Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Respir Res ; 23(1): 193, 2022 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-35902923

RESUMEN

BACKGROUND: CCAAT/Enhancer Binding Protein D (CEBPD), a pleiotropic glucocorticoid-responsive transcription factor, modulates inflammatory responses. Of relevance to asthma, expression of CEBPD in airway smooth muscle (ASM) increases with glucocorticoid exposure. We sought to characterize CEBPD-mediated transcriptomic responses to glucocorticoid exposure in ASM by measuring changes observed after knockdown of CEBPD and its impact on asthma-related ASM function. METHODS: Primary ASM cells derived from four donors were transfected with CEBPD or non-targeting (NT) siRNA and exposed to vehicle control, budesonide (100 nM, 18 h), TNFα (10 ng/ml, 18 h), or both budesonide and TNFα. Subsequently, RNA-Seq was used to measure gene expression levels, and pairwise differential expression results were obtained for exposures versus vehicle and knockdown versus control conditions. Weighted gene co-expression analysis was performed to identify groups of genes with similar expression patterns across the various experimental conditions (i.e., CEBPD knockdown status, exposures). RESULTS: CEBPD knockdown altered expression of 3037 genes under at least one exposure (q-value < 0.05). Co-expression analysis identified sets of 197, 152 and 290 genes that were correlated with CEBPD knockdown status, TNFα exposure status, and both, respectively. JAK-STAT signaling pathway genes, including IL6R and SOCS3, were among those influenced by both TNFα and CEBPD knockdown. Immunoblot assays revealed that budesonide-induced IL-6R protein expression and augmented IL-6-induced STAT3 phosphorylation levels were attenuated by CEBPD knockdown in ASM. CONCLUSIONS: CEBPD modulates glucocorticoid responses in ASM, in part via modulation of IL-6 receptor signaling.


Asunto(s)
Asma , Glucocorticoides , Budesonida/metabolismo , Budesonida/farmacología , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Glucocorticoides/farmacología , Humanos , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
2.
Sci Transl Med ; 14(627): eabf8188, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35020406

RESUMEN

Exacerbations of symptoms represent an unmet need for people with asthma. Bacterial dysbiosis and opportunistic bacterial infections have been observed in, and may contribute to, more severe asthma. However, the molecular mechanisms driving these exacerbations remain unclear. We show here that bacterial lipopolysaccharide (LPS) induces oncostatin M (OSM) and that airway biopsies from patients with severe asthma present with an OSM-driven transcriptional profile. This profile correlates with activation of inflammatory and mucus-producing pathways. Using primary human lung tissue or human epithelial and mesenchymal cells, we demonstrate that OSM is necessary and sufficient to drive pathophysiological features observed in severe asthma after exposure to LPS or Klebsiella pneumoniae. These findings were further supported through blockade of OSM with an OSM-specific antibody. Single-cell RNA sequencing from human lung biopsies identified macrophages as a source of OSM. Additional studies using Osm-deficient murine macrophages demonstrated that macrophage-derived OSM translates LPS signals into asthma-associated pathologies. Together, these data provide rationale for inhibiting OSM to prevent bacterial-associated progression and exacerbation of severe asthma.


Asunto(s)
Asma , Oncostatina M/metabolismo , Animales , Asma/patología , Humanos , Pulmón/patología , Macrófagos/metabolismo , Ratones , Moco , Oncostatina M/genética
3.
Am J Respir Cell Mol Biol ; 66(1): 96-106, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34648729

RESUMEN

In most living cells, the second-messenger roles for adenosine 3',5'-cyclic monophosphate (cAMP) are short-lived, confined to the intracellular space, and tightly controlled by the binary switch-like actions of Gαs (stimulatory G protein)-activated adenylyl cyclase (cAMP production) and cAMP-specific PDE (cAMP breakdown). Here, by using human airway smooth muscle (HASM) cells in culture as a model, we report that activation of the cell-surface ß2AR (ß2-adrenoceptor), a Gs-coupled GPCR (G protein-coupled receptor), evokes cAMP egress to the extracellular space. Increased extracellular cAMP levels ([cAMP]e) are long-lived in culture and are induced by receptor-dependent and receptor-independent mechanisms in such a way as to define a universal response class of increased intracellular cAMP levels ([cAMP]i). We find that HASM cells express multiple ATP-binding cassette (ABC) membrane transporters, with ABCC1 (ABC subfamily member C 1) being the most highly enriched transcript mapped to MRPs (multidrug resistance-associated proteins). We show that pharmacological inhibition or downregulation of ABCC1 with siRNA markedly reduces ß2AR-evoked cAMP release from HASM cells. Furthermore, inhibition of ABCC1 activity or expression decreases basal tone and increases ß-agonist-induced HASM cellular relaxation. These findings identify a previously unrecognized role for ABCC1 in the homeostatic regulation of [cAMP]i in HASM that may be conserved traits of the Gs-GPCRs (Gs-coupled family of GPCRs). Hence, the general features of this activation mechanism may uncover new disease-modifying targets in the treatment of airflow obstruction in asthma. Surprisingly, we find that serum cAMP levels are elevated in a small cohort of patients with asthma as compared with control subjects, which warrants further investigation.


Asunto(s)
AMP Cíclico/metabolismo , Pulmón/citología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Relajación Muscular/fisiología , Miocitos del Músculo Liso/fisiología , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Asma/sangre , Asma/fisiopatología , Cromograninas/metabolismo , AMP Cíclico/sangre , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , ARN Interferente Pequeño/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(45): 28485-28495, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33097666

RESUMEN

The recent discovery of sensory (tastant and odorant) G protein-coupled receptors on the smooth muscle of human bronchi suggests unappreciated therapeutic targets in the management of obstructive lung diseases. Here we have characterized the effects of a wide range of volatile odorants on the contractile state of airway smooth muscle (ASM) and uncovered a complex mechanism of odorant-evoked signaling properties that regulate excitation-contraction (E-C) coupling in human ASM cells. Initial studies established multiple odorous molecules capable of increasing intracellular calcium ([Ca2+]i) in ASM cells, some of which were (paradoxically) associated with ASM relaxation. Subsequent studies showed a terpenoid molecule (nerol)-stimulated OR2W3 caused increases in [Ca2+]i and relaxation of ASM cells. Of note, OR2W3-evoked [Ca2+]i mobilization and ASM relaxation required Ca2+ flux through the store-operated calcium entry (SOCE) pathway and accompanied plasma membrane depolarization. This chemosensory odorant receptor response was not mediated by adenylyl cyclase (AC)/cyclic nucleotide-gated (CNG) channels or by protein kinase A (PKA) activity. Instead, ASM olfactory responses to the monoterpene nerol were predominated by the activity of Ca2+-activated chloride channels (TMEM16A), including the cystic fibrosis transmembrane conductance regulator (CFTR) expressed on endo(sarco)plasmic reticulum. These findings demonstrate compartmentalization of Ca2+ signals dictates the odorant receptor OR2W3-induced ASM relaxation and identify a previously unrecognized E-C coupling mechanism that could be exploited in the development of therapeutics to treat obstructive lung diseases.


Asunto(s)
Anoctamina-1/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Odorantes/metabolismo , Adenilil Ciclasas/metabolismo , Bronquios/metabolismo , Calcio/metabolismo , Células Cultivadas , Humanos , Pulmón/metabolismo , Contracción Muscular/fisiología , Relajación Muscular , Miocitos del Músculo Liso/metabolismo , Receptores Odorantes/genética
5.
FASEB J ; 34(2): 2882-2895, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31908022

RESUMEN

Glucocorticoids are widely used for the suppression of inflammation, but evidence is growing that they can have rapid, non-genomic actions that have been unappreciated. Diverse cell signaling effects have been reported for glucocorticoids, leading us to hypothesize that glucocorticoids alone can swiftly increase the 3',5'-cyclic adenosine monophosphate (cAMP) production. We found that prednisone, fluticasone, budesonide, and progesterone each increased cAMP levels within 3 minutes without phosphodiesterase inhibitors by measuring real-time cAMP dynamics using the cAMP difference detector in situ assay in a variety of immortalized cell lines and primary human airway smooth muscle (HASM) cells. A membrane- impermeable glucocorticoid showed similarly rapid stimulation of cAMP, implying that responses are initiated at the cell surface. siRNA knockdown of Gαs virtually eliminated glucocorticoid-stimulated cAMP responses, suggesting that these drugs activate the cAMP production via a G protein-coupled receptor. Estradiol had small effects on cAMP levels but G protein estrogen receptor antagonists had little effect on responses to any of the glucocorticoids tested. The genomic and non-genomic actions of budesonide were analyzed by RNA-Seq analysis of 24 hours treated HASM, with and without knockdown of Gαs . A 140-gene budesonide signature was identified, of which 48 genes represent a non-genomic signature that requires Gαs signaling. Collectively, this non-genomic cAMP signaling modality contributes to one-third of the gene expression changes induced by glucocorticoid treatment and shifts the view of how this important class of drugs exerts its effects.


Asunto(s)
Cromograninas/metabolismo , AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Miocitos del Músculo Liso/metabolismo , Sistema Respiratorio/metabolismo , Sistemas de Mensajero Secundario/efectos de los fármacos , Línea Celular Transformada , Cromograninas/genética , AMP Cíclico/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Técnicas de Silenciamiento del Gen , Humanos , Miocitos del Músculo Liso/patología , Sistema Respiratorio/patología , Sistemas de Mensajero Secundario/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L345-L355, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31747297

RESUMEN

The nongenomic mechanisms by which glucocorticoids modulate ß2 agonist-induced-bronchodilation remain elusive. Our studies aimed to elucidate mechanisms mediating the beneficial effects of glucocorticoids on agonist-induced bronchodilation. Utilizing human precision-cut lung slices (hPCLS), we measured bronchodilation to formoterol, prostaglandin E2 (PGE2), cholera toxin (CTX), or forskolin in the presence and absence of budesonide. Using cultured human airway smooth muscle (HASM), intracellular cAMP was measured in live cells following exposure to formoterol, PGE2, or forskolin in the presence or absence of budesonide. We showed that simultaneous budesonide administration amplified formoterol-induced bronchodilation and attenuated agonist-induced phosphorylation of myosin light chain, a necessary signaling event mediating force generation. In parallel studies, cAMP levels were augmented by simultaneous exposure of HASM cells to formoterol and budesonide. Budesonide, fluticasone, and prednisone alone rapidly increased cAMP levels, but steroids alone had little effect on bronchodilation in hPCLS. Bronchodilation induced by PGE2, CTX, or forskolin was also augmented by simultaneous exposure to budesonide in hPCLS. Furthermore, HASM cells expressed membrane-bound glucocorticoid receptors that failed to translocate with glucocorticoid stimulation and that potentially mediated the rapid effects of steroids on ß2 agonist-induced bronchodilation. Knockdown of glucocorticoid receptor-α had little effect on budesonide-induced and steroid-dependent augmentation of formoterol-induced cAMP generation in HASM. Collectively, these studies suggest that glucocorticoids amplify cAMP-dependent bronchodilation by directly increasing cAMP levels. These studies identify a molecular mechanism by which the combination of glucocorticoids and ß2 agonists may augment bronchodilation in diseases such as asthma or chronic obstructive pulmonary disease.


Asunto(s)
Bronquios/fisiología , Broncodilatadores/farmacología , Budesonida/farmacología , AMP Cíclico/biosíntesis , Músculo Liso/fisiología , Bronquios/efectos de los fármacos , Carbacol/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Toxina del Cólera/farmacología , Colforsina/farmacología , Dinoprostona/farmacología , Fluticasona/farmacología , Fumarato de Formoterol/farmacología , Humanos , Músculo Liso/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Fosforilación/efectos de los fármacos , Prednisona/farmacología , Receptores de Glucocorticoides/metabolismo
7.
Am J Respir Cell Mol Biol ; 58(4): 530-541, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29262264

RESUMEN

Two cAMP signaling compartments centered on adenylyl cyclase (AC) exist in human airway smooth muscle (HASM) cells, one containing ß2-adrenergic receptor AC6 and another containing E prostanoid receptor AC2. We hypothesized that different PDE isozymes selectively regulate cAMP signaling in each compartment. According to RNA-sequencing data, 18 of 24 PDE genes were expressed in primary HASM cells derived from age- and sex-matched donors with and without asthma. PDE8A was the third most abundant of the cAMP-degrading PDE genes, after PDE4A and PDE1A. Knockdown of PDE8A using shRNA evoked twofold greater cAMP responses to 1 µM forskolin in the presence of 3-isobutyl-1-methylxanthine. Overexpression of AC2 did not alter this response, but overexpression of AC6 increased cAMP responses an additional 80%. We examined cAMP dynamics in live HASM cells using a fluorescence sensor. PF-04957325, a PDE8-selective inhibitor, increased basal cAMP concentrations by itself, indicating a significant basal level of cAMP synthesis. In the presence of an AC inhibitor to reduce basal signaling, PF-04957325 accelerated cAMP production and increased the inhibition of cell proliferation induced by isoproterenol, but it had no effect on cAMP concentrations or cell proliferation regulated by prostaglandin E2. Lipid raft fractionation of HASM cells revealed PDE8A immunoreactivity in buoyant fractions containing caveolin-1 and AC5/6 immunoreactivity. Thus, PDE8 is expressed in lipid rafts of HASM cells, where it specifically regulates ß2-adrenergic receptor AC6 signaling without effects on signaling by the E prostanoid receptors 2/4-AC2 complex. In airway diseases such as asthma and chronic obstructive pulmonary disease, PDE8 may represent a novel therapeutic target to modulate HASM responsiveness and airway remodeling.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Adenilil Ciclasas/metabolismo , Asma/enzimología , AMP Cíclico/metabolismo , Músculo Liso/enzimología , Miocitos del Músculo Liso/enzimología , Receptores Adrenérgicos beta 2/metabolismo , Sistema Respiratorio/enzimología , 3',5'-AMP Cíclico Fosfodiesterasas/genética , Adenilil Ciclasas/genética , Remodelación de las Vías Aéreas (Respiratorias) , Asma/genética , Asma/patología , Asma/fisiopatología , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Humanos , Microdominios de Membrana/enzimología , Microdominios de Membrana/patología , Músculo Liso/patología , Músculo Liso/fisiopatología , Miocitos del Músculo Liso/patología , Receptores Adrenérgicos beta 2/genética , Sistema Respiratorio/patología , Sistema Respiratorio/fisiopatología , Sistemas de Mensajero Secundario , Factores de Tiempo
8.
Nat Biomed Eng ; 2(2): 124-137, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-31015629

RESUMEN

As cells with aberrant force-generating phenotypes can directly lead to disease, cellular force-generation mechanisms are high-value targets for new therapies. Here, we show that single-cell force sensors embedded in elastomers enable single-cell force measurements with ~100-fold improvement in throughput than was previously possible. The microtechnology is scalable and seamlessly integrates with the multi-well plate format, enabling highly parallelized time-course studies. In this regard, we show that airway smooth muscle cells isolated from fatally asthmatic patients have innately greater and faster force-generation capacity in response to stimulation than healthy control cells. By simultaneously tracing agonist-induced calcium flux and contractility in the same cell, we show that the calcium level is ultimately a poor quantitative predictor of cellular force generation. Finally, by quantifying phagocytic forces in thousands of individual human macrophages, we show that force initiation is a digital response (rather than a proportional one) to the proper immunogen. By combining mechanobiology at the single-cell level with high-throughput capabilities, this microtechnology can support drug-discovery efforts for clinical conditions associated with aberrant cellular force generation.


Asunto(s)
Elastómeros/química , Análisis de la Célula Individual/métodos , Asma/patología , Diferenciación Celular , Células Cultivadas , Colorantes Fluorescentes/química , Fumarato de Formoterol/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Microscopía Fluorescente , Contracción Miocárdica/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Fagocitosis/efectos de los fármacos
9.
Int J Mol Sci ; 18(12)2017 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-29186841

RESUMEN

Radiation therapy for the treatment of thoracic malignancies has improved significantly by directing of the proton beam in higher doses on the targeted tumor while normal tissues around the tumor receive much lower doses. Nevertheless, exposure of normal tissues to protons is known to pose a substantial risk in long-term survivors, as confirmed by our work in space-relevant exposures of murine lungs to proton radiation. Thus, radioprotective strategies are being sought. We established that LGM2605 is a potent protector from radiation-induced lung toxicity and aimed in the current study to extend the initial findings of space-relevant, proton radiation-associated late lung damage in mice by looking at acute changes in human lung. We used an ex vivo model of organ culture where tissue slices of donor living human lung were kept in culture and exposed to proton radiation. We exposed donor human lung precision-cut lung sections (huPCLS), pretreated with LGM2605, to 4 Gy proton radiation and evaluated them 30 min and 24 h later for gene expression changes relevant to inflammation, oxidative stress, and cell cycle arrest, and determined radiation-induced senescence, inflammation, and oxidative tissue damage. We identified an LGM2605-mediated reduction of proton radiation-induced cellular senescence and associated cell cycle changes, an associated proinflammatory phenotype, and associated oxidative tissue damage. This is a first report on the effects of proton radiation and of the radioprotective properties of LGM2605 on human lung.


Asunto(s)
Antiinflamatorios/uso terapéutico , Butileno Glicoles/uso terapéutico , Glucósidos/uso terapéutico , Protones/efectos adversos , Neumonitis por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Antiinflamatorios/farmacología , Butileno Glicoles/farmacología , Puntos de Control del Ciclo Celular , Senescencia Celular , Glucósidos/farmacología , Humanos , Pulmón/efectos de los fármacos , Pulmón/efectos de la radiación , Estrés Oxidativo , Neumonitis por Radiación/tratamiento farmacológico , Neumonitis por Radiación/etiología , Protectores contra Radiación/farmacología
10.
Am J Respir Cell Mol Biol ; 55(4): 542-553, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27149505

RESUMEN

Formaldehyde, a common indoor air pollutant, exacerbates asthma and synergizes with allergen to induce airway hyperresponsiveness (AHR) in animal models. The mechanisms mediating formaldehyde-induced AHR remain poorly understood. We posit that formaldehyde modulates agonist-induced contractile response of human airway smooth muscle (HASM) cells to elicit AHR. HASM cells were exposed to formaldehyde or vehicle and agonist-induced intracellular Ca2+ ([Ca2+]i) and myosin light-chain phosphatase (MYPT1) phosphorylation were determined. Air-liquid interface-differentiated human bronchial epithelial (HBE) cells were exposed to formaldehyde or vehicle and cocultured with HASM cells. Agonist-induced [Ca2+]i and MYPT1 phosphorylation were determined in the cocultured HASM cells. Precision-cut human lung slices were exposed to PBS or varying concentrations of formaldehyde, and then carbachol-induced airway narrowing was determined 24 hours after exposure. HASM cells were transfected with nontargeting or nuclear factor erythroid-derived 2, like 2 (Nrf-2)-targeting small interfering RNA and exposed to formaldehyde or vehicle, followed by determination of antioxidant response (quinone oxido-reductase 1 and thioredoxin 1) and basal and agonist-induced MYPT1 phosphorylation. Formaldehyde enhanced the basal Rho-kinase activity and MYPT1 phosphorylation with little effect on agonist-induced [Ca2+]i in HASM cells. Formaldehyde induced Nrf-2-dependent antioxidant response in HASM cells, although the MYPT1 phosphorylation was independent of Nrf-2 induction. Although HBE cells exposed to formaldehyde had little effect on agonist-induced [Ca2+]i or MYPT1 phosphorylation in cocultured HASM cells, formaldehyde enhanced carbachol-induced airway responsiveness in precision-cut human lung slices. In conclusion, formaldehyde induces phosphorylation of the regulatory subunit of MYPT1, independent of formaldehyde-induced Nrf-2 activation in HASM cells. The findings suggest that the Rho kinase-dependent Ca2+ sensitization pathway plays a role in formaldehyde-induced AHR.

11.
Am J Respir Crit Care Med ; 189(4): 437-48, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24417465

RESUMEN

RATIONALE: Respiratory viral infections can result in the establishment of chronic lung diseases. Understanding the early innate immune mechanisms that participate in the development of chronic postviral lung disease may reveal new targets for therapeutic intervention. The intracellular viral sensor protein melanoma differentiation-associated protein 5 (MDA5) sustains the acute immune response to Sendai virus, a mouse pathogen that causes chronic lung inflammation, but its role in the development of postviral chronic lung disease is unknown. OBJECTIVES: To establish the role of MDA5 in the development of chronic lung disease. METHODS: MDA5-deficient or control mice were infected with Sendai virus. The acute inflammatory response was evaluated by profiling chemokine and cytokine expression and by characterizing the composition of the cellular infiltrate. The impact of MDA5 on chronic lung pathology and function was evaluated through histological studies, degree of oxygen saturation, and responsiveness to carbachol. MEASUREMENTS AND MAIN RESULTS: MDA5 deficiency resulted in normal virus replication and in a distinct profile of chemokines and cytokines that associated with acute lung neutropenia and enhanced accumulation of alternatively activated macrophages. Diminished expression of neutrophil-recruiting chemokines was also observed in cells infected with influenza virus, suggesting a key role of MDA5 in driving the early accumulation of neutrophils at the infection site. The biased acute inflammatory response of MDA5-deficient mice led to an enhanced chronic lung inflammation, epithelial cell hyperplasia, airway hyperreactivity, and diminished blood oxygen saturation. CONCLUSIONS: MDA5 modulates the development of chronic lung inflammation by regulating the early inflammatory response in the lung.


Asunto(s)
ARN Helicasas DEAD-box/deficiencia , Neumonía Viral/enzimología , Infecciones por Respirovirus/enzimología , Virus Sendai , Animales , Biomarcadores/metabolismo , Líquido del Lavado Bronquioalveolar/química , Quimiocinas/metabolismo , Enfermedad Crónica , Citocinas/metabolismo , Citometría de Flujo , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1 , Pulmón/enzimología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Neumonía Viral/inmunología , Neumonía Viral/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/patología
12.
Pulm Pharmacol Ther ; 26(2): 229-38, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23154072

RESUMEN

The pharmacological and airways relaxant profiles of PL-3994 (Hept-cyclo(Cys-His-Phe-d-Ala-Gly-Arg-d-Nle-Asp-Arg-Ile-Ser-Cys)-Tyr-[Arg mimetic]-NH(2)), a novel natriuretic peptide receptor-A (NPR-A) agonist, were evaluated. PL-3994, a full agonist, has high affinity for recombinant human (h), dog, or rat NPR-As (K(i)s of 1, 41, and 10 nm, respectively), and produced concentration-dependent cGMP generation in human, dog and rat NPR-As (respective EC(50)s of 2, 3 and 14 nm). PL-3994 has a K(i) of 7 nm for hNPR-C but was without effect on cGMP generation in hNPR-B. PL-3994 (1 µm) was without significant effect against 75 diverse molecular targets. PL-3994 or BNP, a natural NPR ligand, produced concentration-dependent relaxation of pre-contracted guinea-pig trachea (IC(50)s of 42.7 and 10.7 nm, respectively). PL-3994, and also BNP, (0.1 nm-100 µm) elicited a potent, concentration-dependent but small relaxation of pre-contracted human precision-cut lung slices (hPCLS). Intratracheal PL-3994 (1-1000 µg/kg) produced a dose-dependent inhibition of the bronchoconstrictor response evoked by aerosolized methacholine, but was without significant effect on cardiovascular parameters. PL-3994 was resistant to degradation by human neutral endopeptidase (hNEP) (92% remaining after 2 h), whereas the natural ligands, ANP and CNP, were rapidly metabolized (≤1% remaining after 2 h). PL-3994 is a potent, selective NPR agonist, resistant to NEP, with relaxant effects in guinea-pig and human airway smooth muscle systems. PL-3994 has the profile predictive of longer clinical bronchodilator activity than observed previously with ANP, and suggests its potential utility in the treatment of asthma, in addition to being a useful research tool to evaluate NPR biology.


Asunto(s)
Broncodilatadores/farmacología , Neprilisina/metabolismo , Péptidos Cíclicos/farmacología , Piperazinas/farmacología , Receptores del Factor Natriurético Atrial/agonistas , Animales , Perros , Relación Dosis-Respuesta a Droga , Cobayas , Células HEK293 , Humanos , Masculino , Péptido Natriurético Encefálico/farmacología , Péptidos Cíclicos/metabolismo , Ratas , Ratas Wistar , Tráquea/efectos de los fármacos , Tráquea/fisiología
13.
Biochim Biophys Acta ; 1822(10): 1638-42, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22771498

RESUMEN

Airway diseases such as asthma, emphysema, and chronic bronchitis are, in part, characterized by reversible airflow obstruction and inflammation. In severe disease, marked decreases in lung function are associated with airway smooth muscle proliferation and airway neutrophilia. Inhaled glucocorticoids attenuate increased airflow obstruction and airway inflammation that occur, in part, due to increased smooth muscle migration and proliferation, as well as the airway neutrophilia. Glucocorticoids, however, have adverse side effects and, in some patients, are ineffective despite high doses. Recent research has explored the effects of non-traditional steroids on attenuation of inflammation associated with airway diseases. These non-traditional steroids have improved side effect profiles in comparison to glucocorticoid therapy. Our studies assessed effects of dehydroepiandrosterone-3-sulfate (DHEA-S) on migration of both human peripheral blood neutrophils (PMN) and human airway smooth muscle cells (HASM). DHEA-S dose-dependently inhibited chemotaxis of PMN and HASM while having no effect on the phosphorylation levels of Akt, ERK1/2, p38 MAPK or PKC, canonical positive regulators of cell migration. These studies demonstrate direct effects of DHEA-S on cell migration, thereby suggesting that DHEA-S may attenuate airway inflammation and cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Sulfato de Deshidroepiandrosterona/farmacología , Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Células Cultivadas , Glucocorticoides/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Músculo Liso/citología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Curr Opin Pharmacol ; 12(3): 287-92, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22365729

RESUMEN

COPD represents a major respiratory disorder, causing significant morbidity and mortality throughout the world. While therapies exist for COPD, they are not always effective, and many patients experience exacerbations and morbidity despite current therapies. Study of the molecular mechanisms involved in the underlying physiological manifestations of COPD has yielded multiple new targets for therapeutic intervention. In this review, we discuss signaling pathways involved in COPD pathogenesis and review clinical studies of p38 MAPK inhibitors, TNFα inhibitors, and IKK2 inhibitors as potential COPD therapies.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Quinasa I-kappa B/antagonistas & inhibidores , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/uso terapéutico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Humanos , Quinasa I-kappa B/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/inmunología , Sistema Respiratorio/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Immunol Rev ; 242(1): 178-85, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21682745

RESUMEN

Airway smooth muscle (ASM) manifests a hyperresponsive phenotype in airway disorders such as asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis. Current evidence also suggests that ASM modulates immune responses by secreting mediators and expressing cell surface molecules. Such processes amplify or dampen inflammation by inflammatory cells in the airways or by altering cellular responses to viruses, bacteria, or pathogens known to exacerbate airways diseases.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Células Epiteliales/inmunología , Hipersensibilidad/inmunología , Inmunidad Innata , Inmunomodulación , Músculo Liso/inmunología , Sistema Respiratorio/inmunología , Alérgenos/inmunología , Animales , Asma/inmunología , Asma/fisiopatología , Niño , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Células Epiteliales/metabolismo , Humanos , Hipersensibilidad/fisiopatología , Inflamación/inmunología , Interleucina-33 , Interleucinas/biosíntesis , Interleucinas/inmunología , Ratones , Ratones Transgénicos , Músculo Liso/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Sistema Respiratorio/fisiopatología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
16.
J Immunol ; 182(3): 1404-10, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19155487

RESUMEN

The primary function of IL-7 is to promote maturation and survival of T cells. Through microarray expression analysis, we previously observed that human blood eosinophils express mRNA for IL-7R alpha (CD127) and its common gamma chain (CD132). The purpose of this study was to determine whether eosinophils have functional IL-7 receptors and to assess the potential contribution of IL-7 to eosinophilic airway inflammation by evaluating its presence in bronchoalveolar lavage (BAL) fluid of subjects with atopic asthma before and after segmental bronchoprovocation with allergen. Immunoblot analysis revealed that CD127 is present in highly purified human blood eosinophils. Furthermore, eosinophils responded to IL-7 with phosphorylation of STAT5, up-regulation of the activation marker CD69, and prolonged survival. Neutralization of GM-CSF but not IL-5 significantly blunted these functional responses, suggesting that IL-7 mediates its effects by promoting eosinophil release of autologous GM-CSF. Notably, the suppressive effect of anti-GM-CSF on STAT5 phosphorylation occurred within 10 min of eosinophil exposure to IL-7. Thus, IL-7 likely activates eosinophil release of preformed rather than newly synthesized GM-CSF. The biological relevance of IL-7 to eosinophilia in vivo was implicated in a study of airway allergen challenge in patients with allergic asthma. IL-7 concentrations in BAL fluid increased significantly 48 h after segmental allergen challenge and were highly correlated with BAL eosinophils (r = 0.7, p < 0.001). In conclusion, the airway response to allergen is associated with the generation of IL-7, which may contribute to airway inflammation by promoting enhanced eosinophil activation and survival. Activation of eosinophils is a novel function for IL-7.


Asunto(s)
Alérgenos/administración & dosificación , Asma/inmunología , Asma/patología , Eosinofilia/inmunología , Eosinofilia/patología , Mediadores de Inflamación/fisiología , Interleucina-7/fisiología , Adulto , Anticuerpos Monoclonales , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Asma/metabolismo , Comunicación Autocrina/inmunología , Pruebas de Provocación Bronquial/métodos , Supervivencia Celular/inmunología , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Eosinofilia/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Interleucina-7/antagonistas & inhibidores , Interleucina-7/inmunología , Lectinas Tipo C , Masculino , Receptores de Interleucina-7/metabolismo , Receptores de Interleucina-7/fisiología , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA