Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 207(2): 483-492, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34193599

RESUMEN

Alcohol use disorders (AUD) increase susceptibility to respiratory infections by 2- to 4-fold in part because of impaired alveolar macrophage (AM) immune function. Alcohol causes AM oxidative stress, diminishing AM phagocytic capacity and clearance of microbes from the alveolar space. Alcohol increases AM NADPH oxidases (Noxes), primary sources of AM oxidative stress, and reduces peroxisome proliferator-activated receptor γ (PPARγ) expression, a critical regulator of AM immune function. To investigate the underlying mechanisms of these alcohol-induced AM derangements, we hypothesized that alcohol stimulates CCAAT/enhancer-binding protein ß (C/EBPß) to suppress Nox-related microRNAs (miRs), thereby enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. Furthermore, we postulated that pharmacologic PPARγ activation with pioglitazone would inhibit C/EBPß and attenuate alcohol-induced AM dysfunction. AM isolated from human AUD subjects or otherwise healthy control subjects were examined. Compared with control AM, alcohol activated AM C/EBPß, decreased Nox1-related miR-1264 and Nox2-related miR-107, and increased Nox1, Nox2, and Nox4 expression and activity. These alcohol-induced AM derangements were abrogated by inhibition of C/EBPß, overexpression of miR-1264 or miR-107, or pioglitazone treatment. These findings define novel molecular mechanisms of alcohol-induced AM dysfunction mediated by C/EBPß and Nox-related miRs that are amenable to therapeutic targeting with PPARγ ligands. These results demonstrate that PPARγ ligands provide a novel and rapidly translatable strategy to mitigate susceptibility to respiratory infections and related morbidity in individuals with AUD.


Asunto(s)
Alcoholismo/tratamiento farmacológico , Alcoholismo/metabolismo , Etanol/efectos adversos , Macrófagos Alveolares/efectos de los fármacos , Fagocitos/efectos de los fármacos , Pioglitazona/farmacología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/metabolismo , Fagocitos/metabolismo
2.
J Biol Chem ; 295(52): 18051-18064, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33082140

RESUMEN

Evolving evidence suggests that nicotine may contribute to impaired asthma control by stimulating expression of nerve growth factor (NGF), a neurotrophin associated with airway remodeling and airway hyperresponsiveness. We explored the hypothesis that nicotine increases NGF by reducing lung fibroblast (LF) microRNA-98 (miR-98) and PPARγ levels, thus promoting airway remodeling. Levels of NGF, miR-98, PPARγ, fibronectin 1 (FN1), endothelin-1 (EDN1, herein referred to as ET-1), and collagen (COL1A1 and COL3A1) were measured in human LFs isolated from smoking donors, in mouse primary LFs exposed to nicotine (50 µg/ml), and in whole lung homogenates from mice chronically exposed to nicotine (100 µg/ml) in the drinking water. In selected studies, these pathways were manipulated in LFs with miR-98 inhibitor (anti-miR-98), miR-98 overexpression (miR-98 mimic), or the PPARγ agonist rosiglitazone. Compared with unexposed controls, nicotine increased NGF, FN1, ET-1, COL1A1, and COL3A1 expression in human and mouse LFs and mouse lung homogenates. In contrast, nicotine reduced miR-98 levels in LFs in vitro and in lung homogenates in vivo Treatment with anti-miR-98 alone was sufficient to recapitulate increases in NGF, FN1, and ET-1, whereas treatment with a miR-98 mimic significantly suppressed luciferase expression in cells transfected with a luciferase reporter linked to the putative seed sequence in the NGF 3'UTR and also abrogated nicotine-induced increases in NGF, FN1, and ET-1 in LFs. Similarly, rosiglitazone increased miR-98 and reversed nicotine-induced increases in NGF, FN1, and ET-1. Taken together, these findings demonstrate that nicotine-induced increases in NGF and other markers of airway remodeling are negatively regulated by miR-98.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Factor de Crecimiento Nervioso/metabolismo , Nicotina/toxicidad , Hipersensibilidad Respiratoria/patología , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/genética , Agonistas Nicotínicos/toxicidad , PPAR gamma , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/metabolismo
3.
Hepatology ; 69(6): 2455-2470, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30715741

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is a protein that is required for the development and survival of enteric, sympathetic, and catecholaminergic neurons. We previously reported that GDNF is protective against high fat diet (HFD)-induced hepatic steatosis in mice through suppression of hepatic expression of peroxisome proliferator activated receptor-γ and genes encoding enzymes involved in de novo lipogenesis. We also reported that transgenic overexpression of GDNF in mice prevented the HFD-induced liver accumulation of the autophagy cargo-associated protein p62/sequestosome 1 characteristic of impaired autophagy. Here we investigated the effects of GDNF on hepatic autophagy in response to increased fat load, and on hepatocyte mitochondrial fatty acid ß-oxidation and cell survival. GDNF not only prevented the reductions in the liver levels of some key autophagy-related proteins, including Atg5, Atg7, Beclin-1 and LC3A/B-II, seen in HFD-fed control mice, but enhanced their levels after 12 weeks of HFD feeding. In vitro, GDNF accelerated autophagic cargo clearance in primary mouse hepatocytes and a rat hepatocyte cell line, and reduced the phosphorylation of the mechanistic target of rapamycin complex downstream-target p70S6 kinase similar to the autophagy activator rapamycin. GDNF also enhanced mitochondrial fatty acid ß-oxidation in primary mouse and rat hepatocytes, and protected against palmitate-induced lipotoxicity. Conclusion: We demonstrate a role for GDNF in enhancing hepatic autophagy and in potentiating mitochondrial function and fatty acid oxidation. Our studies show that GDNF and its receptor agonists could be useful for enhancing hepatocyte survival and protecting against fatty acid-induced hepatic lipotoxicity.


Asunto(s)
Autofagia/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Hepatocitos/metabolismo , Lipogénesis/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Palmitatos/metabolismo , Animales , Muerte Celular , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Células Hep G2/citología , Células Hep G2/metabolismo , Hepatocitos/citología , Humanos , Lipólisis/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Consumo de Oxígeno/fisiología , Distribución Aleatoria , Ratas , Sensibilidad y Especificidad , Transducción de Señal , Sirolimus/farmacología
4.
Clin Sci (Lond) ; 134(14): 1911-1934, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32537652

RESUMEN

Cystic fibrosis (CF) is a progressive multiorgan autosomal recessive disease with devastating impact on the lungs caused by derangements of the CF transmembrane conductance regulator (CFTR) gene. Morbidity and mortality are caused by the triad of impaired mucociliary clearance, microbial infections and chronic inflammation. Pseudomonas aeruginosa is the main respiratory pathogen in individuals with CF infecting most patients in later stages. Despite its recognized clinical impact, molecular mechanisms that underlie P. aeruginosa pathogenesis and the host response to P. aeruginosa infection remain incompletely understood. The nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) γ (PPARγ), has shown to be reduced in CF airways. In the present study, we sought to investigate the upstream mechanisms repressing PPARγ expression and its impact on airway epithelial host defense. Endoplasmic reticulum-stress (ER-stress) triggered unfolded protein response (UPR) activated by misfolded CFTR and P. aeruginosa infection contributed to attenuated expression of PPARγ. Specifically, the protein kinase RNA (PKR)-like ER kinase (PERK) signaling pathway led to the enhanced expression of the CCAAT-enhancer-binding-protein homologous protein (CHOP). CHOP induction led to the repression of PPARγ expression. Mechanistically, we showed that CHOP induction mediated PPARγ attenuation, impacted the innate immune function of normal and ∆F508 primary airway epithelial cells by reducing expression of antimicrobial peptide (AMP) and paraoxanse-2 (PON-2), as well as enhancing IL-8 expression. Furthermore, mitochondrial reactive oxygen species production (mt-ROS) and ER-stress positive feedforward loop also dysregulated mitochondrial bioenergetics. Additionally, our findings implicate that PPARγ agonist pioglitazone (PIO) has beneficial effect on the host at the multicellular level ranging from host defense to mitochondrial re-energization.


Asunto(s)
Fibrosis Quística/metabolismo , PPAR gamma/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiología , Respuesta de Proteína Desplegada , Células A549 , Arildialquilfosfatasa/metabolismo , Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Estrés del Retículo Endoplásmico , Células Epiteliales/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interleucina-8/metabolismo , Mitocondrias/metabolismo , PPAR gamma/agonistas , Pioglitazona , Infecciones por Pseudomonas/inmunología , Factor de Transcripción CHOP/metabolismo , beta-Defensinas/metabolismo
6.
Am J Respir Cell Mol Biol ; 58(5): 648-657, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29182484

RESUMEN

Pulmonary hypertension (PH) is a progressive disorder that causes significant morbidity and mortality despite existing therapies. PH pathogenesis is characterized by metabolic derangements that increase pulmonary artery smooth muscle cell (PASMC) proliferation and vascular remodeling. PH-associated decreases in peroxisome proliferator-activated receptor γ (PPARγ) stimulate PASMC proliferation, and PPARγ in coordination with PPARγ coactivator 1α (PGC1α) regulates mitochondrial gene expression and biogenesis. To further examine the impact of decreases in PPARγ expression on human PASMC (HPASMC) mitochondrial function, we hypothesized that depletion of either PPARγ or PGC1α perturbs mitochondrial structure and function to stimulate PASMC proliferation. To test this hypothesis, HPASMCs were exposed to hypoxia and treated pharmacologically with the PPARγ antagonist GW9662 or with siRNA against PPARγ or PGC1α for 72 hours. HPASMC proliferation (cell counting), target mRNA levels (qRT-PCR), target protein levels (Western blotting), mitochondria-derived H2O2 (confocal immunofluorescence), mitochondrial mass and fragmentation, and mitochondrial bioenergetic profiling were determined. Hypoxia or knockdown of either PPARγ or PGC1α increased HPASMC proliferation, enhanced mitochondria-derived H2O2, decreased mitochondrial mass, stimulated mitochondrial fragmentation, and impaired mitochondrial bioenergetics. Taken together, these findings provide novel evidence that loss of PPARγ diminishes PGC1α and stimulates derangements in mitochondrial structure and function that cause PASMC proliferation. Overexpression of PGC1α reversed hypoxia-induced HPASMC derangements. This study identifies additional mechanistic underpinnings of PH, and provides support for the notion of activating PPARγ as a novel therapeutic strategy in PH.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR gamma/metabolismo , Anilidas/farmacología , Animales , Hipoxia de la Célula , Proliferación Celular , Células Cultivadas , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/prevención & control , Ratones Endogámicos C57BL , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/patología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Interferencia de ARN
7.
FASEB J ; 31(8): 3608-3621, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28442545

RESUMEN

Pseudomonas aeruginosa is a significant contributor to recalcitrant multidrug-resistant infections, especially in immunocompromised and hospitalized patients. The pathogenic profile of P. aeruginosa is related to its ability to secrete a variety of virulence factors and to promote biofilm formation. Quorum sensing (QS) is a mechanism wherein P. aeruginosa secretes small diffusible molecules, specifically acyl homo serine lactones, such as N-(3-oxo-dodecanoyl)-l-homoserine lactone (3O-C12-HSL), that promote biofilm formation and virulence via interbacterial communication. Strategies that strengthen the host's ability to inhibit bacterial virulence would enhance host defenses and improve the treatment of resistant infections. We have recently shown that peroxisome proliferator-activated receptor γ (PPARγ) agonists are potent immunostimulators that play a pivotal role in host response to virulent P. aeruginosa Here, we show that QS genes in P. aeruginosa (strain PAO1) and 3O-C12-HSL attenuate PPARγ expression in bronchial epithelial cells. PAO1 and 3O-C12-HSL induce barrier derangements in bronchial epithelial cells by lowering the expression of junctional proteins, such as zonula occludens-1, occludin, and claudin-4. Expression of these proteins was restored in cells that were treated with pioglitazone, a PPARγ agonist, before infection with PAO1 and 3O-C12-HSL. Barrier function and bacterial permeation studies that have been performed in primary human epithelial cells showed that PPARγ agonists are able to restore barrier integrity and function that are disrupted by PAO1 and 3O-C12-HSL. Mechanistically, we show that these effects are dependent on the induction of paraoxonase-2, a QS hydrolyzing enzyme, that mitigates the effects of QS molecules. Importantly, our data show that pioglitazone, a PPARγ agonist, significantly inhibits biofilm formation on epithelial cells by a mechanism that is mediated via paraoxonase-2. These findings elucidate a novel role for PPARγ in host defense against P. aeruginosa Strategies that activate PPARγ can provide a therapeutic complement for treatment of resistant P. aeruginosa infections.-Bedi, B., Maurice, N. M., Ciavatta, V. T., Lynn, K. S., Yuan, Z., Molina, S. A., Joo, M., Tyor, W. R., Goldberg, J. B., Koval, M., Hart, C. M., Sadikot, R. T. Peroxisome proliferator-activated receptor-γ agonists attenuate biofilm formation by Pseudomonas aeruginosa.


Asunto(s)
Proteínas Bacterianas/farmacología , Biopelículas/crecimiento & desarrollo , PPAR gamma/agonistas , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/metabolismo , Línea Celular , Células Epiteliales/microbiología , Regulación de la Expresión Génica/fisiología , Humanos , Mutación , Pseudomonas aeruginosa/genética , Percepción de Quorum
8.
Am J Respir Cell Mol Biol ; 56(1): 131-144, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27612006

RESUMEN

Pulmonary hypertension (PH), a serious complication of sickle cell disease (SCD), causes significant morbidity and mortality. Although a recent study determined that hemin release during hemolysis triggers endothelial dysfunction in SCD, the pathogenesis of SCD-PH remains incompletely defined. This study examines peroxisome proliferator-activated receptor γ (PPARγ) regulation in SCD-PH and endothelial dysfunction. PH and right ventricular hypertrophy were studied in Townes humanized sickle cell (SS) and littermate control (AA) mice. In parallel studies, SS or AA mice were gavaged with the PPARγ agonist, rosiglitazone (RSG), 10 mg/kg/day, or vehicle for 10 days. In vitro, human pulmonary artery endothelial cells (HPAECs) were treated with vehicle or hemin for 72 hours, and selected HPAECs were treated with RSG. SS mice developed PH and right ventricular hypertrophy associated with reduced lung levels of PPARγ and increased levels of microRNA-27a (miR-27a), v-ets avian erythroblastosis virus E26 oncogene homolog 1 (ETS1), endothelin-1 (ET-1), and markers of endothelial dysfunction (platelet/endothelial cell adhesion molecule 1 and E selectin). HPAECs treated with hemin had increased ETS1, miR-27a, ET-1, and endothelial dysfunction and decreased PPARγ levels. These derangements were attenuated by ETS1 knockdown, inhibition of miR-27a, or PPARγ overexpression. In SS mouse lung or in hemin-treated HPAECs, activation of PPARγ with RSG attenuated reductions in PPARγ and increases in miR-27a, ET-1, and markers of endothelial dysfunction. In SCD-PH pathogenesis, ETS1 stimulates increases in miR-27a levels that reduce PPARγ and increase ET-1 and endothelial dysfunction. PPARγ activation attenuated SCD-associated signaling derangements, suggesting a novel therapeutic approach to attenuate SCD-PH pathogenesis.


Asunto(s)
Anemia de Células Falciformes/patología , Células Endoteliales/metabolismo , Endotelina-1/metabolismo , Pulmón/patología , MicroARNs/metabolismo , PPAR gamma/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Anemia de Células Falciformes/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hemina/farmacología , Humanos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/fisiopatología , Ligandos , Ratones , Modelos Biológicos , Arteria Pulmonar/patología , Rosiglitazona , Sístole/efectos de los fármacos , Tiazolidinedionas/farmacología , Regulación hacia Arriba/efectos de los fármacos
9.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L371-L383, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28522568

RESUMEN

Pulmonary hypertension (PH) is a progressive disorder whose cellular pathogenesis involves enhanced smooth muscle cell (SMC) proliferation and resistance to apoptosis signals. Existing evidence demonstrates that the tumor suppressor programmed cell death 4 (PDCD4) affects patterns of cell growth and repair responses in the systemic vasculature following experimental injury. In the current study, the regulation PDCD4 and its functional effects on growth and apoptosis susceptibility in pulmonary artery smooth muscle cells were explored. We previously demonstrated that pharmacological activation of the nuclear transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) attenuated hypoxia-induced proliferation of human pulmonary artery smooth muscle cells (HPASMCs) by inhibiting the expression and mitogenic functions of microRNA-21 (miR-21). In the current study, we hypothesize that PPARγ stimulates PDCD4 expression and HPASMC apoptosis by inhibiting miR-21. Our findings demonstrate that PDCD4 is reduced in the mouse lung upon exposure to chronic hypoxia (10% O2 for 3 wk) and in hypoxia-exposed HPASMCs (1% O2). HPASMC apoptosis was reduced by hypoxia, by miR-21 overexpression, or by siRNA-mediated PPARγ and PDCD4 depletion. Activation of PPARγ inhibited miR-21 expression and resultant proliferation, while restoring PDCD4 levels and apoptosis to baseline. Additionally, pharmacological activation of PPARγ with rosiglitazone enhanced PDCD4 protein expression and apoptosis in a dose-dependent manner as demonstrated by increased annexin V detection by flow cytometry. Collectively, these findings demonstrate that PPARγ confers growth-inhibitory signals in hypoxia-exposed HPASMCs through suppression of miR-21 and the accompanying derepression of PDCD4 that augments HPASMC susceptibility to undergo apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR gamma/metabolismo , Arteria Pulmonar/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Anexina A5/genética , Anexina A5/metabolismo , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Miocitos del Músculo Liso/efectos de los fármacos , PPAR gamma/genética , Arteria Pulmonar/efectos de los fármacos , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tiazolidinedionas/farmacología
10.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L599-L608, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130258

RESUMEN

Pulmonary hypertension (PH) is characterized by increased pulmonary vascular resistance, pulmonary vascular remodeling, and increased pulmonary vascular pressures that often result in right ventricular dysfunction, leading to right heart failure. Evidence suggests that reactive oxygen species (ROS) contribute to PH pathogenesis by altering pulmonary vascular cell proliferation and intracellular signaling pathways. However, the role of mitochondrial antioxidants and oxidant-derived stress signaling in the development of hypoxia-induced PH is largely unknown. Therefore, we examined the role of the major mitochondrial redox regulator thioredoxin 2 (Trx2). Levels of Trx2 mRNA and protein were examined in human pulmonary arterial endothelial cells (HPAECs) and smooth muscle cells (HPASMCs) exposed to hypoxia, a common stimulus for PH, for 72 h. Hypoxia decreased Trx2 mRNA and protein levels. In vitro overexpression of Trx2 reduced hypoxia-induced H2O2 production. The effects of increased Trx2 protein level were examined in transgenic mice expressing human Trx2 (TghTrx2) that were exposed to hypoxia (10% O2) for 3 wk. TghTrx2 mice exposed to hypoxia had exacerbated increases in right ventricular systolic pressures, right ventricular hypertrophy, and increased ROS in the lung tissue. Trx2 overexpression did not attenuate hypoxia-induced increases in Trx2 oxidation or Nox4 expression. Expression of a dominant negative C93S Trx2 mutant that mimics Trx2 oxidation exacerbated hypoxia-induced increases in HPASMC H2O2 levels and cell proliferation. In conclusion, Trx2 overexpression failed to attenuate hypoxia-induced HPASMC proliferation in vitro or hypoxia-induced PH in vivo. These findings indicate that strategies to enhance Trx2 expression are unlikely to exert therapeutic effects in PH pathogenesis.


Asunto(s)
Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/metabolismo , Hipoxia/complicaciones , Hipoxia/metabolismo , Mitocondrias/metabolismo , Tiorredoxinas/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Hipertensión Pulmonar/patología , Hipoxia/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mutantes/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Oxidación-Reducción/efectos de los fármacos , Oxígeno/farmacología , Especies Reactivas de Oxígeno/metabolismo
11.
Am J Respir Cell Mol Biol ; 55(1): 35-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26677910

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) γ is critical for alveolar macrophage (AM) function. Chronic alcohol abuse causes AM phagocytic dysfunction and susceptibility to respiratory infections by stimulating nicotinamide adenine dinucleotide oxidases (Nox), transforming growth factor-ß1, and oxidative stress in the AM. Because PPARγ inhibits Nox expression, we hypothesized that alcohol reduces PPARγ, stimulating AM dysfunction. AMs were examined from: (1) patients with alcoholism or control patients; (2) a mouse model of chronic ethanol consumption; (3) PPARγ knockout mice; or (4) MH-S cells exposed to ethanol in vitro. Alcohol reduced AM PPARγ levels and increased Nox1, -2, and -4, transforming growth factor-ß1, oxidative stress, and phagocytic dysfunction. Genetic loss of PPARγ recapitulated, whereas stimulating PPARγ activity attenuated alcohol-mediated alterations in gene expression and phagocytic function, supporting the importance of PPARγ in alcohol-induced AM derangements. Similarly, PPARγ activation in vivo reduced alcohol-mediated impairments in lung bacterial clearance. Alcohol increased levels of microRNA-130a/-301a, which bind to the PPARγ 3' untranslated region to reduce PPARγ expression. MicroRNA-130a/-301a inhibition attenuated alcohol-mediated PPARγ reductions and derangements in AM gene expression and function. Alcohol-induced Toll-like receptor 4 endocytosis was reversed by PPARγ activation. These findings demonstrate that targeting PPARγ provides a novel therapeutic approach for mitigating alcohol-induced AM derangements and susceptibility to lung infection.


Asunto(s)
Etanol/efectos adversos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , PPAR gamma/metabolismo , Animales , Línea Celular , Humanos , Klebsiella/efectos de los fármacos , Ligandos , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fagocitos/efectos de los fármacos , Fagocitos/patología , Rosiglitazona , Tiazolidinedionas/farmacología
12.
Am J Respir Cell Mol Biol ; 54(1): 136-46, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26098770

RESUMEN

Endothelin-1 (ET-1) plays a critical role in endothelial dysfunction and contributes to the pathogenesis of pulmonary hypertension (PH). We hypothesized that peroxisome proliferator-activated receptor γ (PPARγ) stimulates microRNAs that inhibit ET-1 and pulmonary artery endothelial cell (PAEC) proliferation. The objective of this study was to clarify molecular mechanisms by which PPARγ regulates ET-1 expression in vitro and in vivo. In PAECs isolated from patients with pulmonary arterial hypertension, microRNA (miR)-98 expression was reduced, and ET-1 protein levels and proliferation were increased. Similarly, hypoxia reduced miR-98 and increased ET-1 levels and PAEC proliferation in vitro. In vivo, hypoxia reduced miR-98 expression and increased ET-1 and proliferating cell nuclear antigen (PCNA) levels in mouse lung, derangements that were aggravated by treatment with the vascular endothelial growth factor receptor antagonist Sugen5416. Reporter assays confirmed that miR-98 binds directly to the ET-1 3'-untranslated region. Compared with littermate control mice, miR-98 levels were reduced and ET-1 and PCNA expression were increased in lungs from endothelial-targeted PPARγ knockout mice, whereas miR-98 levels were increased and ET-1 and PCNA expression was reduced in lungs from endothelial-targeted PPARγ-overexpression mice. Gain or loss of PPARγ function in PAECs in vitro confirmed that alterations in PPARγ were sufficient to regulate miR-98, ET-1, and PCNA expression. Finally, PPARγ activation with rosiglitazone regimens that attenuated hypoxia-induced PH in vivo and human PAEC proliferation in vitro restored miR-98 levels. The results of this study show that PPARγ regulates miR-98 to modulate ET-1 expression and PAEC proliferation. These results further clarify molecular mechanisms by which PPARγ participates in PH pathogenesis and therapy.


Asunto(s)
Células Endoteliales/metabolismo , Endotelina-1/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , MicroARNs/metabolismo , PPAR gamma/metabolismo , Arteria Pulmonar/metabolismo , Transducción de Señal , Regiones no Traducidas 3' , Animales , Sitios de Unión , Proliferación Celular , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelina-1/genética , Regulación de la Expresión Génica , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipoxia/complicaciones , Hipoxia/genética , Hipoxia/patología , Indoles , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , PPAR gamma/agonistas , PPAR gamma/deficiencia , PPAR gamma/genética , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Pirroles , Interferencia de ARN , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Transfección , Remodelación Vascular
13.
Infect Immun ; 84(7): 1975-1985, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27091928

RESUMEN

The pathogenic profile of Pseudomonas aeruginosa is related to its ability to secrete a variety of virulence factors. Quorum sensing (QS) is a mechanism wherein small diffusible molecules, specifically acyl-homoserine lactones, are produced by P. aeruginosa to promote virulence. We show here that macrophage clearance of P. aeruginosa (PAO1) is enhanced by activation of the nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARγ). Macrophages treated with a PPARγ agonist (pioglitazone) showed enhanced phagocytosis and bacterial killing of PAO1. It is known that PAO1 QS molecules are inactivated by PON-2. QS molecules are also known to inhibit activation of PPARγ by competitively binding PPARγ receptors. In accord with this observation, we found that infection of macrophages with PAO1 inhibited expression of PPARγ and PON-2. Mechanistically, we show that PPARγ induces macrophage paraoxonase 2 (PON-2), an enzyme that degrades QS molecules produced by P. aeruginosa Gene silencing studies confirmed that enhanced clearance of PAO1 in macrophages by PPARγ is PON-2 dependent. Further, we show that PPARγ agonists also enhance clearance of P. aeruginosa from lungs of mice infected with PAO1. Together, these data demonstrate that P. aeruginosa impairs the ability of host cells to mount an immune response by inhibiting PPARγ through secretion of QS molecules. These studies define a novel mechanism by which PPARγ contributes to the host immunoprotective effects during bacterial infection and suggest a role for PPARγ immunotherapy for P. aeruginosa infections.


Asunto(s)
Interacciones Huésped-Patógeno , PPAR gamma/metabolismo , Pseudomonas aeruginosa/inmunología , Animales , Arildialquilfosfatasa/metabolismo , Línea Celular , Células Cultivadas , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Ligandos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Viabilidad Microbiana/inmunología , Modelos Biológicos , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/microbiología , PPAR gamma/agonistas , PPAR gamma/genética , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología
14.
Am J Physiol Lung Cell Mol Physiol ; 306(5): L429-41, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24441868

RESUMEN

Chronic alcohol abuse increases lung oxidative stress and susceptibility to respiratory infections by impairing alveolar macrophage (AM) function. NADPH oxidases (Nox) are major sources of reactive oxygen species in AMs. We hypothesized that treatment with the critical antioxidant glutathione (GSH) attenuates chronic alcohol-induced oxidative stress by downregulating Noxes and restores AM phagocytic function. Bronchoalveolar lavage (BAL) fluid and AMs were isolated from male C57BL/6J mice (8-10 wk) treated ± ethanol in drinking water (20% wt/vol, 12 wk) ± orally gavaged GSH in methylcellulose vehicle (300 mg x kg(-1) x day(-1), during week 12). MH-S cells, a mouse AM cell line, were treated ± ethanol (0.08%, 3 days) ± GSH (500 µM, 3 days or last 1 day of ethanol). BAL and AMs were also isolated from ethanol-fed and control mice ± inoculated airway Klebsiella pneumoniae (200 colony-forming units, 28 h) ± orally gavaged GSH (300 mg/kg, 24 h). GSH levels (HPLC), Nox mRNA (quantitative RT-PCR) and protein levels (Western blot and immunostaining), oxidative stress (2',7'-dichlorofluorescein-diacetate and Amplex Red), and phagocytosis (Staphylococcus aureus internalization) were measured. Chronic alcohol decreased GSH levels, increased Nox expression and activity, enhanced oxidative stress, impaired phagocytic function in AMs in vivo and in vitro, and exacerbated K. pneumonia-induced oxidative stress. Although how oral GSH restored GSH pools in ethanol-fed mice is unknown, oral GSH treatments abrogated the detrimental effects of chronic alcohol exposure and improved AM function. These studies provide GSH as a novel therapeutic approach for attenuating alcohol-induced derangements in AM Nox expression, oxidative stress, dysfunction, and risk for pneumonia.


Asunto(s)
Alcoholismo/inmunología , Antioxidantes/metabolismo , Glutatión/metabolismo , Macrófagos Alveolares/inmunología , NADH NADPH Oxidorreductasas/metabolismo , Alcoholismo/metabolismo , Animales , Antioxidantes/farmacología , Líquido del Lavado Bronquioalveolar/inmunología , Línea Celular , Depresores del Sistema Nervioso Central/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Etanol/farmacología , Glutatión/farmacología , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Fosfoproteínas/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 33(9): 2154-61, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23825363

RESUMEN

OBJECTIVE: On the basis of previous evidence that polymerase delta interacting protein 2 (Poldip2) increases reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (Nox4) activity in vascular smooth muscle cells, we hypothesized that in vivo knockdown of Poldip2 would inhibit reactive oxygen species production and alter vascular function. APPROACH AND RESULTS: Because homozygous Poldip2 deletion is lethal, Poldip2(+/-) mice were used. Poldip2 mRNA and protein levels were reduced by ≈50% in Poldip2(+/-) aorta, with no change in p22phox, Nox1, Nox2, and Nox4 mRNAs. NADPH oxidase activity was also inhibited in Poldip2(+/-) tissue. Isolated aortas from Poldip2(+/-) mice demonstrated impaired phenylephrine and potassium chloride-induced contractions, increased stiffness, and reduced compliance associated with disruption of elastic lamellae and excessive extracellular matrix deposition. Collagen I secretion was elevated in cultured vascular smooth muscle cells from Poldip2(+/-) mice and restored by H2O2 supplementation, suggesting that this novel function of Poldip2 is mediated by reactive oxygen species. Furthermore, Poldip2(+/-) mice were protected against aortic dilatation in a model of experimental aneurysm, an effect consistent with increased collagen secretion. CONCLUSIONS: Poldip2 knockdown reduces H2O2 production in vivo, leading to increases in extracellular matrix, greater vascular stiffness, and impaired agonist-mediated contraction. Thus, unaltered expression of Poldip2 is necessary for vascular integrity and function.


Asunto(s)
Aorta/metabolismo , Aneurisma de la Aorta/prevención & control , Proteínas Mitocondriales/metabolismo , Proteínas Nucleares/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/patología , Aorta/fisiopatología , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Aneurisma de la Aorta/fisiopatología , Presión Sanguínea , Células Cultivadas , Colágeno Tipo I/metabolismo , Grupo Citocromo b/metabolismo , Dilatación Patológica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Tejido Elástico/metabolismo , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Genotipo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Miocitos del Músculo Liso/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Oxidantes/farmacología , Fenotipo , ARN Mensajero/metabolismo , Rigidez Vascular , Vasoconstrictores/farmacología , Vasodilatación
16.
J Immunol ; 188(8): 3648-57, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22412195

RESUMEN

Chronic alcohol abuse is a comorbid variable of acute respiratory distress syndrome. Previous studies showed that, in the lung, chronic alcohol consumption increased oxidative stress and impaired alveolar macrophage (AM) function. NADPH oxidases (Noxes) are the main source of reactive oxygen species in AMs. Therefore, we hypothesized that chronic alcohol consumption increases AM oxidant stress through modulation of Nox1, Nox2, and Nox4 expression. AMs were isolated from male C57BL/6J mice, aged 8-10 wk, which were treated with or without ethanol in drinking water (20% w/v, 12 wk). MH-S cells, a mouse AM cell line, were treated with or without ethanol (0.08%, 3 d) for in vitro studies. Selected cells were treated with apocynin (300 µM), a Nox1 and Nox2 complex formation inhibitor, or were transfected with Nox small interfering RNAs (20-35 nM), before ethanol exposure. Human AMs were isolated from alcoholic and control patients' bronchoalveolar lavage fluid. Nox mRNA levels (quantitative RT-PCR), protein levels (Western blot and immunostaining), oxidative stress (2',7'-dichlorofluorescein-diacetate and Amplex Red analysis), and phagocytosis (Staphylococcus aureus internalization) were measured. Chronic alcohol increased Nox expression and oxidative stress in mouse AMs in vivo and in vitro. Experiments using apocynin and Nox small interfering RNAs demonstrated that ethanol-induced Nox4 expression, oxidative stress, and AM dysfunction were modulated through Nox1 and Nox2 upregulation. Further, Nox1, Nox2, and Nox4 protein levels were augmented in human AMs from alcoholic patients compared with control subjects. Ethanol induces AM oxidative stress initially through upregulation of Nox1 and Nox2 with downstream Nox4 upregulation and subsequent impairment of AM function.


Asunto(s)
Etanol/farmacología , Pulmón/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , NADPH Oxidasas/genética , Acetofenonas/farmacología , Adulto , Alcoholismo/enzimología , Alcoholismo/genética , Alcoholismo/inmunología , Animales , Líquido del Lavado Bronquioalveolar/citología , Línea Celular , Humanos , Isoenzimas/genética , Isoenzimas/inmunología , Pulmón/enzimología , Pulmón/inmunología , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/inmunología , Masculino , Ratones , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/inmunología , Estrés Oxidativo/efectos de los fármacos , Fagocitosis/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/inmunología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Regulación hacia Arriba
17.
J Immunol ; 189(11): 5411-20, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23105142

RESUMEN

Bacterial endotoxin (LPS)-mediated sepsis involves severe, dysregulated inflammation that injures the lungs and other organs, often fatally. Vascular endothelial cells are both key mediators and targets of LPS-induced inflammatory responses. The nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ) exerts anti-inflammatory actions in various cells, but it is unknown whether it modulates inflammation through actions within endothelial cells. To determine whether PPARγ acts within endothelial cells to diminish endotoxemic lung inflammation and injury, we measured inflammatory responses and mediators in mice with endothelial-targeted deletion of PPARγ. Endothelial cell PPARγ (ePPARγ) knockout exacerbated LPS-induced pulmonary inflammation and injury as shown by several measures, including infiltration of inflammatory cells, edema, and production of reactive oxygen species and proinflammatory cytokines, along with upregulation of the LPS receptor TLR4 in lung tissue and increased activation of its downstream signaling pathways. In isolated LPS-stimulated endothelial cells in vitro, absence of PPARγ enhanced the production of numerous inflammatory markers. We hypothesized that the observed in vivo activity of the ligand-activated ePPARγ may arise, in part, from nitrated fatty acids (NFAs), a novel class of endogenous PPARγ ligands. Supporting this idea, we found that treating isolated endothelial cells with physiologically relevant concentrations of the endogenous NFA 10-nitro-oleate reduced LPS-induced expression of a wide range of inflammatory markers in the presence of PPARγ, but not in its absence, and also inhibited neutrophil mobility in a PPARγ-dependent manner. Our results demonstrate a key protective role of ePPARγ against endotoxemic injury and a potential ePPARγ-mediated anti-inflammatory role for NFAs.


Asunto(s)
Endotoxemia/inmunología , Ácidos Grasos/farmacología , Nitrocompuestos/farmacología , PPAR gamma/inmunología , Neumonía/inmunología , Animales , Quimiocina CXCL2/biosíntesis , Quimiocina CXCL2/inmunología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Endotoxemia/inducido químicamente , Endotoxemia/complicaciones , Endotoxemia/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intraperitoneales , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Lipopolisacáridos/administración & dosificación , Pulmón , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/deficiencia , PPAR gamma/genética , Neumonía/inducido químicamente , Neumonía/complicaciones , Neumonía/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
18.
Health Equity ; 7(1): 333-337, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37284533

RESUMEN

The Department of Veterans Affairs (VA) initiative to enhance recruitment of diverse biomedical scientists from Historically Black Colleges and Universities (HBCUs) through the VA Career Development Program has provided a unique opportunity for HBCUs to partner with VA to strengthen diversity recruitment efforts. The Atlanta VA Health Care System and the Morehouse School of Medicine (MSM) enjoy a productive and growing interinstitutional collaboration. The partnership between the Atlanta VA and MSM provides the unique opportunity for MSM to increase research opportunities for faculty and students while providing a pipeline of diverse candidates for the Atlanta VA to enhance recruitment of diverse HCBU biomedical scientists. This relationship led to the creation of an inaugural HBCU Core Recruitment Site (CRS) at MSM and the Atlanta VA. The CRS provides a pathway to identify and recruit young diverse investigators who are eligible to compete for VA Career Development Award funding. This Atlanta VA/MSM CRS initiative established a pipeline program to further enhance diversity in the VA scientific workforce. In this review, the Atlanta VA/MSM CRS is presented as a potential model for maximizing the VA initiative to enhance the recruitment of diverse candidates from HBCUs.

19.
Alcohol ; 106: 30-43, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36328183

RESUMEN

Alcohol use disorders (AUD) cause alveolar macrophage (AM) immune dysfunction and increase risk of lung infections. Excessive alcohol use causes AM oxidative stress, which impairs AM phagocytosis and pathogen clearance from the alveolar space. Alcohol induces expression of NADPH oxidases (Noxes), primary sources of oxidative stress in AM. In contrast, alcohol decreases AM peroxisome proliferator-activated receptor gamma (PPARγ), a critical regulator of AM immune function. To explore the underlying molecular mechanisms for these effects of alcohol, we hypothesized that ethanol promotes CCAAT/enhancer-binding protein beta (C/EBPß)-mediated suppression of Nox-related microRNAs (miRs), in turn enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. We also hypothesized that PPARγ activation with pioglitazone (PIO) would reverse alcohol-induced C/EBPß expression and attenuate AM oxidative stress and phagocytic dysfunction. Cells from the mouse AM cell line (MH-S) were exposed to ethanol in vitro or primary AM were isolated from mice fed ethanol in vivo. Ethanol enhanced C/EBPß expression, decreased Nox 1-related miR-1264 and Nox 2-related miR-107 levels, and increased Nox1, Nox2, and Nox 4 expression in MH-S cells in vitro and mouse AM in vivo. These alcohol-induced AM derangements were abrogated by loss of C/EBPß, overexpression of miRs-1264 or -107, or PIO treatment. These findings identify C/EBPß and Nox-related miRs as novel therapeutic targets for PPARγ ligands, which could provide a translatable strategy to mitigate susceptibility to lung infections in people with a history of AUD. These studies further clarify the molecular underpinnings for a previous clinical trial using short-term PIO treatment to improve AM immunity in AUD individuals.


Asunto(s)
Etanol , Macrófagos Alveolares , MicroARNs , Procesamiento Postranscripcional del ARN , Animales , Ratones , Alcoholismo/tratamiento farmacológico , Alcoholismo/genética , Etanol/efectos adversos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/patología , MicroARNs/genética , MicroARNs/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo
20.
Am J Respir Cell Mol Biol ; 47(5): 718-26, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22904198

RESUMEN

Increased NADP reduced (NADPH) oxidase 4 (Nox4) and reduced expression of the nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ) contribute to hypoxia-induced pulmonary hypertension (PH). To examine the role of Nox4 activity in pulmonary vascular cell proliferation and PH, the current study used a novel Nox4 inhibitor, GKT137831, in hypoxia-exposed human pulmonary artery endothelial or smooth muscle cells (HPAECs or HPASMCs) in vitro and in hypoxia-treated mice in vivo. HPAECs or HPASMCs were exposed to normoxia or hypoxia (1% O(2)) for 72 hours with or without GKT137831. Cell proliferation and Nox4, PPARγ, and transforming growth factor (TGF)ß1 expression were measured. C57Bl/6 mice were exposed to normoxia or hypoxia (10% O(2)) for 3 weeks with or without GKT137831 treatment during the final 10 days of exposure. Lung PPARγ and TGF-ß1 expression, right ventricular hypertrophy (RVH), right ventricular systolic pressure (RVSP), and pulmonary vascular remodeling were measured. GKT137831 attenuated hypoxia-induced H(2)O(2) release, proliferation, and TGF-ß1 expression and blunted reductions in PPARγ in HPAECs and HPASMCs in vitro. In vivo GKT137831 inhibited hypoxia-induced increases in TGF-ß1 and reductions in PPARγ expression and attenuated RVH and pulmonary artery wall thickness but not increases in RVSP or muscularization of small arterioles. This study shows that Nox4 plays a critical role in modulating proliferative responses of pulmonary vascular wall cells. Targeting Nox4 with GKT137831 provides a novel strategy to attenuate hypoxia-induced alterations in pulmonary vascular wall cells that contribute to vascular remodeling and RVH, key features involved in PH pathogenesis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NADPH Oxidasas/antagonistas & inhibidores , Arteria Pulmonar/patología , Pirazoles/farmacología , Piridinas/farmacología , Animales , Hipoxia de la Célula , Células Cultivadas , Células Endoteliales/enzimología , Células Endoteliales/fisiología , Endotelio Vascular/patología , Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Peróxido de Hidrógeno/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/fisiología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Pirazoles/uso terapéutico , Pirazolonas , Piridinas/uso terapéutico , Piridonas , Interferencia de ARN , Rosiglitazona , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Remodelación Ventricular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA