Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Respir Res ; 23(1): 44, 2022 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-35241086

RESUMEN

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is among the top 5 causes of mortality in the world and can develop as a consequence of genetic and/or environmental factors. Current efforts are focused on identifying early life insults and how these contribute to COPD development. In line with this, our study focuses on the influence of early life nicotine exposure and its potential impact on (a) lung pulmonary functions, and (b) elastase-induced emphysema in adulthood. METHODS: To address this hypothesis, we developed a model of 2 hits, delivered at different time points: mouse pups were first exposed to nicotine/placebo in utero and during lactation, and then subsequently received elastase/placebo at the age of 11 weeks. The effect of nicotine pretreatment and elastase instillation was assessed by (a) measurement of pulmonary function at post-elastase day (ped) 21, and (b) transcriptomic profiling at ped3 and 21, and complementary protein determination. Statistical significance was determined by 3- and 2-way ANOVA for pulmonary functions, and RNAseq results were analyzed using the R project. RESULTS: We did not observe any impact of nicotine pre- and early post-natal exposure compared to control samples on lung pulmonary functions in adulthood, as measured by FLEXIVENT technology. After elastase instillation, substantial lung damage was detected by x-ray tomography and was accompanied by loss in body weight at ped3 as well as an increase in cell numbers, inflammatory markers in BAL and lung volume at ped21. Lung functions showed a decrease in elastance and an increase in deep inflation volume and pressure volume (pv) loop area in animals with emphysema at ped21. Nicotine had no effect on elastance and deep inflation volume, but did affect the pv loop area in animals with emphysema at ped21. Extensive transcriptomic changes were induced by elastase at ped3 both in the nicotine-pretreated and the control samples, with several pathways common to both groups, such as for cell cycle, DNA adhesion and DNA damage. Nicotine pretreatment affected the number of lymphocytes present in BAL after elastase instillation and some of the complement pathway related proteins, arguing for a slight modification of the immune response, as well as changes related to general body metabolism. The majority of elastase-induced transcriptomic changes detected at ped3 had disappeared at ped21. In addition, transcriptomic profiling singled out a common gene pool that was independently activated by nicotine and elastase. CONCLUSIONS: Our study reports a broad spectrum of transient transcriptomic changes in mouse emphysema and identifies nicotine as influencing the emphysema-associated immune system response.


Asunto(s)
Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/genética , Esperanza de Vida , Nicotina/efectos adversos , Enfisema Pulmonar/genética , ARN/genética , Animales , Líquido del Lavado Bronquioalveolar/citología , Células Cultivadas , Modelos Animales de Enfermedad , Antígenos H-2 , Antígenos de Histocompatibilidad Clase I/biosíntesis , Ratones , Ratones Endogámicos C57BL , Elastasa Pancreática/toxicidad , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L619-L630, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32022591

RESUMEN

Mouse lung developmental maturation and final alveolarization phase begin at birth. During this dynamic process, alveolar cells modify their morphology and anchorage to the extracellular matrix. In particular, alveolar epithelial cell (AEC) type I undergo cytoplasmic flattening and folding to ensure alveoli lining. We developed FACS conditions for simultaneous isolation of alveolar epithelial and endothelial cells in the absence of specific reporters during the early and middle alveolar phase. We evidenced for the first time a pool of extractable epithelial cell populations expressing high levels of podoplanin at postnatal day (pnd)2, and we confirmed by RT-qPCR that these cells are already differentiated but still immature AEC type I. Maturation causes a decrease in isolation yields, reflecting the morphological changes that these cell populations are undergoing. Moreover, we find that major histocompatibility complex II (MHCII), reported as a good marker of AEC type II, is poorly expressed at pnd2 but highly present at pnd8. Combined experiments using LysoTracker and MHCII demonstrate the de novo acquisition of MCHII in AEC type II during lung alveolarization. The lung endothelial populations exhibit FACS signatures from vascular and lymphatic compartments. They can be concomitantly followed throughout alveolar development and were obtained with a noticeable increased yield at the last studied time point (pnd16). Our results provide new insights into early lung alveolar cell isolation feasibility and represent a valuable tool for pure AEC type I preparation as well as further in vitro two- and three-dimensional studies.


Asunto(s)
Células Epiteliales Alveolares/citología , Células Endoteliales/citología , Células Epiteliales/citología , Pulmón/citología , Alveolos Pulmonares/citología , Animales , Diferenciación Celular/fisiología , Separación Celular/métodos , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Organogénesis/fisiología
3.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L606-L618, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31967849

RESUMEN

Harmful consequences of cigarette smoke (CS) exposure during lung development can already manifest in infancy. In particular, early life exposure to nicotine, the main component of CS, was shown to affect lung development in animal models. We aimed to characterize the effect of nicotine on alveoli formation. We analyzed the kinetics of normal alveolar development during the alveolarization phase and then looked at the effect of nicotine in a mouse model of gestational and early life exposure. Immunohistochemical staining revealed that the wave of cell proliferation [i.e., vascular endothelial cells, alveolar epithelial cells (AEC) type II and mesenchymal cell] occurs at postnatal day (pnd) 8 in control and nicotine-exposed lungs. However, FACS analysis of individual epithelial alveolar cells revealed nicotine-induced transient increase of AEC type I proliferation and decrease of vascular endothelial cell proliferation at pnd8. Furthermore, nicotine increased the percentage of endothelial cells at pnd2. Transcriptomic data also showed significant changes in nicotine samples compared with the controls on cell cycle-associated genes at pnd2 but not anymore at pnd16. Accordingly, the expression of survivin, involved in cell cycle regulation, also follows a different kinetics in nicotine lung extracts. These changes resulted in an increased lung size detected by stereology at pnd16 but no longer in adult age, suggesting that nicotine can act on the pace of lung maturation. Taken together, our results indicate that early life nicotine exposure could be harmful to alveolar development independently from other toxicants contained in CS.


Asunto(s)
Lactancia/efectos de los fármacos , Pulmón/efectos de los fármacos , Exposición Materna/efectos adversos , Nicotina/efectos adversos , Embarazo/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Animales , Animales Recién Nacidos , Ciclo Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
4.
J Pathol ; 235(1): 65-78, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25116588

RESUMEN

Reactive oxygen species (ROS) participate in the pathogenesis of emphysema. Among ROS-producing enzymes, NOX NADPH oxidases are thought to be responsible for tissue injury associated with several lung pathologies. To determine whether NOX2 and/or NOX1 participate in the development of emphysema, their expression patterns were first studied by immunohistochemistry in the lungs of emphysematous patients. Subsequently, we investigated their contribution to elastase-induced emphysema using NOX2- and NOX1-deficient mice. In human lung, NOX2 was mainly detected in macrophages of control and emphysematous lungs, while NOX1 was expressed in alveolar epithelium and bronchial cells. We observed an elevated number of NOX2-positive cells in human emphysematous lungs, as well as increased NOX2 and NOX1 mRNA expression in mouse lungs following elastase exposure. Elastase-induced alveolar airspace enlargement and elastin degradation were prevented in NOX2-deficient mice, but not in NOX1-deficient mice. This protection was independent of inflammation and correlated with reduced ROS production. Concomitantly, an elevation of sirtuin 1 (SIRT1) level and a decrease of matrix metalloproteinase-9 (MMP-9) expression and activity were observed in alveolar macrophages and neutrophils. We addressed the specific role of macrophage-restricted functional NOX2 in elastase-induced lung emphysema using Ncf1 mutant mice and Ncf1 macrophage rescue mice (Ncf1 mutant mice with transgenic expression of Ncf1 only in CD68-positive mononuclear phagocytes; the MN mouse). Compared to WT mice, the lack of functional NOX2 led to decreased elastase-induced ROS production and protected against emphysema. In contrast, ROS production was restored specifically in macrophages from Ncf1 rescue mice and contributes to emphysema. Taken together, our results demonstrate that NOX2 is involved in the pathogenesis of human emphysema and macrophage-specific NOX2 participates in elastase-induced emphysema through the involvement of SIRT1/MMP-9 pathways in mice.


Asunto(s)
Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Enfisema Pulmonar/metabolismo , Sirtuina 1/metabolismo , Animales , Humanos , Inflamación/patología , Pulmón/patología , Macrófagos/patología , Ratones , NADPH Oxidasa 2 , Neutrófilos/patología , Enfisema Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo
5.
Respir Res ; 16: 118, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26415510

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a rapid progressive fibro-proliferative disorder with poor prognosis similar to lung cancer. The pathogenesis of IPF is uncertain, but loss of epithelial cells and fibroblast proliferation are thought to be central processes. Previous reports have shown that BARD1 expression is upregulated in response to hypoxia and associated with TGF-ß signaling, both recognized factors driving lung fibrosis. Differentially spliced BARD1 isoforms, in particular BARD1ß, are oncogenic drivers of proliferation in cancers of various origins. We therefore hypothesized that BARD1 and/or its isoforms might play a role in lung fibrosis. METHODS: We investigated BARD1 expression as a function of TGF-ß in cultured cells, in mice with experimentally induced lung fibrosis, and in lung biopsies from pulmonary fibrosis patients. RESULTS: FL BARD1 and BARD1ß were upregulated in response to TGF-ß in epithelial cells and fibroblasts in vitro and in vivo. Protein and mRNA expression studies showed very low expression in healthy lung tissues, but upregulated expression of full length (FL) BARD1 and BARD1ß in fibrotic tissues. CONCLUSION: Our data suggest that FL BARD1 and BARD1ß might be mediators of pleiotropic effects of TGF-ß. In particular BARD1ß might be a driver of proliferation and of pulmonary fibrosis pathogenesis and progression and represent a target for treatment.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Bleomicina , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Isoformas de Proteínas , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
6.
Respir Res ; 15: 81, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25117627

RESUMEN

OBJECTIVE: Hyperoxia exposure leads to the development of lung injury and bronchial hyperreactivity (BHR) via involvement of nitric oxide (NO) pathway. We aimed at characterizing whether the stimulation of the NO pathway by sildenafil or vasoactive intestinal peptide (VIP) is able to prevent the hyperoxia-induced development of BHR. The respective roles of the preserved lung volume and alveolar architecture, the anti-inflammatory and anti-apoptotic potentials of these treatments in the diminished lung responsiveness were also characterized. MATERIALS AND METHODS: Immature (28-day-old) rats were exposed for 72 hours to room air (Group C), hyperoxia (>95%, Group HC), or hyperoxia with the concomitant administration of vasoactive intestinal peptide (VIP, Group HV) or sildenafil (Group HS). Following exposure, the end-expiratory lung volume (EELV) was assessed plethysmographically. Airway and respiratory tissue mechanics were measured under baseline conditions and following incremental doses of methacholine to assess BHR. Inflammation was assessed by analyzing the bronchoalveolar lavage fluid (BALF), while biochemical and histological analyses were used to characterize the apoptotic and structural changes in the lungs. RESULTS: The BHR, the increased EELV, the aberrant alveolarization, and the infiltration of inflammatory cells into the BALF that developed in Group HC were all suppressed significantly by VIP or sildenafil treatment. The number of apoptotic cells increased significantly in Group HC, with no evidence of statistically significant effects on this adverse change in Groups HS and HV. CONCLUSIONS: These findings suggest that stimulating the NO pathway by sildenafil and VIP exert their beneficial effect against hyperoxia-induced BHR via preserving normal EELV, inhibiting airway inflammation and preserving the physiological lung structure, whereas the antiapoptotic potential of these treatments were not apparent in this process.


Asunto(s)
Hiperreactividad Bronquial/prevención & control , Hiperoxia/prevención & control , Pulmón/fisiología , Piperazinas/uso terapéutico , Sulfonamidas/uso terapéutico , Péptido Intestinal Vasoactivo/uso terapéutico , Animales , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/fisiopatología , Hiperoxia/patología , Hiperoxia/fisiopatología , Pulmón/efectos de los fármacos , Masculino , Piperazinas/farmacología , Purinas/farmacología , Purinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Citrato de Sildenafil , Sulfonamidas/farmacología , Péptido Intestinal Vasoactivo/farmacología
7.
Cells ; 12(19)2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37830589

RESUMEN

(1) Background: Stereological estimations significantly contributed to our understanding of lung anatomy and physiology. Taking stereology fully 3-dimensional facilitates the estimation of novel parameters. (2) Methods: We developed a protocol for the analysis of all airspaces of an entire lung. It includes (i) high-resolution synchrotron radiation-based X-ray tomographic microscopy, (ii) image segmentation using the free machine-learning tool Ilastik and ImageJ, and (iii) calculation of the airspace diameter distribution using a diameter map function. To evaluate the new pipeline, lungs from adult mice with cystic fibrosis (CF)-like lung disease (ßENaC-transgenic mice) or mice with elastase-induced emphysema were compared to healthy controls. (3) Results: We were able to show the distribution of airspace diameters throughout the entire lung, as well as separately for the conducting airways and the gas exchange area. In the pathobiological context, we observed an irregular widening of parenchymal airspaces in mice with CF-like lung disease and elastase-induced emphysema. Comparable results were obtained when analyzing lungs imaged with µCT, sugges-ting that our pipeline is applicable to different kinds of imaging modalities. (4) Conclusions: We conclude that the airspace diameter map is well suited for a detailed analysis of unevenly distri-buted structural alterations in chronic muco-obstructive lung diseases such as cystic fibrosis and COPD.


Asunto(s)
Fibrosis Quística , Enfisema , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Ratones , Animales , Fibrosis Quística/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Enfisema Pulmonar/diagnóstico por imagen , Elastasa Pancreática
8.
Int Immunol ; 23(1): 17-27, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21123830

RESUMEN

BACKGROUND: Intranasal administration of high amount of allergen was shown to induce tolerance and to reverse the allergic phenotype. However, mechanisms of tolerance induction via the mucosal route are still unclear. OBJECTIVES: To characterize the therapeutic effects of intranasal application of ovalbumin (OVA) in a mouse model of bronchial inflammation as well as the cellular and molecular mechanisms leading to protection upon re-exposure to allergen. METHODS: After induction of bronchial inflammation, mice were treated intranasally with OVA and re-exposed to OVA aerosols 10 days later. Bronchoalveolar lavage fluid (BALF), T cell proliferation and cytokine secretion were examined. The respective role of CD4(+)CD25(+) and CD4(+)CD25(-) T cells in the induction of tolerance was analysed. RESULTS: Intranasal treatment with OVA drastically reduced inflammatory cell recruitment into BALF and bronchial hyperresponsiveness upon re-exposure to allergen. Both OVA- specific-proliferation of T cells, T(h)1 and T(h)2 cytokine production from lung and bronchial lymph nodes were inhibited. Transfer of CD4(+)CD25(-) T cells, which strongly expressed membrane-bound transforming growth factor ß (mTGFß), from tolerized mice protected asthmatic recipient mice from subsequent aerosol challenges. The presence of CD4(+)CD25(+)(Foxp3(+)) T cells during the process of tolerization was indispensable to CD4(+)CD25(-) T cells to acquire regulatory properties. Whereas the presence of IL-10 appeared dispensable in this model, the suppression of CD4(+)CD25(-)mTGFß(+) T cells in transfer experiments significantly impaired the down-regulation of airways inflammation. CONCLUSION: Nasal application of OVA in established asthma led to the induction of CD4(+)CD25(-)mTGFß(+) T cells with regulatory properties, able to confer protection upon allergen re-exposure.


Asunto(s)
Asma/terapia , Linfocitos T CD4-Positivos/inmunología , Desensibilización Inmunológica/métodos , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Administración Intranasal , Alérgenos/administración & dosificación , Alérgenos/inmunología , Animales , Asma/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Proliferación Celular , Citocinas/biosíntesis , Evaluación Preclínica de Medicamentos , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología
9.
Antioxidants (Basel) ; 10(11)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34829703

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a noninflammatory progressive lung disease. Oxidative damage is a hallmark of IPF, but the sources and consequences of oxidant generation in the lungs are unclear. In this study, we addressed the link between the H2O2-generating enzyme NADPH oxidase 4 (NOX4) and di-tyrosine (DT), an oxidative post-translational modification in IPF lungs. We performed immunohistochemical staining for DT and NOX4 in pulmonary tissue from patients with IPF and controls using validated antibodies. In the healthy lung, DT showed little or no staining and NOX4 was mostly present in normal vascular endothelium. On the other hand, both markers were detected in several cell types in the IPF patients, including vascular smooth muscle cells and epithelium (bronchial cells and epithelial cells type II). The link between NOX4 and DT was addressed in human fibroblasts deficient for NOX4 activity (mutation in the CYBA gene). Induction of NOX4 by Transforming growth factor beta 1 (TGFß1) in fibroblasts led to moderate DT staining after the addition of a heme-containing peroxidase in control cells but not in the fibroblasts deficient for NOX4 activity. Our data indicate that DT is a histological marker of IPF and that NOX4 can generate a sufficient amount of H2O2 for DT formation in vitro.

10.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L312-22, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20382751

RESUMEN

Bcl-2 is an anti-apoptotic molecule preventing oxidative stress damage and cell death. We have previously shown that Bcl-2 is able to prevent hyperoxia-induced cell death when overexpressed in a murine fibrosarcoma cell line L929. We hypothesized that its specific overexpression in pulmonary epithelial type II cells could prevent hyperoxia-induced lung injury by protecting the epithelial side of the alveolo-capillary barrier. In the present work, we first showed that in vitro Bcl-2 can rescue murine pulmonary epithelial cells (MLE12) from oxygen-induced cell apoptosis, as shown by analysis of LDH release, annexin V/propidium staining, and caspase-3 activity. We then generated transgenic mice overexpressing specifically Bcl-2 in lung epithelial type II cells under surfactant protein C (SP-C) promoter (Tg-Bcl-2) and exposed them to hyperoxia. Bcl-2 did not hinder hyperoxia-induced mitochondria and DNA oxidative damage of type II cell in vivo. Accordingly, lung damage was identical in both Tg-Bcl-2 and littermate mice strains, as measured by lung weight, bronchoalveolar lavage, and protein content. Nevertheless, we observed a significant lower number of TUNEL-positive cells in type II cells isolated from Tg-Bcl-2 mice exposed to hyperoxia compared with cells isolated from littermate mice. In summary, these results show that although Bcl-2 overexpression is able to prevent hyperoxia-induced cell death at single cell level in vitro and ex vivo, it is not sufficient to prevent cell death of parenchymal cells and to protect the lung from acute damage in mice.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Células Epiteliales/metabolismo , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Pulmón/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Lesión Pulmonar Aguda/etiología , Animales , Apoptosis , Muerte Celular , Células Cultivadas , Daño del ADN , Células Epiteliales/clasificación , Hiperoxia/patología , Hiperoxia/fisiopatología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/patología , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Regulación hacia Arriba
11.
Am J Respir Crit Care Med ; 180(10): 972-81, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19661248

RESUMEN

RATIONALE: Hyperoxia-induced acute lung injury has been used for many years as a model of oxidative stress mimicking clinical acute lung injury and the acute respiratory distress syndrome. Excess quantities of reactive oxygen species (ROS) are responsible for oxidative stress-induced lung injury. ROS are produced by mitochondrial chain transport, but also by NADPH oxidase (NOX) family members. Although NOX1 and NOX2 are expressed in the lungs, their precise function has not been determined until now. OBJECTIVES: To determine whether NOX1 and NOX2 contribute in vivo to hyperoxia-induced acute lung injury. METHODS: Wild-type and NOX1- and NOX2-deficient mice, as well as primary lung epithelial and endothelial cells, were exposed to room air or 100% O(2) for 72 hours. MEASUREMENTS AND MAIN RESULTS: Lung injury was significantly prevented in NOX1-deficient mice, but not in NOX2-deficient mice. Hyperoxia-dependent ROS production was strongly reduced in lung sections, in isolated epithelial type II cells, and lung endothelial cells from NOX1-deficient mice. Concomitantly, lung cell death in situ and in primary cells was markedly decreased in NOX1-deficient mice. In wild-type mice, hyperoxia led to phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), two mitogen-activated protein kinases involved in cell death signaling, and to caspase-3 activation. In NOX1-deficient mice, JNK phosphorylation was blunted, and ERK phosphorylation and caspase-3 activation were decreased. CONCLUSIONS: NOX1 is an important contributor to ROS production and cell death of the alveolocapillary barrier during hyperoxia and is an upstream actor in oxidative stress-induced acute lung injury involving JNK and ERK pathways in mice.


Asunto(s)
Hipoxia/complicaciones , Lesión Pulmonar/enzimología , NADPH Oxidasas/fisiología , Animales , Muerte Celular/fisiología , Endotelio/citología , Células Epiteliales/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/citología , Lesión Pulmonar/etiología , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/deficiencia , Fosforilación , Especies Reactivas de Oxígeno/metabolismo
12.
Am J Respir Cell Mol Biol ; 39(4): 458-65, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18458239

RESUMEN

In systemic sclerosis (SSc), a disease characterized by fibrosis of the skin and internal organs, the occurrence of interstitial lung disease is responsible for high morbidity and mortality. We previously demonstrated that proteasome inhibitors (PI) show anti-fibrotic properties in vitro by reducing collagen production and favoring collagen degradation in a c-jun N-terminal kinase (JNK)-dependent manner in human fibroblasts. Therefore, we tested whether PI could control fibrosis development in bleomycin-induced lung injury, which is preceded by massive inflammation. We extended the study to test PI in TSK-1/+ mice, where skin fibrosis develops in the absence of overt inflammation. C57Bl/6 mice received bleomycin intratracheally and were treated or not with PI. Lung inflammation and fibrosis were assessed by histology and quantification of hydroxyproline content, type I collagen mRNA, and TGF-beta at Days 7, 15, and 21, respectively. Histology was used to detect skin fibrosis in TSK-1/+mice. The chymotryptic activity of 20S proteasome was assessed in mice blood. JNK and Smad2 phosphorylation were evaluated by Western blot on lung protein extracts. PI reduced collagen mRNA levels in murine lung fibroblasts, without affecting their viability in vitro. In addition, PI inhibited the chymotryptic activity of proteasome and enhanced JNK and TGF-beta signaling in vivo. PI failed to prevent bleomycin-induced lung inflammation and fibrosis and to attenuate skin fibrosis in TSK-1/+mice. In conclusion, our results provide direct evidence that, despite promising in vitro results, proteasome blockade may not be a strategy easily applicable to control fibrosis development in diseases such as lung fibrosis and scleroderma.


Asunto(s)
Inhibidores de Proteasoma , Fibrosis Pulmonar/patología , Esclerodermia Sistémica/patología , Animales , Bleomicina , Ácidos Borónicos/farmacología , Bortezomib , Células Cultivadas , Colágeno Tipo I/metabolismo , Fibrosis , Hidroxiprolina/metabolismo , Leupeptinas/farmacología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Pirazinas/farmacología , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Transducción de Señal , Piel/metabolismo , Piel/patología , Factor de Crecimiento Transformador beta/metabolismo
13.
Free Radic Biol Med ; 42(7): 1062-74, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17349933

RESUMEN

Bcl-2 is an antiapoptotic molecule that prevents oxidative stress damage and cell death. We investigated the possible protective mechanisms mediated by Bcl-2 during hyperoxia-induced cell death in L929 cells. In these cells, hyperoxia promoted apoptosis without DNA fragmentation. Overexpression of Bcl-2 significantly protected cells from oxygen-induced apoptosis, as shown by measurement of lactate dehydrogenase release, quantification of apoptotic nuclei, and detection of Annexin-V-positive cells. Bcl-2 partially prevented mitochondrial damage and interfered with the mitochondrial proapoptotic signaling pathway: it reduced Bax translocation to mitochondria, decreased the release of cytochrome c, and inhibited caspase 3 activation. However, treatment with the caspase inhibitor Z-VAD.fmk failed to rescue the cells from death, indicating that protection provided by Bcl-2 was due not only to caspase inhibition. Bcl-2 also prevented the release of mitochondrial apoptotic inducing factor, a mediator of caspase-independent apoptosis, correlating with the absence of oligonucleosomal DNA fragmentation. In addition, Bcl-2-overexpressing cells showed significantly higher intracellular amounts of glutathione after 72 h of oxygen exposure. In conclusion, our results demonstrate that the overexpression of Bcl-2 is able to prevent hyperoxia-induced cell death, by affecting mitochondria-dependent apoptotic pathways and increasing intracellular antioxidant compounds.


Asunto(s)
Apoptosis/fisiología , Hiperoxia/fisiopatología , Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular , Citocromos c/metabolismo , Glutatión/metabolismo , Inmunohistoquímica , Ratones , Microscopía Electrónica , Especies Reactivas de Oxígeno/metabolismo
14.
Respir Res ; 7: 45, 2006 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-16566828

RESUMEN

BACKGROUND: As prolonged hyperoxia induces extensive lung tissue damage, we set out to investigate the involvement of endothelin-1 (ET-1) receptors in these adverse changes. METHODS: Experiments were performed on four groups of mice: control animals kept in room air and a group of mice exposed to hyperoxia for 60 h were not subjected to ET-1 receptor blockade, whereas the dual ETA/ETB-receptor blocker tezosantan (TEZ) was administered via an intraperitoneal pump (10 mg/kg/day for 6 days) to other groups of normal and hyperoxic mice. The respiratory system impedance (Zrs) was measured by means of forced oscillations in the anesthetized, paralyzed and mechanically ventilated mice before and after the iv injection of ET-1 (2 microg). Changes in the airway resistance (Raw) and in the tissue damping (G) and elastance (H) of a constant-phase tissue compartment were identified from Zrs by model fitting. RESULTS: The plasma ET-1 level increased in the mice exposed to hyperoxia (3.3 +/- 1.6 pg/ml) relative to those exposed to room air (1.6 +/- 0.3 pg/ml, p < 0.05). TEZ administration prevented the hyperoxia-induced increases in G (13.1 +/- 1.7 vs. 9.6 +/- 0.3 cmH2O/l, p < 0.05) and H (59 +/- 9 vs. 41 +/- 5 cmH2O/l, p < 0.05) and inhibited the lung responses to ET-1. Hyperoxia decreased the reactivity of the airways to ET-1, whereas it elevated the reactivity of the tissues. CONCLUSION: These findings substantiate the involvement of the ET-1 receptors in the physiopathogenesis of hyperoxia-induced lung damage. Dual ET-1 receptor antagonism may well be of value in the prevention of hyperoxia-induced parenchymal damage.


Asunto(s)
Endotelina-1/farmacología , Hiperoxia/sangre , Enfermedades Pulmonares/sangre , Pulmón/efectos de los fármacos , Resistencia de las Vías Respiratorias/efectos de los fármacos , Animales , Endotelina-1/sangre , Femenino , Hiperoxia/metabolismo , Hiperoxia/fisiopatología , Pulmón/metabolismo , Pulmón/fisiopatología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/fisiopatología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Piridinas/farmacología , Receptores de Endotelina/efectos de los fármacos , Receptores de Endotelina/metabolismo , Tetrazoles/farmacología , Factores de Tiempo , Vasodilatadores/farmacología
15.
FEBS Lett ; 579(16): 3411-5, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15922330

RESUMEN

The uncoupling protein-3 (UCP3) is a mitochondrial protein expressed mainly in skeletal muscle. Among several hypotheses for its physiological function, UCP3 has been proposed to prevent excessive production of reactive oxygen species. In the present study, we evaluated the effect of an oxidative stress induced by hyperoxia on UCP3 expression in mouse skeletal muscle and C2C12 myotubes. We found that the hyperoxia-mediated oxidative stress was associated with a 5-fold and 3-fold increase of UCP3 mRNA and protein levels, respectively, in mouse muscle. Hyperoxia also enhanced reactive oxygen species production and UCP3 mRNA expression in C2C12 myotubes. Our findings support the view that both in vivo and in vitro UCP3 may modulate reactive oxygen species production in response to an oxidative stress.


Asunto(s)
Proteínas Portadoras/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Estrés Oxidativo/fisiología , Animales , Proteínas Portadoras/genética , Células Cultivadas , Canales Iónicos , Ratones , Proteínas Mitocondriales , Fibras Musculares Esqueléticas/química , Músculo Esquelético/química , Estrés Oxidativo/genética , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Desacopladora 3
16.
Free Radic Biol Med ; 76: 173-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25151121

RESUMEN

Nicotine contributes to the onset and progression of several pulmonary diseases. Among the various pathophysiological mechanisms triggered by nicotine, oxidative stress and cell death are reported in several cell types. We found that chronic exposure to nicotine (48h) induced NOX1-dependent oxidative stress and apoptosis in primary pulmonary cells. In murine (MLE-12) and human (BEAS-2B) lung epithelial cell lines, nicotine acted as a sensitizer to cell death and synergistically enhanced apoptosis when cells were concomitantly exposed to hyperoxia. The precise signaling pathway was investigated in MLE-12 cells in which NOX1 was abrogated by a specific inhibitor or stably silenced by shRNA. In the early phase of exposure (1h), nicotine mediated intracellular Ca(2+) fluxes and activation of protein kinase C, which in its turn activated NOX1, leading to cellular and mitochondrial oxidative stress. The latter triggered the intrinsic apoptotic machinery by modulating the expression of Bcl-2 and Bax. Overexpression of Bcl-2 completely prevented nicotine's detrimental effects, suggesting Bcl-2as a downstream key regulator in nicotine/NOX1-induced cell damage. These results suggest that NOX1 is a major contributor to the generation of intracellular oxidative stress induced by nicotine and might be an important molecule to target in nicotine-related lung pathologies.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Epiteliales/patología , Pulmón/patología , NADH NADPH Oxidorreductasas/fisiología , Nicotina/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , NADPH Oxidasa 1 , Agonistas Nicotínicos/farmacología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo
17.
Int J Clin Exp Pathol ; 7(2): 537-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24551274

RESUMEN

Reactive oxygen species (ROS) contribute to alveolar cell death in acute respiratory distress syndrome (ARDS) and we previously demonstrated that NOX1-derived ROS contributed to hyperoxia-induced alveolar cell death in mice. The study investigates whether NOX1 expression is modulated in epithelial cells concomitantly to cell death and associated to STAT3 signaling in the exudative phase of ARDS. In addition, the role of STAT3 activation in NOX1-dependent epithelial cell death was confirmed by using a lung epithelial cell line and in mice exposed to hyperoxia. NOX1 expression, cell death and STAT3 staining were evaluated in the lungs of control and ARDS patients by immunohistochemistry. In parallel, a stable NOX1-silenced murine epithelial cell line (MLE12) and NOX1-deficient mice were used to characterize signalling pathways. In the present study, we show that NOX1 is detected in alveolar epithelial cells of ARDS patients in the exudative stage. In addition, increased alveolar epithelial cell death and phosphorylated STAT3 are observed in ARDS patients and associated with NOX1 expression. Phosphorylated STAT3 is also correlated with TUNEL staining. We also confirmed that NOX1-dependent STAT3 activation participates to alveolar epithelial cell death. Silencing and acute inhibition of NOX1 in MLE12 led to decreased cell death and cleaved-caspase 3 induced by hyperoxia. Additionally, hyperoxia-induced STAT3 phosphorylation is dependent on NOX1 expression and associated with cell death in MLE12 and mice. This study demonstrates that NOX1 is involved in human ARDS pathophysiology and is responsible for the damage occurring in alveolar epithelial cells at least in part via STAT3 signalling pathways.


Asunto(s)
Células Epiteliales/enzimología , Hiperoxia/enzimología , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasas/metabolismo , Alveolos Pulmonares/enzimología , Síndrome de Dificultad Respiratoria/enzimología , Factor de Transcripción STAT3/metabolismo , Animales , Estudios de Casos y Controles , Caspasa 3/metabolismo , Muerte Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Humanos , Hiperoxia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADH NADPH Oxidorreductasas/deficiencia , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Fosforilación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Alveolos Pulmonares/patología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/patología , Transducción de Señal , Factores de Tiempo , Transfección
18.
Antioxid Redox Signal ; 15(3): 607-19, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21391892

RESUMEN

UNLABELLED: The pathogenesis of pulmonary fibrosis is linked to oxidative stress, possibly generated by the reactive oxygen species (ROS) generating NADPH oxidase NOX4. Epithelial cell death is a crucial early step in the development of the disease, followed only later by the fibrotic stage. We demonstrate that in lungs of patients with idiopathic lung fibrosis, there is strong expression of NOX4 in hyperplastic alveolar type II cells. AIM: To study a possible causative role of NOX4 in the death of alveolar cells, we have generated NOX4-deficient mice. RESULTS: Three weeks after administration of bleomycin, wild-type (WT) mice developed massive fibrosis, whereas NOX4-deficient mice displayed almost normal lung histology, and only little Smad2 phosphorylation and accumulation of myofibroblasts. However, the protective effects of NOX4 deficiency preceded the fibrotic stage. Indeed, at day 7 after bleomycin, lungs of WT mice showed massive increase in epithelial cell apoptosis and inflammation. In NOX4-deficient mice, no increase in apoptosis was observed, whereas inflammation was comparable to WT. In vitro, NOX4-deficient primary alveolar epithelial cells exposed to transforming growth factor-ß(1) did not generate ROS and were protected from apoptosis. Acute treatment with the NOX inhibitors also blunted transforming growth factor-ß(1)-induced apoptosis. CONCLUSION: ROS generation by NOX4 is a key player in epithelial cell death leading to pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , NADPH Oxidasas/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bleomicina/farmacología , Células Cultivadas , Expresión Génica , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Estrés Oxidativo/genética , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/patología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L619-29, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17575013

RESUMEN

Oxygen-based therapies expose lung to elevated levels of ROS and induce lung cell damage and inflammation. Injured cells are replaced through increased proliferation and differentiation of epithelial cells and fibroblasts. Failure to modulate these processes leads to excessive cell proliferation, collagen deposition, fibrosis, and chronic lung disease. Poly(ADP-ribose) polymerase-1 (PARP-1) is activated in response to DNA damage and participates in DNA repair, genomic integrity, and cell death. In this study, we evaluated the role of PARP-1 in lung repair during recovery after acute hyperoxia exposure. We exposed PARP-1 -/- and wild-type mice for 64 h to 100% hyperoxia and let them recover in air for 5-21 days. PARP-1-deficient mice exhibited significantly higher lung cell hyperplasia and proliferation than PARP-1 +/+ animals after 5 and 10 days of recovery. This was accompanied by an increased inflammatory response in PARP-1 -/- compared with wild-type animals, characterized by neutrophil infiltration and increased IL-6 levels in bronchoalveolar lavages. These lesions were reversible, since the extent of the hyperplastic regions was reduced after 21 days of recovery and did not result in fibrosis. In vitro, lung primary fibroblasts derived from PARP-1 -/- mice showed a higher proliferative response than PARP-1 +/+ cells during air recovery after hyperoxia-induced growth arrest. Altogether, these results reveal an essential role of PARP-1 in the control of cell repair and tissue remodeling after hyperoxia-induced lung injury.


Asunto(s)
Hiperoxia/patología , Pulmón/enzimología , Pulmón/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Cicatrización de Heridas , Aire , Animales , Caspasa 3/metabolismo , Proliferación Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/metabolismo , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Hiperoxia/inducido químicamente , Hiperplasia , Inmunohistoquímica , Inflamación , Pulmón/citología , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA