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










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(9)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38732272

RESUMEN

Lung branching morphogenesis relies on intricate epithelial-mesenchymal interactions and signaling networks. Still, the interplay between signaling and energy metabolism in shaping embryonic lung development remains unexplored. Retinoic acid (RA) signaling influences lung proximal-distal patterning and branching morphogenesis, but its role as a metabolic modulator is unknown. Hence, this study investigates how RA signaling affects the metabolic profile of lung branching. We performed ex vivo lung explant culture of embryonic chicken lungs treated with DMSO, 1 µM RA, or 10 µM BMS493. Extracellular metabolite consumption/production was evaluated by using 1H-NMR spectroscopy. Mitochondrial respiration and biogenesis were also analyzed. Proliferation was assessed using an EdU-based assay. The expression of crucial metabolic/signaling components was examined through Western blot, qPCR, and in situ hybridization. RA signaling stimulation redirects glucose towards pyruvate and succinate production rather than to alanine or lactate. Inhibition of RA signaling reduces lung branching, resulting in a cystic-like phenotype while promoting mitochondrial function. Here, RA signaling emerges as a regulator of tissue proliferation and lactate dehydrogenase expression. Furthermore, RA governs fatty acid metabolism through an AMPK-dependent mechanism. These findings underscore RA's pivotal role in shaping lung metabolism during branching morphogenesis, contributing to our understanding of lung development and cystic-related lung disorders.


Asunto(s)
Metabolismo Energético , Pulmón , Morfogénesis , Transducción de Señal , Tretinoina , Animales , Tretinoina/metabolismo , Tretinoina/farmacología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/embriología , Metabolismo Energético/efectos de los fármacos , Morfogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Embrión de Pollo , Proliferación Celular/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Pollos
2.
Respir Res ; 24(1): 42, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36740669

RESUMEN

BACKGROUND: Clinical and experimental evidence shows lung fluid volume as a modulator of fetal lung growth with important value in treating fetal lung hypoplasia. Thus, understanding the mechanisms underlying these morphological dynamics has been the topic of multiple investigations with, however, limited results, partially due to the difficulty of capturing or recapitulating these movements in the lab. In this sense, this study aims to establish an ex vivo model allowing the study of lung fluid function in branching morphogenesis and identify the subsequent molecular/ cellular mechanisms. METHODS: Ex vivo lung explant culture was selected as a model to study branching morphogenesis, and intraluminal injections were performed to change the composition of lung fluid. Distinct chloride (Cl-) concentrations (5.8, 29, 143, and 715 mM) or Cl- channels inhibitors [antracene-9-carboxylic acid (A9C), cystic fibrosis transmembrane conductance regulator inhibitor172 (CFTRinh), and calcium-dependent Cl- channel inhibitorA01 (CaCCinh)] were injected into lung lumen at two timepoints, day0 (D0) and D2. At D4, morphological and molecular analyses were performed in terms of branching morphogenesis, spatial distribution (immunofluorescence), and protein quantification (western blot) of mechanoreceptors (PIEZO1 and PIEZO2), neuroendocrine (bombesin, ghrelin, and PGP9.5) and smooth muscle [alpha-smooth muscle actin (α-SMA) and myosin light chain 2 (MLC2)] markers. RESULTS: For the first time, we described effective intraluminal injections at D0 and D2 and demonstrated intraluminal movements at D4 in ex vivo lung explant cultures. Through immunofluorescence assay in in vivo and ex vivo branching morphogenesis, we show that PGP9.5 colocalizes with PIEZO1 and PIEZO2 receptors. Fetal lung growth is increased at higher [Cl-], 715 mM Cl-, through the overexpression of PIEZO1, PIEZO2, ghrelin, bombesin, MLC2, and α-SMA. In contrast, intraluminal injection of CFTRinh or CaCCinh decreases fetal lung growth and the expression of PIEZO1, PIEZO2, ghrelin, bombesin, MLC2, and α-SMA. Finally, the inhibition of PIEZO1/PIEZO2 by GsMTx4 decreases branching morphogenesis and ghrelin, bombesin, MLC2, and α-SMA expression in an intraluminal injection-independent manner. CONCLUSIONS: Our results identify PIEZO1/PIEZO2 expressed in neuroendocrine cells as a regulator of fetal lung growth induced by lung fluid.


Asunto(s)
Bombesina , Cloruros , Bombesina/metabolismo , Bombesina/farmacología , Ghrelina/farmacología , Pulmón/metabolismo , Mecanotransducción Celular , Morfogénesis , Proteínas de la Membrana
3.
Toxicol Lett ; 376: 13-19, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36638931

RESUMEN

Electronic cigarette usage has significantly expanded among young people and pregnant women in the last decade. Although there are already some data regarding the short- and long-term consequences of e-cigarettes on human health, their effect on embryo and lung development still needs to be fully disclosed. In this sense, this study describes, for the first time, the impact of electronic cigarette aerosol on early lung development. For this purpose, ex vivo chick (Gallus gallus) embryonic lungs were cultured in vitro for 48 h in e-cigarette aerosol exposed-medium or unexposed medium. Chick lung explants were also cultured in a cigarette smoke-exposed medium for comparison purposes. Lung explants were morphologically analyzed to assess the impact on lung growth. Additionally, TNF-α levels were determined in the supernatant as a marker of pro-inflammatory response. The results suggest that electronic cigarette aerosol impairs lung growth and promotes lung inflammation. However, its impact on early lung growth seems less detrimental than conventional cigarette smoke. This work provides significant data regarding the impact of e-cig aerosol, adding to the efforts to fully understand its effect on embryo development. The validation of these effects may eventually lead to new tobacco control recommendations for pregnant women.


Asunto(s)
Fumar Cigarrillos , Sistemas Electrónicos de Liberación de Nicotina , Adolescente , Animales , Femenino , Humanos , Embarazo , Aerosoles , Pollos , Pulmón , Nicotiana
4.
Cell Biosci ; 11(1): 134, 2021 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-34274010

RESUMEN

BACKGROUND: Lung branching morphogenesis is characterized by epithelial-mesenchymal interactions that ultimately define the airway conducting system. Throughout this process, energy and structural macromolecules are necessary to sustain the high proliferative rates. The extensive knowledge of the molecular mechanisms underlying pulmonary development contrasts with the lack of data regarding the embryonic lung metabolic requirements. Here, we studied the metabolic profile associated with the early stages of chicken pulmonary branching. METHODS: In this study, we used an ex vivo lung explant culture system and analyzed the consumption/production of extracellular metabolic intermediates associated with glucose catabolism (alanine, lactate, and acetate) by 1H-NMR spectroscopy in the culture medium. Then, we characterized the transcript levels of metabolite membrane transporters (glut1, glut3, glut8, mct1, mct3, mct4, and mct8) and glycolytic enzymes (hk1, hk2, pfk1, ldha, ldhb, pdha, and pdhb) by qPCR. ldha and ldhb mRNA spatial localization was determined by in situ hybridization. Proliferation was analyzed by directly assessing DNA synthesis using an EdU-based assay. Additionally, we performed western blot to analyze LDHA and LDHT protein levels. Finally, we used a Clark-Type Electrode to assess the lung explant's respiratory capacity. RESULTS: Glucose consumption decreases, whereas alanine, lactate, and acetate production progressively increase as branching morphogenesis proceeds. mRNA analysis revealed variations in the expression levels of key enzymes and transporters from the glycolytic pathway. ldha and ldhb displayed a compartment-specific expression pattern that resembles proximal-distal markers. In addition, high proliferation levels were detected at active branching sites. LDH protein expression levels suggest that LDHB may account for the progressive rise in lactate. Concurrently, there is a stable oxygen consumption rate throughout branching morphogenesis. CONCLUSIONS: This report describes the temporal metabolic changes that accompany the early stages of chicken lung branching morphogenesis. Overall, the embryonic chicken lung seems to shift to a glycolytic lactate-based metabolism as pulmonary branching occurs. Moreover, this metabolic rewiring might play a crucial role during lung development.

5.
Sci Rep ; 10(1): 14688, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32895458

RESUMEN

Benign prostate hyperplasia is a dysfunctional disease with an elevated prevalence. Despite the accepted impact of aging and testosterone (TES) in its pathophysiology, its aetiology remains unknown. Recent studies described that serotonin (5-HT) inhibits benign prostate growth through the modulation of the androgen receptor, in the presence of TES. Accordingly, this work aimed to determine the impact of castration and TES replacement in plasmatic and prostatic 5-HT regulation. C57BL/6 mice were submitted to surgical castration and divided into three groups, continually exposed to either vehicle or different TES doses for 14 days. Plasmatic 5-HT concentration was measured before and after castration, and after TES reintroduction. Finally, total prostatic weight and intra-prostatic 5-HT were determined in the different groups. Our results demonstrate that mice prostate exhibits high 5-HT tissue levels and that intra-prostatic total 5-HT was independent of castration or TES reintroduction, in all studied groups. Also, 5-HT plasmatic concentration significantly increased after castration and then normalized after TES administration. Our findings revealed that mice prostate has a high 5-HT content and that total prostatic 5-HT levels do not depend on androgens' action. On the other hand, castration induced a significant increase in plasmatic 5-HT concentration, raising the hypothesis that androgens might be regulating the production of extra-prostatic 5-HT.


Asunto(s)
Hipogonadismo/patología , Próstata/patología , Hiperplasia Prostática/patología , Serotonina/análisis , Testosterona/farmacología , Animales , Castración , Modelos Animales de Enfermedad , Hipogonadismo/sangre , Hipogonadismo/metabolismo , Masculino , Ratones Endogámicos C57BL , Próstata/efectos de los fármacos , Próstata/metabolismo , Hiperplasia Prostática/sangre , Hiperplasia Prostática/metabolismo , Serotonina/sangre , Serotonina/metabolismo , Testosterona/administración & dosificación , Testosterona/metabolismo
6.
Biomolecules ; 10(1)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31963453

RESUMEN

Retinoic acid (RA) is a key molecular player in embryogenesis and adult tissue homeostasis. In embryo development, RA plays a crucial role in the formation of different organ systems, namely, the respiratory system. During lung development, there is a spatiotemporal regulation of RA levels that assures the formation of a fully functional organ. RA signaling influences lung specification, branching morphogenesis, and alveolarization by regulating the expression of particular target genes. Moreover, cooperation with other developmental pathways is essential to shape lung organogenesis. This review focuses on the events regulated by retinoic acid during lung developmental phases and pulmonary vascular development; also, it aims to provide a snapshot of RA interplay with other well-known regulators of lung development.


Asunto(s)
Pulmón/irrigación sanguínea , Pulmón/crecimiento & desarrollo , Tretinoina/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Pulmón/embriología , Pulmón/metabolismo , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/metabolismo , Arteria Pulmonar/embriología , Arteria Pulmonar/crecimiento & desarrollo , Arteria Pulmonar/metabolismo , Venas Pulmonares/embriología , Venas Pulmonares/crecimiento & desarrollo , Venas Pulmonares/metabolismo , Transducción de Señal , Tretinoina/análisis
7.
Methods Mol Biol ; 2019: 209-224, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31359399

RESUMEN

Retinoic acid modulates numerous cellular events, namely, proliferation, differentiation, apoptosis, and patterning, hence influencing both embryo development and adult homeostasis. In vitro explant culture is a valuable technique for studying the impact of growth factors and signaling molecules, such as retinoic acid, in organ development since tissue architecture is maintained. This technique allows controlled supplementation of culture medium and straightforward analysis of its effect on morphogenesis. This chapter describes the detailed protocol for culturing embryonic chick lung explants and testing the impact of retinoic acid in branching and patterning, based on morphometric and molecular analysis.


Asunto(s)
Pulmón/crecimiento & desarrollo , Morfogénesis/efectos de los fármacos , Tretinoina/farmacología , Animales , Tipificación del Cuerpo/efectos de los fármacos , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hibridación in Situ , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Técnicas de Cultivo de Órganos , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXB1/genética
8.
Cell Physiol Biochem ; 45(1): 1-14, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29310117

RESUMEN

BACKGROUND: Congenital diaphragmatic hernia (CDH) is a life-threatening developmental anomaly, intrinsically combining severe pulmonary hypoplasia and hypertension. During development, signal transducers and activators of transcription (STAT) are utilized to elicit cell growth, differentiation, and survival. METHODS: We used the nitrofen-induced CDH rat model. At selected gestational time points, lungs were divided into two experimental groups, i.e., control or CDH. We performed immunohistochemistry and western blotting analysis to investigate the developmental expression profile of the complete family of STATs (STAT1-6), plus specific STATs activation (p-STAT3, p-STAT6) and regulation by SOCS (SOCS3) in normal lungs against those of diseased lungs. The normal fetal lung explants were treated with piceatannol (STAT3 inhibitor) in vitro followed by morphometrical analysis. RESULTS: Molecular profiling of STATs during the lung development revealed distinct early and late expression signatures. Experimental CDH altered the STATs expression, activation, and regulation in the fetal lungs. In particular, STAT3 and STAT6 were persistently over-expressed and early over-activated. Piceatannol treatment dose-dependently stimulated the fetal lung growth. CONCLUSION: These findings suggest that STATs play an important role during normal fetal lung development and CDH pathogenesis. Moreover, functionally targeting STAT signaling modulates fetal lung growth, which highlights that STAT3 and STAT6 signaling might be promising therapeutic targets in reducing or preventing pulmonary hypoplasia in CDH.


Asunto(s)
Pulmón/crecimiento & desarrollo , Factores de Transcripción STAT/metabolismo , Animales , Femenino , Desarrollo Fetal/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hernias Diafragmáticas Congénitas/inducido químicamente , Hernias Diafragmáticas Congénitas/patología , Inmunohistoquímica , Pulmón/metabolismo , Éteres Fenílicos/toxicidad , Ratas , Ratas Sprague-Dawley , Factores de Transcripción STAT/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/metabolismo , Estilbenos/farmacología , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
9.
Sci Rep ; 7(1): 15428, 2017 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-29133842

RESUMEN

Aging and testosterone almost inexorably cause benign prostatic hyperplasia (BPH) in Human males. However, etiology of BPH is largely unknown. Serotonin (5-HT) is produced by neuroendocrine prostatic cells and presents in high concentration in normal prostatic transition zone, but its function in prostate physiology is unknown. Previous evidence demonstrated that neuroendocrine cells and 5-HT are decreased in BPH compared to normal prostate. Here, we show that 5-HT is a strong negative regulator of prostate growth. In vitro, 5-HT inhibits rat prostate branching through down-regulation of androgen receptor (AR). This 5-HT's inhibitory mechanism is also present in human cells of normal prostate and BPH, namely in cell lines expressing AR when treated with testosterone. In both models, 5-HT's inhibitory mechanism was replicated by specific agonists of 5-Htr1a and 5-Htr1b. Since peripheral 5-HT production is specifically regulated by tryptophan hydroxylase 1(Tph1), we showed that Tph1 knockout mice present higher prostate mass and up-regulation of AR when compared to wild-type, whereas 5-HT treatment restored the prostate weight and AR levels. As 5-HT is decreased in BPH, we present here evidence that links 5-HT depletion to BPH etiology through modulation of AR. Serotoninergic prostate pathway should be explored as a new therapeutic target for BPH.


Asunto(s)
Próstata/patología , Hiperplasia Prostática/patología , Receptores Androgénicos/metabolismo , Serotonina/metabolismo , Animales , Animales Recién Nacidos , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Noqueados , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Técnicas de Cultivo de Órganos , Tamaño de los Órganos/efectos de los fármacos , Próstata/citología , Próstata/efectos de los fármacos , Próstata/metabolismo , Hiperplasia Prostática/tratamiento farmacológico , Hiperplasia Prostática/genética , Ratas , Receptores de Serotonina 5-HT1/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Agonistas del Receptor de Serotonina 5-HT1/uso terapéutico , Testosterona/metabolismo , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo , Regulación hacia Arriba
10.
J Physiol ; 593(15): 3301-11, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26096456

RESUMEN

KEY POINTS: Retinoic acid (RA) and ghrelin levels are altered in human hypoplastic lungs when compared to healthy lungs. Although considerable data have been obtained about RA, ghrelin and bombesin in the congenital diaphragmatic hernia (CDH) rat model, neuroendocrine factors have never been associated with the RA signalling pathway in this animal model. In this study, the interaction between neuroendocrine factors and RA was explored in the CDH rat model. The authors found that normal fetal lung explants treated with RA, bombesin and ghrelin showed an increase in lung growth. Hypoplastic lungs presented higher expression levels of the RA receptors α and γ. Moreover bombesin and ghrelin supplementation, in vitro, to normal lungs increased RA receptor α/γ expression whereas administration of bombesin and ghrelin antagonists to normal and hypoplastic lungs decreased it. These data reveal for the first time that there is a link between neuroendocrine factors and RA, and that neuroendocrine factors sensitise the lung to the RA action through RA receptor modulation. ABSTRACT: Congenital diaphragmatic hernia (CDH) is characterised by a spectrum of lung hypoplasia and consequent pulmonary hypertension, leading to high morbidity and mortality rates. Moreover, CDH has been associated with an increase in the levels of pulmonary neuroendocrine factors, such as bombesin and ghrelin, and a decrease in the action of retinoic acid (RA). The present study aimed to elucidate the interaction between neuroendocrine factors and RA. In vitro analyses were performed on Sprague-Dawley rat embryos. Normal lung explants were treated with bombesin, ghrelin, a bombesin antagonist, a ghrelin antagonist, dimethylsulfoxide (DMSO), RA dissolved in DMSO, bombesin plus RA and ghrelin plus RA. Hypoplastic lung explants (nitrofen model) were cultured with bombesin, ghrelin, bombesin antagonist or ghrelin antagonist. The lung explants were analysed morphometrically, and retinoic acid receptor (RAR) α, ß and γ expression levels were assessed via Western blotting. Immunohistochemistry analysis of RAR was performed in normal and hypoplastic lungs 17.5 days post-conception (dpc). Compared with the controls, hypoplastic lungs exhibited significantly higher RARα/γ expression levels. Furthermore considering hypoplastic lungs, bombesin and ghrelin antagonists decreased RARα/γ expression. Normal lung explants (13.5 dpc) treated with RA, bombesin plus RA, ghrelin plus RA, bombesin or ghrelin exhibited increased lung growth. Moreover, bombesin and ghrelin increased RARα/γ expression levels, whereas the bombesin and ghrelin antagonists decreased RARα/γ expression. This study demonstrates for the first time that neuroendocrine factors function as lung growth regulators, sensitising the lung to the action of RA through up-regulation of RARα and RARγ.


Asunto(s)
Bombesina/farmacología , Ghrelina/farmacología , Hernias Diafragmáticas Congénitas/metabolismo , Pulmón/metabolismo , Receptor alfa X Retinoide/metabolismo , Receptor gamma X Retinoide/metabolismo , Animales , Bombesina/antagonistas & inhibidores , Ghrelina/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Pulmón/embriología , Ratas , Ratas Sprague-Dawley , Receptor alfa X Retinoide/genética , Receptor gamma X Retinoide/genética
11.
Cell Physiol Biochem ; 35(1): 104-15, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25547331

RESUMEN

UNLABELLED: BACKGROUND/ AIMS: The knowledge of the molecular network that governs fetal lung branching is an essential step towards the discovery of novel therapeutic targets against pulmonary pathologies. Lung consists of two highly branched systems: airways and vasculature. Ephrins and its receptors, Eph, have been implicated in cardiovascular development, angiogenesis and vascular remodeling. This study aims to clarify the role of these factors during lung morphogenesis. METHODS: Ephrins-B1, -B2 and receptor EphB4 expression pattern was assessed in fetal rat lungs between 15.5 and 21.5 days post-conception, by immunohistochemistry. Fetal rat lungs were harvested at 13.5 dpc, cultured during 4 days and treated with increasing doses of ephrins-B1 and -B2 and the activity of key signaling pathways was assessed. RESULTS: Ephrin-B1 presents mesenchymal expression, whereas ephrin-B2 and its receptor EphB4 were expressed by the epithelium. Both ephrins stimulated pulmonary branching. Moreover, while ephrin-B1 did not affect the pathways studied, ephrin-B2 supplementation decreased activity of JNK, ERK and STAT. This study characterizes the expression pattern of ephrins-B1, -B2 and EphB4 receptor throughout rat lung development. CONCLUSION: Our data highlight a possible role of ephrins as molecular stimulators of lung morphogenesis. Moreover, it supports the idea that classical vascular factors might play a role as airway growth promoters.


Asunto(s)
Efrina-B1/metabolismo , Efrina-B2/metabolismo , Pulmón/crecimiento & desarrollo , Animales , Células Cultivadas , Desarrollo Embrionario , Efrina-B1/genética , Efrina-B1/farmacología , Efrina-B2/genética , Efrina-B2/farmacología , Epitelio/metabolismo , Femenino , Feto/metabolismo , Feto/patología , Técnicas In Vitro , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfogénesis , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor EphB4/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
12.
PLoS One ; 8(6): e67607, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826327

RESUMEN

The glycoprotein 130 (gp130) dependent family of cytokines comprises interleukin-6 (IL-6), IL-11, leukemia inhibitory factor (LIF), cardiotrophin-like cytokine (CLC), ciliary neurotrophic factor (CNTF), cardiotrophin-1 (CT-1) and oncostatin M (OSM). These cytokines share the membrane gp130 as a common signal transducer. Recently, it was demonstrated that IL-6 promotes, whereas LIF inhibits fetal lung branching. Thus, in this study, the effects on fetal lung morphogenesis of the other classical members of the gp130-type cytokines (IL-11, CLC, CNTF, CT-1 and OSM) were investigated. We also provide the first description of these cytokines and their common gp130 receptor protein expression patterns during rat lung development. Fetal rat lung explants were cultured in vitro with increasing concentrations of IL-11, CLC, CNTF, CT-1 and OSM. Treated lung explants were morphometrically analyzed and assessed for MAPK, PI3K/AKT and STAT3 signaling modifications. IL-11, which similarly to IL-6 acts through a gp130 homodimer receptor, significantly stimulated lung growth via p38 phosphorylation. On the other hand, CLC, CNTF, CT-1 and OSM, whose receptors are gp130 heterodimers, inhibited lung growth acting in different signal-transducing pathways. Thus, the present study demonstrated that although cytokines of the gp130 family share a common signal transducer, there are specific biological activities for each cytokine on lung development. Indeed, cytokine signaling through gp130 homodimers stimulate, whereas cytokine signaling through gp130 heterodimers inhibit lung branching.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Citocinas/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Animales , Apoptosis , Proliferación Celular , Femenino , Espacio Intracelular/metabolismo , Pulmón/citología , Modelos Biológicos , Ratas , Ratas Sprague-Dawley , Transducción de Señal
13.
PLoS One ; 7(1): e30517, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291973

RESUMEN

BACKGROUND: Leukemia inhibitory factor (LIF) and interleukin-6 (IL-6) are members of the family of the glycoprotein 130 (gp130)-type cytokines. These cytokines share gp130 as a common signal transducer, which explains why they show some functional redundancy. Recently, it was demonstrated that IL-6 promotes fetal lung branching. Additionally, LIF has been implicated in developmental processes of some branching organs. Thus, in this study LIF expression pattern and its effects on fetal rat lung morphogenesis were assessed. METHODOLOGY/PRINCIPAL FINDINGS: LIF and its subunit receptor LIFRα expression levels were evaluated by immunohistochemistry and western blot in fetal rat lungs of different gestational ages, ranging from 13.5 to 21.5 days post-conception. Throughout all gestational ages studied, LIF was constitutively expressed in pulmonary epithelium, whereas LIFRα was first mainly expressed in the mesenchyme, but after pseudoglandular stage it was also observed in epithelial cells. These results point to a LIF epithelium-mesenchyme cross-talk, which is known to be important for lung branching process. Regarding functional studies, fetal lung explants were cultured with increasing doses of LIF or LIF neutralizing antibodies during 4 days. MAPK, AKT, and STAT3 phosphorylation in the treated lung explants was analyzed. LIF supplementation significantly inhibited lung growth in spite of an increase in p44/42 phosphorylation. On the other hand, LIF inhibition significantly stimulated lung growth via p38 and Akt pathways. CONCLUSIONS/SIGNIFICANCE: The present study describes that LIF and its subunit receptor LIFRα are constitutively expressed during fetal lung development and that they have an inhibitory physiological role on fetal lung branching.


Asunto(s)
Feto/metabolismo , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/fisiología , Pulmón/embriología , Pulmón/metabolismo , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Factor Inhibidor de Leucemia/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/fisiología , Técnicas de Cultivo de Órganos , Organogénesis/genética , Organogénesis/fisiología , Embarazo , Alveolos Pulmonares/embriología , Alveolos Pulmonares/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Mol Med ; 18: 231-43, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22113494

RESUMEN

Antenatal stimulation of lung growth is a reasonable approach to treat congenital diaphragmatic hernia (CDH), a disease characterized by pulmonary hypoplasia and hypertension. Several evidences from the literature demonstrated a possible involvement of renin-angiotensin system (RAS) during fetal lung development. Thus, the expression pattern of renin, angiotensin-converting enzyme, angiotensinogen, type 1 (AT1) and type 2 (AT2) receptors of angiotensin II (ANGII) was assessed by immunohisto-chemistry throughout gestation, whereas the function of RAS in the fetal lung was evaluated using fetal rat lung explants. These were morphometrically analyzed and intracellular pathway alterations assessed by Western blot. In nitrofen-induced CDH model, pregnant rats were treated with saline or PD-123319. In pups, lung growth, protein/DNA ratio, radial saccular count, epithelial differentiation and lung maturation, vascular morphometry, right ventricular hypertrophy and overload molecular markers, gasometry and survival time were evaluated. Results demonstrated that all RAS components were constitutively expressed in the lung during gestation and that ANGII had a stimulatory effect on lung branching, mediated by AT1 receptor, through p44/42 and Akt phosphorylation. This stimulatory effect on lung growth was mimicked by AT2-antagonist (PD-123319) treatment. In vivo antenatal PD-123319 treatment increased lung growth, ameliorated indirect parameters of pulmonary hypertension, improved lung function and survival time in nonventilated CDH pups, without maternal or fetal deleterious effects. Therefore, this study demonstrated a local and physiologically active RAS during lung morphogenesis. Moreover, selective inhibition of AT2 receptor is presented as a putative antenatal therapy for CDH.


Asunto(s)
Feto/metabolismo , Hernias Diafragmáticas Congénitas , Sistema Renina-Angiotensina/efectos de los fármacos , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/uso terapéutico , Angiotensinógeno/metabolismo , Animales , Femenino , Hernia Diafragmática/tratamiento farmacológico , Imidazoles/farmacología , Inmunohistoquímica , Peptidil-Dipeptidasa A/metabolismo , Embarazo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo
15.
Peptides ; 32(12): 2474-83, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22015267

RESUMEN

Apelin and its receptor APJ constitute a signaling pathway best recognized as an important regulator of cardiovascular homeostasis. This multifunctional peptidergic system is currently being described to be involved in embryonic events which extend into vascular, ocular and heart development. Additionally, it is highly expressed in pulmonary tissue. Therefore, the aim of this study was to investigate the role of apelinergic system during fetal lung development. Immunohistochemistry and Western blot analysis were used to characterize apelin and APJ expression levels and cellular localization in normal fetal rat lungs, at five different gestational ages as well as in the adult. Fetal rat lung explants were cultured in vitro with increasing doses of apelin. Treated lung explants were morphometrically analyzed and assessed for MAPK signaling modifications. Both components of the apelinergic system are constitutively expressed in the developing lung, with APJ exhibiting monomeric, dimeric and oligomeric forms in the pulmonary tissue. Pulmonary epithelium also displayed constitutive nuclear localization of the receptor. Fetal apelin expression is higher than adult expression. Apelin supplementation inhibitory effect on branching morphogenesis was associated with a dose dependent decrease in p38 and JNK phosphorylation. The results presented provide the first evidence of the presence of an apelinergic system operating in the developing lung. Our findings also suggest that apelin inhibits fetal lung growth by suppressing p38 and JNK signaling pathways.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/química , Pulmón/química , Sistema de Señalización de MAP Quinasas , Receptores Acoplados a Proteínas G/química , Factores de Edad , Animales , Apelina , Receptores de Apelina , Western Blotting , Núcleo Celular/química , Embrión de Mamíferos/química , Embrión de Mamíferos/embriología , Desarrollo Embrionario , Células Epiteliales/química , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Edad Gestacional , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/farmacología , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Morfogénesis , Fosforilación , Embarazo , Ratas , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos
16.
PLoS One ; 6(3): e17660, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21412430

RESUMEN

BACKGROUND: Fibroblast growth factors (FGF) are essential key players during embryonic development. Through their specific cognate receptors (FGFR) they activate intracellular cascades, finely regulated by modulators such as Sprouty. Several FGF ligands (FGF1, 2, 7, 9, 10 and 18) signaling through the four known FGFRs, have been implicated in lung morphogenesis. Although much is known about mammalian lung, so far, the avian model has not been explored for lung studies. METHODOLOGY/PRINCIPAL FINDINGS: In this study we provide the first description of fgf10, fgfr1-4 and spry2 expression patterns in early stages of chick lung development by in situ hybridization and observe that they are expressed similarly to their mammalian counterparts. Furthermore, aiming to determine a role for FGF signaling in chick lung development, in vitro FGFR inhibition studies were performed. Lung explants treated with an FGF receptor antagonist (SU5402) presented an impairment of secondary branch formation after 48 h of culture; moreover, abnormal lung growth with a cystic appearance of secondary bronchi and reduction of the mesenchymal tissue was observed. Branching and morphometric analysis of lung explants confirmed that FGFR inhibition impaired branching morphogenesis and induced a significant reduction of the mesenchyme. CONCLUSIONS/SIGNIFICANCE: This work demonstrates that FGFRs are essential for the epithelial-mesenchymal interactions that determine epithelial branching and mesenchymal growth and validate the avian embryo as a good model for pulmonary studies, namely to explore the FGF pathway as a therapeutic target.


Asunto(s)
Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Transducción de Señal , Animales , Embrión de Pollo , Dimetilsulfóxido/farmacología , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Pulmón/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
17.
Peptides ; 29(12): 2150-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18789988

RESUMEN

Ghrelin and obestatin are two proteins that originate from post-translational processing of the preproghrelin peptide. Various authors claim an opposed role of ghrelin and obestatin in several systems. Preproghrelin mRNA is significantly expressed in airway epithelium throughout lung development, predominantly during the earliest stages. The aim of this study was to evaluate the role of ghrelin and obestatin in fetal lung development in vitro. Immunohistochemistry studies were performed at different gestational ages in order to clarify the expression pattern of ghrelin, GHS-R1a, obestatin and GPR39 during fetal lung development. Fetal rat lung explants were harvested at 13.5 days post-conception (dpc) and cultured during 4 days with increasing doses of total ghrelin, acylated ghrelin, desacyl-ghrelin, ghrelin antagonist (D-Lys(3)-GHRP-6) or obestatin. Immunohistochemistry studies demonstrated that ghrelin, GHS-R1a, obestatin and GPR39 proteins were expressed in primitive rat lung epithelium throughout all studied gestational ages. Total and acylated ghrelin supplementation significantly increased the total number of peripheral airway buds, whereas desacyl-ghrelin induced no effect. Moreover, GHS-R1a antagonist significantly decreased lung branching. Finally, obestatin supplementation induced no significant effect in the measured parameters. The present study showed that ghrelin has a positive effect in fetal lung development through its GHS-R1a receptor, whereas obestatin has no effect on lung branching.


Asunto(s)
Edad Gestacional , Ghrelina/metabolismo , Pulmón/embriología , Hormonas Peptídicas/metabolismo , Mucosa Respiratoria/embriología , Animales , Femenino , Ghrelina/antagonistas & inhibidores , Ghrelina/farmacología , Pulmón/metabolismo , Hormonas Peptídicas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ghrelina/metabolismo , Mucosa Respiratoria/metabolismo , Técnicas de Cultivo de Tejidos
18.
Endocrinology ; 149(9): 4367-73, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18511508

RESUMEN

The pathogenesis of pulmonary hypertension (PH) includes an inflammatory response. Thymulin, a zinc-dependent thymic hormone, has important immunobiological effects by inhibiting various proinflammatory cytokines and chemokines. We investigated morphological and hemodynamic effects of thymulin administration in a rat model of monocrotaline (MCT)-induced PH, as well as the pattern of proinflammatory cytokine gene expression and the intracellular pathways involved. Adult Wistar rats received an injection of MCT (60 mg/kg, sc) or an equal volume of saline. One day after, the animals randomly received during 3 wk an injection of saline, vehicle (zinc plus carboxymethyl cellulose), or thymulin (100 ng/kg, sc, daily). At d 23-25, the animals were anesthetized for hemodynamic recordings, whereas heart and lungs were collected for morphometric and molecular analysis. Thymulin prevented morphological, hemodynamic, and inflammatory cardiopulmonary profile characteristic of MCT-induced PH, whereas part of these effects were also observed in MCT-treated animals injected with the thymulin's vehicle containing zinc. The pulmonary thymulin effect was likely mediated through suppression of p38 pathway.


Asunto(s)
Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/prevención & control , Interleucina-6/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Monocrotalina , Factor Tímico Circulante/farmacología , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Corazón/anatomía & histología , Corazón/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/genética , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Pulmón/anatomía & histología , Pulmón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratas , Ratas Wistar , Factor Tímico Circulante/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
19.
Pediatr Pulmonol ; 43(7): 680-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18500730

RESUMEN

Fetal lung hypoplasia is a common finding in several fetal conditions such as congenital diaphragmatic hernia (CDH). Interestingly, previous studies have demonstrated that hypoplastic lungs have the ability to recover to normal size, when relieved from mechanical factors. However, the underlying mechanisms remain largely unknown. Recently, interleukin-6 (IL-6) has been involved in catch-up growth phenomenon in children. Thus, we hypothesized that IL-6 could mediate fetal growth recover from hypoplastic lungs. Control and nitrofen-induced hypoplastic lung explants were cultured either in normal conditions or with IL-6 neutralizing antibodies. The total number of peripheral airway buds, epithelial perimeter, and total explant area were analyzed and daily branching rates were calculated. Additionally, IL-6 mRNA and protein expression was assessed both in qualitative (by in situ hybridization and immunohistochemistry) and in quantitative (by real-time PCR and Western blot) approaches, in control and hypoplastic lungs (nitrofen and CDH groups). Nitrofen-induced hypoplastic lungs showed in vitro, out of systemic environment, the ability to recover from hypoplasia and presented daily branching rates significantly higher than controls. Blocking IL-6 activity significantly diminished the intrinsic capacity of hypoplastic fetal lungs to recover from hypoplasia and attenuated their daily branching rates. Although more exacerbated in CDH, both nitrofen-exposed lungs presented significant IL-6 mRNA and protein over-expression throughout all studied gestational ages. The present study suggests, for the first time, that fetal lung is able to recover from growth retardation through a way that resembles the catch-up growth phenomenon, and it seems to be, at least partially, orchestrated by intrinsic mechanisms implicating IL-6.


Asunto(s)
Hernia Diafragmática/complicaciones , Interleucina-6/análisis , Enfermedades Pulmonares/metabolismo , Pulmón/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Hernias Diafragmáticas Congénitas , Pulmón/crecimiento & desarrollo , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/etiología , Éteres Fenílicos/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Pediatr Surg ; 42(7): 1231-7, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17618886

RESUMEN

BACKGROUND/AIM: In congenital diaphragmatic hernia (CDH), there is pulmonary neuroendocrine cell (PNEC) hyperplasia and Clara (nonendocrine) cell hypoplasia, the meaning of which remains unknown. In embryonic/fetal lung, an intricate cross talk between Notch pathway and basic helix-loop-helix transcription factors Mash1 and Hes1 determines the balance between endocrine and nonendocrine epithelial cell fate. Differences at the molecular level in pulmonary epithelial cell differentiation between control and CDH hypoplastic lungs were investigated. MATERIAL AND METHODS: The nitrofen-induced CDH rat model was used. At 15.5 days postconception (dpc), fetuses were assigned to 2 experimental groups: control and nitrofen (exposed to nitrofen, without CDH), whereas at 17.5, 19.5, and 21.5 dpc, fetuses were assigned to 3 experimental groups: control, nitrofen, and CDH (exposed to nitrofen, with CDH). The fetal lungs were processed for expression quantification of CC10, Hes1, Mash1, and Dll1 by real-time polymerase chain reaction. RESULTS: In control fetuses, expression of all studied genes increased with gestational age. In nitrofen-exposed fetal lungs, endocrine cell marker Mash1 was downregulated only at the earliest studied gestational age, whereas Dll1 expression levels were significantly increased in the CDH group at 19.5 and 21.5 dpc. Regarding nonendocrine markers, Hes1 presented increased expression at 15.5 and 19.5 dpc, whereas CC10 was downregulated at 17.5 and 19.5 dpc but not at term. CONCLUSIONS: This study suggests that PNEC hyperplasia in CDH fetal lung is likely because of Notch signaling deregulation, whereas Clara cell hypoplasia in CDH lungs could be a consequence of protein synthesis delay, reflecting a functional maturation hindrance and not a cell fate commitment problem.


Asunto(s)
Diferenciación Celular , Células Epiteliales/patología , Enfermedades Fetales/patología , Hernia Diafragmática/complicaciones , Enfermedades Pulmonares/patología , Análisis de Varianza , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Enfermedades Fetales/inducido químicamente , Hernia Diafragmática/inducido químicamente , Hernias Diafragmáticas Congénitas , Proteínas de Homeodominio/metabolismo , Pulmón/anomalías , Pulmón/efectos de los fármacos , Pulmón/patología , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/congénito , Éteres Fenílicos , Embarazo , Efectos Tardíos de la Exposición Prenatal , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción HES-1 , Uteroglobina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...