Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Am J Respir Crit Care Med ; 206(4): 459-475, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35687485

RESUMEN

Rationale: A prevailing paradigm recognizes idiopathic pulmonary fibrosis (IPF) originating from various alveolar epithelial cell (AEC) injuries, and there is a growing appreciation of AEC aging as a key driver of the pathogenesis. Despite this progress, it is incompletely understood what main factor(s) contribute to the worsened alveolar epithelial aging in lung fibrosis. It remains a challenge how to dampen AEC aging and thereby mitigate the disease progression. Objectives: To determine the role of AEC CD38 (cluster of differentiation 38) in promoting cellular aging and lung fibrosis. Methods: We used single-cell RNA sequencing, real-time PCR, flow cytometry, and Western blotting. Measurements and Main Results: We discovered a pivotal role of CD38, a cardinal nicotinamide adenine dinucleotide (NAD) hydrolase, in AEC aging and its promotion of lung fibrosis. We found increased CD38 expression in IPF lungs that inversely correlated with the lung functions of patients. CD38 was primarily located in the AECs of human lung parenchyma and was markedly induced in IPF AECs. Similarly, CD38 expression was elevated in the AECs of fibrotic lungs of young mice and further augmented in those of old mice, which was in accordance with a worsened AEC aging phenotype and an aggravated lung fibrosis in the old animals. Mechanistically, we found that CD38 elevation downregulated intracellular NAD, which likely led to the aging promoting impairment of the NAD-dependent cellular and molecular activities. Furthermore, we demonstrated that genetic and pharmacological inactivation of CD38 improved these NAD dependent events and ameliorated bleomycin-induced lung fibrosis. Conclusions: Our study suggests targeting alveolar CD38 as a novel and effective therapeutic strategy to treat this pathology.


Asunto(s)
Células Epiteliales Alveolares , Fibrosis Pulmonar Idiopática , Envejecimiento , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina , Senescencia Celular/genética , Humanos , Fibrosis Pulmonar Idiopática/genética , Pulmón/patología , Ratones , NAD/metabolismo
2.
Am J Respir Cell Mol Biol ; 64(1): 115-125, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33074715

RESUMEN

Augmented glycolysis due to metabolic reprogramming in lung myofibroblasts is critical to their profibrotic phenotype. The primary glycolysis byproduct, lactate, is also secreted into the extracellular milieu, together with which myofibroblasts and macrophages form a spatially restricted site usually described as fibrotic niche. Therefore, we hypothesized that myofibroblast glycolysis might have a non-cell autonomous effect through lactate regulating the pathogenic phenotype of alveolar macrophages. Here, we demonstrated that there was a markedly increased lactate in the conditioned media of TGF-ß1 (transforming growth factor-ß1)-induced lung myofibroblasts and in the BAL fluids (BALFs) from mice with TGF-ß1- or bleomycin-induced lung fibrosis. Importantly, the media and BALFs promoted profibrotic mediator expression in macrophages. Mechanistically, lactate induced histone lactylation in the promoters of the profibrotic genes in macrophages, consistent with the upregulation of this epigenetic modification in these cells in the fibrotic lungs. The lactate inductions of the histone lactylation and profibrotic gene expression were mediated by p300, as evidenced by their diminished concentrations in p300-knockdown macrophages. Collectively, our study establishes that in addition to protein, lipid, and nucleic acid molecules, a metabolite can also mediate intercellular regulations in the setting of lung fibrosis. Our findings shed new light on the mechanism underlying the key contribution of myofibroblast glycolysis to the pathogenesis of lung fibrosis.


Asunto(s)
Histonas/metabolismo , Lactatos/metabolismo , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Miofibroblastos/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Células Cultivadas , Humanos , Indoles/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Piridonas/farmacología
3.
Eur Respir J ; 58(6)2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34049949

RESUMEN

AIM: We investigated the mechanisms by which N1-(ß-d-ribofuranosyl)-5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase (AMPK), decreases lung injury and mortality when administered to mice post exposure to bromine gas (Br2). METHODS: We exposed male C57BL/6 mice and heme oxygenase-1 (HO-1)-deficient (HO-1-/-) and corresponding wild-type (WT) littermate mice to Br2 (600 ppm for 45 or 30 min, respectively) in environmental chambers and returned them to room air. AICAR was administered 6 h post exposure (10 mg·kg-1, intraperitoneal). We assessed survival, indices of lung injury, high mobility group box 1 (HMGB1) in the plasma, HO-1 levels in lung tissues and phosphorylation of AMPK and its upstream liver kinase B1 (LKB1). Rat alveolar type II epithelial (L2) cells and human club-like epithelial (H441) cells were also exposed to Br2 (100 ppm for 10 min). After 24 h we measured apoptosis and necrosis, AMPK and LKB1 phosphorylation, and HO-1 expression. RESULTS: There was a marked downregulation of phosphorylated AMPK and LKB1 in lung tissues and in L2 and H441 cells post exposure. AICAR increased survival in C57BL/6 but not in HO-1-/- mice. In WT mice, AICAR decreased lung injury and restored phosphorylated AMPK and phosphorylated LKB1 to control levels and increased HO-1 levels in both lung tissues and cells exposed to Br2. Treatment of L2 and H441 cells with small interfering RNAs against nuclear factor erythroid 2-related factor 2 or HO-1 abrogated the protective effects of AICAR. CONCLUSIONS: Our data indicate that the primary mechanism for the protective action of AICAR in toxic gas injury is the upregulation of lung HO-1 levels.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Lesión Pulmonar Aguda , Lesión Pulmonar Aguda/inducido químicamente , Aminoimidazol Carboxamida/análogos & derivados , Animales , Hemo-Oxigenasa 1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ribonucleótidos/farmacología
4.
Am J Respir Cell Mol Biol ; 63(4): 478-489, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32551949

RESUMEN

Although endoplasmic reticulum (ER) unfolded protein response (UPRER) is well known, mitochondrial unfolded protein response (UPRmt) has not been recognized in alveolar epithelial cells. Furthermore, ER stress and mitochondrial dysfunction are frequently encountered in alveolar epithelial cells from an array of lung disorders. However, these two scenarios have been often regarded as separate mechanisms contributing to the pathogeneses. It is unclear whether there is interplay between these two phenomena or an integrator that couples these two signaling cascades in the stressed alveolar epithelial cells from those pathologies. In this study, we defined UPRmt in alveolar epithelial cells and identified ATF4 (activating transcription factor 4), but not ATF5, as the key regulator of UPRmt. We found that UPRER led to UPRmt and mitochondrial dysfunction in an ATF4-dependent manner. In contrast, mitochondrial stresses did not activate UPRER. We found that alveolar epithelial ATF4 and UPRmt were induced in aged mice with experimental pulmonary fibrosis as well as in patients with idiopathic pulmonary fibrosis. Finally, we found that the inducible expression of ATF4 in mouse alveolar epithelial cells aggravated pulmonary UPRmt, lung inflammation, body weight loss, and death upon bleomycin-induced lung injury. In conclusion, ER stress induces ATF4-dependent UPRmt and mitochondrial dysfunction, indicating a novel mechanism by which ER stress contributes to the pathogeneses of a variety of pulmonary disorders.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Células Epiteliales Alveolares/metabolismo , Mitocondrias/metabolismo , Respuesta de Proteína Desplegada/fisiología , Células Epiteliales Alveolares/fisiología , Animales , Apoptosis/fisiología , Línea Celular , Estrés del Retículo Endoplásmico/fisiología , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/fisiopatología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Transducción de Señal/fisiología
5.
J Immunol ; 200(9): 3218-3230, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29563175

RESUMEN

Rapid initiation and timely resolution of inflammatory response in macrophages are synergistic events that are known to be equally critical to optimal host defense against pathogen infections. However, the regulation of these processes, in particular by a specific cellular metabolic program, has not been well understood. In this study, we found that IFN regulatory factor 2 (IRF2) underwent an early degradation in a proteasome-mediated pathway in LPS-treated mouse macrophages, followed by a later recovery of the expression via transactivation. We showed that IRF2 was anti-inflammatory in that knockdown of this protein promoted the production of LPS-induced proinflammatory mediators. Mechanistically, although IRF2 apparently did not target the proximal cytoplasmic signaling events upon LPS engagements, it inhibited HIF-1α-dependent expression of glycolytic genes and thereby cellular glycolysis, sequential events necessary for the IRF2 anti-inflammatory activity. We found that macrophages in endotoxin tolerant state demonstrated deficiency in LPS-augmented glycolysis, which was likely caused by failed downregulation of IRF2 and the ensuing upregulation of the glycolytic genes in these cells. In contrast to observations with LPS, knockdown of IRF2 decreased IL-4-induced macrophage alternative activation. The pro-IL-4 activity of IRF2 was dependent on KLF4, a key mediator of the alternative activation, which was transcriptionally induced by IRF2. In conclusion, our data suggest that IRF2 is an important regulator of the proinflammatory response in macrophages by controlling HIF-1α-dependent glycolytic gene expression and glycolysis. This study also indicates IRF2 as a novel therapeutic target to treat inflammatory disorders associated with dysregulations of macrophage activations.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Glucólisis/fisiología , Inflamación/inmunología , Factor 2 Regulador del Interferón/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Factor 4 Similar a Kruppel , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL
6.
Am J Respir Cell Mol Biol ; 60(2): 167-178, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30183330

RESUMEN

Profound impairment in cellular oxygen consumption, referred to as cytopathic dysoxia, is one of the pathological hallmarks in the lungs of patients with pathogen-induced acute lung injury (ALI). However, the underlying mechanism for this functional defect remains largely unexplored. In this study, we found that primary mouse alveolar epithelial cells (AECs) conducted robust fatty acid oxidation (FAO). More importantly, FAO was strikingly impaired in AECs of mice with LPS-induced ALI. The metabolic deficiency in these cells was likely due to decreased expression of key mediators involved in FAO and mitochondrial bioenergenesis, such as peroxisome proliferator-activated receptor γ coactivator (PGC)-1α, carnitine palmitoyltransferase 1A, and medium-chain acyl-CoA dehydrogenase (CAD). We found that treatment of alveolar epithelial line MLE-12 cells with BAL fluids from mice with ALI decreased FAO, and this effect was largely replicated in MLE-12 cells treated with the proinflammatory cytokine TNF-α, which was consistent with downregulations of PGC-1α, carnitine palmitoyltransferase 1A, long-chain CAD, and medium-chain CAD in the same treated cells. Furthermore, we found that the BAL fluids from ALI mice and TNF-α inhibited MLE-12 bioenergenesis and promoted cell apoptosis. In delineation of the role of FAO in ALI in vivo, we found that conditional ablation of AEC PGC-1α aggravated LPS-induced ALI. In contrast, fenofibrate, an activator of the PPAR-α/PGC-1α cascade, protected mice from this pathology. In summary, these data suggest that FAO is essential to AEC bioenergenesis and functional homeostasis. This study also indicates that FAO impairment-induced AEC dysfunction is an important contributing factor to the pathogenesis of ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Células Epiteliales Alveolares/metabolismo , Ácidos Grasos/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Líquido del Lavado Bronquioalveolar , Línea Celular , Metabolismo Energético/genética , Fenofibrato/farmacología , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Lipopolisacáridos/toxicidad , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Oxidación-Reducción , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
7.
Am J Respir Cell Mol Biol ; 61(4): 492-500, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30943369

RESUMEN

It has been increasingly recognized lately that aberrant cellular metabolism plays an important role in the pathogenesis of pulmonary fibrosis. In our previous systemic studies, we found that human lung myofibroblasts undergo glutaminolytic reprogramming, which is mediated by an increased expression of glutaminase (Gls) 1. We showed that augmented glutaminolysis critically regulates collagen production by promoting its stabilization in human lung myofibroblasts. Our study indicates that lung fibroblast Gls1 is a promising therapeutic target for this disease. In this investigation, we primarily focused on delineating the in vivo role of fibroblast Gls1 in mouse models of pulmonary fibrosis and determining the efficacy of Gls1 inhibition in treating this pathology. We now show that fibroblast Gls1 is upregulated in fibrotic mouse lungs. We present evidence that mice with ablation of fibroblast Gls1 are protected from bleomycin-induced lung fibrosis. We show that the Gls1 inhibitor, CB-839, is therapeutically efficacious in treating both bleomycin- and transforming growth factor-ß1-induced pulmonary fibrosis. Our study has thus established a solid rationale for advancing Gls1 inhibitors, particularly CB-839, to the next stage of testing in the treatment of this disease.


Asunto(s)
Fibroblastos/enzimología , Glutaminasa/antagonistas & inhibidores , Fibrosis Pulmonar/tratamiento farmacológico , Animales , Bencenoacetamidas/farmacología , Bleomicina/toxicidad , Línea Celular , Colágeno/biosíntesis , Inducción Enzimática , Glutaminasa/genética , Pulmón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/enzimología , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Tiadiazoles/farmacología , Factor de Crecimiento Transformador beta1/farmacología
8.
Am J Respir Cell Mol Biol ; 58(3): 378-390, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29019707

RESUMEN

Glutaminolysis is the metabolic process of glutamine, aberration of which has been implicated in several pathogeneses. Although we and others recently found a diversity of metabolic dysregulation in organ fibrosis, it is unknown if glutaminolysis regulates the profibrotic activities of myofibroblasts, the primary effector in this pathology. In this study, we found that lung myofibroblasts demonstrated significantly augmented glutaminolysis that was mediated by elevated glutaminase 1 (Gls1). Inhibition of glutaminolysis by specific Gls1 inhibitors CB-839 and BPTES as well as Gls1 siRNA blunted the expression of collagens but not that of fibronectin, elastin, or myofibroblastic marker smooth muscle actin-α. We found that glutaminolysis enhanced collagen translation and stability, which were mediated by glutaminolysis-dependent mTOR complex 1 activation and collagen proline hydroxylation, respectively. Furthermore, we found that the amount of the glutaminolytic end product α-ketoglutarate (α-KG) was increased in myofibroblasts. Similar to glutaminolysis, α-KG activated mTOR complex 1 and promoted the expression of collagens but not of fibronectin, elastin, or smooth muscle actin-α. α-KG also remarkably inhibited collagen degradation in fibroblasts. Taken together, our studies identified a previously unrecognized mechanism by which a major metabolic program regulates the exuberant production of collagens in myofibroblasts and suggest that glutaminolysis is a novel therapeutic target for treating organ fibrosis, including idiopathic pulmonary fibrosis.


Asunto(s)
Glutamina/metabolismo , Ácidos Cetoglutáricos/metabolismo , Miofibroblastos/metabolismo , Prolina/química , Fibrosis Pulmonar/patología , Serina-Treonina Quinasas TOR/metabolismo , Actinas/biosíntesis , Animales , Bencenoacetamidas/farmacología , Células Cultivadas , Colágeno/biosíntesis , Modelos Animales de Enfermedad , Elastina/biosíntesis , Activación Enzimática/fisiología , Fibronectinas/biosíntesis , Glutaminasa/antagonistas & inhibidores , Glutaminasa/genética , Glutaminasa/metabolismo , Humanos , Hidroxilación , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño/genética , Sulfuros/farmacología , Tiadiazoles/farmacología
9.
Am J Respir Cell Mol Biol ; 66(2): 117-119, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34758277
10.
Am J Respir Cell Mol Biol ; 56(2): 168-178, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27635790

RESUMEN

Cellular senescence has been implicated in diverse pathologies. However, there is conflicting evidence regarding the role of this process in tissue fibrosis. Although dysregulation of microRNAs is a key mechanism in the pathogenesis of lung fibrosis, it is unclear whether microRNAs function by regulating cellular senescence in the disease. In this study, we found that miR-34a demonstrated greater expression in the lungs of patients with idiopathic pulmonary fibrosis and in mice with experimental pulmonary fibrosis, with its primary localization in lung fibroblasts. More importantly, miR-34a was up-regulated significantly in both human and mouse lung myofibroblasts. We found that mice with miR-34a ablation developed more severe pulmonary fibrosis than did wild-type animals after fibrotic lung injury. Mechanistically, we found that miR-34a induced a senescent phenotype in lung fibroblasts because this microRNA increased senescence-associated ß-galactosidase activity, enhanced expression of senescence markers, and decreased cell proliferative capacities. Consistently, we found that primary lung fibroblasts from fibrotic lungs of miR-34a-deficient mice had a diminished senescent phenotype and enhanced resistance to apoptosis as compared with those from wild-type animals. We also identified multiple miR-34a targets that likely mediated its activities in inducing senescence in lung fibroblasts. In conclusion, our data suggest that miR-34a functions through a negative feedback mechanism to restrain fibrotic response in the lungs by promoting senescence of pulmonary fibroblasts.


Asunto(s)
Senescencia Celular , Fibroblastos/patología , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , MicroARNs/metabolismo , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos C57BL , MicroARNs/genética , Miofibroblastos/metabolismo , Miofibroblastos/patología , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
11.
Am J Physiol Lung Cell Mol Physiol ; 313(5): L834-L844, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28798256

RESUMEN

Metabolic reprogramming has been intrinsically linked to macrophage activation. Alveolar macrophages are known to play an important role in the pathogenesis of pulmonary fibrosis. However, systematic characterization of expression profile in these cells is still lacking. Furthermore, main metabolic programs and their regulation of cellular phenotype are completely unknown. In this study, we comprehensively analyzed the expression profile and main metabolic programs in alveolar macrophages from mice with or without experimental pulmonary fibrosis. We found that alveolar macrophages from both bleomycin and active TGF-ß1-induced fibrotic mouse lungs demonstrated a primarily profibrotic M2-like profile that was distinct from the well-defined M1 or any of the M2 subtypes. More importantly, we found that fibrotic lung alveolar macrophages assumed augmented glycolysis, which was likely attributed to enhanced expression of multiple key glycolytic mediators. We also found that fatty acid oxidation was upregulated in these cells. However, the profibrotic M2-like profile of fibrotic lung alveolar macrophages was not dependent on fatty acid oxidation and synthesis or lipolysis, but instead on glycolysis, in contrast to the typical IL-4-induced macrophages M(IL-4). Additionally, glutaminolysis, a key metabolic program that has been implicated in numerous pathologies, was not required for the profibrotic M2-like phenotype of these macrophages. In summary, our study identifies a unique expression and metabolic profile in alveolar macrophages from fibrotic lungs and suggests glycolytic inhibition as an effective antifibrotic strategy in treating lung fibrosis.


Asunto(s)
Macrófagos Alveolares/metabolismo , Fibrosis Pulmonar/metabolismo , ARN/metabolismo , Animales , Bleomicina/farmacología , Glucólisis/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Ratones Endogámicos C57BL , Fenotipo , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología
12.
Am J Physiol Lung Cell Mol Physiol ; 312(3): L415-L424, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27979858

RESUMEN

Idiopathic pulmonary fibrosis is a well-known age-related disease. However, much less recognized has been the aging associated pathogenesis of this disorder. As we and others previously showed that dysregulation of micro-RNAs (miRNAs) was an important mechanism involved in pulmonary fibrosis, the role of these molecules in this pathology in the aged population has not been investigated (Cushing L, Kuang PP, Qian J, Shao F, Wu J, Little F, Thannickal VJ, Cardoso WV, Lü J. Am J Respir Cell Mol Biol 45: 287-294, 2011; Liu G, Friggeri A, Yang Y, Milosevic J, Ding Q, Thannickal VJ, Kaminski N, Abraham E. J Exp Med 207: 1589-1597, 2010; Pandit KV, Corcoran D, Yousef H, Yarlagadda M, Tzouvelekis A, Gibson KF, Konishi K, Yousem SA, Singh M, Handley D, Richards T, Selman M, Watkins SC, Pardo A, Ben-Yehudah A, Bouros D, Eickelberg O, Ray P, Benos PV, Kaminski N. Am J Respir Crit Care Med 182: 220-229, 2010). In this study, by using a lung fibrosis model established in old mice, we found that ablation of miR-34a protected aged animals from developing experimental lung fibrosis. miR-34a was upregulated in lung epithelial cells, but not in lung fibroblasts of aged mice, and miR-34a expression was further increased in epithelial cells of the fibrotic lungs of these old animals. We found that miR-34a induced dysfunctions in alveolar epithelial cells (AECs), as evidenced by increased cellular senescence and apoptosis and mitochondrial aberrations. More importantly, these abnormalities were attenuated in AECs of the fibrotic lungs of aged miR-34a-/- mice. We found that miR-34a targeted Sirt1, a master anti-aging regulator, and two key cell cycle modulators, E2F3 and cyclin E2, in lung epithelial cells, and the repression of these targets was relieved in miR-34a-deficient AECs. In summary, our data suggest that elevated AEC miR-34a plays a critical role in the pathogenesis of pulmonary fibrosis in the aged population. Our study also indicates miR-34a to be a more precise miRNA target for treating this disease that overwhelmingly affects people of advanced age.


Asunto(s)
Envejecimiento/genética , Envejecimiento/patología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , MicroARNs/metabolismo , Animales , Apoptosis/genética , Bleomicina , Línea Celular , Proliferación Celular , Senescencia Celular , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/prevención & control , Ratones , MicroARNs/genética , Mitocondrias/metabolismo , Fenotipo , Regulación hacia Arriba/genética
13.
J Gen Virol ; 98(6): 1489-1499, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28590239

RESUMEN

miR-146a was reported to participate in various pathophysiological conditions in mammals, such as inflammation and immune responses, oncogenesis and cell damage. However, its function in low vertebrates has not been well elucidated. In this study, we characterized the expression profiles and functions of miR-146a in fish cells during iridovirus infection. We found that the reported fish miR-146a genes encoded an identical mature sequence, which shared high similarity with its mammalian orthologues, suggesting a putative functional conservation of miR-146a between fish and other vertebrates. Using a well-established infection model of Singapore grouper iridovirus (SGIV) in fathead minnow cells, we found that SGIV infection induced the expression of miR-146a to a dramatic extent. More importantly, we found that miR-146a promoted SGIV propagation, as demonstrated by higher expression of viral genes and increased virus titres in miR-146a-overexpressing cells. Mechanistically, we found that miR-146a overexpression suppressed, while miR-146a knockdown promoted, NF-κB activation and SGIV-induced cell apoptosis, two major cellular events involved in SGIV infection. Our study suggested that the induction of miR-146a by SGIV infection may function through a feed-forward mechanism to promote viral infection by restraining anti-viral cellular responses.


Asunto(s)
Apoptosis , Cyprinidae/virología , Interacciones Huésped-Patógeno , MicroARNs/metabolismo , FN-kappa B/metabolismo , Ranavirus/crecimiento & desarrollo , Animales , Células Cultivadas , Carga Viral
14.
J Immunol ; 194(12): 6082-9, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25964487

RESUMEN

The M1 and M2 polarized phenotypes dictate distinctive roles for macrophages as they participate in inflammatory disorders. There has been growing interest in the role of cellular metabolism in macrophage polarization. However, it is currently unclear whether different aspects of a specific metabolic program coordinately regulate this cellular process. In this study, we found that pyruvate dehydrogenase kinase 1 (PDK1), a key regulatory enzyme in glucose metabolism, plays an important role in the differential activation of macrophages. Knockdown of PDK1 diminished M1, whereas it enhanced M2 activation of macrophages. Mechanistically, PDK1 knockdown led to diminished aerobic glycolysis in M1 macrophages, which likely accounts for the attenuated inflammatory response in these cells. Furthermore, we found that mitochondrial respiration is enhanced during and required by the early activation of M2 macrophages. Suppression of glucose oxidation, but not that of fatty acids, inhibits this process. Consistent with its inhibitory role in early M2 activation, knockdown of PDK1 enhanced mitochondrial respiration in macrophages. Our data suggest that two arms of the glucose metabolism synergistically regulate the differential activation of macrophages. Our findings also highlight the central role of PDK1 in this event via controlling glycolysis and glucose oxidation.


Asunto(s)
Glucosa/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Bacterias/inmunología , Diferenciación Celular/inmunología , Ácidos Grasos , Técnicas de Silenciamiento del Gen , Glucólisis , Lipopolisacáridos/inmunología , Activación de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/microbiología , Ratones , Mitocondrias/metabolismo , Oxidación-Reducción , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Transducción de Señal , Receptor Toll-Like 2/metabolismo
15.
Am J Respir Cell Mol Biol ; 54(6): 843-52, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26600197

RESUMEN

Although microRNAs (miRs) have been well recognized to play an important role in the pathogenesis of organ fibrosis, there is a lack of evidence as to whether miRs directly regulate the differentiation of myofibroblasts, the putative effector cells during pathological fibrogenesis. In this study, we found that levels of miR-27a-3p were up-regulated in transforming growth factor-ß1-treated human lung fibroblasts in a Smad2/3-dependent manner and in fibroblasts isolated from lungs of mice with experimental pulmonary fibrosis. However, both basal and transforming growth factor-ß1-induced expression of miR-27a-3p were reduced in lung fibroblasts from patients with idiopathic pulmonary fibrosis compared with that from normal control subjects. Overexpression of miR-27a-3p inhibited, whereas knockdown of miR-27a-3p enhanced, the differentiation of lung fibroblasts into myofibroblasts. We found that miR-27a-3p directly targeted the phenotypic marker of myofibroblasts, α-smooth muscle actin, and two key Smad transcription factors, Smad2 and Smad4. More importantly, we found that therapeutic expression of miR-27a-3p in mouse lungs through lentiviral delivery diminished bleomycin-induced lung fibrosis. In conclusion, our data suggest that miR-27a-3p functions via a negative-feedback mechanism in inhibiting lung fibrosis. This study also indicates that targeting miR-27a-3p is a novel therapeutic approach to treat fibrotic organ disorders, including lung fibrosis.


Asunto(s)
Diferenciación Celular , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , MicroARNs/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Actinas/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Células HEK293 , Humanos , Lentivirus/metabolismo , Ratones Endogámicos C57BL , MicroARNs/genética , Miofibroblastos/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
16.
J Biol Chem ; 290(1): 46-55, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25406319

RESUMEN

There has been fast growing evidence showing that glycolysis plays a critical role in the activation of immune cells. Enhanced glycolysis leads to increased formation of intracellular lactate that is exported to the extracellular environment by monocarboxylate transporter 4 (MCT4). Although the biological activities of extracellular lactate have been well studied, it is less understood how the lactate export is regulated or whether lactate export affects glycolysis during inflammatory activation. In this study, we found that MCT4 is up-regulated by TLR2 and TLR4, but not TLR3 agonists in a variety of macrophages. The increased expression of MCT4 was mediated by MYD88 in a NF-κB-dependent manner. Furthermore, we found that MCT4 is required for macrophage activation upon TLR2 and TLR4 stimulations, as evidenced by attenuated expression of proinflammatory mediators in macrophages with MCT4 knockdown. Mechanistically, we found that MCT4 knockdown leads to enhanced intracellular accumulation of lactate and decreased glycolysis in LPS-treated macrophages. We found that LPS-induced expression of key glycolytic enzymes hexokinase 2 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 is diminished in macrophages with MCT4 knockdown. Our data suggest that MCT4 up-regulation represents a positive feedback mechanism in macrophages to maintain a high glycolytic rate that is essential to a fully activated inflammatory response.


Asunto(s)
Glucólisis/genética , Macrófagos Alveolares/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Proteínas Musculares/genética , Animales , Transporte Biológico , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Glucólisis/inmunología , Hexoquinasa/genética , Hexoquinasa/inmunología , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ácido Láctico/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Transportadores de Ácidos Monocarboxílicos/inmunología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/inmunología , Proteínas Musculares/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/inmunología , Cultivo Primario de Células , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
17.
J Immunol ; 193(1): 327-334, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24835395

RESUMEN

Although microRNAs were shown to participate in innate immune responses, it is not completely understood how they regulate negative immunomodulatory events. IL-10 is an important anti-inflammatory mediator that prevents excessive inflammation and associated immunological pathologies. Although the regulation of IL-10 expression has been well studied at both the transcriptional and translational levels, it is less clear how microRNAs control IL-10 expression during inflammation. In this study, we found that miR-27a is downregulated in macrophages following stimulation through TLR2 and TLR4, but not TLR3. Upregulation of miR-27a enhanced the expression of proinflammatory cytokines in TLR2/4-activated macrophages. Conversely, knockdown of miR-27a diminished cytokine expression. Mechanistically, we found that miR-27a negatively regulates IL-10 expression; upregulation of miR-27a decreases, whereas downregulation of miR-27a increases, IL-10 expression in activated macrophages. Likely due to the decreased expression of IL-10, upregulation of miR-27a diminished IL-10-dependent STAT3 phosphorylation in TLR4-activated macrophages. Consistent with IL-10 being a potential mediator for the role of miR-27a in the immune response, blocking IL-10 abolished the enhancing effect of miR-27a on TLR4-activated inflammation. In conclusion, our study identified miR-27a downregulation as a negative-regulatory mechanism that prevents overly exuberant TLR2- and TLR4-driven inflammatory responses.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interleucina-10/inmunología , Macrófagos Peritoneales/inmunología , MicroARNs/inmunología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Lipopolisacáridos/toxicidad , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/patología , Ratones , Factor de Transcripción STAT3/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología
18.
Cell Mol Life Sci ; 72(18): 3507-19, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26026420

RESUMEN

The pathogenesis of pulmonary fibrosis is a complicated and complex process that involves phenotypic abnormalities of a variety of cell types and dysregulations of multiple signaling pathways. There are numerous genetic, epigenetic and post-transcriptional mechanisms that have been identified to participate in the pathogenesis of this disease. However, efficacious therapeutics developed from these studies have been disappointingly limited. In the past several years, a group of new molecules, i.e., non-coding RNAs (ncRNAs), has been increasingly appreciated to have critical roles in the pathological progression of lung fibrosis. In this review, we summarize the recent findings on the roles of ncRNAs in the pathogenesis of this disorder. We analyze the translational potential of this group of molecules in treating lung fibrosis. We also discuss challenges and future opportunities of studying and utilizing ncRNAs in lung fibrosis.


Asunto(s)
Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , ARN no Traducido/genética , Animales , Humanos , Transducción de Señal/genética
19.
Am J Respir Crit Care Med ; 192(12): 1462-74, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26284610

RESUMEN

RATIONALE: Dysregulation of cellular metabolism has been shown to participate in several pathologic processes. However, the role of metabolic reprogramming is not well appreciated in the pathogenesis of organ fibrosis. OBJECTIVES: To determine if glycolytic reprogramming participates in the pathogenesis of lung fibrosis and assess the therapeutic potential of glycolytic inhibition in treating lung fibrosis. METHODS: A cell metabolism assay was performed to determine glycolytic flux and mitochondrial respiration. Lactate levels were measured to assess glycolysis in fibroblasts and lungs. Glycolytic inhibition by genetic and pharmacologic approaches was used to demonstrate the critical role of glycolysis in lung fibrosis. MEASUREMENTS AND MAIN RESULTS: Augmentation of glycolysis is an early and sustained event during myofibroblast differentiation, which is dependent on the increased expression of critical glycolytic enzymes, in particular, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3). Augmented glycolysis contributes to the stabilization of hypoxia-inducible factor 1-α, a master regulator of glycolytic enzymes implicated in organ fibrosis, by increasing cellular levels of tricarboxylic acid cycle intermediate succinate in lung myofibroblasts. Inhibition of glycolysis by the PFKFB3 inhibitor 3PO or genomic disruption of the PFKFB3 gene blunted the differentiation of lung fibroblasts into myofibroblasts, and attenuated profibrotic phenotypes in myofibroblasts isolated from the lungs of patients with idiopathic pulmonary fibrosis. Inhibition of glycolysis by 3PO demonstrates therapeutic benefit in bleomycin-induced and transforming growth factor-ß1-induced lung fibrosis in mice. CONCLUSIONS: Our data support the novel concept of glycolytic reprogramming in the pathogenesis of lung fibrosis and provide proof-of-concept that targeting this pathway may be efficacious in treating fibrotic disorders, such as idiopathic pulmonary fibrosis.


Asunto(s)
Glucólisis/fisiología , Fibrosis Pulmonar Idiopática/metabolismo , Miofibroblastos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL
20.
Eur J Immunol ; 44(7): 2085-95, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24723426

RESUMEN

Long noncoding RNAs (lncRNAs), once thought to be transcriptional noise, have been recently shown to regulate a variety of biological processes. However, there is not much knowledge regarding their roles in the inflammatory response. In this study, we performed human lncRNA microarray assays and identified a number of lncRNAs that demonstrated altered expression in response to LPS stimulation. Of these lncRNAs, lnc-IL7R, which overlaps with the 3'untranslated region (3'UTR) of the human interleukin-7 receptor α-subunit gene (IL7R) gene, was significantly upregulated in LPS-treated cells. Functionally, lnc-IL7R was capable of diminishing the LPS-induced inflammatory response, demonstrated by elevated expression of LPS-induced E-selectin, VCAM-1, IL-6, and IL-8 in lnc-IL7R knockdown cells. Mechanistically, we found that lnc-IL7R knockdown diminished trimethylation of histone H3 at lysine 27 (H3K27me3), a hallmark of silent transcription, at the proximal promoters of the inflammatory mediators. Our data suggest that lnc-IL7R contributes another layer of complexity in regulation of the inflammatory response.


Asunto(s)
Inflamación/etiología , ARN Largo no Codificante/fisiología , Receptores de Interleucina-7/genética , Células Cultivadas , Selectina E/genética , Histonas/metabolismo , Humanos , Lipopolisacáridos/farmacología , Metilación , Molécula 1 de Adhesión Celular Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA