Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Allergy Clin Immunol ; 149(4): 1445-1457.e5, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34624393

RESUMEN

BACKGROUND: Orosomucoid 1-like protein 3 (ORMDL3), a transmembrane protein localized in the endoplasmic reticulum (ER), has been genetically associated with chronic obstructive pulmonary disease (COPD), in addition to childhood-onset asthma. However, the functional role of ORMDL3 in the pathogenesis of COPD is still unknown. OBJECTIVE: Because cigarette smoke is the major risk factor for COPD, we aimed to investigate the role of ORMDL3 in cigarette smoke-induced human airway smooth muscle cell (HASMC) injury. METHODS: The mRNA and protein expression of ORMDL3 was examined in HASMCs from nonsmokers and smokers without or with COPD. Knockdown of ORMDL3 in primary healthy HASMCs was performed using small interfering RNA before exposure to cigarette smoke medium (CSM) for 24 hours. Inflammatory, proliferative/apoptotic, ER stress, and mitochondrial markers were evaluated. RESULTS: Elevation of ORMDL3 mRNA and protein expression was observed in HASMCs of smokers without or with COPD. CSM caused significant upregulation of ORMDL3 expression in healthy nonsmokers. ORMDL3 knockdown regulated CSM-induced inflammation, cell proliferation, and apoptosis. Silencing ORMDL3 led to reduction of CSM-induced ER stress via inhibition of unfolded protein response pathways such as activating transcription factor 6 and protein kinase RNA-like ER kinase. ORMDL3 was also involved in CSM-induced mitochondrial dysfunction via the mitochondrial fission process. CONCLUSIONS: We report the induction of ORMDL3 in HASMCs after cigarette smoke exposure. ORMDL3 may mediate cigarette smoke-induced activation of unfolded protein response pathways during airway smooth muscle cell injury.


Asunto(s)
Asma , Fumar Cigarrillos , Enfermedad Pulmonar Obstructiva Crónica , Asma/metabolismo , Niño , Fumar Cigarrillos/efectos adversos , Estrés del Retículo Endoplásmico , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Miocitos del Músculo Liso/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , ARN Mensajero/metabolismo , Nicotiana
2.
Am J Respir Cell Mol Biol ; 67(4): 471-481, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35763375

RESUMEN

Mitochondrial dysfunction has been reported in chronic obstructive pulmonary disease (COPD). Transfer of mitochondria from mesenchymal stem cells to airway smooth muscle cells (ASMCs) can attenuate oxidative stress-induced mitochondrial damage. It is not known whether mitochondrial transfer can occur between structural cells in the lungs or what role this may have in modulating bioenergetics and cellular function in healthy and COPD airways. Here, we show that ASMCs from both healthy ex-smokers and subjects with COPD can exchange mitochondria, a process that happens, at least partly, via extracellular vesicles. Exposure to cigarette smoke induces mitochondrial dysfunction and leads to an increase in the donation of mitochondria by ASMCs, suggesting that the latter may be a stress response mechanism. Healthy ex-smoker ASMCs that receive mitochondria show increases in mitochondrial biogenesis and respiration and a reduction in cell proliferation, irrespective of whether the mitochondria are transferred from healthy ex-smoker or COPD ASMCs. Our data indicate that mitochondrial transfer between structural cells is a homeostatic mechanism for the regulation of bioenergetics and cellular function within the airways and may represent an endogenous mechanism for reversing the functional consequences of mitochondrial dysfunction in diseases such as COPD.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica , Metabolismo Energético , Humanos , Pulmón/metabolismo , Mitocondrias/metabolismo , Músculo Liso , Enfermedad Pulmonar Obstructiva Crónica/metabolismo
3.
Respir Res ; 21(1): 262, 2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046036

RESUMEN

BACKGROUND: Mitochondrial damage and dysfunction have been reported in airway and quadriceps muscle cells of patients with chronic obstructive pulmonary disease (COPD). We determined the concomitance of mitochondrial dysfunction in these cells in COPD. METHODS: Bronchial biopsies were obtained from never- and ex-smoker volunteers and COPD patients (GOLD Grade 2) and quadriceps muscle biopsies from the same volunteers in addition to COPD patients at GOLD Grade 3/4 for measurement of mitochondrial function. RESULTS: Decreased mitochondrial membrane potential (ΔΨm), increased mitochondrial reactive oxygen species (mtROS) and decreased superoxide dismutase 2 (SOD2) levels were observed in mitochondria isolated from bronchial biopsies from Grade 2 patients compared to healthy never- and ex-smokers. There was a significant correlation between ΔΨm and FEV1 (% predicted), transfer factor of the lung for carbon monoxide (TLCOC % predicted), 6-min walk test and maximum oxygen consumption. In addition, ΔΨm was also associated with decreased expression levels of electron transport chain (ETC) complex proteins I and II. In quadriceps muscle of Grade 2 COPD patients, a significant increase in total ROS and mtROS was observed without changes in ΔΨm, SOD2 or ETC complex protein expression. However, quadriceps muscle of GOLD Grade 3/4 COPD patients showed an increased mtROS and decreased SOD2 and ETC complex proteins I, II, III and V expression. CONCLUSIONS: Mitochondrial dysfunction in the airways, but not in quadriceps muscle, is associated with airflow obstruction and exercise capacity in Grade 2 COPD. Oxidative stress-induced mitochondrial dysfunction in the quadriceps may result from similar disease processes occurring in the lungs.


Asunto(s)
Bronquios/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Músculo Cuádriceps/metabolismo , Anciano , Bronquios/patología , Femenino , Humanos , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Persona de Mediana Edad , Mitocondrias/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Músculo Cuádriceps/patología , Especies Reactivas de Oxígeno/metabolismo
4.
J Allergy Clin Immunol ; 141(5): 1634-1645.e5, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28911970

RESUMEN

BACKGROUND: Oxidative stress-induced mitochondrial dysfunction can contribute to inflammation and remodeling in patients with chronic obstructive pulmonary disease (COPD). Mesenchymal stem cells protect against lung damage in animal models of COPD. It is unknown whether these effects occur through attenuating mitochondrial dysfunction in airway cells. OBJECTIVE: We sought to examine the effect of induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) on oxidative stress-induce mitochondrial dysfunction in human airway smooth muscle cells (ASMCs) in vitro and in mouse lungs in vivo. METHODS: ASMCs were cocultured with iPSC-MSCs in the presence of cigarette smoke medium (CSM), and mitochondrial reactive oxygen species (ROS) levels, mitochondrial membrane potential (ΔΨm), and apoptosis were measured. Conditioned medium from iPSC-MSCs and transwell cocultures were used to detect any paracrine effects. The effect of systemic injection of iPSC-MSCs on airway inflammation and hyperresponsiveness in ozone-exposed mice was also investigated. RESULTS: Coculture of iPSC-MSCs with ASMCs attenuated CSM-induced mitochondrial ROS, apoptosis, and ΔΨm loss in ASMCs. iPSC-MSC-conditioned medium or transwell cocultures with iPSC-MSCs reduced CSM-induced mitochondrial ROS but not ΔΨm or apoptosis in ASMCs. Mitochondrial transfer from iPSC-MSCs to ASMCs was observed after direct coculture and was enhanced by CSM. iPSC-MSCs attenuated ozone-induced mitochondrial dysfunction, airway hyperresponsiveness, and inflammation in mouse lungs. CONCLUSION: iPSC-MSCs offered protection against oxidative stress-induced mitochondrial dysfunction in human ASMCs and in mouse lungs while reducing airway inflammation and hyperresponsiveness. These effects are, at least in part, dependent on cell-cell contact, which allows for mitochondrial transfer, and paracrine regulation. Therefore iPSC-MSCs show promise as a therapy for oxidative stress-dependent lung diseases, such as COPD.


Asunto(s)
Pulmón/patología , Células Madre Mesenquimatosas/patología , Mitocondrias/patología , Enfermedades Mitocondriales/patología , Estrés Oxidativo/fisiología , Animales , Apoptosis/fisiología , Técnicas de Cocultivo/métodos , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Pulmón/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Especies Reactivas de Oxígeno/metabolismo , Humo/efectos adversos , Nicotiana/efectos adversos
5.
Eur Respir J ; 50(5)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29191950

RESUMEN

Chronic obstructive pulmonary disease (COPD) airways are characterised by thickening of airway smooth muscle, partly due to airway smooth muscle cell (ASMC) hyperplasia. Metabolic reprogramming involving increased glycolysis and glutamine catabolism supports the biosynthetic and redox balance required for cellular growth. We examined whether COPD ASMCs show a distinct metabolic phenotype that may contribute to increased growth.We performed an exploratory intracellular metabolic profile analysis of ASMCs from healthy nonsmokers, healthy smokers and COPD patients, under unstimulated or growth conditions of transforming growth factor (TGF)-ß and fetal bovine serum (FBS).COPD ASMCs showed impaired energy balance and accumulation of the glycolytic product lactate, glutamine, fatty acids and amino acids compared to controls in unstimulated and growth conditions. Fatty acid oxidation capacity was reduced under unstimulated conditions. TGF-ß/FBS-stimulated COPD ASMCs showed restoration of fatty acid oxidation capacity, upregulation of the pentose phosphate pathway product ribose-5-phosphate and of nucleotide biosynthesis intermediates, and increased levels of the glutamine catabolite glutamate. In addition, TGF-ß/FBS-stimulated COPD ASMCs showed a higher reduced-to-oxidised glutathione ratio and lower mitochondrial oxidant levels. Inhibition of glycolysis and glutamine depletion attenuated TGF-ß/FBS-stimulated growth of COPD ASMCs.Changes in glycolysis, glutamine and fatty acid metabolism may lead to increased biosynthesis and redox balance, supporting COPD ASMC growth.


Asunto(s)
Bronquios/citología , Miocitos del Músculo Liso/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fumar/efectos adversos , Factor de Crecimiento Transformador beta/farmacología
6.
Respir Res ; 18(1): 194, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162108

RESUMEN

BACKGROUND: Patients with severe asthma have increased airway remodelling and elevated numbers of circulating fibrocytes with enhanced myofibroblastic differentiation capacity, despite being treated with high doses of corticosteroids, and long acting ß2-adrenergic receptor (AR) agonists (LABAs). We determined the effect of ß2-AR agonists, alone or in combination with corticosteroids, on fibrocyte function. METHODS: Non-adherent non-T cells from peripheral blood mononuclear cells isolated from healthy subjects and patients with non-severe or severe asthma were treated with the ß2-AR agonist, salmeterol, in the presence or absence of the corticosteroid dexamethasone. The number of fibrocytes (collagen I+/CD45+ cells) and differentiating fibrocytes (α-smooth muscle actin+ cells), and the expression of CC chemokine receptor 7 and of ß2-AR were determined using flow cytometry. The role of cyclic adenosine monophosphate (cAMP) was elucidated using the cAMP analogue 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP) and the phosphodiesterase type IV (PDE4) inhibitor, rolipram. RESULTS: Salmeterol reduced the proliferation, myofibroblastic differentiation and CCR7 expression of fibrocytes from healthy subjects and non-severe asthma patients. Fibrocytes from severe asthma patients had a lower baseline surface ß2-AR expression and were relatively insensitive to salmeterol but not to 8-Br-cAMP or rolipram. Dexamethasone increased ß2-AR expression and enhanced the inhibitory effect of salmeterol on severe asthma fibrocyte differentiation. CONCLUSIONS: Fibrocytes from patients with severe asthma are relatively insensitive to the inhibitory effects of salmeterol, an effect which is reversed by combination with corticosteroids.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Asma/fisiopatología , Fibroblastos/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Xinafoato de Salmeterol/farmacología , Índice de Severidad de la Enfermedad , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Adulto , Asma/tratamiento farmacológico , Asma/inmunología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Fibroblastos/fisiología , Humanos , Leucocitos Mononucleares/fisiología , Masculino , Persona de Mediana Edad , Xinafoato de Salmeterol/uso terapéutico , Resultado del Tratamiento
7.
J Immunol ; 192(10): 4913-4920, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24733848

RESUMEN

Oxidative stress, a pathogenetic factor in many conditions, including chronic obstructive pulmonary disease, arises due to accumulation of reactive oxygen species and defective antioxidant defenses in the lungs. The latter is due, at least in part, to impaired activation of NF-E2-related factor 2 (Nrf2), a transcription factor involved in the activation of antioxidant and cytoprotective genes. The bromodomain and extraterminal (BET) proteins, Brd2, Brd3, Brd4, and BrdT, bind to acetylated lysine residues on histone or nonhistone proteins recruiting transcriptional regulators and thus activating or repressing gene transcription. We investigated whether BET proteins modulate the regulation of Nrf2-dependent gene expression in primary human airway smooth muscle cells and the human monocytic cell line, THP-1. Inhibition of BET protein bromodomains using the inhibitor JQ1+ or attenuation of Brd2 and Brd4 expression using small interfering RNA led to activation of Nrf2-dependent transcription and expression of the antioxidant proteins heme oxygenase-1, NADPH quinone oxidoreductase 1, and glutamate-cysteine ligase catalytic subunit. Also, JQ1+ prevented H2O2-induced intracellular reactive oxygen species production. By coimmunoprecipitation, BET proteins were found to be complexed with Nrf2, whereas chromatin-immunoprecipitation studies indicated recruitment of Brd2 and Brd4 to Nrf2-binding sites on the promoters of heme oxygenase-1 and NADPH quinone oxidoreductase 1. BET proteins, particularly Brd2 and Brd4, may play a key role in the regulation of Nrf2-dependent antioxidant gene transcription and are hence an important target for augmenting antioxidant responses in oxidative stress-mediated diseases.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/inmunología , Hemo-Oxigenasa 1/inmunología , NAD(P)H Deshidrogenasa (Quinona)/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Proteínas Nucleares/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Factores de Transcripción/inmunología , Transcripción Genética/inmunología , Azepinas/farmacología , Línea Celular Tumoral , Proteínas Cromosómicas no Histona , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Hemo-Oxigenasa 1/genética , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/genética , Proteínas Nucleares/genética , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Estrés Oxidativo/inmunología , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Transcripción Genética/genética , Triazoles/farmacología
8.
Am J Respir Crit Care Med ; 191(1): 54-62, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25411910

RESUMEN

RATIONALE: Patients with severe asthma (SA) are less responsive to the beneficial effects of corticosteroid (CS) therapy, and relative CS insensitivity has been shown in airway smooth muscle cells (ASMC) from patients with SA. OBJECTIVES: We investigated whether there was a defect in the actions of the glucocorticoid receptor (GR) underlying the ability of CS to suppress the inflammatory response in ASMC of patients with SA. ASMC from healthy subjects (n = 10) and subjects with severe (n = 8) and nonsevere asthma (N-SA; n = 8) were cultured from endobronchial biopsies. MEASUREMENTS AND MAIN RESULTS: GR expression in ASMC from SA and N-SA was reduced compared with that from healthy subjects by 49% (P < 0.01). Although baseline levels of nuclear GR were similar, GR nuclear translocation induced by dexamethasone (10(-7) M) in SA was 60% of that measured in either healthy subjects or subjects with N-SA. Tumor necrosis factor (TNF)-α induced greater nuclear factor (NF)-κB (p65) mRNA expression in ASMC from subjects with SA (5.6- vs. 2.0-fold; P < 0.01), whereas baseline and TNF-α-induced nuclear translocation and dexamethasone-mediated suppression of p65 expression were similar between groups. Dexamethasone, although not modulating TNF-α-induced p65 nuclear translocation, attenuated p65 recruitment to the CCL11 promoter in the healthy and N-SA groups, but this suppressive effect was impaired in subjects with SA. CONCLUSIONS: Decreased GR expression with impaired nuclear translocation in ASMC, associated with reduced dexamethasone-mediated attenuation of p65 recruitment to NF-κB-dependent gene promoters, may underlie CS insensitivity of severe asthma.


Asunto(s)
Corticoesteroides/uso terapéutico , Asma/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Receptores de Glucocorticoides/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Corticoesteroides/inmunología , Corticoesteroides/farmacología , Adulto , Asma/tratamiento farmacológico , Asma/inmunología , Resistencia a Medicamentos , Femenino , Humanos , Masculino , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Receptores de Glucocorticoides/inmunología , Receptores de Glucocorticoides/metabolismo , Sistema Respiratorio/inmunología , Sistema Respiratorio/fisiopatología , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología
9.
J Allergy Clin Immunol ; 135(5): 1186-95.e1-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25488691

RESUMEN

BACKGROUND: Patients with severe asthma are less responsive to corticosteroid therapy and show increased airway remodeling. The mesenchymal progenitors, fibrocytes, may be involved in the remodeling of asthmatic airways. We propose that fibrocytes in severe asthma are different from those in nonsevere asthma. OBJECTIVES: To examine the survival, myofibroblastic differentiation, and C-C chemokine receptor 7 (CCR7) expression in blood fibrocytes from patients with severe and nonsevere asthma and study the effect of corticosteroids on fibrocyte function. METHODS: The nonadherent non-T-cell fraction of blood mononuclear cells was isolated from healthy subjects and patients with nonsevere and severe asthma. Total and differentiating fibrocytes were identified by their expression of CD45, collagen I, and α-smooth muscle actin using flow cytometry. The expression of CCR7 and of the glucocorticoid receptor was measured by using flow cytometry. RESULTS: Increased numbers of circulating fibrocytes, with greater myofibroblastic differentiation potential, were observed in patients with severe asthma. Dexamethasone induced apoptosis, leading to reduction in the number of cultured fibrocytes and total nonadherent non-T cells from healthy subjects and patients with nonsevere asthma but not from patients with severe asthma. Dexamethasone reduced CCR7 expression in fibrocytes from patients with nonsevere asthma but not from patients with severe asthma. Glucocorticoid receptor expression was attenuated in fibrocytes from patients with severe asthma. CONCLUSIONS: Patients with severe asthma have elevated numbers of circulating fibrocytes that show enhanced myofibroblastic differentiation and that are less responsive to the effects of corticosteroids.


Asunto(s)
Corticoesteroides/uso terapéutico , Antiasmáticos/uso terapéutico , Asma/diagnóstico , Asma/tratamiento farmacológico , Resistencia a Medicamentos , Fenotipo , Corticoesteroides/farmacología , Adulto , Antiasmáticos/farmacología , Apoptosis/efectos de los fármacos , Estudios de Casos y Controles , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Células del Tejido Conectivo/citología , Células del Tejido Conectivo/efectos de los fármacos , Células del Tejido Conectivo/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores CCR7/metabolismo , Receptores de Glucocorticoides/metabolismo , Índice de Severidad de la Enfermedad
10.
J Allergy Clin Immunol ; 136(3): 769-80, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25828268

RESUMEN

BACKGROUND: Inflammation and oxidative stress play critical roles in patients with chronic obstructive pulmonary disease (COPD). Mitochondrial oxidative stress might be involved in driving the oxidative stress-induced pathology. OBJECTIVE: We sought to determine the effects of oxidative stress on mitochondrial function in the pathophysiology of airway inflammation in ozone-exposed mice and human airway smooth muscle (ASM) cells. METHODS: Mice were exposed to ozone, and lung inflammation, airway hyperresponsiveness (AHR), and mitochondrial function were determined. Human ASM cells were isolated from bronchial biopsy specimens from healthy subjects, smokers, and patients with COPD. Inflammation and mitochondrial function in mice and human ASM cells were measured with and without the presence of the mitochondria-targeted antioxidant MitoQ. RESULTS: Mice exposed to ozone, a source of oxidative stress, had lung inflammation and AHR associated with mitochondrial dysfunction and reflected by decreased mitochondrial membrane potential (ΔΨm), increased mitochondrial oxidative stress, and reduced mitochondrial complex I, III, and V expression. Reversal of mitochondrial dysfunction by the mitochondria-targeted antioxidant MitoQ reduced inflammation and AHR. ASM cells from patients with COPD have reduced ΔΨm, adenosine triphosphate content, complex expression, basal and maximum respiration levels, and respiratory reserve capacity compared with those from healthy control subjects, whereas mitochondrial reactive oxygen species (ROS) levels were increased. Healthy smokers were intermediate between healthy nonsmokers and patients with COPD. Hydrogen peroxide induced mitochondrial dysfunction in ASM cells from healthy subjects. MitoQ and Tiron inhibited TGF-ß-induced ASM cell proliferation and CXCL8 release. CONCLUSIONS: Mitochondrial dysfunction in patients with COPD is associated with excessive mitochondrial ROS levels, which contribute to enhanced inflammation and cell hyperproliferation. Targeting mitochondrial ROS represents a promising therapeutic approach in patients with COPD.


Asunto(s)
Antioxidantes/farmacología , Mitocondrias/metabolismo , Músculo Liso/metabolismo , Compuestos Organofosforados/farmacología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Sistema Respiratorio/metabolismo , Ubiquinona/análogos & derivados , Adulto , Anciano , Remodelación de las Vías Aéreas (Respiratorias)/genética , Animales , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/tratamiento farmacológico , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/patología , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Músculo Liso/efectos de los fármacos , Músculo Liso/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Estrés Oxidativo/efectos de los fármacos , Ozono , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/genética , Neumonía/patología , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/patología , Transducción de Señal , Fumar/metabolismo , Fumar/fisiopatología , Ubiquinona/farmacología
11.
Front Cell Dev Biol ; 12: 1360079, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38495619

RESUMEN

Mitochondria are key metabolic hubs involved in cellular energy production and biosynthesis. ATP is generated primarily by glucose and fatty acid oxidation through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) in the mitochondria. During OXPHOS there is also production of reactive oxygen species (ROS), which are involved in the regulation of cellular function. Mitochondria are also central in the regulating cell survival and death, particularly in the intrinsic apoptosis pathway. Severe asthma is a heterogeneous disease driven by various immune mechanisms. Severe eosinophilic asthma entails a type 2 inflammatory response and peripheral and lung eosinophilia, associated with severe airflow obstruction, frequent exacerbations and poor response to treatment. Mitochondrial dysfunction and altered metabolism have been observed in airway epithelial and smooth muscle cells from patients with asthma. However, the role of mitochondria in the development of eosinophilia and eosinophil-mediated inflammation in severe asthma is unknown. In this review, we discuss the currently limited literature on the role of mitochondria in eosinophil function and how it is regulated by asthma-relevant cytokines, including interleukin (IL)-5 and granulocyte-macrophage colony-stimulating factor (GM-CSF), as well as by corticosteroid drugs. Moreover, we summarise the evidence on the role of mitochondria in the regulation of eosinophils apoptosis and eosinophil extracellular trap formation. Finally, we discuss the possible role of altered mitochondrial function in eosinophil dysfunction in severe asthma and suggest possible research avenues in order to better understand their role in disease pathogenesis, and identify novel therapeutic targets.

12.
Vitam Horm ; 124: 341-366, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38408802

RESUMEN

This article discusses the physiological and anatomical changes of adrenal gland with age and the effects this has overall on how the organ responds to stress. Physiological changes entail a decrease in adrenocorticoid hormone secretion however cortisol levels remain intact leading to a disruptive stress response. Additionally, loss of zonation of the organ also occurs. Both characteristics in combination with chronic stress affect overall health. Complex interplay between adrenal aging and stress responsiveness is confounded further by the impact they expel on other systems, such as the thyroid hormone. The body undergoes age-related transformations modifying rate of cellular growth, differentiation, senescence, and hormone production. Given the multiplicity and complexity of hormones, their production must be considered to develop appropriate interventions to mitigate its effect on age related diseases in health.


Asunto(s)
Glándulas Suprarrenales , Hormonas , Humanos , Hormonas Tiroideas , Envejecimiento
14.
J Allergy Clin Immunol ; 130(4): 877-85.e5, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22947346

RESUMEN

BACKGROUND: Patients with severe asthma are less responsive to the beneficial effects of corticosteroid therapy. OBJECTIVE: We investigated whether corticosteroid insensitivity was present in airway smooth muscle cells (ASMCs) of patients with severe asthma. METHODS: ASMCs cultured from bronchial biopsy specimens of nonasthmatic control subjects (n = 12) and patients with nonsevere (n = 10) or severe (n = 10) asthma were compared for the effect of dexamethasone on suppression of TNF-α- and IFN-γ-induced CCL11 (eotaxin), CXCL8 (IL-8), and CX3CL1 (fractalkine) expression. The mechanisms of corticosteroid insensitivity are also determined. RESULTS: CCL11 release was higher in ASMCs of patients with nonsevere but not severe asthma and nonasthmatic control subjects; CXCL8 and CX3CL1 release were similar in all groups. In patients with severe asthma, dexamethasone caused less suppression of CCL11 and CXCL8 release induced by TNF-α. Dexamethasone potentiated TNF-α- and IFN-γ-induced CX3CL1 release equally in all 3 groups. TNF-α-induced phosphorylated p38 mitogen-activated protein kinase levels were increased in ASMCs from patients with severe asthma compared with those from patients with nonsevere asthma and nonasthmatic subjects, whereas TNF-α-induced phosphorylated c-Jun N-terminal kinase and phosphorylated extracellular signal-related kinase levels were increased in all asthmatic groups. A p38 inhibitor increased the inhibitory effect of dexamethasone. CONCLUSIONS: ASMCs of patients with severe asthma are corticosteroid insensitive; this might be secondary to heightened p38 mitogen-activated protein kinase levels.


Asunto(s)
Asma/tratamiento farmacológico , Bronquios/efectos de los fármacos , Quimiocinas/genética , Dexametasona/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Adulto , Asma/inmunología , Bronquios/inmunología , Células Cultivadas , Quimiocinas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción ReIA/fisiología
15.
Nutrients ; 15(22)2023 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-38004225

RESUMEN

Cardiovascular disease (CVD) that includes myocardial infarction and stroke, is the leading cause of mortality worldwide. Atherosclerosis, the primary underlying cause of CVD, can be controlled by pharmacological and dietary interventions, including n-3 polyunsaturated fatty acid (PUFA) supplementation. n-3 PUFA supplementation, primarily consisting of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), has shown promise in reducing atherosclerosis by modulating risk factors, including triglyceride levels and vascular inflammation. n-3 PUFAs act by replacing pro-inflammatory fatty acid types in cell membranes and plasma lipids, by regulating transcription factor activity, and by inducing epigenetic changes. EPA and DHA regulate cellular function through shared and differential molecular mechanisms. Large clinical studies on n-3 PUFAs have reported conflicting findings, causing confusion among the public and health professionals. In this review, we discuss important factors leading to these inconsistencies, in the context of atherosclerosis, including clinical study design and the differential effects of EPA and DHA on cell function. We propose steps to improve clinical and basic experimental study design in order to improve supplement composition optimization. Finally, we propose that understanding the factors underlying the poor response to n-3 PUFAs, and the development of molecular biomarkers for predicting response may help towards a more personalized treatment.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Ácidos Grasos Omega-3 , Humanos , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/uso terapéutico , Ácido Eicosapentaenoico/farmacología , Ácido Eicosapentaenoico/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/prevención & control , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Ácidos Grasos Insaturados , Ácidos Grasos , Aterosclerosis/tratamiento farmacológico
16.
Front Immunol ; 14: 1201658, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37520564

RESUMEN

The airway epithelium comprises of different cell types and acts as a physical barrier preventing pathogens, including inhaled particles and microbes, from entering the lungs. Goblet cells and submucosal glands produce mucus that traps pathogens, which are expelled from the respiratory tract by ciliated cells. Basal cells act as progenitor cells, differentiating into different epithelial cell types, to maintain homeostasis following injury. Adherens and tight junctions between cells maintain the epithelial barrier function and regulate the movement of molecules across it. In this review we discuss how abnormal epithelial structure and function, caused by chronic injury and abnormal repair, drives airway disease and specifically asthma and chronic obstructive pulmonary disease (COPD). In both diseases, inhaled allergens, pollutants and microbes disrupt junctional complexes and promote cell death, impairing the barrier function and leading to increased penetration of pathogens and a constant airway immune response. In asthma, the inflammatory response precipitates the epithelial injury and drives abnormal basal cell differentiation. This leads to reduced ciliated cells, goblet cell hyperplasia and increased epithelial mesenchymal transition, which contribute to impaired mucociliary clearance and airway remodelling. In COPD, chronic oxidative stress and inflammation trigger premature epithelial cell senescence, which contributes to loss of epithelial integrity and airway inflammation and remodelling. Increased numbers of basal cells showing deregulated differentiation, contributes to ciliary dysfunction and mucous hyperproduction in COPD airways. Defective antioxidant, antiviral and damage repair mechanisms, possibly due to genetic or epigenetic factors, may confer susceptibility to airway epithelial dysfunction in these diseases. The current evidence suggests that a constant cycle of injury and abnormal repair of the epithelium drives chronic airway inflammation and remodelling in asthma and COPD. Mechanistic understanding of injury susceptibility and damage response may lead to improved therapies for these diseases.


Asunto(s)
Asma , Enfermedad Pulmonar Obstructiva Crónica , Trastornos Respiratorios , Humanos , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Pulmón/metabolismo , Inflamación
17.
Am J Respir Crit Care Med ; 184(8): 894-903, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21799075

RESUMEN

RATIONALE: Aberrant airway smooth muscle cell (ASMC) function and overexpression of transforming growth factor (TGF)-ß, which modulates ASMC proliferative and inflammatory function and induces oxidant release, are features of asthma. Nuclear factor E2-related factor 2 (Nrf2) activates antioxidant genes conferring protection against oxidative stress. OBJECTIVES: To determine the role of Nrf2 in ASMCs and its modulation by TGF-ß, and compare Nrf2 activity in ASMCs from subjects with severe and nonsevere asthma and healthy subjects. METHODS: ASMCs were cultured from airways of subjects without asthma, and from airway biopsies from patients with severe and nonsevere asthma. We studied Nrf2 activation on antioxidant gene expression and proliferation, the effect of TGF-ß on Nrf2 transcriptional activity, and the impact of Nrf2 activation on TGF-ß­mediated proliferation and IL-6 release. Nrf2­antioxidant response elements binding and Nrf2-dependent antioxidant gene expression was determined in asthmatic ASMCs. MEASUREMENTS AND MAIN RESULTS: Activation of Nrf2 led to up-regulation of the antioxidant genes heme oxygenase (HO)-1, NAD(P)H:quinone oxidoreductase, and manganese superoxide dismutase, and a reduction in proliferation. TGF-ß reduced Nrf2-mediated antioxidant gene transcription through induction of activating transcription factor-3 expression. Nrf2 activation attenuated TGF-ß­mediated reduction in HO-1,ASMC proliferation, and IL-6 release. Nrf2­antioxidant response elements binding was reduced in ASMCs from patients with severe asthma compared with ASMCs from patients with nonsevere asthma and normal subjects. HO-1 expression was reduced in ASMCs from patients with both nonsevere and severe asthma compared with healthy subjects. CONCLUSIONS: Nrf2 regulates antioxidant responses and proliferation in ASMCs and is inactivated by TGF-ß. Nrf2 reduction may underlie compromised antioxidant protection and aberrant ASM function in asthma.


Asunto(s)
Asma/metabolismo , Proliferación Celular , Músculo Liso/metabolismo , Factor de Transcripción NF-E2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adenoviridae/genética , Antioxidantes/metabolismo , Proliferación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Isotiocianatos , Músculo Liso/citología , Factor de Transcripción NF-E2/antagonistas & inhibidores , ARN Interferente Pequeño , Sulfóxidos , Tiocianatos/farmacología , Transfección , Factor de Crecimiento Transformador beta/farmacología
18.
Mol Aspects Med ; 85: 101026, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34625291

RESUMEN

The lungs are exposed to reactive oxygen species oxygen (ROS) produced as a result of inhalation of oxygen, as well as smoke and other air pollutants. Cell metabolism and the NADPH oxidases (Nox) generate low levels of intracellular ROS that act as signal transduction mediators by inducing oxidative modifications of histones, enzymes and transcription factors. Redox signalling is also regulated by localised production and sensing of ROS in mitochondria, the endoplasmic reticulum (ER) and inside the nucleus. Intracellular ROS are maintained at low levels through the action of a battery of enzymatic and non-enzymatic antioxidants. Asthma is a heterogeneous airway inflammatory disease with different immune endotypes; these include atopic or non-atopic Th2 type immune response associated with eosinophilia, or a non-Th2 response associated with neutrophilia. Airway remodelling and hyperresponsiveness accompany the inflammatory response in asthma. Over-production of ROS resulting from infiltrating immune cells, particularly eosinophils and neutrophils, and a concomitant impairment of antioxidant responses lead to development of oxidative stress in asthma. Oxidative stress is augmented in severe asthma and during exacerbations, as well as by air pollution and obesity, and causes oxidative damage of tissues promoting airway inflammation and hyperresponsiveness. Furthermore, deregulated Nox activity, mitochondrial dysfunction, ER stress and/or oxidative DNA damage, resulting from exposure to irritants, inflammatory mediators or obesity, may lead to redox-dependent changes in cell signalling. ROS play a central role in airway epithelium-mediated sensing, development of innate and adaptive immune responses, and airway remodelling and hyperresponsiveness. Nonetheless, antioxidant compounds have proven clinically ineffective as therapeutic agents for asthma, partly due to issues with stability and in vivo metabolism of these compounds. The compartmentalised nature of ROS production and sensing, and the role of ROS in homeostatic responses and in the action of corticosteroids and ß2-adrenergic receptor agonists, adds another layer of complexity to antioxidant therapy development. Nox inhibitors and mitochondrial-targeted antioxidants are in clinical development for a number of diseases but they have not yet been investigated in asthma. A better understanding of the complex role of ROS in the pathogenesis of asthma will highlight new opportunities for more targeted and effective redox therapies.


Asunto(s)
Antioxidantes , Asma , Remodelación de las Vías Aéreas (Respiratorias) , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Asma/tratamiento farmacológico , Humanos , NADPH Oxidasas/metabolismo , Obesidad , Estrés Oxidativo , Oxígeno , Especies Reactivas de Oxígeno/metabolismo
19.
Am J Respir Cell Mol Biol ; 45(4): 746-52, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21297080

RESUMEN

Hydrogen sulfide (H(2)S) is synthesized intracellularly by the enzymes cystathionine-γ-lyase and cystathionine-ß-synthase (CBS), and is proposed to be a gasotransmitter with effects in modulating inflammation and cellular proliferation. We determined a role of H(2)S in airway smooth muscle (ASM) function. ASM were removed from resection or transplant donor lungs and were placed in culture. Proliferation of ASM was induced by FCS and the proinflammatory cytokine, IL-1ß. Proliferation of ASM and IL-8 release were measured by bromodeoxyuridine incorporation and ELISA, respectively. Exposure of ASM to H(2)S "donors" inhibited this proliferation and IL-8 release. Methemoglobin, a scavenger of endogenous H(2)S, increased DNA synthesis induced by FCS and IL-1ß. In addition, methemoglobin increased IL-8 release induced by FCS, but not by IL-1ß, indicating a role for endogenous H(2)S in these systems. Inhibition of CBS, but not cystathionine-γ-lyase, reversed the inhibitory effect of H(2)S on proliferation and IL-8 release, indicating that this is dependent on CBS. CBS mRNA and protein expression were inhibited by H(2)S donors, and were increased by methemoglobin, indicating that CBS is the main enzyme responsible for endogenous H(2)S production. Finally, we found that exogenous H(2)S inhibited the phosphorylation of extracellular signal-regulated kinase-1/2 and p38, which could represent a mechanism by which H(2)S inhibited cellular proliferation and IL-8 release. In summary, H(2)S production provides a novel mechanism for regulation of ASM proliferation and IL-8 release. Therefore, regulation of H(2)S may represent a novel approach to controlling ASM proliferation and cytokine release that is found in patients with asthma.


Asunto(s)
Bronquios/metabolismo , Proliferación Celular , Sulfuro de Hidrógeno/metabolismo , Interleucina-8/metabolismo , Miocitos del Músculo Liso/metabolismo , Bronquios/efectos de los fármacos , Bronquios/inmunología , Bronquios/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cistationina betasintasa/antagonistas & inhibidores , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-1beta/metabolismo , Metahemoglobina/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfolinas/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Compuestos Organotiofosforados/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Suero/metabolismo , Sulfuros/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L295-304, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21131394

RESUMEN

Reactive oxygen species (ROS) are generated as a result of normal cellular metabolism, mainly through the mitochondria and peroxisomes, but their release is enhanced by the activation of oxidant enzymes such as NADPH oxidases or downregulation of endogenous antioxidant enzymes such as manganese-superoxide dismutase (MnSOD) and catalase. Transforming growth factor-ß (TGF-ß), found to be overexpressed in airway smooth muscle (ASM) from asthmatic and chronic obstructive pulmonary disease patients, may be a pivotal regulator of abnormal ASM cell (ASMC) function in these diseases. An important effect of TGF-ß on ASMC inflammatory responses is the induction of IL-6 release. TGF-ß also triggers intracellular ROS release in ASMCs by upregulation of NADPH oxidase 4 (Nox4). However, the effect of TGF-ß on the expression of key antioxidant enzymes and subsequently on oxidant/antioxidant balance is unknown. Moreover, the role of redox-dependent pathways in the mediation of the proinflammatory effects of TGF-ß in ASMCs is unclear. In this study, we show that TGF-ß induced the expression of Nox4 while at the same time inhibiting the expression of MnSOD and catalase. This change in oxidant/antioxidant enzymes was accompanied by elevated ROS levels and IL-6 release. Further studies revealed a role for Smad3 and phosphatidyl-inositol kinase-mediated pathways in the induction of oxidant/antioxidant imbalance and IL-6 release. The changes in oxidant/antioxidant enzymes and IL-6 release were reversed by the antioxidants N-acetyl-cysteine (NAC) and ebselen through inhibition of Smad3 phosphorylation, indicating redox-dependent activation of Smad3 by TGF-ß. Moreover, these findings suggest a potential role for NAC in preventing TGF-ß-mediated pro-oxidant and proinflammatory responses in ASMCs. Knockdown of Nox4 using small interfering RNA partially prevented the inhibition of MnSOD but had no effect on catalase and IL-6 expression. These findings provide novel insights into redox regulation of ASM function by TGF-ß.


Asunto(s)
Catalasa/metabolismo , Interleucina-6/biosíntesis , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/metabolismo , Músculos Respiratorios/efectos de los fármacos , Músculos Respiratorios/metabolismo , Superóxido Dismutasa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Secuencia de Bases , Catalasa/genética , Células Cultivadas , Cartilla de ADN/genética , Expresión Génica/efectos de los fármacos , Humanos , Modelos Biológicos , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/farmacología , Músculos Respiratorios/citología , Transducción de Señal , Proteínas Smad/metabolismo , Superóxido Dismutasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA