Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Respir Res ; 25(1): 137, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38521949

RESUMO

Publications utilizing precision cut lung slices (PCLS) steadily increased from the 1970's, with a significant increase in 2010, to tripling by 2023. PCLS have been used to study a vast array of pulmonary diseases and exposures to pathogens and toxicants to understand pathogenesis of disease but also to examine basic cellular mechanisms that underly lung biology. This Special Issue will highlight new, exciting, and novel research using PCLS, while acknowledging the substantial fund of knowledge that has been gained using this platform.


Assuntos
Pneumopatias , Pulmão , Humanos , Pulmão/patologia , Pneumopatias/diagnóstico , Pneumopatias/tratamento farmacológico , Pneumopatias/patologia , Técnicas de Cultura de Órgãos
2.
Am J Respir Cell Mol Biol ; 68(4): 417-429, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36662576

RESUMO

TAS2Rs (bitter taste receptors) are GPCRs (G protein-coupled receptors) expressed on human airway smooth muscle (HASM) cells; when activated by receptor agonists they evoke marked airway relaxation. In both taste and HASM cells, TAS2Rs activate a canonical Gßγ-mediated stimulation of Ca2+ release from intracellular stores by activation of PLCß (phospholipase Cß). Alone, this [Ca2+]i signaling does not readily account for relaxation, particularly since bronchoconstrictive agonists acting at Gq-coupled receptors also increase [Ca2+]i. We established that TAS2R14 activation in HASM promotes relaxation through F-actin (filamentous actin) severing. This destabilization of actin was from agonist-promoted activation (dephosphorylation) of cofilin, which was pertussis toxin sensitive. Cofilin dephosphorylation was due to TAS2R-mediated deactivation of LIM domain kinase. The link between early receptor action and the distal cofilin dephosphorylation was found to be the polarity protein partitioning defective 3 (Par3), a known binding partner with PLCß that inhibits LIM kinase. The physiologic relevance of this pathway was assessed using knock-downs of cofilin and Par3 in HASM cells and in human precision-cut lung slices. Relaxation by TAS2R14 agonists was ablated with knock-down of either protein as assessed by magnetic twisting cytometry in isolated cells or intact airways in the slices. Blocking [Ca2+]i release by TAS2R14 inhibited agonist-promoted cofilin dephosphorylation, confirming a role for [Ca2+]i in actin-modifying pathways. These results further elucidate the mechanistic basis of TAS2R-mediated HASM relaxation and point toward nodal points that may act as asthma or chronic obstructive pulmonary disease response modifiers or additional targets for novel bronchodilators.


Assuntos
Actinas , Asma , Receptores Acoplados a Proteínas G , Humanos , Actinas/metabolismo , Asma/metabolismo , Quinases Lim/metabolismo , Pulmão/metabolismo , Relaxamento Muscular/fisiologia , Receptores Acoplados a Proteínas G/metabolismo
3.
Am J Respir Cell Mol Biol ; 69(2): 172-181, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37098126

RESUMO

Rhinoviruses (RVs) evoke as many as 85% of acute asthma exacerbations in children and 50% in adults and can induce airway hyperresponsiveness and decrease efficacy of current therapeutics to provide symptom relief. Using human precision-cut lung slices (hPCLSs), primary human air-liquid interface-differentiated airway epithelial cells (HAECs), and human airway smooth muscle (HASM) as preclinical experimental models, we demonstrated that RV-C15 attenuates agonist-induced bronchodilation. Specifically, airway relaxation to formoterol and cholera toxin, but not forskolin (Fsk), was attenuated following hPCLS exposure to RV-C15. In isolated HASM cells, exposure to conditioned media from RV-exposed HAECs decreased cellular relaxation in response to isoproterenol and prostaglandin E2, but not Fsk. Additionally, cAMP generation elicited by formoterol and isoproterenol, but not Fsk, was attenuated following HASM exposure to RV-C15-conditioned HAEC media. HASM exposure to RV-C15-conditioned HAEC media modulated expression of components of relaxation pathways, specifically GNAI1 and GRK2. Strikingly, similar to exposure to intact RV-C15, hPCLS exposed to UV-inactivated RV-C15 showed markedly attenuated airway relaxation in response to formoterol, suggesting that the mechanism(s) of RV-C15-mediated loss of bronchodilation is independent of virus replication pathways. Further studies are warranted to identify soluble factor(s) regulating the epithelial-driven smooth muscle loss of ß2-adrenergic receptor function.


Assuntos
Infecções por Enterovirus , Rhinovirus , Adulto , Criança , Humanos , Rhinovirus/fisiologia , Isoproterenol/farmacologia , Músculo Liso/metabolismo , Pulmão/metabolismo , Fumarato de Formoterol/farmacologia , Fumarato de Formoterol/metabolismo , Colforsina/farmacologia , Relaxamento Muscular
4.
Respir Res ; 24(1): 238, 2023 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-37773065

RESUMO

BACKGROUND: Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY: While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the ß2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS: Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.


Assuntos
Obstrução das Vias Respiratórias , Asma , Criança , Adulto , Humanos , Rhinovirus/fisiologia , Remodelação das Vias Aéreas , Células Endoteliais/metabolismo , Broncodilatadores , Asma/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 323(2): L142-L151, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35787178

RESUMO

Epinephrine (EPI), an endogenous catecholamine involved in the body's fight-or-flight responses to stress, activates α1-adrenergic receptors (α1ARs) expressed on various organs to evoke a wide range of physiological functions, including vasoconstriction. In the smooth muscle of human bronchi, however, the functional role of EPI on α1ARs remains controversial. Classically, evidence suggests that EPI promotes bronchodilation by stimulating ß2-adrenergic receptors (ß2ARs). Conventionally, the selective ß2AR agonism of EPI was thought to be, in part, due to a predominance of ß2ARs and/or a sparse, or lack of α1AR activity in human airway smooth muscle (HASM) cells. Surprisingly, we find that HASM cells express a high abundance of ADRA1B (the α1AR subtype B) and identify a spontaneous "switch-like" activation of α1ARs that evokes intracellular calcium, myosin light chain phosphorylation, and HASM cell shortening. The switch-like responses, and related EPI-induced biochemical and mechanical signals, emerged upon pharmacological inhibition of ß2ARs and/or under experimental conditions that induce ß2AR tachyphylaxis. EPI-induced procontractile effects were abrogated by an α1AR antagonist, doxazosin mesylate (DM). These data collectively uncover a previously unrecognized feed-forward mechanism driving bronchospasm via two distinct classes of G protein-coupled receptors (GPCRs) and provide a basis for reexamining α1AR inhibition for the management of stress/exercise-induced asthma and/or ß2-agonist insensitivity in patients with difficult-to-control, disease subtypes.


Assuntos
Miócitos de Músculo Liso , Receptores Adrenérgicos beta 2 , Agonistas Adrenérgicos beta , Brônquios , Broncodilatadores/farmacologia , Epinefrina/farmacologia , Humanos , Músculo Liso , Receptores Adrenérgicos alfa 1
6.
Am J Respir Cell Mol Biol ; 62(1): 43-48, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31340135

RESUMO

The soluble guanylyl cyclase (sGC)-cyclic guanosine monophosphate signaling pathway evokes vascular smooth muscle relaxation; whether this pathway mediates airway smooth muscle relaxation remains controversial. We posit that sGC activators are equi-effective as ß-agonists in reversing contractile agonist-induced airway smooth muscle shortening. To provide clarity, we tested the efficacy of sGC stimulator and activator drugs, BAY 41-2272 and BAY 60-2270, respectively, in reversing bronchoconstriction of human small airways using human precision-cut lung slices (hPCLS). Both BAY drugs reversed carbachol-induced bronchoconstriction to a maximal degree comparable to that of formoterol. Moreover, the sGC drugs remained effective bronchodilators despite formoterol-induced desensitization of the airways. Analysis of the hPCLS after their activation by sGC or ß2-adrenergic receptor agonist showed distinct cyclic nucleotide accumulation in the hPCLS. Collectively, these data suggest that cAMP and cyclic guanosine monophosphate pathways are equi-effective for reversing carbachol-induced bronchoconstriction in the human airway via separate and distinct second messenger pathways. This should open the door for future studies to test whether sGC-targeted drugs alone or in combination can serve as effective bronchodilators in asthma and chronic obstructive pulmonary disease.


Assuntos
Broncodilatadores/farmacologia , Músculo Liso/efeitos dos fármacos , Sistema Respiratório/efeitos dos fármacos , Guanilil Ciclase Solúvel/metabolismo , Asma/tratamento farmacológico , Asma/metabolismo , Broncoconstrição/efeitos dos fármacos , GMP Cíclico/metabolismo , Humanos , Contração Muscular/efeitos dos fármacos , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Sistema Respiratório/metabolismo , Transdução de Sinais/efeitos dos fármacos , Traqueia/efeitos dos fármacos , Traqueia/metabolismo
7.
Am J Respir Cell Mol Biol ; 62(3): 310-318, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31533004

RESUMO

Rhinovirus (RV) exposure evokes exacerbations of asthma that markedly impact morbidity and mortality worldwide. The mechanisms by which RV induces airway hyperresponsiveness (AHR) or by which specific RV serotypes differentially evoke AHR remain unknown. We posit that RV infection evokes AHR and inflammatory mediator release, which correlate with degrees of RV infection. Furthermore, we posit that rhinovirus C-induced AHR requires paracrine or autocrine mediator release from epithelium that modulates agonist-induced calcium mobilization in human airway smooth muscle. In these studies, we used an ex vivo model to measure bronchoconstriction and mediator release from infected airways in human precision cut lung slices to understand how RV exposure alters airway constriction. We found that rhinovirus C15 (RV-C15) infection augmented carbachol-induced airway narrowing and significantly increased release of IP-10 (IFN-γ-induced protein 10) and MIP-1ß (macrophage inflammatory protein-1ß) but not IL-6. RV-C15 infection of human airway epithelial cells augmented agonist-induced intracellular calcium flux and phosphorylation of myosin light chain in co-cultured human airway smooth muscle to carbachol, but not after histamine stimulation. Our data suggest that RV-C15-induced structural cell inflammatory responses are associated with viral load but that inflammatory responses and alterations in agonist-mediated constriction of human small airways are uncoupled from viral load of the tissue.


Assuntos
Sinalização do Cálcio , Infecções por Enterovirus/fisiopatologia , Enterovirus/fisiologia , Músculo Liso/virologia , Hipersensibilidade Respiratória/etiologia , Asma/virologia , Carbacol/farmacologia , Células Cultivadas , Quimiocina CXCL10/metabolismo , Enterovirus/genética , Enterovirus/isolamento & purificação , Infecções por Enterovirus/virologia , Histamina/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Contração Muscular/efeitos dos fármacos , Músculo Liso/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Viral/análise , Hipersensibilidade Respiratória/virologia , Carga Viral
8.
Proc Natl Acad Sci U S A ; 113(17): E2355-62, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27071111

RESUMO

Asthma is defined by airway inflammation and hyperresponsiveness, and contributes to morbidity and mortality worldwide. Although bronchodilation is a cornerstone of treatment, current bronchodilators become ineffective with worsening asthma severity. We investigated an alternative pathway that involves activating the airway smooth muscle enzyme, soluble guanylate cyclase (sGC). Activating sGC by its natural stimulant nitric oxide (NO), or by pharmacologic sGC agonists BAY 41-2272 and BAY 60-2770, triggered bronchodilation in normal human lung slices and in mouse airways. Both BAY 41-2272 and BAY 60-2770 reversed airway hyperresponsiveness in mice with allergic asthma and restored normal lung function. The sGC from mouse asthmatic lungs displayed three hallmarks of oxidative damage that render it NO-insensitive, and identical changes to sGC occurred in human lung slices or in human airway smooth muscle cells when given chronic NO exposure to mimic the high NO in asthmatic lung. Our findings show how allergic inflammation in asthma may impede NO-based bronchodilation, and reveal that pharmacologic sGC agonists can achieve bronchodilation despite this loss.


Assuntos
Antiasmáticos/farmacologia , Asma/tratamento farmacológico , Benzoatos/farmacologia , Compostos de Bifenilo/farmacologia , Broncodilatadores/farmacologia , Guanilato Ciclase/efeitos dos fármacos , Hidrocarbonetos Fluorados/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Animais , Antiasmáticos/uso terapêutico , Asma/enzimologia , Asma/fisiopatologia , Benzoatos/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Hiper-Reatividade Brônquica/tratamento farmacológico , Hiper-Reatividade Brônquica/enzimologia , Broncodilatadores/uso terapêutico , Técnicas de Cocultura , GMP Cíclico/metabolismo , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Hidrocarbonetos Fluorados/uso terapêutico , Pulmão/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/efeitos dos fármacos , Músculo Liso/enzimologia , Óxido Nítrico/farmacologia , Pirazóis/uso terapêutico , Piridinas/uso terapêutico , Solubilidade , Traqueia/efeitos dos fármacos
9.
Am J Respir Cell Mol Biol ; 58(1): 89-98, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28853915

RESUMO

A cardinal feature of asthma is airway hyperresponsiveness (AHR) to spasmogens, many of which activate G protein-coupled receptors (GPCRs) on airway smooth muscle (ASM) cells. Asthma subtypes associated with allergy are characterized by eosinophilic inflammation in the lung due to the type 2 immune response to allergens and proinflammatory mediators that promote AHR. The degree to which intrinsic abnormalities of ASM contribute to this phenotype remains unknown. The regulators of G protein signaling (RGS) proteins are a large group of intracellular proteins that inhibit GPCR signaling pathways. RGS2- and RGS5-deficient mice develop AHR spontaneously. Although RGS4 is upregulated in ASM from patients with severe asthma, the effects of increased RGS4 expression on AHR in vivo are unknown. Here, we examined the impact of forced RGS4 overexpression in lung on AHR using transgenic (Tg) mice. Tg RGS4 was expressed in bronchial epithelium and ASM in vivo, and protein expression in lung was increased at least 4-fold in Tg mice compared with wild-type (WT) mice. Lung slices from Tg mice contracted less in response to the m3 muscarinic receptor agonist methacholine compared with the WT, although airway resistance in live, unchallenged mice of both strains was similar. Tg mice were partially protected against AHR induced by fungal allergen challenge due to weakened contraction signaling in ASM and reduced type 2 cytokine (IL-5 and IL-13) levels in Tg mice compared with the WT. These results provide support for the hypothesis that increasing RGS4 expression and/or function could be a viable therapeutic strategy for asthma.


Assuntos
Asma/imunologia , Brônquios/imunologia , Regulação da Expressão Gênica/imunologia , Pulmão/imunologia , Proteínas RGS/imunologia , Mucosa Respiratória/imunologia , Animais , Asma/genética , Asma/patologia , Brônquios/patologia , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Proteínas RGS/genética , Mucosa Respiratória/patologia
10.
Am J Respir Cell Mol Biol ; 58(4): 530-541, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29262264

RESUMO

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.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Adenilil Ciclases/metabolismo , Asma/enzimologia , AMP Cíclico/metabolismo , Músculo Liso/enzimologia , Miócitos de Músculo Liso/enzimologia , Receptores Adrenérgicos beta 2/metabolismo , Sistema Respiratório/enzimologia , 3',5'-AMP Cíclico Fosfodiesterases/genética , Adenilil Ciclases/genética , Remodelação das Vias Aéreas , Asma/genética , Asma/patologia , Asma/fisiopatologia , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Humanos , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/patologia , Músculo Liso/patologia , Músculo Liso/fisiopatologia , Miócitos de Músculo Liso/patologia , Receptores Adrenérgicos beta 2/genética , Sistema Respiratório/patologia , Sistema Respiratório/fisiopatologia , Sistemas do Segundo Mensageiro , Fatores de Tempo
11.
Respir Res ; 19(1): 208, 2018 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-30373568

RESUMO

Rhinovirus (RV) exposure has been implicated in childhood development of wheeze evoking asthma and exacerbations of underlying airways disease. Studies such as the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) and Childhood Origins of ASThma (COAST) have identified RV as a pathogen inducing severe respiratory disease. RVs also modulate airway hyperresponsiveness (AHR), a key characteristic of such diseases. Although potential factors underlying mechanisms by which RV induces AHR have been postulated, the precise mechanisms of AHR following RV exposure remain elusive.A challenge to RV-related research stems from inadequate models for study. While human models raise ethical concerns and are relatively difficult in terms of subject recruitment, murine models are limited by susceptibility of infection to the relatively uncommon minor group (RV-B) serotypes, strains that are generally associated with infrequent clinical respiratory virus infections. Although a transgenic mouse strain that has been developed has enhanced susceptibility for infection with the common major group (RV-A) serotypes, few studies have focused on RV in the context of allergic airways disease rather than understanding RV-induced AHR. Recently, the receptor for the virulent RV-C CDHR3, was identified, but a dearth of studies have examined RV-C-induced effects in humans.Currently, the mechanisms by which RV infections modulate airway smooth muscle (ASM) shortening or excitation-contraction coupling remain elusive. Further, only one study has investigated the effects of RV on bronchodilatory mechanisms, with only speculation as to mechanisms underlying RV-mediated modulation of bronchoconstriction.


Assuntos
Hipersensibilidade Respiratória/fisiopatologia , Hipersensibilidade Respiratória/virologia , Rhinovirus/isolamento & purificação , Rhinovirus/fisiologia , Poluentes Atmosféricos/efeitos adversos , Animais , Asma/epidemiologia , Asma/fisiopatologia , Asma/virologia , Técnicas de Cocultura , Humanos , Hipersensibilidade Respiratória/epidemiologia
13.
PLoS Pathog ; 11(9): e1005122, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26336095

RESUMO

Human respiratory syncytial virus (RSV) is a major cause of severe respiratory illness in children and susceptible adults. RSV blocks the development of the innate antiviral immune response and can grow to high titers in the respiratory tract. Here we demonstrate that immunostimulatory defective viral genomes (iDVGs) that are naturally generated during RSV replication are strong inducers of the innate antiviral response to RSV in mice and humans. In mice, RSV iDVGs stimulated the expression of antiviral genes, restricted viral replication, and prevented weight loss and lung inflammation. In human cells, the antiviral response to RSV iDVGs was dominated by the expression of IFN-λ1 over IFN-ß and was driven by rapid intranuclear accumulation of the transcription factor IRF1. RSV iDVGs were detected in respiratory secretions of hospitalized patients, and their amount positively correlated with the level of expression of antiviral genes in the samples. Infection of explanted human lung tissue from different donors revealed that most humans can respond to RSV iDVGs and that the rate of accumulation of iDVGs during infection directly correlates with the quality of the antiviral response. Taken together, our data establish iDVGs as primary triggers of robust antiviral responses to RSV and provide the first evidence for an important biological role for naturally occurring iDVGs during a paramyxovirus infection in humans.


Assuntos
Genoma Viral , Interações Hospedeiro-Patógeno , Interferon beta/agonistas , Interleucinas/agonistas , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Feminino , Regulação Viral da Expressão Gênica , Humanos , Imunidade Inata , Interferon beta/antagonistas & inibidores , Interferon beta/genética , Interferon beta/metabolismo , Interferons , Interleucinas/antagonistas & inibidores , Interleucinas/genética , Interleucinas/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos Endogâmicos BALB C , Nasofaringe/imunologia , Nasofaringe/patologia , Nasofaringe/virologia , Interferência de RNA , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/isolamento & purificação , Técnicas de Cultura de Tecidos , Células Vero , Tropismo Viral , Replicação Viral
14.
J Immunol ; 191(6): 2900-6, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23956426

RESUMO

Allergic asthma, a chronic respiratory disorder marked by inflammation and recurrent airflow obstruction, is associated with elevated levels of IL-5 family cytokines and elevated numbers of eosinophils (EOS). IL-5 family cytokines elongate peripheral blood EOS (EOS(PB)) viability, recruit EOS(PB) to the airways, and, at higher concentrations, induce degranulation and reactive oxygen species generation. Although airway EOS (EOS(A)) remain signal ready in that GM-CSF treatment induces degranulation, treatment of EOS(A) with IL-5 family cytokines no longer confers a survival advantage. Because the IL-5 family receptors have common signaling capacity, but are uncoupled from EOS(A) survival, whereas other IL-5 family induced endpoints remain functional, we tested the hypothesis that EOS(A) possess a JAK/STAT-specific regulatory mechanism (because JAK/STAT signaling is critical to EOS survival). We found that IL-5 family-induced STAT3 and STAT5 phosphorylation is attenuated in EOS(A) relative to blood EOS from airway allergen-challenged donors. However, IL-5 family-induced ERK1/2 phosphorylation is not altered between EOS(A) and EOS from airway allergen-challenged donors. These observations suggest EOS(A) possess a regulatory mechanism for suppressing STAT signaling distinct from ERK1/2 activation. Furthermore, we found, in EOS(PB), IL-5 family cytokines induce members of the suppressors of cytokine signaling (SOCS) genes, CISH and SOCS1. Additionally, following allergen challenge, EOS(A) express significantly more CISH and SOCS1 mRNA and CISH protein than EOS(PB) counterparts. In EOS(PB), long-term pretreatment with IL-5 family cytokines, to varying degrees, attenuates IL-5 family-induced STAT5 phosphorylation. These data support a model in which IL-5 family cytokines trigger a selective downregulation mechanism in EOS(A) for JAK/STAT pathways.


Assuntos
Asma/imunologia , Eosinófilos/imunologia , Fatores de Transcrição STAT/imunologia , Transdução de Sinais/imunologia , Proteínas Supressoras da Sinalização de Citocina/imunologia , Asma/metabolismo , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Eosinófilos/metabolismo , Humanos , Immunoblotting , Interleucina-5/imunologia , Interleucina-5/metabolismo , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição STAT/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo
15.
Am J Respir Crit Care Med ; 189(4): 437-48, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24417465

RESUMO

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.


Assuntos
RNA Helicases DEAD-box/deficiência , Pneumonia Viral/enzimologia , Infecções por Respirovirus/enzimologia , Vírus Sendai , Animais , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/química , Quimiocinas/metabolismo , Doença Crônica , Citocinas/metabolismo , Citometria de Fluxo , Imunidade Inata , Helicase IFIH1 Induzida por Interferon , Pulmão/enzimologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Reação em Cadeia da Polimerase em Tempo Real , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia
16.
Immunol Rev ; 242(1): 178-85, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21682745

RESUMO

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.


Assuntos
Remodelação das Vias Aéreas/imunologia , Células Epiteliais/imunologia , Hipersensibilidade/imunologia , Imunidade Inata , Imunomodulação , Músculo Liso/imunologia , Sistema Respiratório/imunologia , Alérgenos/imunologia , Animais , Asma/imunologia , Asma/fisiopatologia , Criança , Fibrose Cística/imunologia , Fibrose Cística/fisiopatologia , Células Epiteliais/metabolismo , Humanos , Hipersensibilidade/fisiopatologia , Inflamação/imunologia , Interleucina-33 , Interleucinas/biossíntese , Interleucinas/imunologia , Camundongos , Camundongos Transgênicos , Músculo Liso/metabolismo , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Sistema Respiratório/fisiopatologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
17.
Am J Respir Cell Mol Biol ; 50(4): 678-83, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24219573

RESUMO

Bitter taste receptors (TAS2Rs) have recently been found to be expressed on human airway smooth muscle (HASM), and their activation results in marked relaxation. These agents have been proposed as a new class of bronchodilators in the treatment of obstructive lung diseases because they act via a different mechanism than ß-agonists. The TAS2R signal transduction pathway in HASM has multiple elements that are potentially subject to regulation by inflammatory, genetic, and epigenetic mechanisms associated with asthma. To address this, expression, signaling, and physiologic functions of the three major TAS2Rs (subtypes 10, 14, and 31) on HASM were studied. Transcript expression of these TAS2Rs was not decreased in HASM cells derived from donors with asthma compared with those without asthma (n = 6 from each group). In addition, intracellular calcium ([Ca(2+)]i) signaling using TAS2R subtype-specific agonists (diphenhydramine, chloroquine, saccharin, and flufenamic acid) was not impaired in the cells derived from donors with asthma, nor was the response to quinine, which activates all three subtypes. HASM cell mechanics measured by magnetic twisting cytometry revealed equivalent TAS2R-mediated relaxation of methacholine-treated cells between the two groups. Human precision-cut lung slices treated with IL-13 caused a decrease in ß-agonist (formoterol)-mediated relaxation of carbachol-contracted airways compared with control slices. In contrast, TAS2R-mediated relaxation was unaffected by IL-13. We conclude that TAS2R expression or function is unaffected in HASM cells derived from patients with asthma or the IL-13 inflammatory environment.


Assuntos
Asma/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratório/metabolismo , Paladar , Agonistas Adrenérgicos beta/farmacologia , Asma/fisiopatologia , Broncoconstrição , Broncodilatadores/farmacologia , Sinalização do Cálcio , Estudos de Casos e Controles , Células Cultivadas , Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-13/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/fisiopatologia
19.
Biochim Biophys Acta ; 1822(10): 1638-42, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22771498

RESUMO

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.


Assuntos
Movimento Celular/efeitos dos fármacos , Sulfato de Desidroepiandrosterona/farmacologia , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Células Cultivadas , Glucocorticoides/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Músculo Liso/citologia , Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Pulm Pharmacol Ther ; 26(2): 229-38, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23154072

RESUMO

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.


Assuntos
Broncodilatadores/farmacologia , Neprilisina/metabolismo , Peptídeos Cíclicos/farmacologia , Piperazinas/farmacologia , Receptores do Fator Natriurético Atrial/agonistas , Animais , Cães , Relação Dose-Resposta a Droga , Cobaias , Células HEK293 , Humanos , Masculino , Peptídeo Natriurético Encefálico/farmacologia , Peptídeos Cíclicos/metabolismo , Ratos , Ratos Wistar , Traqueia/efeitos dos fármacos , Traqueia/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA