expression in a lung mesenchyme-specific manner are not available, which becomes a critical barrier to the study of lung mesenchymal biology and the related pulmonary diseases. RESULTS: We have identified a mouse Tbx4 gene enhancer that contains conserved DNA sequences across many vertebrate species with lung or lung-like gas exchange organ. We then generate a mouse line to express rtTA/LacZ under the control of the Tbx4 lung enhancer, and therefore a Tet-On inducible transgenic system to target lung mesenchymal cells at different developmental stages. By combining a Tbx4-rtTA driven Tet-On inducible Cre expression mouse line with a Cre reporter mouse line, the spatial-temporal patterns of Tbx4 lung enhancer targeted lung mesenchymal cells were defined. Pulmonary endothelial cells and vascular smooth muscle cells were targeted by the Tbx4-rtTA driver line prior to E11.5 and E15.5, respectively, while other subtypes of lung mesenchymal cells including airway smooth muscle cells, fibroblasts, pericytes could be targeted during the entire developmental stage. CONCLUSIONS: Developmental lung mesenchymal cells can be specifically marked by Tbx4 lung enhancer activity. With our newly created Tbx4 lung enhancer-driven Tet-On inducible system, lung mesenchymal cells can be specifically and differentially targeted in vivo for the first time by controlling the doxycycline induction time window. This novel system provides a unique tool to study lung mesenchymal cell lineages and gene functions in lung mesenchymal development, injury repair, and regeneration in mice.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Células Epiteliales/citología , Pulmón/embriología , Mesodermo/embriología , Proteínas de Dominio T Box/genética , Animales , Diferenciación Celular , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Pulmón/citología , Ratones , Modelos Animales , Miocitos del Músculo Liso/citología , Miofibroblastos/citología , Organogénesis/genética , Análisis de Secuencia de ADN
11.
Adv Ther ; 41(2): 534-552, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38110652

RESUMEN

The implementation of artificial intelligence (AI) and machine learning (ML) techniques in healthcare has garnered significant attention in recent years, especially as a result of their potential to revolutionize personalized medicine. Despite advances in the treatment and management of asthma, a significant proportion of patients continue to suffer acute exacerbations, irrespective of disease severity and therapeutic regimen. The situation is further complicated by the constellation of factors that influence disease activity in a patient with asthma, such as medical history, biomarker phenotype, pulmonary function, level of healthcare access, treatment compliance, comorbidities, personal habits, and environmental conditions. A growing body of work has demonstrated the potential for AI and ML to accurately predict asthma exacerbations while also capturing the entirety of the patient experience. However, application in the clinical setting remains mostly unexplored, and important questions on the strengths and limitations of this technology remain. This review presents an overview of the rapidly evolving landscape of AI and ML integration into asthma management by providing a snapshot of the existing scientific evidence and proposing potential avenues for future applications.


Asunto(s)
Inteligencia Artificial , Asma , Humanos , Aprendizaje Automático , Asma/diagnóstico , Asma/tratamiento farmacológico , Medicina de Precisión , Gravedad del Paciente
12.
Dev Biol ; 350(1): 112-26, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21129374

RESUMEN

The proper level of proliferation and differentiation along the proximodistal axis is crucial for lung organogenesis. Elucidation of the factors that control these processes will therefore provide important insights into embryonic lung development and regeneration. Eya1 is a transcription factor/protein phosphatase that regulates cell lineage specification and proliferation. Yet its functions during lung development are unknown. In this paper we show that Eya1(-/-) lungs are severely hypoplastic with reduced epithelial branching and increased mesenchymal cellularity. Eya1 is expressed at the distal epithelial tips of branching tubules as well as in the surrounding distal mesenchyme. Eya1(-/-) lung epithelial cells show loss of progenitor cell markers with increased expression of differentiation markers and cell cycle exit. In addition, Eya1(-/-) embryos and newborn mice exhibit severe defects in the smooth muscle component of the bronchi and major pulmonary vessels with decreased Fgf10 expression. These defects lead to rupture of the major vessels and hemorrhage into the lungs after birth. Treatment of Eya1(-/-) epithelial explants in culture with recombinant Fgf10 stimulates epithelial branching. Since Shh expression and activity are abnormally increased in Eya1(-/-) lungs, we tested whether genetically lowering Shh activity could rescue the Eya1(-/-) lung phenotype. Indeed, genetic reduction of Shh partially rescues Eya1(-/-) lung defects while restoring Fgf10 expression. This study provides the first evidence that Eya1 regulates Shh signaling in embryonic lung, thus ensuring the proper level of proliferation and differentiation along the proximodistal axis of epithelial, mesenchymal and endothelial cells. These findings uncover novel functions for Eya1 as a critical upstream coordinator of Shh-Fgf10 signaling during embryonic lung development. We conclude, therefore, that Eya1 function is critical for proper coordination of lung epithelial, mesenchymal and vascular development.


Asunto(s)
Vasos Sanguíneos/embriología , Péptidos y Proteínas de Señalización Intracelular/genética , Pulmón/embriología , Pulmón/enzimología , Mesodermo/embriología , Morfogénesis/genética , Proteínas Nucleares/genética , Proteínas Tirosina Fosfatasas/genética , Mucosa Respiratoria/embriología , Animales , Vasos Sanguíneos/enzimología , Ciclo Celular , Diferenciación Celular , Eliminación de Gen , Genes Letales , Proteínas Hedgehog/metabolismo , Pulmón/irrigación sanguínea , Mesodermo/enzimología , Ratones , Ratones Noqueados , Mucosa Respiratoria/citología , Mucosa Respiratoria/enzimología , Células Madre/citología , Células Madre/fisiología
13.
Dev Biol ; 353(2): 242-58, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21385574

RESUMEN

Six1 is a member of the six-homeodomain family of transcription factors. Six1 is expressed in multiple embryonic cell types and plays important roles in proliferation, differentiation and survival of precursor cells of different organs, yet its function during lung development was hitherto unknown. Herein we show that Six1(-/-) lungs are severely hypoplastic with greatly reduced epithelial branching and increased mesenchymal cellularity. Six1 is expressed at the distal epithelial tips of branching tubules as well as in the surrounding distal mesenchyme. Six1(-/-) lung epithelial cells show increased expression of differentiation markers, but loss of progenitor cell markers. Six1 overexpression in MLE15 lung epithelial cells in vitro inhibited cell differentiation, but increases the expression of progenitor cell markers. In addition, Six1(-/-) embryos and newborn mice exhibit mesenchymal overproliferation, decreased Fgf10 expression and severe defects in the smooth muscle component of the bronchi and major pulmonary vessels. These defects lead to rupture of major vessels in mutant lungs after birth. Treatment of Six1(-/-) epithelial explants in culture with recombinant Fgf10 protein restores epithelial branching. As Shh expression is abnormally increased in Six1(-/-) lungs, we also treated mutant mesenchymal explants with recombinant Shh protein and found that these explants were competent to respond to Shh and continued to grow in culture. Furthermore, inhibition of Shh signaling with cyclopamine stimulated Six1(-/-) lungs to grow and branch in culture. This study provides the first evidence for the requirement of Six1 in coordinating Shh-Fgf10 signaling in embryonic lung to ensure proper levels of proliferation and differentiation along the proximodistal axis of epithelial, mesenchymal and endothelial cells. These findings uncover novel and essential functions for Six1 as a critical coordinator of Shh-Fgf10 signaling during embryonic lung development. We propose that Six1 is hence critical for coordination of proper lung epithelial, mesenchymal and vascular development.


Asunto(s)
Proteínas de Homeodominio/fisiología , Pulmón/embriología , Actinas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/citología , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/farmacología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/farmacología , Proteínas Hedgehog/fisiología , Proteínas de Homeodominio/genética , Pulmón/anomalías , Pulmón/irrigación sanguínea , Pulmón/crecimiento & desarrollo , Mesodermo/embriología , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Ratones Transgénicos , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Morfogénesis/fisiología , Miocitos del Músculo Liso/citología , Mucosa Respiratoria/embriología , Mucosa Respiratoria/crecimiento & desarrollo , Mucosa Respiratoria/metabolismo , Transducción de Señal/efectos de los fármacos , Alcaloides de Veratrum/farmacología
14.
Dev Biol ; 333(2): 238-50, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19559694

RESUMEN

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.


Asunto(s)
Cadherinas/metabolismo , Células Epiteliales/citología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Pulmón/embriología , MicroARNs/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Pulmón/citología , Ratones , Modelos Biológicos
15.
Nat Commun ; 11(1): 726, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32024828

RESUMEN

Hyperspectral fluorescence imaging is gaining popularity for it enables multiplexing of spatio-temporal dynamics across scales for molecules, cells and tissues with multiple fluorescent labels. This is made possible by adding the dimension of wavelength to the dataset. The resulting datasets are high in information density and often require lengthy analyses to separate the overlapping fluorescent spectra. Understanding and visualizing these large multi-dimensional datasets during acquisition and pre-processing can be challenging. Here we present Spectrally Encoded Enhanced Representations (SEER), an approach for improved and computationally efficient simultaneous color visualization of multiple spectral components of hyperspectral fluorescence images. Exploiting the mathematical properties of the phasor method, we transform the wavelength space into information-rich color maps for RGB display visualization. We present multiple biological fluorescent samples and highlight SEER's enhancement of specific and subtle spectral differences, providing a fast, intuitive and mathematical way to interpret hyperspectral images during collection, pre-processing and analysis.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Espectrometría de Fluorescencia/métodos , Algoritmos , Animales , Animales Modificados Genéticamente , Color , Embrión no Mamífero , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Procesamiento de Imagen Asistido por Computador/normas , Ratones Endogámicos C57BL , Microscopía Confocal/métodos , Relación Señal-Ruido , Pez Cebra/embriología , Pez Cebra/genética
16.
Stem Cells ; 26(11): 2902-11, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18719226

RESUMEN

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.


Asunto(s)
Líquido Amniótico/citología , Células Epiteliales/citología , Pulmón/citología , Mucosa Respiratoria/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Quimiocina CXCL12/metabolismo , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Humanos , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Lesión Pulmonar/terapia , Masculino , Mesodermo/citología , Ratones , Ratones Desnudos , Microinyecciones , Naftalenos , Surfactantes Pulmonares/metabolismo , Receptores CXCR4/metabolismo , Trasplante de Células Madre , Factores de Transcripción
17.
Biomed Opt Express ; 9(2): 780-790, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29552412

RESUMEN

Hyperspectral endoscopic imaging has the potential to enhance clinical diagnostics and outcome. Most commercial endoscopes utilize imaging fiber bundles to transmit the collected signal from the patient to the medical operator. These bundles consist of several fiber cores surrounded by a cladding layer creating comb structure-like artifacts, which complicate further analysis, both spatially and spectrally. Here we present an optical fiber pattern removal algorithm which we applied to hyperspectral bronchoscopic images robustly and quantitatively without the need for specific optical or electrical hardware. We validate the performance of the pattern removal by using a novel hyperspectral phasor approach. This algorithm can be generalized to all forms of fiber bundle hyperspectral endoscopy.

18.
J Transl Med ; 4: 50, 2006 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-17129373

RESUMEN

BACKGROUND: Current clinical and histopathological criteria used to define the prognosis of melanoma patients are inadequate for accurate prediction of clinical outcome. We investigated whether genome screening by means of high-throughput gene microarray might provide clinically useful information on patient survival. METHODS: Forty-three tumor tissues from 38 patients with stage III and stage IV melanoma were profiled with a 17,500 element cDNA microarray. Expression data were analyzed using significance analysis of microarrays (SAM) to identify genes associated with patient survival, and supervised principal components (SPC) to determine survival prediction. RESULTS: SAM analysis revealed a set of 80 probes, corresponding to 70 genes, associated with survival, i.e. 45 probes characterizing longer and 35 shorter survival times, respectively. These transcripts were included in a survival prediction model designed using SPC and cross-validation which allowed identifying 30 predicting probes out of the 80 associated with survival. CONCLUSION: The longer-survival group of genes included those expressed in immune cells, both innate and acquired, confirming the interplay between immunological mechanisms and the natural history of melanoma. Genes linked to immune cells were totally lacking in the poor-survival group, which was instead associated with a number of genes related to highly proliferative and invasive tumor cells.

19.
Cancer Res ; 74(21): 5999-6009, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25164011

RESUMEN

Heparan sulfate proteoglycans (HSPG) play a critical role in the interaction of tumor cells and their microenvironment. HSPG activity is dictated by sulfation patterns controlled by sulfotransferases, which add sulfate groups, and sulfatases (Sulf), which remove 6-O-sulfates. Here, we report altered expression of these enzymes in human neuroblastoma cells with higher levels of Sulf-2 expression, a specific feature of MYCN-amplified cells (MYCN-A cells) that represent a particularly aggressive subclass. Sulf-2 overexpression in neuroblastoma cells lacking MYCN amplification (MYCN-NA cells) increased their in vitro survival. Mechanistic investigations revealed evidence of a link between Sulf-2 expression and MYCN pathogenicity in vitro and in vivo. Analysis of Sulf-2 protein expression in 65 human neuroblastoma tumors demonstrated a higher level of Sulf-2 expression in MYCN-A tumors than in MYCN-NA tumors. In two different patient cohorts, we confirmed the association in expression patterns of Sulf-2 and MYCN and determined that Sulf-2 overexpression predicted poor outcomes in a nonindependent manner with MYCN. Our findings define Sulf-2 as a novel positive regulator of neuroblastoma pathogenicity that contributes to MYCN oncogenicity. Cancer Res; 74(21); 5999-6009. ©2014 AACR.


Asunto(s)
Proteoglicanos de Heparán Sulfato/metabolismo , Neuroblastoma/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Sulfotransferasas/biosíntesis , Línea Celular Tumoral , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/patología , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Transducción de Señal/genética , Sulfatasas , Sulfotransferasas/genética , Microambiente Tumoral/genética
20.
PLoS One ; 8(8): e71679, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967234

RESUMEN

The potential for amniotic fluid stem cell (AFSC) treatment to inhibit the progression of fibrotic lung injury has not been described. We have previously demonstrated that AFSC can attenuate both acute and chronic-fibrotic kidney injury through modification of the cytokine environment. Fibrotic lung injury, such as in Idiopathic Pulmonary Fibrosis (IPF), is mediated through pro-fibrotic and pro-inflammatory cytokine activity. Thus, we hypothesized that AFSC treatment might inhibit the progression of bleomycin-induced pulmonary fibrosis through cytokine modulation. In particular, we aimed to investigate the effect of AFSC treatment on the modulation of the pro-fibrotic cytokine CCL2, which is increased in human IPF patients and is correlated with poor prognoses, advanced disease states and worse fibrotic outcomes. The impacts of intravenous murine AFSC given at acute (day 0) or chronic (day 14) intervention time-points after bleomycin injury were analyzed at either day 3 or day 28 post-injury. Murine AFSC treatment at either day 0 or day 14 post-bleomycin injury significantly inhibited collagen deposition and preserved pulmonary function. CCL2 expression increased in bleomycin-injured bronchoalveolar lavage (BAL), but significantly decreased following AFSC treatment at either day 0 or at day 14. AFSC were observed to localize within fibrotic lesions in the lung, showing preferential targeting of AFSC to the area of fibrosis. We also observed that MMP-2 was transiently increased in BAL following AFSC treatment. Increased MMP-2 activity was further associated with cleavage of CCL2, rendering it a putative antagonist for CCL2/CCR2 signaling, which we surmise is a potential mechanism for CCL2 reduction in BAL following AFSC treatment. Based on this data, we concluded that AFSC have the potential to inhibit the development or progression of fibrosis in a bleomycin injury model during both acute and chronic remodeling events.


Asunto(s)
Líquido Amniótico/citología , Lavado Broncoalveolar , Quimiocina CCL2/metabolismo , Fibrosis Pulmonar/metabolismo , Células Madre/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina/efectos adversos , Quimiotaxis/inmunología , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Feto , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Modelos Biológicos , Embarazo , Proteolisis , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/terapia , Trasplante de Células Madre , Células Madre/inmunología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA