Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(48): e2308342120, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-37983492

RESUMEN

COVID-19 pneumonia causes acute lung injury and acute respiratory distress syndrome (ALI/ARDS) characterized by early pulmonary endothelial and epithelial injuries with altered pulmonary diffusing capacity and obstructive or restrictive physiology. Growth hormone-releasing hormone receptor (GHRH-R) is expressed in the lung and heart. GHRH-R antagonist, MIA-602, has been reported to modulate immune responses to bleomycin lung injury and inflammation in granulomatous sarcoidosis. We hypothesized that MIA-602 would attenuate rVSV-SARS-CoV-2-induced pulmonary dysfunction and heart injury in a BSL-2 mouse model. Male and female K18-hACE2tg mice were inoculated with SARS-CoV-2/USA-WA1/2020, BSL-2-compliant recombinant VSV-eGFP-SARS-CoV-2-Spike (rVSV-SARS-CoV-2), or PBS, and lung viral load, weight loss, histopathology, and gene expression were compared. K18-hACE2tg mice infected with rVSV-SARS-CoV-2 were treated daily with subcutaneous MIA-602 or vehicle and conscious, unrestrained plethysmography performed on days 0, 3, and 5 (n = 7 to 8). Five days after infection mice were killed, and blood and tissues collected for histopathology and protein/gene expression. Both native SARS-CoV-2 and rVSV-SARS-CoV-2 presented similar patterns of weight loss, infectivity (~60%), and histopathologic changes. Daily treatment with MIA-602 conferred weight recovery, reduced lung perivascular inflammation/pneumonia, and decreased lung/heart ICAM-1 expression compared to vehicle. MIA-602 rescued altered respiratory rate, increased expiratory parameters (Te, PEF, EEP), and normalized airflow parameters (Penh and Rpef) compared to vehicle, consistent with decreased airway inflammation. RNASeq followed by protein analysis revealed heightened levels of inflammation and end-stage necroptosis markers, including ZBP1 and pMLKL induced by rVSV-SARS-CoV-2, that were normalized by MIA-602 treatment, consistent with an anti-inflammatory and pro-survival mechanism of action in this preclinical model of COVID-19 pneumonia.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Ratones , Masculino , Femenino , Animales , SARS-CoV-2 , COVID-19/patología , Pulmón/patología , Inflamación/patología , Síndrome de Dificultad Respiratoria/patología , Pérdida de Peso , Ratones Transgénicos , Modelos Animales de Enfermedad
2.
Exp Lung Res ; 49(1): 152-164, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37584484

RESUMEN

Purpose: Growth hormone-releasing hormone (GHRH) is a 44-amino acid peptide that regulates growth hormone (GH) secretion. We hypothesized that GHRH receptor (GHRH-R) in alveolar type 2 (AT2) cells could modulate pro-inflammatory and possibly subsequent pro-fibrotic effects of lipopolysaccharide (LPS) or cytokines, such that AT2 cells could participate in lung inflammation and fibrosis. Methods: We used human alveolar type 2 (iAT2) epithelial cells derived from induced pluripotent stem cells (iPSC) to investigate how GHRH-R modulates gene and protein expression. We tested iAT2 cells' gene expression in response to LPS or cytokines, seeking whether these mechanisms caused endogenous production of pro-inflammatory molecules or mesenchymal markers. Quantitative real-time PCR (RT-PCR) and Western blotting were used to investigate differential expression of epithelial and mesenchymal markers. Result: Incubation of iAT2 cells with LPS increased expression of IL1-ß and TNF-α in addition to mesenchymal genes, including ACTA2, FN1 and COL1A1. Alveolar epithelial cell gene expression due to LPS was significantly inhibited by GHRH-R peptide antagonist MIA-602. Incubation of iAT2 cells with cytokines like those in fibrotic lungs similarly increased expression of genes for IL1-ß, TNF-α, TGFß-1, Wnt5a, smooth muscle actin, fibronectin and collagen. Expression of mesenchymal proteins, such as N-cadherin and vimentin, were also elevated after prolonged exposure to cytokines, confirming epithelial production of pro-inflammatory molecules as an important mechanism that might lead to subsequent fibrosis. Conclusion: iAT2 cells clearly expressed the GHRH-R. Exposure to LPS or cytokines increased iAT2 cell production of pro-inflammatory factors. GHRH-R antagonist MIA-602 inhibited pro-inflammatory gene expression, implicating iAT2 cell GHRH-R signaling in lung inflammation and potentially in fibrosis.


Asunto(s)
Neumonía , Fibrosis Pulmonar , Humanos , Células Epiteliales Alveolares/metabolismo , Factor de Necrosis Tumoral alfa , Lipopolisacáridos/farmacología , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Inflamación , Citocinas
3.
Stem Cell Reports ; 17(7): 1699-1713, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35750043

RESUMEN

Conjunctival epithelial cells, which express viral-entry receptors angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine type 2 (TMPRSS2), constitute the largest exposed epithelium of the ocular surface tissue and may represent a relevant viral-entry route. To address this question, we generated an organotypic air-liquid-interface model of conjunctival epithelium, composed of basal, suprabasal, and superficial epithelial cells, and fibroblasts, which could be maintained successfully up to day 75 of differentiation. Using single-cell RNA sequencing (RNA-seq), with complementary imaging and virological assays, we observed that while all conjunctival cell types were permissive to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome expression, a productive infection did not ensue. The early innate immune response to SARS-CoV-2 infection in conjunctival cells was characterised by a robust autocrine and paracrine NF-κB activity, without activation of antiviral interferon signalling. Collectively, these data enrich our understanding of SARS-CoV-2 infection at the human ocular surface, with potential implications for the design of preventive strategies and conjunctival transplantation.


Asunto(s)
COVID-19 , Células Epiteliales/metabolismo , Humanos , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Receptores Virales/metabolismo , SARS-CoV-2
4.
Cells ; 9(10)2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096674

RESUMEN

Growth hormone-releasing hormone (GHRH) is secreted primarily from the hypothalamus, but other tissues, including the lungs, produce it locally. GHRH stimulates the release and secretion of growth hormone (GH) by the pituitary and regulates the production of GH and hepatic insulin-like growth factor-1 (IGF-1). Pituitary-type GHRH-receptors (GHRH-R) are expressed in human lungs, indicating that GHRH or GH could participate in lung development, growth, and repair. GHRH-R antagonists (i.e., synthetic peptides), which we have tested in various models, exert growth-inhibitory effects in lung cancer cells in vitro and in vivo in addition to having anti-inflammatory, anti-oxidative, and pro-apoptotic effects. One antagonist of the GHRH-R used in recent studies reviewed here, MIA-602, lessens both inflammation and fibrosis in a mouse model of bleomycin lung injury. GHRH and its peptide agonists regulate the proliferation of fibroblasts through the modulation of extracellular signal-regulated kinase (ERK) and Akt pathways. In addition to downregulating GH and IGF-1, GHRH-R antagonist MIA-602 inhibits signaling pathways relevant to inflammation, including p21-activated kinase 1-signal transducer and activator of transcription 3/nuclear factor-kappa B (PAK1-STAT3/NF-κB and ERK). MIA-602 induces fibroblast apoptosis in a dose-dependent manner, which is an effect that is likely important in antifibrotic actions. Taken together, the novel data reviewed here show that GHRH is an important peptide that participates in lung homeostasis, inflammation, wound healing, and cancer; and GHRH-R antagonists may have therapeutic potential in lung diseases.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/metabolismo , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/fisiopatología , Pulmón/metabolismo , Pulmón/fisiopatología , Animales , Respiración de la Célula , Humanos , Pulmón/patología , Enfermedades Pulmonares/patología , Estrés Oxidativo , Transducción de Señal
5.
J Bone Miner Res ; 35(8): 1452-1469, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32181934

RESUMEN

Cysteine-rich with epidermal growth factor (EGF)-like domains 2 (CRELD2) is an endoplasmic reticulum (ER)-resident chaperone highly activated under ER stress in conditions such as chondrodysplasias; however, its role in healthy skeletal development is unknown. We show for the first time that cartilage-specific deletion of Creld2 results in disrupted endochondral ossification and short limbed dwarfism, whereas deletion of Creld2 in bone results in osteopenia, with a low bone density and altered trabecular architecture. Our study provides the first evidence that CRELD2 promotes the differentiation and maturation of skeletal cells by modulating noncanonical WNT4 signaling regulated by p38 MAPK. Furthermore, we show that CRELD2 is a novel chaperone for the receptor low-density lipoprotein receptor-related protein 1 (LRP1), promoting its transport to the cell surface, and that LRP1 directly regulates WNT4 expression in chondrocytes through TGF-ß1 signaling. Therefore, our data provide a novel link between an ER-resident chaperone and the essential WNT signaling pathways active during skeletal differentiation that could be applicable in other WNT-responsive tissues. © 2020 American Society for Bone and Mineral Research. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research..


Asunto(s)
Moléculas de Adhesión Celular , Proteínas de la Matriz Extracelular , Diferenciación Celular , Condrocitos , Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Vía de Señalización Wnt
6.
Respir Physiol Neurobiol ; 274: 103367, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31911201

RESUMEN

PURPOSE: Hypoxemia limits exercise in some patients with interstitial lung disease (ILD). High levels of supplemental oxygen during exercise might allow physical training at a higher level and more effective pulmonary rehabilitation (PR). Our goals were to use graded cardiopulmonary exercise testing (CPET) to determine whether hyperoxia (FIO2≈1.0) increased exercise tolerance in patients with mild to moderate ILD. METHODS: We studied 6 patients with ILD, including idiopathic pulmonary fibrosis (IPF) and nonspecific interstitial pneumonia (NSIP). The study population included 3 females and 3 males (age 69 ± 5 [SD] years; FVC 61 ± 14 %; absolute DLCO 53 ± 19 %). Subjects underwent 2 ramped (15 W/min) CPET protocols on a cycle ergometer (Jaeger Oxycon Pro™, CareFusion Respiratory Care) breathing either air or oxygen (FIO2≈1.0) from a Douglas bag in random order. RESULTS: Minute ventilation (VE) increased significantly during CPET breathing air (pre CPET, 18 ± 2 [SEM] L/min; post CPET, 47 ± 6; P = 0.01), but it did not increase significantly breathing oxygen (pre CPET, 15 ± 3 [SEM]; post CPET, 29 ± 9; P = 0.06). Likewise, carbon dioxide production (VCO2) increased significantly during CPET breathing air (pre CPET, 450 ± 93 [SEM] mL/min; post CPET, 1311 ± 200; P = 0.01), but it did not increase significantly breathing oxygen (pre CPET, 369 ± 129; post CPET, 847 ± 832; P = 0.09). Exercise time during CPET did not differ significantly (P = 0.34) in air (5.6 ± 0.9 [SEM] min) or oxygen (7.0 ± 1.8). Increases in heart rate (HR) and Borg dyspnea index (BDI) after CPET were not affected by breathing oxygen. CONCLUSION: Exercise-induced increases in VE and VCO2 were prevented by breathing pure oxygen during CPET, demonstrating both decreased ventilatory drive and more efficient exercise at achieved workloads. Hyperoxia could enhance the ability of patients with ILD to train at higher workloads, resulting in more effective rehabilitation.


Asunto(s)
Disnea/prevención & control , Prueba de Esfuerzo , Tolerancia al Ejercicio/fisiología , Hiperoxia/inducido químicamente , Fibrosis Pulmonar Idiopática/fisiopatología , Enfermedades Pulmonares Intersticiales/fisiopatología , Consumo de Oxígeno/fisiología , Oxígeno/administración & dosificación , Anciano , Femenino , Humanos , Neumonías Intersticiales Idiopáticas/fisiopatología , Masculino , Persona de Mediana Edad
7.
PLoS Genet ; 15(7): e1008215, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31260448

RESUMEN

The unfolded protein response (UPR) is a conserved cellular response to the accumulation of proteinaceous material in endoplasmic reticulum (ER), active both in health and disease to alleviate cellular stress and improve protein folding. Multiple epiphyseal dysplasia (EDM5) is a genetic skeletal condition and a classic example of an intracellular protein aggregation disease, whereby mutant matrilin-3 forms large insoluble aggregates in the ER lumen, resulting in a specific 'disease signature' of increased expression of chaperones and foldases, and alternative splicing of the UPR effector XBP1. Matrilin-3 is expressed exclusively by chondrocytes thereby making EDM5 a perfect model system to study the role of protein aggregation in disease. In order to dissect the role of XBP1 signalling in aggregation-related conditions we crossed a p.V194D Matn3 knock-in mouse model of EDM5 with a mouse line carrying a cartilage specific deletion of XBP1 and analysed the resulting phenotype. Interestingly, the growth of mice carrying the Matn3 p.V194D mutation compounded with the cartilage specific deletion of XBP1 was severely retarded. Further phenotyping revealed increased intracellular retention of amyloid-like aggregates of mutant matrilin-3 coupled with dramatically decreased cell proliferation and increased apoptosis, suggesting a role of XBP1 signalling in protein accumulation and/or degradation. Transcriptomic analysis of chondrocytes extracted from wild type, EDM5, Xbp1-null and compound mutant lines revealed that the alternative splicing of Xbp1 is crucial in modulating levels of protein aggregation. Moreover, through detailed transcriptomic comparison with a model of metaphyseal chondrodysplasia type Schmid (MCDS), an UPR-related skeletal condition in which XBP1 was removed without overt consequences, we show for the first time that the differentiation-state of cells within the cartilage growth plate influences the UPR resulting from retention of a misfolded mutant protein and postulate that modulation of XBP1 signalling pathway presents a therapeutic target for aggregation related conditions in cells undergoing proliferation.


Asunto(s)
Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/patología , Proteína 1 de Unión a la X-Box/genética , Empalme Alternativo , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Perfilación de la Expresión Génica , Humanos , Proteínas Matrilinas/química , Proteínas Matrilinas/genética , Ratones , Osteocondrodisplasias/metabolismo , Agregado de Proteínas , Transducción de Señal , Respuesta de Proteína Desplegada , Proteína 1 de Unión a la X-Box/metabolismo
8.
Cells ; 7(12)2018 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-30486409

RESUMEN

We have previously shown nuclear respiratory factor 1 (NRF1)-mediated transcriptional programming of mitobiogenesis contributes to estrogen-induced breast cancer through modulating cell cycle progression. In this study, we report a new role of NRF1 that goes beyond that of programming mitobiogenesis. Specifically, we report a novel oncogenic function of NRF1 supporting its causative role in breast cancer development and progression. The gain of NRF1 and/or treatment with 17ß-estradiol (E2) produced heterogeneous breast cancer stem cell (BCSC)-like subsets composed of more than 10 distinct cell sub-populations. Flow sorting combined with confocal imaging of markers for pluripotency, epithelial mesenchymal transition (EMT), and BCSCs phenotypically confirmed that the BCSC-like subset arise from cell re-programming. Thus, we determined the molecular actions of NRF1 on its target gene CXCR4 because of its known role in the acquisition of the BCSC-like subset through EMT. CXCR4 was activated by NRF1 in a redox-dependent manner during malignant transformation. An NRF1-induced BCSC-like subset was able to form xenograft tumors in vivo, while inhibiting transcription of CXCR4 prevented xenograft tumor growth. Consistent with our observation of NRF1-driven breast tumorigenesis in the experimental model, higher protein levels of NRF1 were also found in human breast cancer tissue specimens. This highly novel role of NRF1 in the stochastic acquisition of BCSC-like subsets and their progression to a malignant phenotype may open an entirely new research direction targeting NRF1 signaling in invasive breast cancer. Our discovery of targeting transcriptional activation of CXCR4 to inhibit NRF1-induced oncogenic transformation provides a mechanistic explanation for estrogen-dependent breast carcinogenesis and opens new avenues in strategic therapeutics to fight breast cancer.

9.
Respir Care ; 59(12): 1872-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25185149

RESUMEN

INTRODUCTION: Pulmonary rehabilitation is effective for patients with COPD, but its benefit is less clearly established in idiopathic pulmonary fibrosis (IPF), especially in regard to levels of physical activity and health-related quality of life. The objectives were to determine whether pulmonary rehabilitation increased physical activity as assessed by the International Physical Activity Questionnaire (IPAQ), and improved quality of life and symptoms as assessed by the St George respiratory questionnaire for IPF (SGRQ-I) and the Borg dyspnea index (BDI). METHODS: Subjects who met current criteria for IPF were randomized to a 3-month pulmonary rehabilitation program (n = 11) or to a control group (n = 10). The rehabilitation group participated in twice-weekly, 90-min exercise sessions (24 total sessions). The control group maintained its preceding, normal physical activity. All subjects underwent 6-min walk tests to assess the postexertion BDI. The SGRQ-I and a 5-point self-assessment of health were completed at baseline, after 3 months of intervention or observation, and after 3-month follow-up. All subjects completed the IPAQ weekly. RESULTS: Subjects in the rehabilitation group maintained significantly higher levels of physical activity throughout the 3-month rehabilitation program (rehabilitation: 51,364 ± 57,713 [mean ± SD] metabolic equivalent of task-minutes; control: 20,832 ± 37,155, P = .027 by 2-tailed Mann-Whitney test). SGRQ-I symptom domain scores improved considerably by -9 ± 22 in the rehabilitation group, whereas in the control group they worsened (16 ± 12 rehabilitation compared with control, P = .013 by 2-tailed Mann-Whitney test). During the 3-month follow-up, self-reported physical activity levels in the rehabilitation group were 14,428 ± 8,884 metabolic equivalent of task-minutes and in the control group 16,923 ± 32,620 (P = .17 by 2-tailed Mann-Whitney test), demonstrating substantial reversal of activity in the rehabilitation group. BDI scores after 6-min walk tests did not change significantly. CONCLUSIONS: A 3-month rehabilitation program significantly improved symptoms (SGRQ-I) and physical activity levels (IPAQ) in subjects with IPF while they participated actively in the program. (ClinicalTrials.gov registration NCT01118221.).


Asunto(s)
Terapia por Ejercicio , Fibrosis Pulmonar Idiopática/rehabilitación , Actividad Motora/fisiología , Calidad de Vida , Anciano , Disnea/etiología , Prueba de Esfuerzo , Femenino , Humanos , Fibrosis Pulmonar Idiopática/complicaciones , Fibrosis Pulmonar Idiopática/fisiopatología , Masculino , Equivalente Metabólico , Persona de Mediana Edad , Capacidad de Difusión Pulmonar , Encuestas y Cuestionarios , Capacidad Vital , Caminata/fisiología
10.
Lung ; 192(3): 367-76, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24705678

RESUMEN

BACKGROUND: Patients with idiopathic pulmonary fibrosis (IPF) have severely limited exercise capacity due to dyspnea, hypoxemia, and abnormal lung mechanics. This pilot study was designed to determine whether pulmonary rehabilitation were efficacious in improving the 6-min walk test (6-MWT) distance, exercise oxygen uptake, respiratory muscle strength [maximum inspiratory pressure (MIP)], and dyspnea in patients with IPF. Underlying physiological mechanisms and effects of the intervention were investigated. METHODS: Subjects were randomly assigned to a 3-month pulmonary rehabilitation program (n = 11) or to a control group (n = 10). All subjects initially underwent the 6-MWT and constant load exercise gas exchange studies. RESULTS: Subjects in the rehabilitation group increased treadmill exercise [metabolic equivalent of task-minutes] over the first 14 sessions. Beneficial effects on physical function resulted in those who completed rehabilitation. Subjects who completed the program increased cycle ergometer time and maintained exercise oxygen consumption (exercise VO(2)) at the baseline level over 3 months, while the control group suffered a significant decrease in exercise VO(2). Rehabilitation subjects also increased their MIP. Plasma lactate doubled and brain natriuretic peptide levels increased significantly after exercise, as did the plasma amino acids glutamic acid, arginine, histidine, and methionine. These changes were associated with significant decreases in arterial oxygen saturation and increases in 15-F(2t)-isoprostanes after exercise. CONCLUSIONS: Pulmonary rehabilitation effectively maintained exercise oxygen uptake over 3 months and lengthened constant load exercise time in patients with moderately severe IPF. Exercise endurance on cycle ergometry testing was limited by dyspnea and severe hypoxemia associated with systemic oxidant stress.


Asunto(s)
Terapia por Ejercicio , Tolerancia al Ejercicio , Fibrosis Pulmonar Idiopática/rehabilitación , Pulmón/fisiopatología , Terapia por Inhalación de Oxígeno , Anciano , Biomarcadores/sangre , Disnea/fisiopatología , Disnea/rehabilitación , Prueba de Esfuerzo , Florida , Humanos , Hipoxia/fisiopatología , Hipoxia/rehabilitación , Fibrosis Pulmonar Idiopática/sangre , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/fisiopatología , Persona de Mediana Edad , Fuerza Muscular , Consumo de Oxígeno , Proyectos Piloto , Recuperación de la Función , Pruebas de Función Respiratoria , Músculos Respiratorios/fisiopatología , Factores de Tiempo , Resultado del Tratamiento
11.
Oncotarget ; 5(1): 131-9, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24318044

RESUMEN

TSC2 (Tuberous sclerosis complex 2) is an important tumour suppressor gene, mutations within which are linked to the development of tuberous sclerosis and implicated in multiple tumour types. TSC2 protein complexes with TSC1 and blocks the ability of the Rheb (Ras homolog enriched in brain) GTPase to activate mTOR (mammalian target of rapamycin), a crucial signal transducer which regulates protein synthesis and cell growth. Here, we report the characterisation of a novel isoform of TSC2 which is under direct control of the ligand-activated androgen receptor. TSC2 isoform A (TSC2A) is derived from an internal androgen-regulated alternative promoter and encodes a 508-amino acid cytoplasmic protein corresponding to the C-terminal region of full-length TSC2, lacking the interaction domain for TSC1 and containing an incomplete interaction domain required for Rheb inactivation. Expression of TSC2A is induced in response to androgens and full-length TSC2 is co-ordinately down-regulated, indicating an androgen-driven switch in TSC2 protein isoforms. In contrast to the well-characterised suppressive effect on cell proliferation of full-length TSC2 protein, both LNCaP and HEK293 cells over-expressing TSC2 isoform A proliferate more rapidly (measured by MTT assays) and have increased levels of cells in S-phase (measured by both Edu staining and FACS analysis). Our work indicates, for the first time, a novel role for this well-known tumour suppressor gene, which encodes an activator of cell proliferation in response to androgen stimulation.


Asunto(s)
Receptores Androgénicos/genética , Proteínas Supresoras de Tumor/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética , Transfección , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/metabolismo
12.
Exp Lung Res ; 36(1): 45-56, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20128681

RESUMEN

The authors investigated the mechanisms by which hypoxia regulates glutathione (GSH) in lung epithelial cells, and specifically whether the mitogen-activated protein kinase (MAPK) system is involved in the response to hypoxia. Hypoxia decreased cellular GSH content and appeared to decrease the effect of N-acetylcysteine on repletion of GSH after hypoxia. Hypoxia decreased 2 key enzyme activities that regulate GSH synthesis, glutamate cysteine ligase (GCL) (E.C. 6.3.2.2) and glutathione synthase (GS) (E.C. 6.3.2.3). No hypoxia-dependent change occurred in GCL or GS protein expression on Western blots. When epithelial cells were transfected with an adenoviral vector that caused over expression of human catalase protein (Ad.Cat or Ad.mCat), GCL and GS activities did not decrease in hypoxia. Inhibition of p38(MAPK) (using SB203580) or extracellular signal-regulated kinase (ERK; PD98059) prevented the hypoxia-dependent decrease in GCL and GS activity. To seek in vivo correlation, the authors assayed total glutathione in lungs and livers from MK2(-/-) (homozygous knockout) mice. MK2(-/-) mice are presumably unable to phosphorylate heat shock protein 27 (Hsp27) normally, because of absent kinase (MK2) activity. Liver GSH content (expressed per mg protein) was 20% less in MK2(-/-) mice than in nontransgenic Black 6 controls. Down-regulation of lung GSH content in hypoxia depends on peroxide tone of the cell and the p38(MAPK) system.


Asunto(s)
Glutamato-Cisteína Ligasa/biosíntesis , Glutatión Sintasa/biosíntesis , Glutatión/análisis , Hipoxia/metabolismo , Pulmón/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Catalasa/genética , Regulación hacia Abajo , Células Epiteliales/química , Células Epiteliales/metabolismo , Glutatión/biosíntesis , Humanos , Pulmón/química , Pulmón/citología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados
13.
Lung ; 188(2): 115-23, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20012639

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a form of idiopathic interstitial pneumonia characterized by temporally and spatially heterogeneous fibroblast proliferation and poor prognosis. No therapies have been shown in randomized clinical trials (RCT) to influence survival. Twenty-nine subjects were assigned randomly in a pilot study to a double-blind, placebo-controlled, RCT to test sildenafil in patients with IPF with forced vital capacity 40-90% and diffusing capacity 30-90% of predicted. During the 6-month experimental treatment period, patients underwent 6-min walk tests and estimation of dyspnea using the Borg scale at baseline (0 months), 3 months, and 6 months. Participants had moderate impairment of pulmonary function, and there were no significant differences between placebo (n = 15) and sildenafil (n = 14)-treated groups. Sildenafil did not significantly increase 6-min walk test distance (mean distance +/- SD after 6-month protocol: placebo 355 +/- 82 m, sildenafil 324 +/- 41 m; p = 0.256) nor did it lessen dyspnea after exercise (mean Borg score after 6-month protocol: placebo 3.4 +/- 1.6, sildenafil 4.1 +/- 2.3; p = 0.492). Adverse reactions were few and minor in nature. In this trial, sildenafil did not significantly increase 6-min walk test distance or decrease the Borg dyspnea index in patients with clinically typical IPF. This trial was registered at clinicaltrials.gov as NCT00359736.


Asunto(s)
Tolerancia al Ejercicio/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico , Piperazinas/uso terapéutico , Sulfonas/uso terapéutico , Vasodilatadores/uso terapéutico , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Disnea/tratamiento farmacológico , Disnea/etiología , Disnea/fisiopatología , Prueba de Esfuerzo , Femenino , Humanos , Fibrosis Pulmonar Idiopática/complicaciones , Fibrosis Pulmonar Idiopática/fisiopatología , Masculino , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa/efectos adversos , Proyectos Piloto , Piperazinas/efectos adversos , Capacidad de Difusión Pulmonar/efectos de los fármacos , Purinas/efectos adversos , Purinas/uso terapéutico , Citrato de Sildenafil , Sulfonas/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Vasodilatadores/efectos adversos , Capacidad Vital/efectos de los fármacos
15.
Exp Lung Res ; 34(5): 245-62, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18465403

RESUMEN

The 27-kDa heat shock protein (Hps27) is phosphorylated in a way that appears to regulate antioxidant defenses by mitogen-activated protein kinase (MAPK)-activated protein kinase 2 (MK2), a component of the p38(MAPK) pathway. To investigate the role of Hsp27 in cellular resistance to oxidant stress, lung cells (A549) were incubated with MAPK inhibitors to investigate the pathway's role in antioxidant defense. Cells were harvested for measurement of reduced gluthathione and glutathione disulfide (GSH and GSSH); or, exposed to 2,3-dimethoxy-1,4-napthoquinone (DMNQ). Inhibition of MAPK with SB203580 decreased total cellular glutathione (mean +/- SE): Vehicle, 150 +/- 20 mu M; SB203580, 57 +/- 10* (*P < .01). Inhibition of MAPK tripled [GSSG]/[GSH]: Vehicle, 0.29 +/- 0.09; SB203580, 1.06 +/- 0.43* (*P > .05; n = 6 per group). Hsp27 protein content did not change significantly after MAPK inhibition: Vehicle 2.20 +/- 0.24 ng/mg protein; SB203580, 2.03 +/- 0.34 (P > .05). Transfection of epithelial cells with wild-type (pcDNA-HA-Hsp27) or phosphomimic (pcDNA-HA-Hsp27-S3D) vector increased Hsp27 protein, which significantly protected cells from oxidant stress. Inhibition of the MAPK system, including p38(MAPK), results in cellular oxidant stress. Hsp27, which is phosphorylated by MK2 in the MAPK pathway, protects epithelial cells from oxidant stress.


Asunto(s)
Células Epiteliales/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Mucosa Respiratoria/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adenocarcinoma Papilar , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares , Naftoquinonas/farmacología , Estrés Oxidativo , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Mucosa Respiratoria/efectos de los fármacos , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
16.
Exp Lung Res ; 30(8): 653-71, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15700545

RESUMEN

Bacterial lipopolysaccharide (LPS) causes acute lung injury (ALI) and contributes to inflammation in the acute respiratory distress syndrome (ARDS) and sepsis, making mechanisms of resistance to LPS critically important in clinical settings. The authors postulated that intratracheal administration of a plasmid (pcDNA3. 0-rTNFalpha) encoding rat tumor necrosis factor-alpha (TNF-alpha) would increase resistance of mice to LPS-induced ALI or mortality. They investigated the time course and dose-response for development of LPS-induced ALI in C57/BL6 mice and sought possible protective effects of 100 microg pcDNA3.0-rTNFalpha intratracheally 1, 2, or 3 weeks before LPS challenge. Lung myeloperoxidase (MPO) activity and alveolar lavage fluid (BALF) cell counts increased significantly 48 hours after intraperitoneal (IP) LPS challenges. After pcDNA3.0-rTNFalpha pretreatment, mice challenged with LPS had lower lung/body weight ratios than mice treated with pcDNA3.0; however, other indices of lung injury did not differ. Survival of mice challenged with lethal IP LPS 2 weeks after intratracheal pcDNA3.0-rTNFalpha vector improved significantly, compared to mice pretreated with the control vector, pcDNA3.0. However, pcDNA3.0-pretreated mice tolerated LPS challenge less well than saline-pretreated controls. LPS causes neutrophilic lung injury and mortality, but pcDNA3.0-TNFalpha does not prevent ALI due to LPS. Intratracheal pcDNA3.0-rTNFalpha pretreatment significantly improves survival of mice after LPS challenge, compared to those pretreated with pcDNA3.0.


Asunto(s)
Endotoxinas/toxicidad , Lipopolisacáridos/toxicidad , Pulmón/efectos de los fármacos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Escherichia coli/inmunología , Vectores Genéticos/genética , Inyecciones Intraperitoneales , Intubación Intratraqueal , Lipopolisacáridos/administración & dosificación , Longevidad/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Peroxidasa/metabolismo , Plásmidos/genética , Síndrome de Dificultad Respiratoria/mortalidad , Síndrome de Dificultad Respiratoria/patología , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/genética
17.
Am J Physiol Lung Cell Mol Physiol ; 285(1): L189-98, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12665464

RESUMEN

Both NADH dehydrogenase (complex I) and aconitase are inactivated partially in vitro by superoxide (O2-.) and other oxidants that cause loss of iron from enzyme cubane (4Fe-4S) centers. We tested whether hypoxia-reoxygenation (H-R) by itself would decrease lung epithelial cell NADH dehydrogenase, aconitase, and succinate dehydrogenase (SDH) activities and whether transfection with adenoviral vectors expressing MnSOD (Ad.MnSOD) would inhibit oxidative enzyme inactivation and thus confirm a mechanism involving O2-. Human lung carcinoma cells with alveolar epithelial cell characteristics (A549 cells) were exposed to <1% O2-5% CO2 (hypoxia) for 24 h followed by air-5% CO2 for 24 h (reoxygenation). NADH dehydrogenase activity was assayed in submitochondrial particles; aconitase and SDH activities were measured in cell lysates. H-R significantly decreased NADH dehydrogenase, aconitase, and SDH activities. Ad.MnSOD increased mitochondrial MnSOD substantially and prevented the inhibitory effects of H-R on enzyme activities. Addition of alpha-ketoglutarate plus aspartate, but not succinate, to medium prevented cytotoxicity due to 2,3-dimethoxy-1,4-naphthoquinone. After hypoxia, cells displayed significantly increased dihydrorhodamine fluorescence, indicating increased mitochondrial oxidant production. Inhibition of NADH dehydrogenase, aconitase, and SDH activities during reoxygenation are due to excess O2-. produced in mitochondria, because enzyme inactivation can be prevented by overexpression of MnSOD.


Asunto(s)
Mitocondrias/enzimología , Daño por Reperfusión/metabolismo , Mucosa Respiratoria/enzimología , Superóxido Dismutasa/metabolismo , Aconitato Hidratasa/metabolismo , Adenocarcinoma Papilar , Adenoviridae/genética , Ácido Aspártico/farmacología , Ditiotreitol/farmacología , Complejo I de Transporte de Electrón , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Células Epiteliales/citología , Células Epiteliales/enzimología , Regulación Enzimológica de la Expresión Génica , Vectores Genéticos , Humanos , Ácidos Cetoglutáricos/farmacología , Luz , NADH Deshidrogenasa/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Estrés Oxidativo/efectos de la radiación , Oxígeno/farmacología , Protectores contra Radiación/farmacología , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/citología , Rodaminas , Succinato Deshidrogenasa/metabolismo , Ácido Succínico/farmacología , Superóxido Dismutasa/genética , Transfección , Células Tumorales Cultivadas
18.
Free Radic Biol Med ; 33(7): 1010, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12361810

RESUMEN

Nitrotyrosine (NO(2)Tyr) formation is a hallmark of acute and chronic inflammation and has been detected in a wide variety of human pathologies. However, the mechanisms responsible for this posttranslational protein modification remain elusive. While NO(2)Tyr has been considered a marker of peroxynitrite (ONOO(-)) formation previously, there is growing evidence that heme-protein peroxidase activity, in particular neutrophil-derived myeloperoxidase (MPO), significantly contributes to NO(2)Tyr formation in vivo via the oxidation of nitrite (NO(2)(-)) to nitrogen dioxide (.NO(2)). Coronary arteries from a patient with coronary artery disease, liver and lung tissues from a sickle cell disease patient, and an open lung biopsy from a lung transplant patient undergoing rejection were analyzed immunohistochemically to map relative tissue distributions of MPO and NO(2)Tyr. MPO immunodistribution was concentrated along the subendothelium in coronary tissue and hepatic veins as well as in the alveolar epithelial compartment of lung tissue from patients with sickle cell disease or acute rejection. MPO immunoreactivity strongly colocalized with NO(2)Tyr formation, which was similarly distributed in the subendothelial and epithelial regions of these tissues. The extracellular matrix protein fibronectin (FN), previously identified as a primary site of MPO association in vascular inflammatory reactions, proved to be a major target protein for tyrosine nitration, with a strong colocalization of MPO, NO(2)Tyr, and tissue FN occurring. Finally, lung tissue from MPO(-/-) mice, having tissue inflammatory responses stimulated by intraperitoneal zymosan administration, revealed less subendothelial NO(2)Tyr immunoreactivity than tissue from wild-type mice, confirming the significant role that MPO plays in catalyzing tissue nitration reactions. These observations reveal that (i) sequestration of neutrophil-derived MPO in vascular endothelial and alveolar epithelial compartments is an important aspect of MPO distribution and action in vivo, (ii) MPO-catalyzed NO(2)Tyr formation occurs in diverse vascular and pulmonary inflammatory pathologies, and (iii) extracellular matrix FN is an important target of tyrosine nitration in these inflammatory processes.


Asunto(s)
Inflamación/fisiopatología , Peroxidasa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Vasos Coronarios/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/deficiencia , Peroxidasa/genética
19.
Exp Lung Res ; 28(5): 373-89, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12097231

RESUMEN

This study tested the hypothesis that hypoxia exposure predisposed lung epithelial cells to reactive oxygen species-(ROS) mediated cellular injury. Human lung carcinoma cells (ATCC line H441) having epithelial characteristics (including lamellar bodies, surfactant protein [SP]-A, and SP-B) were cultured in air (air/5% CO(2)) or hypoxia (< 1% O(2)/5% CO(2)) for 0 to 24 hours before imposition of oxidant stress. Cellular manganese superoxide dismutase (MnSOD) activity (units/mg protein) decreased significantly after 24 hours of hypoxia. In normoxic culture after hypoxia, the cells produced increased ROS, detected as dichlorofluorescein (DCF) fluorescence and H(2)O(2) accumulation in medium. Antioxidants N-acetylcysteine (N-Ac) and ebselen inhibited increased DCF fluorescence after hypoxia. To test their ability to tolerate oxidant stress, some cells were incubated with antimycin A (100 microM) and trifluorocarbonylcyanide phenylhydrazone (10 microM) (anti A + FCCP), a mitochondrial complex III inhibitor and respiratory chain uncoupler, which together increase mitochondrial superoxide (O(2)(-)) and H(2)O(2) production. Lung epithelial cells preexposed to hypoxia released more lactate dehydrogenase (LDH) than normoxic controls in response to increased O(2)(-) production. Increased LDH release from hypoxia-preexposed cells treated with anti A + FCCP was inhibited by 1 mM N-Ac. Rotenone and myxothiazole increased DCF oxidation more in hypoxic than in normoxic cells, suggesting that mitochondrial electron transport complex I may have been altered by hypoxia preexposure.


Asunto(s)
Hipoxia de la Célula/fisiología , Células Epiteliales/metabolismo , Oxígeno/farmacología , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Adenocarcinoma Papilar , Adenosina Trifosfato/metabolismo , Anexina A5/análisis , Apoptosis/fisiología , Dióxido de Carbono/farmacología , Dimerización , Células Epiteliales/química , Etidio , Fluoresceínas , Colorantes Fluorescentes , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Neoplasias Pulmonares , Mitocondrias/metabolismo , Necrosis , Estrés Oxidativo/fisiología , Consumo de Oxígeno/fisiología , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Células Tumorales Cultivadas , Desacopladores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA