Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clín. salud ; 34(2): 85-90, jul. 2023. tab, graf
Article in English | IBECS | ID: ibc-223209

ABSTRACT

Rationale: Pregnant women are especially vulnerable to mental health problems, including stress, anxiety, and depression. This risk has been increased during the COVID-19 pandemic and differences in psychological symptoms in pregnancy and postpartum before and during COVID-19 exists. Mental health problems can have adverse effects on both the woman, and the neonate, including miscarriages, premature births, low birth weight, and higher rates of cesarean sections and instrument-assisted deliveries. Aim: To evaluate the prevalence of psychological symptoms of pregnant women before and during the COVID-19 pandemic. Method: A cross-sectional study was carried out to assess the psychological profile in a sample of pregnant women selected before the pandemic and a sample of women studied during the first wave of the pandemic. A total of 122 women were selected prior to the pandemic and 181 women during the COVID-19 outbreak. The Symptom Checklist-90 Revised (SCL-90-R) was used to assess depression and anxiety during pregnancy and in the postpartum period. Results: The prevalence of symptoms of depression and anxiety during pregnancy were higher in the sample of women studied during the pandemic (SCL-90-R: M = 54.6 vs. 42.6 and M = 62.6 vs. 51.7 respectively). In the postpartum the difference between both samples of women was even higher for depression and anxiety (SCL-90-R: M = 50.4 vs. 35.0 and M = 51.3 vs. 36.0 respectively). Being a pregnant woman at the COVID-19 outbreak was directly associated with a higher score of depression symptoms (aOR = 8.67, 95% CI [3.26, 23.02], p < .001). Anxiety during childbirth was more frequently reported by women before the pandemic (aOR = 5.13, 95% CI [2.53, 10.44], p < .001). The variable stage (before /during pandemic) was also associated with having a clinical SCL-90-R score above 70 (aOR = 7.61, 95% CI [2.7, 21.47], p < .001). (AU)


Justificación: Las mujeres embarazadas son especialmente vulnerables a los problemas de salud mental, como el estrés, la ansiedad y la depresión. Este riesgo se ha incrementado durante la pandemia de COVID-19, habiendo diferencias en los síntomas psicológicos en el embarazo y el puerperio antes y después de la pandemia. Los problemas de salud mental pueden tener efectos perjudiciales tanto en la mujer como en el recién nacido, como abortos espontáneos, partos prematuros, bajo peso al nacer y tasas más altas de cesáreas y partos asistidos. Objetivo: Evaluar la prevalencia de síntomas psicológicos de gestantes antes y durante la pandemia de COVID-19. Método: Se realizó un estudio transversal para evaluar el perfil psicológico en una muestra de mujeres embarazadas seleccionadas antes de la pandemia y una muestra de mujeres estudiadas durante la primera ola de la pandemia. Se seleccionó un total de 122 mujeres antes de la pandemia y 181 mujeres durante el brote de COVID-19. Se utilizó la Inventario de Verificación de Síntomas-90 Revisada (SCL-90-R) para evaluar la depresión y la ansiedad durante el embarazo y el puerperio. Resultados: La prevalencia de síntomas de depresión y ansiedad durante el embarazo fue mayor en la muestra de mujeres estudiadas durante la pandemia (SCL-90-R: M = 54.6 vs. 42.6 y M = 62.6 vs. 51.7 respectivamente). En el postparto la diferencia entre ambas muestras de mujeres fue aún mayor para depresión y ansiedad (SCL-90-R: M = 50.4 vs. 35.0 y M = 51.3 vs. 36.0 respectivamente). Ser mujer embarazada en el brote de COVID-19 se asoció directamente con una mayor puntuación de síntomas de depresión (aOR = 8.67, IC 95% [3.26, 23.02, p < .001). Las mujeres manifestaron ansiedad durante el parto con mayor frecuencia antes de la pandemia (aOR = 5.13, IC 95% 2.53, 10.44], p < .001). La variable estadio (antes/durante la pandemia) también se asoció con tener una puntuación clínica SCL-90-R superior a 70 (aOR = 7.61, IC 95% [2.7, 21.47], p < .001). (AU)


Subject(s)
Humans , Female , Pregnancy , Pregnancy/psychology , Pandemics , Coronavirus Infections/epidemiology , Coronavirus Infections/psychology , Postpartum Period/psychology , Behavioral Symptoms/epidemiology , Cross-Sectional Studies , Anxiety , Depression , Mental Health , Prevalence , Spain
2.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1049-L1060, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30892080

ABSTRACT

Cellular senescence is a biological process by which cells lose their capacity to proliferate yet remain metabolically active. Although originally considered a protective mechanism to limit the formation of cancer, it is now appreciated that cellular senescence also contributes to the development of disease, including common respiratory ailments such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. While many factors have been linked to the development of cellular senescence, mitochondrial dysfunction has emerged as an important causative factor. In this study, we uncovered that the mitochondrial biogenesis pathway driven by the mammalian target of rapamycin/peroxisome proliferator-activated receptor-γ complex 1α/ß (mTOR/PGC-1α/ß) axis is markedly upregulated in senescent lung epithelial cells. Using two different models, we show that activation of this pathway is associated with other features characteristic of enhanced mitochondrial biogenesis, including elevated number of mitochondrion per cell, increased oxidative phosphorylation, and augmented mitochondrial reactive oxygen species (ROS) production. Furthermore, we found that pharmacological inhibition of the mTORC1 complex with rapamycin not only restored mitochondrial homeostasis but also reduced cellular senescence to bleomycin in lung epithelial cells. Likewise, mitochondrial-specific antioxidant therapy also effectively inhibited mTORC1 activation in these cells while concomitantly reducing mitochondrial biogenesis and cellular senescence. In summary, this study provides a mechanistic link between mitochondrial biogenesis and cellular senescence in lung epithelium and suggests that strategies aimed at blocking the mTORC1/PGC-1α/ß axis or reducing ROS-induced molecular damage could be effective in the treatment of senescence-associated lung diseases.


Subject(s)
Cellular Senescence/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Respiratory Mucosa/metabolism , Animals , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Bleomycin/pharmacology , Cell Line , Idiopathic Pulmonary Fibrosis/pathology , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Oxidative Stress/physiology , Pulmonary Disease, Chronic Obstructive/pathology , Rats , Reactive Oxygen Species/metabolism , Respiratory Mucosa/cytology , Sirolimus/pharmacology
3.
Sci Rep ; 8(1): 14418, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30258080

ABSTRACT

The function of most long noncoding RNAs (lncRNAs) is unknown. However, recent studies reveal important roles of lncRNAs in regulating cancer-related pathways. Human antisense lncRNA-NKX2-1-AS1 partially overlaps the NKX2-1/TTF1 gene within chromosomal region 14q13.3. Amplification of this region and/or differential expression of genes therein are associated with cancer progression. Herein we show higher levels of NKX2-AS1 and NKX2-1 in lung adenocarcinomas relative to non-tumor controls but no correlation between NKX2-1-AS1 and NKX2-1 levels across specimens, or with amplification of the 14q13.3 region, suggesting that NKX2-1-AS1 and NKX2-1 are independently regulated. Loss-and-gain of function experiments showed that NKX2-1-AS1 does not regulate NKX2-1 expression, or nearby genes, but controls genes in trans. Genes up-regulated by NKX2-1-AS1-knockdown belong to cell adhesion and PD-L1/PD-1 checkpoint pathways. NKX2-1-AS1 negatively regulates endogenous CD274/PD-L1, a known target of NKX2-1, and the transcriptional activity of -1kb-CD274 promoter-reporter construct. Furthermore, NKX2-1-AS1 interferes with NKX2-1 protein binding to the CD274-promoter, likely by NKX2-1 protein-NKX2-1-AS1 interactions. Finally, NKX2-1-AS1 negatively regulates cell migration and wound healing, but not proliferation or apoptosis. These findings support potential roles of NKX2-1-AS1 in limiting motility and immune system evasion of lung carcinoma cells, highlighting a novel mechanism that may influence tumorigenic capabilities of lung epithelial cells.


Subject(s)
B7-H1 Antigen/metabolism , Cell Movement , Neoplasm Proteins/metabolism , RNA, Antisense/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Thyroid Nuclear Factor 1/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Humans , Neoplasm Proteins/genetics , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Thyroid Nuclear Factor 1/genetics
4.
Clin Exp Metastasis ; 35(3): 149-165, 2018 03.
Article in English | MEDLINE | ID: mdl-29909489

ABSTRACT

Distinct members of the Ets family of transcription factors act as positive or negative regulators of genes involved in cellular proliferation, development, and tumorigenesis. In human lung cancer, increased ETS1 expression is associated with poor prognosis and metastasis. We tested whether ETS1 contributes to lung tumorigenesis by binding to Twist1, a gene involved in tumor cell motility and dissemination. We used a mouse lung cancer model with metastasis driven by conditionally activated Kras and concurrent tumor suppressor Lkb1 loss (KrasG12D/ Lkb1-/- model) and a similar model of lung cancer that does not metastasize, driven by conditionally activated Kras alone (KrasG12D model). We show that Ets1 and Twist1 gene expression differs between KrasG12D tumors (low Ets1 and Twist1 expression) and KrasG12D/Lkb1-/- tumors (high Ets1 and Twist1 expression). In human lung tumors, ETS1 and TWIST1 expression positively correlates and low combined ETS1 and TWIST1 levels are associated with improved survival compared to high levels. Using mouse cell lines derived from KrasG12D and KrasG12D/Lkb1-/- mouse models and the human lung cancer (A549) cell line, we show that ETS1 regulates Twist1 expression. Chromatin immunoprecipitation assays confirm binding of ETS1 to the Twist1 promoter. Overexpression studies show that ETS1 transactivates Twist1 promoter activity in mouse and human cells. Silencing endogenous Ets1 by siRNA in mouse cell lines decreases Twist1 mRNA levels, decreases invasion, and increases cell growth. Ets1 and Twist1 are at the crossroad of several signaling pathways in cancer. Understanding their regulation may inform the development of therapies to impair lung tumor metastasis.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Proto-Oncogene Protein c-ets-1/genetics , Twist-Related Protein 1/genetics , AMP-Activated Protein Kinases , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction
5.
Am J Respir Cell Mol Biol ; 55(3): 407-18, 2016 09.
Article in English | MEDLINE | ID: mdl-27064756

ABSTRACT

Airway epithelial cell responses are critical to the outcome of lung infection. In this study, we aimed to identify unique contributions of epithelial cells during lung infection. To differentiate genes induced selectively in epithelial cells during pneumonia, we compared genome-wide expression profiles from three sorted cell populations: epithelial cells from uninfected mouse lungs, epithelial cells from mouse lungs with pneumococcal pneumonia, and nonepithelial cells from those same infected lungs. Of 1,166 transcripts that were more abundant in epithelial cells from infected lungs compared with nonepithelial cells from the same lungs or from epithelial cells of uninfected lungs, 32 genes were identified as highly expressed secreted products. Especially strong signals included two related secreted and transmembrane (Sectm) 1 genes, Sectm1a and Sectm1b. Refinement of sorting strategies suggested that both Sectm1 products were induced predominantly in conducting airway epithelial cells. Sectm1 was induced during the early stages of pneumococcal pneumonia, and mutation of NF-κB RelA in epithelial cells did not diminish its expression. Instead, type I IFN signaling was necessary and sufficient for Sectm1 induction in lung epithelial cells, mediated by signal transducer and activator of transcription 1. For target cells, Sectm1a bound to myeloid cells preferentially, in particular Ly6G(bright)CD11b(bright) neutrophils in the infected lung. In contrast, Sectm1a did not bind to neutrophils from uninfected lungs. Sectm1a increased expression of the neutrophil-attracting chemokine CXCL2 by neutrophils from the infected lung. We propose that Sectm1a is an epithelial product that sustains a positive feedback loop amplifying neutrophilic inflammation during pneumococcal pneumonia.


Subject(s)
Epithelial Cells/metabolism , Membrane Proteins/metabolism , Neutrophil Activation , Neutrophils/metabolism , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/pathology , Signal Transduction , Animals , Chemokine CXCL2/biosynthesis , Electric Conductivity , Epithelial Cells/microbiology , Gene Expression Regulation , Interferon Type I/metabolism , Lung/microbiology , Lung/pathology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Pneumonia, Pneumococcal/genetics , Recombinant Proteins/metabolism , Streptococcus pneumoniae/physiology
6.
Exp Cell Res ; 335(1): 115-22, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25959509

ABSTRACT

Podoplanin (PDPN) is a transmembrane glycoprotein that promotes tumor cell migration, invasion, and cancer metastasis. In fact, PDPN expression is induced in many types of cancer. Thus, PDPN has emerged as a functionally relevant cancer biomarker and chemotherapeutic target. PDPN contains 2 intracellular serine residues that are conserved between species ranging from mouse to humans. Recent studies indicate that protein kinase A (PKA) can phosphorylate PDPN in order to inhibit cell migration. However, the number and identification of specific residues phosphorylated by PKA have not been defined. In addition, roles of other kinases that may phosphorylate PDPN to control cell migration have not been investigated. We report here that cyclin dependent kinase 5 (CDK5) can phosphorylate PDPN in addition to PKA. Moreover, results from this study indicate that PKA and CDK5 cooperate to phosphorylate PDPN on both intracellular serine residues to decrease cell motility. These results provide new insight into PDPN phosphorylation dynamics and the role of PDPN in cell motility. Understanding novel mechanisms of PDPN intracellular signaling could assist with designing novel targeted chemotherapeutic agents and procedures.


Subject(s)
Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase 5/metabolism , Membrane Glycoproteins/metabolism , Serine/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Membrane Glycoproteins/genetics , Mice , Phosphorylation , Protein Structure, Tertiary , Serine/genetics
7.
Stem Cell Reports ; 4(5): 873-85, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25843048

ABSTRACT

Induced pluripotent stem cells (iPSCs) provide an inexhaustible source of cells for modeling disease and testing drugs. Here we develop a bioinformatic approach to detect differences between the genomic programs of iPSCs derived from diseased versus normal human cohorts as they emerge during in vitro directed differentiation. Using iPSCs generated from a cohort carrying mutations (PiZZ) in the gene responsible for alpha-1 antitrypsin (AAT) deficiency, we find that the global transcriptomes of PiZZ iPSCs diverge from normal controls upon differentiation to hepatic cells. Expression of 135 genes distinguishes PiZZ iPSC-hepatic cells, providing potential clues to liver disease pathogenesis. The disease-specific cells display intracellular accumulation of mutant AAT protein, resulting in increased autophagic flux. Furthermore, we detect beneficial responses to the drug carbamazepine, which further augments autophagic flux, but adverse responses to known hepatotoxic drugs. Our findings support the utility of iPSCs as tools for drug development or prediction of toxicity.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , alpha 1-Antitrypsin/metabolism , Alleles , Autophagy/drug effects , Carbamazepine/toxicity , Cell Differentiation , Cells, Cultured , DNA Methylation , Enzyme-Linked Immunosorbent Assay , Epigenomics , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Liver Diseases/metabolism , Liver Diseases/pathology , Mutation , Oligonucleotide Array Sequence Analysis , Transcriptome , alpha 1-Antitrypsin/analysis , alpha 1-Antitrypsin/genetics
8.
Respir Res ; 16: 22, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25763778

ABSTRACT

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.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Lung/metabolism , MicroRNAs/metabolism , NFI Transcription Factors/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins v-myb/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , 5' Flanking Region , Animals , Binding Sites , Cell Line , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, Reporter , Mice , MicroRNAs/genetics , NFI Transcription Factors/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Oncogene Proteins v-myb/genetics , Phosphorylation , Promoter Regions, Genetic , Thyroid Nuclear Factor 1 , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Transfection
9.
An. psicol ; 30(1): 114-122, ene. 2014. tab, graf
Article in Spanish | IBECS | ID: ibc-118900

ABSTRACT

El objetivo de este estudio ha sido comprobar la posibilidad de modulación de variables de personalidad (tales como algunos de los Cinco Grandes Factores, o la personalidad resistente) a través de la aplicación de un programa de afrontamiento del estrés. Para ello, han participado 26 personas del ámbito universitario con alto estrés percibido, distribuidas en dos grupos (grupo de tratamiento y grupo de control). Los instrumentos de evaluación seleccionados se clasificaron en dos grupos: variables psicológicas y emocionales y variables de personalidad. Los resultados encontrados mediantes un ANOVA de medidas repetidas mostraron que existía inter-acción momento x grupo en las variables optimismo, extraversión y responsabilidad, correspondientes al Modelo de los Cinco Grandes Factores, mostrando un incremento de las puntuaciones en estas variables en el grupo terapia y no encontrándose dicha interacción en el grupo control. Las puntuaciones en los componentes de neuroticismo, amabilidad, apertura a la experiencia y personalidad resistente no se modificaron en ninguno de los dos grupos. Este estudio apoya la idea de que modificando determinados parámetros emocionales relacionados con el estrés mediante terapia cognitivo-conductual se pueden ver modulados algunos factores de personalidad


The objective of this study was to assess the possibility of varying personality variables (such as some of the Big Five factors, or hardiness) through the application of a stress doping therapy. Twenty-six people participated from universities with high perceived stress, divided into two groups (treatment group and control group). The assessment instruments selected were classified into two groups: psychological and emotional variables and personality variables. The results showed that after the program, the experimental group showed a decline in scores of stress, psychopathology and worries. On the other hand, personality variables, interaction was found in the variables moment x group, optimism, extraversion factor and accountability, corresponding to the model of the Big Five factors showed increased scores on these variables. However, these changes were not in the control group. The hardy personality scores did not change in either group. This study tests the modulation of certain factors of personality through cognitive-behavioral therapy for stress management, after which beneficial effects are obtained


Subject(s)
Humans , Personality Disorders/therapy , Cognitive Behavioral Therapy/methods , Stress, Psychological/therapy , Adaptation, Psychological , Psychometrics/instrumentation
10.
Am J Physiol Lung Cell Mol Physiol ; 306(5): L405-19, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24375798

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/metabolism , Idiopathic Pulmonary Fibrosis/physiopathology , Respiratory Mucosa/physiology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Fetus/metabolism , Gene Expression Regulation, Developmental/physiology , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Male , Mesoderm/metabolism , Mesoderm/physiology , Mice , Mice, Mutant Strains , Middle Aged , Pregnancy , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Species Specificity , Transcription Factors/genetics
11.
J Biol Chem ; 288(17): 12215-21, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23530051

ABSTRACT

Podoplanin (PDPN) is a transmembrane receptor that affects the activities of Rho, ezrin, and other proteins to promote tumor cell motility, invasion, and metastasis. PDPN is found in many types of cancer and may serve as a tumor biomarker and chemotherapeutic target. The intracellular region of PDPN contains only two serines, and these are conserved in mammals including mice and humans. We generated cells from the embryos of homozygous null Pdpn knock-out mice to investigate the relevance of these serines to cell growth and migration on a clear (PDPN-free) background. We report here that one or both of these serines can be phosphorylated by PKA (protein kinase A). We also report that conversion of these serines to nonphosphorylatable alanine residues enhances cell migration, whereas their conversion to phosphomimetic aspartate residues decreases cell migration. These results indicate that PKA can phosphorylate PDPN to decrease cell migration. In addition, we report that PDPN expression in fibroblasts causes them to facilitate the motility and viability of neighboring melanoma cells in coculture. These findings shed new light on how PDPN promotes cell motility, its role in tumorigenesis, and its utility as a functionally relevant biomarker and chemotherapeutic target.


Subject(s)
Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Line, Tumor , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/genetics , Fibroblasts/pathology , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Phosphorylation/genetics , Serine/genetics , Serine/metabolism
12.
J Biol Chem ; 287(44): 37282-95, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955271

ABSTRACT

The Grainyhead family of transcription factors controls morphogenesis and differentiation of epithelial cell layers in multicellular organisms by regulating cell junction- and proliferation-related genes. Grainyhead-like 2 (Grhl2) is expressed in developing mouse lung epithelium and is required for normal lung organogenesis. The specific epithelial cells expressing Grhl2 and the genes regulated by Grhl2 in normal lungs are mostly unknown. In these studies we identified the NK2-homeobox 1 transcription factor (Nkx2-1) as a direct transcriptional target of Grhl2. By binding and transcriptional assays and by confocal microscopy we showed that these two transcription factors form a positive feedback loop in vivo and in cell lines and are co-expressed in lung bronchiolar and alveolar type II cells. The morphological changes observed in flattening lung alveolar type II cells in culture are associated with down-regulation of Grhl2 and Nkx2-1. Reduction of Grhl2 in lung epithelial cell lines results in lower expression levels of Nkx2-1 and of known Grhl2 target genes. By microarray analysis we identified that in addition to Cadherin1 and Claudin4, Grhl2 regulates other cell interaction genes such as semaphorins and their receptors, which also play a functional role in developing lung epithelium. Impaired collective cell migration observed in Grhl2 knockdown cell monolayers is associated with reduced expression of these genes and may contribute to the altered epithelial phenotype reported in Grhl2 mutant mice. Thus, Grhl2 functions at the nexus of a novel regulatory network, connecting lung epithelial cell identity, migration, and cell-cell interactions.


Subject(s)
Alveolar Epithelial Cells/physiology , Cell Differentiation , Morphogenesis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Cell Shape , Chromatin Immunoprecipitation , Gene Expression , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Lung/cytology , Lung/embryology , Mice , Nuclear Proteins/genetics , Phalloidine/metabolism , Phenotype , Promoter Regions, Genetic , Protein Binding , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcriptional Activation , Transcriptome
13.
J Immunol ; 189(5): 2450-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844121

ABSTRACT

Pneumonia results from bacteria in the alveoli. The alveolar epithelium consists of type II cells, which secrete surfactant and associated proteins, and type I cells, which constitute 95% of the surface area and meet anatomic and structural needs. Other than constitutively expressed surfactant proteins, it is unknown whether alveolar epithelial cells have distinct roles in innate immunity. Because innate immunity gene induction depends on NF-κB RelA (also known as p65) during pneumonia, we generated a murine model of RelA mutated throughout the alveolar epithelium. In response to LPS, only 2 of 84 cytokine transcripts (CCL20 and CXCL5) were blunted in lungs of mutants, suggesting that a very limited subset of immune mediators is selectively elaborated by the alveolar epithelium. Lung CCL20 induction required epithelial RelA regardless of stimulus, whereas lung CXCL5 expression depended on RelA after instillation of LPS but not pneumococcus. RelA knockdown in vitro suggested that CXCL5 induction required RelA in type II cells but not type I cells. Sorted cell populations from mouse lungs revealed that CXCL5 was induced during pneumonia in type I cells, which did not require RelA. TLR2 and STING were also induced in type I cells, with RelA essential for TLR2 but not STING. To our knowledge, these data are the first direct demonstration that type I cells, which constitute the majority of the alveolar surface, mount innate immune responses during bacterial infection. These are also, to our knowledge, the first evidence for entirely RelA-independent pathways of innate immunity gene induction in any cell during pneumonia.


Subject(s)
Immunity, Innate , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Animals , Immunity, Innate/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Pneumonia, Pneumococcal/genetics , Pulmonary Alveoli/metabolism , Respiratory Mucosa/metabolism , Streptococcus pneumoniae/immunology , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Transcriptional Activation/immunology
14.
Cell Stem Cell ; 10(4): 398-411, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22482505

ABSTRACT

Two populations of Nkx2-1(+) progenitors in the developing foregut endoderm give rise to the entire postnatal lung and thyroid epithelium, but little is known about these cells because they are difficult to isolate in a pure form. We demonstrate here the purification and directed differentiation of primordial lung and thyroid progenitors derived from mouse embryonic stem cells (ESCs). Inhibition of TGFß and BMP signaling, followed by combinatorial stimulation of BMP and FGF signaling, can specify these cells efficiently from definitive endodermal precursors. When derived using Nkx2-1(GFP) knockin reporter ESCs, these progenitors can be purified for expansion in culture and have a transcriptome that overlaps with developing lung epithelium. Upon induction, they can express a broad repertoire of markers indicative of lung and thyroid lineages and can recellularize a 3D lung tissue scaffold. Thus, we have derived a pure population of progenitors able to recapitulate the developmental milestones of lung/thyroid development.


Subject(s)
Cell Separation , Embryonic Stem Cells/cytology , Lung/cytology , Thyroid Gland/cytology , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/metabolism , Lung/embryology , Lung/metabolism , Mice , Mice, Transgenic , Signal Transduction/physiology , Thyroid Gland/embryology , Thyroid Gland/metabolism , Tissue Scaffolds
15.
PLoS One ; 7(1): e29907, 2012.
Article in English | MEDLINE | ID: mdl-22242187

ABSTRACT

The homeodomain transcription factor Nkx2-1 is essential for normal lung development and homeostasis. In lung tumors, it is considered a lineage survival oncogene and prognostic factor depending on its expression levels. The target genes directly bound by Nkx2-1, that could be the primary effectors of its functions in the different cellular contexts where it is expressed, are mostly unknown. In embryonic day 11.5 (E11.5) mouse lung, epithelial cells expressing Nkx2-1 are predominantly expanding, and in E19.5 prenatal lungs, Nkx2-1-expressing cells are predominantly differentiating in preparation for birth. To evaluate Nkx2-1 regulated networks in these two cell contexts, we analyzed genome-wide binding of Nkx2-1 to DNA regulatory regions by chromatin immunoprecipitation followed by tiling array analysis, and intersected these data to expression data sets. We further determined expression patterns of Nkx2-1 developmental target genes in human lung tumors and correlated their expression levels to that of endogenous NKX2-1. In these studies we uncovered differential Nkx2-1 regulated networks in early and late lung development, and a direct function of Nkx2-1 in regulation of the cell cycle by controlling the expression of proliferation-related genes. New targets, validated in Nkx2-1 shRNA transduced cell lines, include E2f3, Cyclin B1, Cyclin B2, and c-Met. Expression levels of Nkx2-1 direct target genes identified in mouse development significantly correlate or anti-correlate to the levels of endogenous NKX2-1 in a dosage-dependent manner in multiple human lung tumor expression data sets, supporting alternative roles for Nkx2-1 as a transcriptional activator or repressor, and direct regulator of cell cycle progression in development and tumors.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Lung Neoplasms/genetics , Lung/embryology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle/genetics , Cell Proliferation , Chromatin Immunoprecipitation , Conserved Sequence , Down-Regulation/genetics , Humans , Lung/metabolism , Lung Neoplasms/pathology , Mice , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Binding/genetics , Reproducibility of Results , Signal Transduction/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/genetics
16.
J Biol Chem ; 285(3): 2152-64, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19906647

ABSTRACT

Epigenetic regulation of transcription plays an important role in cell-specific gene expression by altering chromatin structure and access of transcriptional regulators to DNA binding sites. Surfactant protein B (Sftpb) is a developmentally regulated lung epithelial gene critical for lung function. Thyroid transcription factor 1 (Nkx2-1) regulates Sftpb gene expression in various species. We show that Nkx2-1 binds to the mouse Sftpb (mSftpb) promoter in the lung. In a mouse lung epithelial cell line (MLE-15), Nkx2-1 knockdown reduces Sftpb expression, and mutation of Nkx2-1 cis-elements significantly reduces mSftpb promoter activity. Whether chromatin structure modulates Nkx2-1 regulation of Sftpb transcription is unknown. We found that DNA methylation of the mSftpb promoter inversely correlates with known patterns of Sftpb expression in vivo. The mSftpb promoter activity can be manipulated by altering its cytosine methylation status in vitro. Nkx2-1 activation of the mSftpb promoter is impaired by DNA methylation. The unmethylated Sftpb promoter shows an active chromatin structure enriched in the histone modification H3K4me3 (histone 3-lysine 4 trimethylated). The ATP-dependent chromatin remodeling protein Brg1 is recruited to the Sftpb promoter in Sftpb-expressing, but not in non-expressing tissues and cell lines. Brg1 knockdown in MLE-15 cells greatly decreases H3K4me3 levels at the Sftpb promoter region and expression of the Sftpb gene. Brg1 can be co-immunoprecipitated with Nkx2-1 protein. Last, Nkx2-1 and Brg1 with intact ATPase activity are required for mSftpb promoter activation in vitro. Our findings suggest that DNA methylation and chromatin modifications cooperate with Nkx2-1 to regulate Sftpb gene cell specific expression.


Subject(s)
Epigenesis, Genetic , Nuclear Proteins/metabolism , Pulmonary Surfactant-Associated Protein B/genetics , Transcription Factors/metabolism , Transcription, Genetic , Adenosine Triphosphatases/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line , Chromatin/metabolism , CpG Islands/genetics , DNA Helicases/metabolism , DNA Methylation , Decitabine , Epithelial Cells/metabolism , Female , Humans , Lung/cytology , Male , Mice , Promoter Regions, Genetic/genetics , Rats , Thyroid Nuclear Factor 1 , Transcriptional Activation/drug effects
17.
Carcinogenesis ; 30(8): 1433-42, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19483189

ABSTRACT

Caveolin-1 protein has been called a 'conditional tumor suppressor' because it can either suppress or enhance tumor progression depending on cellular context. Caveolin-1 levels are dynamic in non-small-cell lung cancer, with increased levels in metastatic tumor cells. We have shown previously that transactivation of an erythroblastosis virus-transforming sequence (ETS) cis-element enhances caveolin-1 expression in a murine lung epithelial cell line. Based on high sequence homology between the murine and human caveolin-1 promoters, we proposed that ETS proteins might regulate caveolin-1 expression in human lung tumorigenesis. We confirm that caveolin-1 is not detected in well-differentiated primary lung tumors. Polyoma virus enhancer activator 3 (PEA3), a pro-metastatic ETS protein in breast cancer, is expressed at low levels in well-differentiated tumors and high levels in poorly differentiated tumors. Conversely, Net, a known ETS repressor, is expressed at high levels in the nucleus of well-differentiated primary tumor cells. In tumor cells in metastatic lymph node sites, caveolin-1 and PEA3 are highly expressed, whereas Net is now expressed in the cytoplasm. We studied transcriptional regulation of caveolin-1 in two human lung cancer cell lines, Calu-1 (high caveolin-1 expressing) and NCI-H23 (low caveolin-1 expressing). Chromatin immunoprecipitation-binding assays and small interfering RNA experiments show that PEA3 is a transcriptional activator in Calu-1 cells and that Net is a transcriptional repressor in NCI-H23 cells. These results suggest that Net may suppress caveolin-1 transcription in primary lung tumors and that PEA3 may activate caveolin-1 transcription in metastatic lymph nodes.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Caveolin 1/genetics , Gene Expression Regulation, Neoplastic/physiology , Lung Neoplasms/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caveolin 1/metabolism , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Cytoplasm/metabolism , Disease Progression , Enzyme-Linked Immunosorbent Assay , Humans , Immunoenzyme Techniques , Luciferases/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphatic Metastasis , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ets , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Tumor Cells, Cultured
18.
Proc Am Thorac Soc ; 6(1): 1-10, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19131525

ABSTRACT

The Division of Lung Diseases of the National Heart, Lung, and Blood Institute (NHLBI) recently held a workshop to identify gaps in our understanding and treatment of childhood lung diseases and to define strategies to enhance translational research in this field. Leading experts with diverse experience in both laboratory and patient-oriented research reviewed selected areas of pediatric lung diseases, including perinatal programming and epigenetic influences; mechanisms of lung injury, repair, and regeneration; pulmonary vascular disease; sleep and control of breathing; and the application of novel translational methods to enhance personalized medicine. This report summarizes the proceedings of this workshop and provides recommendations for emphasis on targeted areas for future investigation. The priority areas identified for research in pediatric pulmonary diseases included: (1) epigenetic and environmental influences on lung development that program pediatric lung diseases; (2) injury, regeneration, and repair in the developing lung; (3) pulmonary vascular disease in children; (4) development and adaptation of ventilatory responses to postnatal life; (5) nonatopic wheezing: aberrant large airway development or injury?; (6) strategies to improve assessment, diagnosis, and treatment of pediatric respiratory diseases; and (7) predictive and personalized medicine for children.


Subject(s)
Biomedical Research/organization & administration , Pediatrics , Respiration Disorders , Humans , National Institutes of Health (U.S.) , Respiration Disorders/diagnosis , Respiration Disorders/genetics , Respiration Disorders/physiopathology , Respiration Disorders/therapy , United States
19.
Pediatr Pulmonol ; 44(1): 2-13, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19086051

ABSTRACT

The Division of Lung Diseases of the National Heart, Lung and Blood Institute (NHLBI) recently held a workshop to identify gaps in our understanding and treatment of childhood lung diseases and to define strategies to enhance translational research in this field. Leading experts with diverse experience in both laboratory and patient-oriented research reviewed selected areas of pediatric lung diseases, including perinatal programming and epigenetic influences; mechanisms of lung injury, repair, and regeneration; pulmonary vascular disease (PVD); sleep and control of breathing; and the application of novel translational methods to enhance personalized medicine. This report summarizes the proceedings of this workshop and provides recommendations for emphasis on targeted areas for future investigation. The priority areas identified for research in pediatric pulmonary diseases included: (1) epigenetic and environmental influences on lung development that program pediatric lung diseases, (2) injury, regeneration, and repair in the developing lung, (3) PVD in children, (4) development and adaptation of ventilatory responses to postnatal life, (5) nonatopic wheezing: aberrant large airway development or injury? (6) strategies to improve assessment, diagnosis, and treatment of pediatric respiratory diseases, and (7) predictive and personalized medicine for children.


Subject(s)
Biomedical Research , Respiratory Tract Diseases , Child , Humans , National Heart, Lung, and Blood Institute (U.S.) , United States
20.
J Clin Invest ; 118(11): 3725-37, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18924607

ABSTRACT

Mucin-type O-glycans (O-glycans) are highly expressed in vascular ECs. However, it is not known whether they are important for vascular development. To investigate the roles of EC O-glycans, we generated mice lacking T-synthase, a glycosyltransferase encoded by the gene C1galt1 that is critical for the biosynthesis of core 1-derived O-glycans, in ECs and hematopoietic cells (termed here EHC T-syn(-/-) mice). EHC T-syn(-/-) mice exhibited embryonic and neonatal lethality associated with disorganized and blood-filled lymphatic vessels. Bone marrow transplantation and EC C1galt1 transgene rescue demonstrated that lymphangiogenesis specifically requires EC O-glycans, and intestinal lymphatic microvessels in EHC T-syn(-/-) mice expressed a mosaic of blood and lymphatic EC markers. The level of O-glycoprotein podoplanin was significantly reduced in EHC T-syn(-/-) lymphatics, and podoplanin-deficient mice developed blood-filled lymphatics resembling EHC T-syn(-/-) defects. In addition, postnatal inactivation of C1galt1 caused blood/lymphatic vessel misconnections that were similar to the vascular defects in the EHC T-syn(-/-) mice. One consequence of eliminating T-synthase in ECs and hematopoietic cells was that the EHC T-syn(-/-) pups developed fatty liver disease, because of direct chylomicron deposition via misconnected portal vein and intestinal lymphatic systems. Our studies therefore demonstrate that EC O-glycans control the separation of blood and lymphatic vessels during embryonic and postnatal development, in part by regulating podoplanin expression.


Subject(s)
Endothelial Cells/immunology , Fatty Liver/immunology , Galactosyltransferases/deficiency , Lymphatic Vessels/immunology , Microvessels/immunology , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Fatty Liver/metabolism , Galactosyltransferases/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/ultrastructure , Mice , Mice, Transgenic , Microvessels/metabolism , Microvessels/ultrastructure , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...