Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Development ; 146(9)2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30944105

RESUMO

Although the Hippo-yes-associated protein (Yap) pathway has been implicated in lung development, the specific roles for Yap and its nucleocytoplasmic shuttling in the developing airway and alveolar compartments remain elusive. Moreover, conflicting results from expression studies and differences in the lung phenotypes of Yap and Hippo kinase null mutants caused controversy over the dynamics and significance of Yap subcellular localization in the developing lung. Here, we show that the aberrant morphogenesis of Yap-deficient lungs results from the disruption of developmental events specifically in distal epithelial progenitors. We also show that activation of nuclear Yap is enough to fulfill the Yap requirements to rescue abnormalities in these lungs. Remarkably, we found that Yap nucleocytoplasmic shuttling is largely dispensable in epithelial progenitors for both branching morphogenesis and sacculation. However, if maintained transcriptionally active in airways, nuclear Yap profoundly alters proximal-distal identity and halts epithelial differentiation. Taken together, these observations provide novel insights into the crucial importance of Hippo-Yap signaling in the lung prenatally.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Imunofluorescência , Via de Sinalização Hippo , Hibridização In Situ , Masculino , Camundongos , Morfogênese/genética , Morfogênese/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Proteínas de Sinalização YAP
2.
PLoS Genet ; 11(5): e1005238, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26020233

RESUMO

Differentiation of lung vascular smooth muscle cells (vSMCs) is tightly regulated during development or in response to challenges in a vessel specific manner. Aberrant vSMCs specifically associated with distal pulmonary arteries have been implicated in the pathogenesis of respiratory diseases, such as pulmonary arterial hypertension (PAH), a progressive and fatal disease, with no effective treatment. Therefore, it is highly relevant to understand the underlying mechanisms of lung vSMC differentiation. miRNAs are known to play critical roles in vSMC maturation and function of systemic vessels; however, little is known regarding the role of miRNAs in lung vSMCs. Here, we report that miR-29 family members are the most abundant miRNAs in adult mouse lungs. Moreover, high levels of miR-29 expression are selectively associated with vSMCs of distal vessels in both mouse and human lungs. Furthermore, we have shown that disruption of miR-29 in vivo leads to immature/synthetic vSMC phenotype specifically associated with distal lung vasculature, at least partially due to the derepression of KLF4, components of the PDGF pathway and ECM-related genes associated with synthetic phenotype. Moreover, we found that expression of FBXO32 in vSMCs is significantly upregulated in the distal vasculature of miR-29 null lungs. This indicates a potential important role of miR-29 in smooth muscle cell function by regulating FBXO32 and SMC protein degradation. These results are strongly supported by findings of a cell autonomous role of endogenous miR-29 in promoting SMC differentiation in vitro. Together, our findings suggested a vessel specific role of miR-29 in vSMC differentiation and function by targeting several key negative regulators.


Assuntos
Diferenciação Celular/genética , Hipertensão Pulmonar/genética , MicroRNAs/genética , Artéria Pulmonar/metabolismo , Animais , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Camundongos , MicroRNAs/antagonistas & inibidores , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Artéria Pulmonar/crescimento & desenvolvimento , Artéria Pulmonar/patologia , Proteínas Ligases SKP Culina F-Box/biossíntese , Proteínas Ligases SKP Culina F-Box/genética
3.
Am J Respir Cell Mol Biol ; 52(4): 397-408, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25211015

RESUMO

Decades of studies have shown evolutionarily conserved molecular networks consisting of transcriptional factors, diffusing growth factors, and signaling pathways that regulate proper lung development. Recently, microRNAs (miRNAs), small, noncoding regulatory RNAs, have been integrated into these networks. Significant advances have been made in characterizing the developmental stage- or cell type-specific miRNAs during lung development by using approaches such as genome-wide profiling and in situ hybridization. Results from gain- or loss-of-function studies revealed pivotal roles of protein components of the miRNA pathway and individual miRNAs in regulating proliferation, apoptosis, differentiation, and morphogenesis during lung development. Aberrant expression or functions of these components have been associated with pulmonary disorders, suggesting their involvement in pathogenesis of these diseases. Moreover, genetically modified mice generated in these studies have become useful models of human lung diseases. Challenges in this field include characterization of collective function and responsible targets of miRNAs specifically expressed during lung development, and translation of these basic findings into clinically relevant information for better understanding of human diseases. The goal of this review is to discuss the recent progress on the understanding of how the miRNA pathway regulates lung development, how dysregulation of miRNA activities contributes to pathogenesis of related pulmonary diseases, and to identify relevant questions and future directions.


Assuntos
Pneumopatias/metabolismo , MicroRNAs/fisiologia , Animais , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Pulmão/irrigação sanguínea , Pulmão/embriologia , Pulmão/metabolismo , Pulmão/patologia , Pneumopatias/genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/fisiologia , Interferência de RNA , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
4.
Biochem Cell Biol ; 93(2): 109-18, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25454218

RESUMO

Pulmonary fibrosis is a pathological condition in which lungs become scarred due to the excess extracellular matrix (ECM) deposition and structural alterations in the interstitium of lung parenchyma. Many patients with interstitial lung diseases (ILDs) caused by long-term exposure to toxic substances, chronic infections, or autoimmune responses develop fibrosis. Etiologies for many ILDs are unknown, such as idiopathic pulmonary fibrosis (IPF), a devastating, relentless form of pulmonary fibrosis with a median survival of 2-3 years. Despite several decades of research, factors that initiate and sustain the fibrotic response in lungs remain unclear and there is no effective treatment to block progression of fibrosis. Here we summarize recent findings on the antifibrotic activity of miR-29, a small noncoding regulatory RNA, in the pathogenesis of fibrosis by regulating ECM production and deposition, and epithelial-mesenchymal transition (EMT). We also describe interactions of miR-29 with multiple profibrotic and inflammatory pathways. Finally, we review the antifibrotic activity of miR-29 in animal models of fibrosis and highlight miR-29 as a promising therapeutic reagent or target for the treatment of pulmonary fibrosis.


Assuntos
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrose Pulmonar Idiopática/genética , MicroRNAs/metabolismo , Animais , Colágeno/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos
5.
Respir Res ; 16: 22, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25763778

RESUMO

BACKGROUND: The transcription factor NK2 homeobox 1 (Nkx2-1) plays essential roles in epithelial cell proliferation and differentiation in mouse and human lung development and tumorigenesis. A better understanding of genes and pathways downstream of Nkx2-1 will clarify the multiple roles of this critical lung factor. Nkx2-1 regulates directly or indirectly numerous protein-coding genes; however, there is a paucity of information about Nkx2-1-regulated microRNAs (miRNAs). METHODS AND RESULTS: By miRNA array analyses of mouse epithelial cell lines in which endogenous Nkx2-1 was knocked-down, we revealed that 29 miRNAs were negatively regulated including miR-200c, and 39 miRNAs were positively regulated by Nkx2-1 including miR-1195. Mouse lungs lacking functional phosphorylated Nkx2-1 showed increased expression of miR-200c and alterations in the expression of other top regulated miRNAs. Moreover, chromatin immunoprecipitation assays showed binding of NKX2-1 protein to regulatory regions of these miRNAs. Promoter reporter assays indicated that 1kb of the miR-200c 5' flanking region was transcriptionally active but did not mediate Nkx2-1- repression of miR-200c expression. 3'UTR reporter assays support a direct regulation of the predicted targets Nfib and Myb by miR-200c. CONCLUSIONS: These studies suggest that Nkx2-1 controls the expression of specific miRNAs in lung epithelial cells. In particular, we identified a regulatory link between Nkx2-1, the known tumor suppressor miR-200c, and the developmental and oncogenic transcription factors Nfib and Myb, adding new players to the regulatory mechanisms driven by Nkx2-1 in lung epithelial cells that may have implications in lung development and tumorigenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Pulmão/metabolismo , MicroRNAs/metabolismo , Fatores de Transcrição NFI/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas v-myb/metabolismo , Fatores de Transcrição/metabolismo , Regiões 3' não Traduzidas , Região 5'-Flanqueadora , Animais , Sítios de Ligação , Linhagem Celular , Transformação Celular Neoplásica/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Reporter , Camundongos , MicroRNAs/genética , Fatores de Transcrição NFI/genética , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Oncogênicas v-myb/genética , Fosforilação , Regiões Promotoras Genéticas , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica , Transfecção
6.
Am J Respir Cell Mol Biol ; 51(2): 273-83, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24617895

RESUMO

Sonic hedgehog (Shh) is expressed and secreted from the embryonic lung epithelium and acts on the adjacent mesenchymal cells via its receptor Patched (Ptch)/Smoothened (Smo) and transcriptional effectors Gli proteins. Genetic studies showed that the Shh pathway plays critical roles in mouse lung development. However, little is known about microRNAs (miRNAs) downstream of Shh in embryonic lungs. Here we profiled miRNAs in embryonic lung cultures treated with cyclopamine, a specific Smo antagonist or with Smo agonist by next-generation of sequencing. We then performed functional screening to examine whether some of these miRNAs can modulate the induction of Gli-responsive luciferase by Shh treatment. These analyses revealed that expression of miR-326 and its host gene, Arrestin ß1, is selectively enriched in embryonic lung mesenchymal cells and is specifically influenced by Shh activity. Furthermore, functional analyses showed that miR-326 acts as a negative modulator for Shh signaling by directly targeting Smo and Gli2. Together, these findings suggest a novel miR-326-negative feedback loop in regulating the activity of Shh signaling.


Assuntos
Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Pulmão/metabolismo , MicroRNAs/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Arrestinas/genética , Arrestinas/metabolismo , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas Hedgehog/genética , Sequenciamento de Nucleotídeos em Larga Escala , Fatores de Transcrição Kruppel-Like/genética , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Camundongos , MicroRNAs/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Receptor Smoothened , Transfecção , Alcaloides de Veratrum/farmacologia , Proteína Gli2 com Dedos de Zinco , beta-Arrestinas
7.
Am J Physiol Lung Cell Mol Physiol ; 306(5): L405-19, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24375798

RESUMO

Chronic injury of alveolar lung epithelium leads to epithelial disintegrity in idiopathic pulmonary fibrosis (IPF). We had reported earlier that Grhl2, a transcriptional factor, maintains alveolar epithelial cell integrity by directly regulating components of adherens and tight junctions and thus hypothesized an important role of GRHL2 in pathogenesis of IPF. Comparison of GRHL2 distribution at different stages of human lung development showed its abundance in developing lung epithelium and in adult lung epithelium. However, GRHL2 is detected in normal human lung mesenchyme only at early fetal stage (week 9). Similar mesenchymal reexpression of GRHL2 was also observed in IPF. Immunofluorescence analysis in serial sections from three IPF patients revealed at least two subsets of alveolar epithelial cells (AEC), based on differential GRHL2 expression and the converse fluorescence intensities for epithelial vs. mesenchymal markers. Grhl2 was not detected in mesenchyme in intraperitoneal bleomycin-induced injury as well as in spontaneously occurring fibrosis in double-mutant HPS1 and HPS2 mice, whereas in contrast in a radiation-induced fibrosis model, with forced Forkhead box M1 (Foxm1) expression, an overlap of Grhl2 with a mesenchymal marker was observed in fibrotic regions. Grhl2's role in alveolar epithelial cell plasticity was confirmed by altered Grhl2 gene expression analysis in IPF and further validated by in vitro manipulation of its expression in alveolar epithelial cell lines. Our findings reveal important pathophysiological differences between human IPF and specific mouse models of fibrosis and support a crucial role of GRHL2 in epithelial activation in lung fibrosis and perhaps also in epithelial plasticity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fibrose Pulmonar Idiopática/fisiopatologia , Mucosa Respiratória/fisiologia , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Masculino , Mesoderma/metabolismo , Mesoderma/fisiologia , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade , Gravidez , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Especificidade da Espécie , Fatores de Transcrição/genética
8.
J Biol Chem ; 287(8): 5979-87, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22187428

RESUMO

Tnrc6 family members (Tnrc6a/b/c) are key components of the RNA-induced silencing complex in microRNA (miRNA)-mediated gene suppression. Here, we show that Tnrc6a, also known as GW182, is selectively expressed in the yolk sac endoderm and that gene trap disruption of GW182 leads to growth arrest and apoptosis. We found that targets of miRNAs highly expressed in the yolk sac are significantly derepressed in GW182(gt/gt) mutant mice, although levels of miRNAs are not altered. Specifically, growth arrest and apoptosis phenotype are associated with significant derepression of Cdkn1a (p21), Cdkn1c (P27), Lats1, Lats2, Rb1, Rbl, Bim, and Pten, known targets of miRNAs from miR-17/20/93/106 clusters highly expressed in yolk sac endoderm. Together, these data strongly suggest that GW182 is an essential functional component in the RNA-induced silencing complex for miRNA-mediated gene silencing in vivo, and selectively regulation of miRNA activity plays an important role in the proper development of yolk sac.


Assuntos
Autoantígenos/metabolismo , Endoderma/metabolismo , MicroRNAs/genética , Saco Vitelino/embriologia , Animais , Apoptose/genética , Autoantígenos/genética , Sequência de Bases , Ciclo Celular/genética , Linhagem Celular , Endoderma/citologia , Inativação Gênica , Hematopoese/genética , Camundongos , Fatores de Tempo
9.
Sci Rep ; 13(1): 7090, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37127654

RESUMO

Biogenesis of organelles requires targeting of a subset of proteins to specific subcellular domains by signal peptides or mechanisms controlling mRNA localization and local translation. How local distribution and translation of specific mRNAs for organelle biogenesis is achieved remains elusive and likely to be dependent on the cellular context. Here we identify Trinucleotide repeat containing-6a (Tnrc6a), a component of the miRNA pathway, distinctively localized to apical granules of differentiating airway multiciliated cells (MCCs) adjacent to centrioles. In spite of being enriched in TNRC6A and the miRNA-binding protein AGO2, they lack enzymes for mRNA degradation. Instead, we found these apical granules enriched in components of the mRNA translation machinery and newly synthesized proteins suggesting that they are specific hubs for target mRNA localization and local translation in MCCs. Consistent with this, Tnrc6a loss of function prevented formation of these granules and led to a broad reduction, rather than stabilization of miRNA targets. These included downregulation of key genes involved in ciliogenesis and was associated with defective multicilia formation both in vivo and in primary airway epithelial cultures. Similar analysis of Tnrc6a disruption in yolk sac showed stabilization of miRNA targets, highlighting the potential diversity of these mechanisms across organs.


Assuntos
Centríolos , MicroRNAs , Centríolos/metabolismo , MicroRNAs/genética , Proteínas/metabolismo , Epitélio/metabolismo , RNA Mensageiro/metabolismo
10.
J Neurosci ; 31(43): 15407-15, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22031887

RESUMO

Dysfunctional neural control of airway smooth muscle (ASM) is involved in inflammatory diseases, such as asthma. However, neurogenesis in the lung is poorly understood. This study uses mouse models to investigate developmental mechanisms of ASM innervation, a process that is highly coordinated with ASM formation during lung branching morphogenesis. We show that brain-derived neurotrophic factor (BDNF) is an essential ASM-derived signal for innervation. Although BDNF mRNA expression is temporally dissociated with ASM formation and innervation, BDNF protein is coordinately produced through post-transcriptional suppression by miR-206. Using a combination of chemical and genetic approaches to modulate sonic hedgehog (Shh) signaling, a pathway essential for lung branching and ASM formation, we show that Shh signaling blocks miR-206 expression, which in turn increases BDNF protein expression. Together, our work uncovers a functional cascade that involves Shh, miR-206 and BDNF to coordinate ASM formation and innervation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas Hedgehog/metabolismo , MicroRNAs/metabolismo , Músculo Liso/fisiologia , Sistema Respiratório/anatomia & histologia , Sistema Respiratório/inervação , Fatores Etários , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas Hedgehog/genética , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Músculo Liso/inervação , RNA Mensageiro/metabolismo , Sistema Respiratório/embriologia , Transdução de Sinais/genética , Tubulina (Proteína)/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(7): 2319-24, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19168627

RESUMO

We have shown that smoking impacts bronchial airway gene expression and that heterogeneity in this response associates with smoking-related disease risk. In this study, we sought to determine whether microRNAs (miRNAs) play a role in regulating the airway gene expression response to smoking. We examined whole-genome miRNA and mRNA expression in bronchial airway epithelium from current and never smokers (n = 20) and found 28 miRNAs to be differentially expressed (P < 0.05) with the majority being down-regulated in smokers. We further identified a number of mRNAs whose expression level is highly inversely correlated with miRNA expression in vivo. Many of these mRNAs contain potential binding sites for the differentially expressed miRNAs in their 3'-untranslated region (UTR) and are themselves affected by smoking. We found that either increasing or decreasing the levels of mir-218 (a miRNA that is strongly affected by smoking) in both primary bronchial epithelial cells and H1299 cells was sufficient to cause a corresponding decrease or increase in the expression of predicted mir-218 mRNA targets, respectively. Further, mir-218 expression is reduced in primary bronchial epithelium exposed to cigarette smoke condensate (CSC), and alteration of mir-218 levels in these cells diminishes the induction of the predicted mir-218 target MAFG in response to CSC. These data indicate that mir-218 levels modulate the airway epithelial gene expression response to cigarette smoke and support a role for miRNAs in regulating host response to environmental toxins.


Assuntos
Epitélio/metabolismo , Regulação da Expressão Gênica , MicroRNAs/genética , Fumar , Traqueia/metabolismo , Regiões 3' não Traduzidas , Adulto , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Risco
12.
iScience ; 25(8): 104751, 2022 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-35942101

RESUMO

Differences in ciliary morphology and dynamics among multiciliated cells of the respiratory tract contribute to efficient mucociliary clearance. Nevertheless, little is known about how these phenotypic differences are established. We show that Prominin 1 (Prom1), a transmembrane protein widely used as stem cell marker, is crucial to this process. During airway differentiation, Prom1 becomes restricted to multiciliated cells, where it is expressed at distinct levels along the proximal-distal axis of the airways. Prom1 is induced by Notch in multiciliated cells, and Notch inactivation abolishes this gradient of expression. Prom1 was not required for multicilia formation, but when inactivated resulted in longer cilia that beat at a lower frequency. Disruption of Notch resulted in opposite effects and suggested that Notch fine-tunes Prom1 levels to regulate the multiciliated cell phenotype and generate diversity among these cells. This mechanism could contribute to the innate defense of the lung and help prevent pulmonary disease.

13.
Am J Respir Cell Mol Biol ; 45(2): 287-94, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20971881

RESUMO

MicroRNAs (miRNA) are small regulatory RNAs that control gene expression by translational suppression and destabilization of target mRNAs. There is increasing evidence that miRNAs regulate genes associated with fibrosis in organs, such as the heart, kidney, liver, and the lung. In a large-scale screening for miRNAs potentially involved in bleomycin-induced fibrosis, we found expression of miR-29 family members significantly reduced in fibrotic lungs. Analysis of normal lungs showed the presence of miR-29 in subsets of interstitial cells of the alveolar wall, pleura, and at the entrance of the alveolar duct, known sites of pulmonary fibrosis. miR-29 levels inversely correlated with the expression levels of profibrotic target genes and the severity of the fibrosis. To study the impact of miR-29 down-regulation in the lung interstitium, we characterized gene expression profiles of human fetal lung fibroblast IMR-90 cells in which endogenous miR-29 was knocked down. This confirmed the derepression of reported miR-29 targets, including several collagens, but also revealed up-regulation of a large number of previously unrecognized extracellular matrix-associated and remodeling genes. Moreover, we found that miR-29 is suppressed by transforming growth factor (TGF)-ß1 in these cells, and that many fibrosis-associated genes up-regulated by TGF-ß1 are derepressed by miR-29 knockdown. Interestingly, a comparison of TGF-ß1 and miR-29 targets revealed that miR-29 controls an additional subset of fibrosis-related genes, including laminins and integrins, independent of TGF-ß1. Together, these strongly suggest a role of miR-29 in the pathogenesis of pulmonary fibrosis. miR-29 may be a potential new therapeutic target for this disease.


Assuntos
Biomarcadores/metabolismo , Pulmão/metabolismo , MicroRNAs/genética , Fibrose Pulmonar/genética , Fator de Crescimento Transformador beta1/farmacologia , Regiões 3' não Traduzidas , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Northern Blotting , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Hibridização In Situ , Luciferases/metabolismo , Pulmão/citologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Breast Cancer Res ; 13(2): R24, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21375733

RESUMO

INTRODUCTION: microRNA (miRNA) are short, noncoding RNA that negatively regulate gene expression and may play a causal role in invasive breast cancer. Since many genetic aberrations of invasive disease are detectable in early stages, we hypothesized that miRNA expression dysregulation and the predicted changes in gene expression might also be found in early breast neoplasias. METHODS: Expression profiling of 365 miRNA by real-time quantitative polymerase chain reaction assay was combined with laser capture microdissection to obtain an epithelium-specific miRNA expression signature of normal breast epithelium from reduction mammoplasty (RM) (n = 9) and of paired samples of histologically normal epithelium (HN) and ductal carcinoma in situ (DCIS) (n = 16). To determine how miRNA may control the expression of codysregulated mRNA, we also performed gene expression microarray analysis in the same paired HN and DCIS samples and integrated this with miRNA target prediction. We further validated several target pairs by modulating the expression levels of miRNA in MCF7 cells and measured the expression of target mRNA and proteins. RESULTS: Thirty-five miRNA were aberrantly expressed between RM, HN and DCIS. Twenty-nine miRNA and 420 mRNA were aberrantly expressed between HN and DCIS. Combining these two data sets with miRNA target prediction, we identified two established target pairs (miR-195:CCND1 and miR-21:NFIB) and tested several novel miRNA:mRNA target pairs. Overexpression of the putative tumor suppressor miR-125b, which is underexpressed in DCIS, repressed the expression of MEMO1, which is required for ErbB2-driven cell motility (also a target of miR-125b), and NRIP1/RIP140, which modulates the transcriptional activity of the estrogen receptor. Knockdown of the putative oncogenic miRNA miR-182 and miR-183, both highly overexpressed in DCIS, increased the expression of chromobox homolog 7 (CBX7) (which regulates E-cadherin expression), DOK4, NMT2 and EGR1. Augmentation of CBX7 by knockdown of miR-182 expression, in turn, positively regulated the expression of E-cadherin, a key protein involved in maintaining normal epithelial cell morphology, which is commonly lost during neoplastic progression. CONCLUSIONS: These data provide the first miRNA expression profile of normal breast epithelium and of preinvasive breast carcinoma. Further, we demonstrate that altered miRNA expression can modulate gene expression changes that characterize these early cancers. We conclude that miRNA dysregulation likely plays a substantial role in early breast cancer development.


Assuntos
Neoplasias da Mama/genética , Carcinoma Intraductal não Infiltrante/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/biossíntese , Carcinoma Intraductal não Infiltrante/metabolismo , Carcinoma Intraductal não Infiltrante/patologia , Linhagem Celular Tumoral , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Ferroproteínas não Heme/biossíntese , Complexo Repressor Polycomb 1 , RNA Mensageiro/biossíntese , Proteínas Repressoras/biossíntese
15.
J Cell Biol ; 175(2): 209-15, 2006 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-17060494

RESUMO

Toll receptors in Drosophila melanogaster function in morphogenesis and host defense. Mammalian orthologues of Toll, the Toll-like receptors (TLRs), have been studied extensively for their essential functions in controlling innate and adaptive immune responses. We report that TLR8 is dynamically expressed during mouse brain development and localizes to neurons and axons. Agonist stimulation of TLR8 in cultured cortical neurons causes inhibition of neurite outgrowth and induces apoptosis in a dissociable manner. Our evidence indicates that such TLR8-mediated neuronal responses do not involve the canonical TLR-NF-kappaB signaling pathway. These findings reveal novel functions for TLR8 in the mammalian nervous system that are distinct from the classical role of TLRs in immunity.


Assuntos
Apoptose , Neuritos/fisiologia , Neurônios/fisiologia , Receptor 8 Toll-Like/fisiologia , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Quinase I-kappa B/metabolismo , Técnicas Imunoenzimáticas , Hibridização In Situ , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Neurônios/citologia , Transdução de Sinais , Receptor 8 Toll-Like/genética
16.
Sci Rep ; 11(1): 10799, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34031475

RESUMO

The impact of prematurity on human development and neonatal diseases, such as bronchopulmonary dysplasia, has been widely reported. However, little is known about the effects of prematurity on the programs of stem cell self-renewal and differentiation of the upper respiratory epithelium, which is key for adaptation to neonatal life. We developed a minimally invasive methodology for isolation of neonatal basal cells from nasopharyngeal (NP) aspirates and performed functional analysis in organotypic cultures to address this issue. We show that preterm NP progenitors have a markedly distinct molecular signature of abnormal proliferation and mitochondria quality control compared to term progenitors. Preterm progenitors had lower oxygen consumption at baseline and were unable to ramp up consumption to the levels of term cells when challenged. Although they formed a mucociliary epithelium, ciliary function tended to decline in premature cells as they differentiated, compared to term cells. Together, these differences suggested increased sensitivity of preterm progenitors to environmental stressors under non-homeostatic conditions.


Assuntos
Displasia Broncopulmonar/patologia , Perfilação da Expressão Gênica/métodos , Nasofaringe/citologia , Oxigênio/metabolismo , Células-Tronco/citologia , Displasia Broncopulmonar/genética , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Nasofaringe/metabolismo , Análise de Sequência de RNA , Células-Tronco/metabolismo
17.
Elife ; 82019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31631837

RESUMO

Notch signaling regulates cell fate selection during development in multiple organs including the lung. Previous studies on the role of Notch in the lung focused mostly on Notch pathway core components or receptor-specific functions. It is unclear, however, how Jagged or Delta-like ligands collectively or individually (Jag1, Jag2, Dll1, Dll4) influence differentiation of airway epithelial progenitors. Using mouse genetic models we show major differences in Jag and Dll in regulation and establishment of cell fate. Jag ligands had a major impact in balancing distinct cell populations in conducting airways, but had no role in the establishment of domains and cellular abundance in the neuroendocrine (NE) microenvironment. Surprisingly, Dll ligands were crucial in restricting cell fate and size of NE bodies and showed an overlapping role with Jag in differentiation of NE-associated secretory (club) cells. These mechanisms may potentially play a role in human conditions that result in aberrant NE differentiation, including NE hyperplasias and cancer.


Assuntos
Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pulmão/citologia , Proteínas de Membrana/metabolismo , Proteínas Serrate-Jagged/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Microambiente Celular , Ligantes , Camundongos , Sistemas Neurossecretores/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
18.
Genesis ; 46(1): 8-18, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18196599

RESUMO

Heparan sulfate (HS) proteoglycans modulate the biological activity of a number of growth factors in development, homeostasis, and cancer. Specific modifications of HS chains by HS biosynthetic enzymes have been implicated in growth factor signaling in multiple aspects of organogenesis. Although the role of HS 6-O-sulfotransferases has been described in processes such as trachea formation in Drosophila and vasculogenesis in zebrafish, little is known about how HS 6-O-sulfotransferases (Hs6st1-3 in mice) influence mouse development. To address this issue, we generated a conditionally mutant Hs6st1 mouse line and then generated mice with systemic inactivation of Hs6st1. Hs6st1-null pups were viable and grossly normal at birth. The lack of obvious abnormalities in lung, liver, and kidney, which express high levels of Hs6st1 during development, suggests that at least during embryonic life, the loss of Hs6st1 function may be compensated for by mechanisms involving other HS modifying enzymes. During early adulthood, however, Hs6st1-null mice failed to thrive and exhibited growth retardation, body weight loss, enlargement of airspaces in the lung and, in some cases, lethality. Our results suggest a potentially critical role for HS 6-O sulfation by Hs6st1 in postnatal processes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Sulfotransferases/genética , Sulfotransferases/fisiologia , Alelos , Animais , Cruzamentos Genéticos , Feminino , Técnicas Genéticas , Genótipo , Heparitina Sulfato/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Mutantes , Mutação , Transdução de Sinais
19.
J Cell Biochem ; 103(6): 1886-94, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18022820

RESUMO

Fgf10 has a prominent role in organogenesis. In the developing lung, Fgf10 is dynamically expressed in the distal mesenchyme from where it diffuses and activates its epithelial receptor, Fgfr2b, to trigger budding. Little is known about how Fgf10 expression is regulated. Here we have identified a mouse lung-specific mesenchymal cell line, MLg, which expresses endogenous Fgf10 and responds to known regulators of Fgf10 in a way that is reminiscent of the early lung. To gain insights into the mechanisms involved in the transcriptional regulation of Fgf10 in these cells, we have cloned and analyzed approximately a 4.5 kb region of the mouse Fgf10 promoter. Promoter deletion analysis and Luciferase reporter assays revealed an upstream region of the Fgf10 promoter with selective enhancer activity in the MLg, but not in the non-lung-derived cell line NIH3T3. Our data suggest that a potential lung mesenchyme-specific enhancer may exist within this region of the Fgf10 promoter.


Assuntos
Fator 10 de Crescimento de Fibroblastos/biossíntese , Mesoderma/fisiologia , Animais , Linhagem Celular , Elementos Facilitadores Genéticos , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Pulmão/citologia , Mesoderma/citologia , Camundongos , Regiões Promotoras Genéticas
20.
Gene Expr Patterns ; 8(2): 124-39, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18023262

RESUMO

To identify genes expressed during initiation of lung organogenesis, we generated transcriptional profiles of the prospective lung region of the mouse foregut (mid-foregut) microdissected from embryos at three developmental stages between embryonic day 8.5 (E8.5) and E9.5. This period spans from lung specification of foregut cells to the emergence of the primary lung buds. We identified a number of known and novel genes that are temporally regulated as the lung bud forms. Genes that regulate transcription, including DNA binding factors, co-factors, and chromatin remodeling genes, are the main functional groups that change during lung bud formation. Members of key developmental transcription and growth factor families, not previously described to participate in lung organogenesis, are expressed in the mid-foregut during lung bud induction. These studies also show early expression in the mid-foregut of genes that participate in later stages of lung development. This characterization of the mid-foregut transcriptome provides new insights into molecular events leading to lung organogenesis.


Assuntos
Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Perfilação da Expressão Gênica , Pulmão/embriologia , Pulmão/metabolismo , Animais , Sistema Digestório/citologia , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/citologia , Camundongos , Modelos Biológicos , Organogênese , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA