Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Respir Res ; 25(1): 137, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38521949

ABSTRACT

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.


Subject(s)
Lung Diseases , Lung , Humans , Lung/pathology , Lung Diseases/diagnosis , Lung Diseases/drug therapy , Lung Diseases/pathology , Organ Culture Techniques
2.
Respir Res ; 24(1): 238, 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37773065

ABSTRACT

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.


Subject(s)
Airway Obstruction , Asthma , Child , Adult , Humans , Rhinovirus/physiology , Airway Remodeling , Endothelial Cells/metabolism , Bronchodilator Agents , Asthma/metabolism
4.
Am J Respir Cell Mol Biol ; 69(2): 172-181, 2023 08.
Article in English | MEDLINE | ID: mdl-37098126

ABSTRACT

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.


Subject(s)
Enterovirus Infections , Rhinovirus , Adult , Child , Humans , Rhinovirus/physiology , Isoproterenol/pharmacology , Muscle, Smooth/metabolism , Lung/metabolism , Formoterol Fumarate/pharmacology , Formoterol Fumarate/metabolism , Colforsin/pharmacology , Muscle Relaxation
5.
Am J Respir Cell Mol Biol ; 68(4): 417-429, 2023 04.
Article in English | MEDLINE | ID: mdl-36662576

ABSTRACT

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.


Subject(s)
Actins , Asthma , Receptors, G-Protein-Coupled , Humans , Actins/metabolism , Asthma/metabolism , Lim Kinases/metabolism , Lung/metabolism , Muscle Relaxation/physiology , Receptors, G-Protein-Coupled/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 323(2): L142-L151, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35787178

ABSTRACT

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.


Subject(s)
Myocytes, Smooth Muscle , Receptors, Adrenergic, beta-2 , Adrenergic beta-Agonists , Bronchi , Bronchodilator Agents/pharmacology , Epinephrine/pharmacology , Humans , Muscle, Smooth , Receptors, Adrenergic, alpha-1
7.
Minerva Med ; 113(1): 4-16, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33496164

ABSTRACT

Airway smooth muscle plays a pivotal role in modulating bronchomotor tone. Modulation of contractile and relaxation signaling is critical to alleviate the airway hyperresponsiveness (AHR) associated with asthma. Emerging studies examining the phenotype of ASM in the context of asthma provide rich avenues to develop more effective therapeutics to attenuate the AHR associated with the disease.


Subject(s)
Asthma , Muscle, Smooth , Humans , Muscle Contraction , Signal Transduction
8.
Sci Rep ; 11(1): 8205, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859248

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors are widely expressed in the central nervous system. However, their presence and function at extraneuronal sites is less well characterized. In the present study, we examined the expression of NMDA receptor subunit mRNA and protein in human pulmonary artery (HPA) by quantitative polymerase chain reaction (PCR), immunohistochemistry and immunoblotting. We demonstrate that both GluN1 and GluN2 subunit mRNAs are expressed in HPA. In addition, GluN1 and GluN2 (A-D) subunit proteins are expressed by human pulmonary artery smooth muscle cells (HPASMCs) in vitro and in vivo. These subunits localize on the surface of HPASMCs and form functional ion channels as evidenced by whole-cell patch-clamp electrophysiology and reduced phenylephrine-induced contractile responsiveness of human pulmonary artery by the NMDA receptor antagonist MK801 under hypoxic condition. HPASMCs also express high levels of serine racemase and vesicular glutamate transporter 1, suggesting a potential source of endogenous agonists for NMDA receptor activation. Our findings show HPASMCs express functional NMDA receptors in line with their effect on pulmonary vasoconstriction, and thereby suggest a novel therapeutic target for pharmacological modulations in settings associated with pulmonary vascular dysfunction.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cells, Cultured , Humans , Lung/blood supply , Lung/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Vasoconstriction/genetics
9.
Redox Biol ; 39: 101832, 2021 02.
Article in English | MEDLINE | ID: mdl-33360351

ABSTRACT

A subset of asthmatics develop a severe form of the disease whose etiology involves airway inflammation along with inherent drivers that remain ill-defined. To address this, we studied human airway smooth muscle cells (HASMC), whose relaxation drives airway bronchodilation and whose dysfunction contributes to airway obstruction and hypersensitivity in severe asthma. Because HASMC relaxation can be driven by the NO-soluble guanylyl cyclase (sGC)-cGMP signaling pathway, we questioned if HASMC from severe asthma donors might possess inherent defects in their sGC or in redox enzymes that support sGC function. We analyzed HASMC primary lines derived from 17 severe asthma and 16 normal donors and corresponding lung tissue samples regarding sGC activation by NO or by pharmacologic agonists, and also determined expression levels of sGC α1 and ß1 subunits, supporting redox enzymes, and related proteins. We found a majority of the severe asthma donor HASMC (12/17) and lung samples primarily expressed a dysfunctional sGC that was NO-unresponsive and had low heterodimer content and high Hsp90 association. This sGC phenotype correlated with lower expression levels of the supporting redox enzymes cytochrome b5 reductase, catalase, and thioredoxin-1, and higher expression of heme oxygenases 1 and 2. Together, our work reveals that severe asthmatics are predisposed toward defective NO-sGC-cGMP signaling in their airway smooth muscle due to an inherent sGC dysfunction, which in turn is associated with inherent changes in the cell redox enzymes that impact sGC maturation and function.


Subject(s)
Asthma , Guanylate Cyclase , Cyclic GMP/metabolism , Humans , Nitric Oxide , Oxidation-Reduction , Signal Transduction , Soluble Guanylyl Cyclase/genetics , Soluble Guanylyl Cyclase/metabolism
10.
Am J Respir Cell Mol Biol ; 62(3): 310-318, 2020 03.
Article in English | MEDLINE | ID: mdl-31533004

ABSTRACT

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.


Subject(s)
Calcium Signaling , Enterovirus Infections/physiopathology , Enterovirus/physiology , Muscle, Smooth/virology , Respiratory Hypersensitivity/etiology , Asthma/virology , Carbachol/pharmacology , Cells, Cultured , Chemokine CXCL10/metabolism , Enterovirus/genetics , Enterovirus/isolation & purification , Enterovirus Infections/virology , Histamine/pharmacology , Humans , Inflammation Mediators/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/metabolism , Phosphorylation , Protein Processing, Post-Translational , RNA, Viral/analysis , Respiratory Hypersensitivity/virology , Viral Load
11.
Am J Respir Cell Mol Biol ; 62(1): 43-48, 2020 01.
Article in English | MEDLINE | ID: mdl-31340135

ABSTRACT

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.


Subject(s)
Bronchodilator Agents/pharmacology , Muscle, Smooth/drug effects , Respiratory System/drug effects , Soluble Guanylyl Cyclase/metabolism , Asthma/drug therapy , Asthma/metabolism , Bronchoconstriction/drug effects , Cyclic GMP/metabolism , Humans , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/metabolism , Nitric Oxide/metabolism , Respiratory System/metabolism , Signal Transduction/drug effects , Trachea/drug effects , Trachea/metabolism
12.
Sci Signal ; 12(597)2019 09 03.
Article in English | MEDLINE | ID: mdl-31481522

ABSTRACT

Asthma is a chronic allergic inflammatory airway disease caused by aberrant immune responses to inhaled allergens, which leads to airway hyperresponsiveness (AHR) to contractile stimuli and airway obstruction. Blocking T helper 2 (TH2) differentiation represents a viable therapeutic strategy for allergic asthma, and strong TCR-mediated ERK activation blocks TH2 differentiation. Here, we report that targeting diacylglycerol (DAG) kinase zeta (DGKζ), a negative regulator of DAG-mediated cell signaling, protected against allergic asthma by simultaneously reducing airway inflammation and AHR though independent mechanisms. Targeted deletion of DGKζ in T cells decreased type 2 inflammation without reducing AHR. In contrast, loss of DGKζ in airway smooth muscle cells decreased AHR but not airway inflammation. T cell-specific enhancement of ERK signaling was only sufficient to limit type 2 airway inflammation, not AHR. Pharmacological inhibition of DGK diminished both airway inflammation and AHR in mice and also reduced bronchoconstriction of human airway samples in vitro. These data suggest that DGK is a previously unrecognized therapeutic target for asthma and reveal that the inflammatory and AHR components of asthma are not as interdependent as generally believed.


Subject(s)
Asthma/immunology , Diacylglycerol Kinase/immunology , Inflammation/immunology , Respiratory Hypersensitivity/immunology , Animals , Asthma/enzymology , Asthma/genetics , Bronchoconstriction/drug effects , Bronchoconstriction/genetics , Bronchoconstriction/immunology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Enzyme Inhibitors/pharmacology , Humans , Inflammation/enzymology , Inflammation/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , Piperidines/pharmacology , Quinazolinones/pharmacology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/drug effects , Th2 Cells/enzymology , Th2 Cells/immunology
13.
Clin Chest Med ; 40(1): 51-57, 2019 03.
Article in English | MEDLINE | ID: mdl-30691716

ABSTRACT

Airway smooth muscle is the primary cell mediating bronchomotor tone. The milieu created in the asthmatic lung modulates airway smooth muscle contractility and relaxation. Experimental findings suggest intrinsic abnormalities in airway smooth muscle derived from patients with asthma in comparison with airway smooth muscle from those without asthma. These changes to excitation-contraction pathways may underlie airway hyperresponsiveness and increased airway resistance associated with asthma.


Subject(s)
Airway Resistance/immunology , Asthma/immunology , Bronchoconstriction/immunology , Bronchodilator Agents/immunology , Muscle, Smooth/metabolism , Humans
14.
J Clin Invest ; 129(3): 1387-1401, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30645205

ABSTRACT

Allergen immunotherapy for patients with allergies begins with weekly escalating doses of allergen under medical supervision to monitor and treat IgE mast cell-mediated anaphylaxis. There is currently no treatment to safely desensitize mast cells to enable robust allergen immunotherapy with therapeutic levels of allergen. Here, we demonstrated that liposomal nanoparticles bearing an allergen and a high-affinity glycan ligand of the inhibitory receptor CD33 profoundly suppressed IgE-mediated activation of mast cells, prevented anaphylaxis in Tg mice with mast cells expressing human CD33, and desensitized mice to subsequent allergen challenge for several days. We showed that high levels of CD33 were consistently expressed on human skin mast cells and that the antigenic liposomes with CD33 ligand prevented IgE-mediated bronchoconstriction in slices of human lung. The results demonstrated the potential of exploiting CD33 to desensitize mast cells to provide a therapeutic window for administering allergen immunotherapy without triggering anaphylaxis.


Subject(s)
Allergens/immunology , Anaphylaxis/prevention & control , Desensitization, Immunologic , Immunoglobulin E/immunology , Mast Cells/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Anaphylaxis/genetics , Anaphylaxis/immunology , Anaphylaxis/pathology , Animals , Bronchoconstriction/genetics , Bronchoconstriction/immunology , Humans , Immunoglobulin E/genetics , Mast Cells/pathology , Mice , Mice, Transgenic , Sialic Acid Binding Ig-like Lectin 3/genetics
15.
Respir Res ; 19(1): 208, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30373568

ABSTRACT

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.


Subject(s)
Respiratory Hypersensitivity/physiopathology , Respiratory Hypersensitivity/virology , Rhinovirus/isolation & purification , Rhinovirus/physiology , Air Pollutants/adverse effects , Animals , Asthma/epidemiology , Asthma/physiopathology , Asthma/virology , Coculture Techniques , Humans , Respiratory Hypersensitivity/epidemiology
16.
J Leukoc Biol ; 104(1): 205-214, 2018 07.
Article in English | MEDLINE | ID: mdl-29733456

ABSTRACT

The asthmatic airways are highly susceptible to inflammatory injury by air pollutants such as ozone (O3 ), characterized by enhanced activation of eosinophilic granulocytes and a failure of immune protective mechanisms. Eosinophil activation during asthma exacerbation contributes to the proinflammatory oxidative stress by high levels of nitric oxide (NO) production and extracellular DNA release. Surfactant protein-D (SP-D), an epithelial cell product of the airways, is a critical immune regulatory molecule with a multimeric structure susceptible to oxidative modifications. Using recombinant proteins and confocal imaging, we demonstrate here that SP-D directly bound to the membrane and inhibited extracellular DNA trap formation by human and murine eosinophils in a concentration and carbohydrate-dependent manner. Combined allergic airway sensitization and O3 exposure heightened eosinophilia and nos2 mRNA (iNOS) activation in the lung tissue and S-nitrosylation related de-oligomerisation of SP-D in the airways. In vitro reproduction of the iNOS action led to similar effects on SP-D. Importantly, S-nitrosylation abolished the ability of SP-D to block extracellular DNA trap formation. Thus, the homeostatic negative regulatory feedback between SP-D and eosinophils is destroyed by the NO-rich oxidative lung tissue environment in asthma exacerbations.


Subject(s)
Asthma/immunology , Eosinophils/immunology , Extracellular Traps/immunology , Oxidative Stress/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Animals , Asthma/metabolism , Cells, Cultured , Eosinophils/drug effects , Eosinophils/metabolism , Extracellular Traps/metabolism , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Mice , Oxidants, Photochemical/toxicity , Oxidative Stress/drug effects , Ozone/toxicity
18.
Am J Respir Cell Mol Biol ; 58(1): 89-98, 2018 01.
Article in English | MEDLINE | ID: mdl-28853915

ABSTRACT

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.


Subject(s)
Asthma/immunology , Bronchi/immunology , Gene Expression Regulation/immunology , Lung/immunology , RGS Proteins/immunology , Respiratory Mucosa/immunology , Animals , Asthma/genetics , Asthma/pathology , Bronchi/pathology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Lung/pathology , Mice , Mice, Transgenic , RGS Proteins/genetics , Respiratory Mucosa/pathology
19.
Am J Respir Cell Mol Biol ; 58(4): 530-541, 2018 04.
Article in English | MEDLINE | ID: mdl-29262264

ABSTRACT

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.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adenylyl Cyclases/metabolism , Asthma/enzymology , Cyclic AMP/metabolism , Muscle, Smooth/enzymology , Myocytes, Smooth Muscle/enzymology , Receptors, Adrenergic, beta-2/metabolism , Respiratory System/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Adenylyl Cyclases/genetics , Airway Remodeling , Asthma/genetics , Asthma/pathology , Asthma/physiopathology , Case-Control Studies , Cell Proliferation , Cells, Cultured , Humans , Membrane Microdomains/enzymology , Membrane Microdomains/pathology , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/pathology , Receptors, Adrenergic, beta-2/genetics , Respiratory System/pathology , Respiratory System/physiopathology , Second Messenger Systems , Time Factors
20.
Nat Biomed Eng ; 2(2): 124-137, 2018 02.
Article in English | MEDLINE | ID: mdl-31015629

ABSTRACT

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.


Subject(s)
Elastomers/chemistry , Single-Cell Analysis/methods , Asthma/pathology , Cell Differentiation , Cells, Cultured , Fluorescent Dyes/chemistry , Formoterol Fumarate/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Myocardial Contraction/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phagocytosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...