Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1354-66, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26432865

ABSTRACT

High mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) protein that binds Toll-like receptors (e.g., TLR4) and the receptor for advanced glycated end products (RAGE). The direct effects of HMGB1 on airway structural cells are not fully known. As epithelial cell responses are fundamental drivers of asthma, including abnormal repair-restitution linked to changes in extracellular matrix (ECM) synthesis, we tested the hypothesis that HMGB1 promotes bronchial epithelial cell wound repair via TLR4 and/or RAGE signaling that regulates ECM (fibronectin and the γ2-chain of laminin-5) and integrin protein abundance. To assess impact of HMGB1 we used molecular and pharmacological inhibitors of RAGE or TLR4 signaling in scratch wound, immunofluorescence, and immunoblotting assays to assess wound repair, ECM synthesis, and phosphorylation of intracellular signaling. HMGB1 increased wound closure, and this effect was attenuated by blocking RAGE and TLR4 signaling. HMGB1-induced fibronectin and laminin-5 (γ2 chain) was diminished by blocking RAGE and/or blunting TLR4 signaling. Similarly, induction of α3-integrin receptor for fibronectin and laminin-5 was also diminished by blocking TLR4 signaling and RAGE. Lastly, rapid and/or sustained phosphorylation of SMAD2, ERK1/2, and JNK signaling modulated HMGB1-induced wound closure. Our findings suggest a role for HMGB1 in human airway epithelial cell repair and restitution via multiple pathways mediated by TLR4 and RAGE that underpin increased ECM synthesis and modulation of cell-matrix adhesion.


Subject(s)
Bronchi/pathology , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , HMGB1 Protein/metabolism , Wound Healing , Aged , Animals , Cell Line , Extracellular Matrix Proteins/metabolism , Humans , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Protein Biosynthesis , Pulmonary Disease, Chronic Obstructive/pathology , Receptor for Advanced Glycation End Products/metabolism , Smad2 Protein/metabolism , Tissue Donors , Toll-Like Receptor 4/metabolism
3.
Am J Physiol Lung Cell Mol Physiol ; 308(12): L1253-64, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25888577

ABSTRACT

Human airway smooth muscle (HASM) exhibits enhanced contractility in asthma. Inflammation is associated with airway hypercontractility, but factors that underpin these features are not fully elucidated. Glutamate toxicity associated with increased plasma glutamate concentrations was observed in airway inflammation, suggesting that multisubunit glutamate receptors, N-methyl-d-aspartate receptors (NMDA-R) contribute to airway hyperreactivity. We tested the hypothesis that HASM expresses NMDA-R subunits that can form functional receptors to mediate contractile responses to specific extracellular ligands. In cultured HASM cells, we measured NMDA-R subunit mRNA and protein abundance by quantitative PCR, immunoblotting, flow cytometry, and epifluorescence immunocytochemistry. We measured mRNA for a number of NMDA-R subunits, including the obligatory NR1 subunit, which we confirmed to be present as a protein. In vitro and ex vivo functional NMDA-R activation in HASM cells was measured using intracellular calcium flux (fura-2 AM), collagen gel contraction assays, and murine thin-cut lung slices (TCLS). NMDA, a pharmacological glutamate analog, induced cytosolic calcium mobilization in cultured HASM cells. We detected three different temporal patterns of calcium response, suggesting the presence of heterogeneous myocyte subpopulations. NMDA-R activation also induced airway contraction in murine TCLS and soft collagen gels seeded with HASM cells. Responses in cells, lung slices, and collagen gels were mediated by NMDA-R, as they could be blocked by (2R)-amino-5-phosphonopentanoate, a specific NMDA-R inhibitor. In summary, we reveal the presence of NMDA-R in HASM that mediate contractile responses via glutamatergic mechanisms. These findings suggest that accumulation of glutamate-like ligands for NMDA-R associated with airway inflammation contributes directly to airway hyperreactivity.


Subject(s)
Muscle Contraction/physiology , Myocytes, Smooth Muscle/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Respiratory System/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Female , Flow Cytometry , Fura-2/metabolism , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, N-Methyl-D-Aspartate/genetics , Respiratory System/cytology , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Physiol Lung Cell Mol Physiol ; 307(10): L765-74, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25217661

ABSTRACT

Emerging epidemiological evidence reveals a link between lung disease and exposure to indoor pollutants such as perfluorinated compounds (PFCs). PFC exposure during critical developmental stages may increase asthma susceptibility. Thus, in a murine model, we tested the hypothesis that early life and continued exposure to two ubiquitous household PFCs, perfluorooctanoic acid (PFOA) and perflurooctanesulfonic acid (PFOS), can induce lung dysfunction that exacerbates allergen-induced airway hyperresponsiveness (AHR) and inflammation. Balb/c mice were exposed to PFOA or PFOS (4 mg/kg chow) from gestation day 2 to 12 wk of age by feeding pregnant and nursing dams, and weaned pups. Some pups were also sensitized and challenged with ovalbumin (OVA). We assessed lung function and inflammatory cell and cytokine expression in the lung and examined bronchial goblet cell number. PFOA, but not PFOS, without the OVA sensitization/challenge induced AHR concomitant with a 25-fold increase of lung macrophages. PFOA exposure did not affect OVA-induced lung inflammatory cell number. In contrast, PFOS exposure inhibited OVA-induced lung inflammation, decreasing total cell number in lung lavage by 68.7%. Interferon-γ mRNA in the lung was elevated in all PFC-exposed groups. Despite these effects, neither PFOA nor PFOS affected OVA-induced AHR. Our data do not reveal PFOA or PFOS exposure as a risk factor for more severe allergic asthma-like symptoms, but PFOA alone can induce airway inflammation and alter airway function.


Subject(s)
Air Pollutants/toxicity , Alkanesulfonic Acids/toxicity , Asthma/immunology , Caprylates/toxicity , Fluorocarbons/toxicity , Goblet Cells/immunology , Lung/immunology , Maternal Exposure/adverse effects , Animals , Asthma/chemically induced , Asthma/pathology , Female , Goblet Cells/pathology , Interferon-gamma/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Pregnancy
5.
Biochim Biophys Acta ; 1843(7): 1259-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24637330

ABSTRACT

HMG-CoA reductase, the proximal rate-limiting enzyme in the mevalonate pathway, is inhibited by statins. Beyond their cholesterol lowering impact, statins have pleiotropic effects and their use is linked to improved lung health. We have shown that mevalonate cascade inhibition induces apoptosis and autophagy in cultured human airway mesenchymal cells. Here, we show that simvastatin also induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) in these cells. We tested whether coordination of ER stress, autophagy and apoptosis determines survival or demise of human lung mesenchymal cells exposed to statin. We observed that simvastatin exposure activates UPR (activated transcription factor 4, activated transcription factor 6 and IRE1α) and caspase-4 in primary human airway fibroblasts and smooth muscle cells. Exogenous mevalonate inhibited apoptosis, autophagy and UPR, but exogenous cholesterol was without impact, indicating that sterol intermediates are involved with mechanisms mediating statin effects. Caspase-4 inhibition decreased simvastatin-induced apoptosis, whereas inhibition of autophagy by ATG7 or ATG3 knockdown significantly increased cell death. In BAX(-/-)/BAK(-/-) murine embryonic fibroblasts, simvastatin-triggered apoptotic and UPR events were abrogated, but autophagy flux was increased leading to cell death via necrosis. Our data indicate that mevalonate cascade inhibition, likely associated with depletion of sterol intermediates, can lead to cell death via coordinated apoptosis, autophagy, and ER stress. The interplay between these pathways appears to be principally regulated by autophagy and Bcl-2-family pro-apoptotic proteins. These findings uncover multiple mechanisms of action of statins that could contribute to refining the use of such agent in treatment of lung disease.


Subject(s)
Autophagy/drug effects , Fibroblasts/drug effects , Mevalonic Acid/pharmacology , Myocytes, Smooth Muscle/drug effects , Unfolded Protein Response/drug effects , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics , Animals , Apoptosis/drug effects , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Caspases, Initiator/genetics , Caspases, Initiator/metabolism , Cell Survival , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Simvastatin/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Unfolded Protein Response/genetics , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
6.
Pulm Pharmacol Ther ; 26(1): 132-44, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22981423

ABSTRACT

Asthma is characterized in part by variable airflow obstruction and non-specific hyperresponsiveness to a variety of bronchoconstrictors, both of which are mediated by the airway smooth muscle (ASM). The ASM is also involved in the airway inflammation and airway wall remodeling observed in asthma. For all these reasons, the ASM provides an important target for the treatment of asthma. Several classes of drugs were developed decades ago which targeted the ASM - including ß-agonists, anti-cholinergics, anti-histamines and anti-leukotrienes - but no substantially new class of drug has appeared recently. In this review, we summarize the on-going work of several laboratories aimed at producing novel targets and/or tools for the treatment of asthma. These range from receptors and ion channels on the ASM plasmalemma, to intracellular effectors (particularly those related to cyclic nucleotide signaling, calcium-homeostasis and phosphorylation cascades), to anti-IgE therapy and outright destruction of the ASM itself.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Muscle, Smooth/drug effects , Airway Obstruction/drug therapy , Airway Obstruction/pathology , Airway Remodeling/drug effects , Animals , Asthma/physiopathology , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/physiopathology , Drug Design , Humans , Inflammation/drug therapy , Inflammation/pathology , Molecular Targeted Therapy , Muscle, Smooth/metabolism
7.
Pulm Pharmacol Ther ; 26(1): 42-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22939888

ABSTRACT

The biological responses of airway smooth muscle (ASM) are diverse, in part due to ASM phenotype plasticity. ASM phenotype plasticity refers to the ability of ASM cells to change the degree of a variety of functions, including contractility, proliferation, migration and secretion of inflammatory mediators. This plasticity occurs due to intrinsic or acquired abnormalities in ASM cells, and these abnormalities or predisposition of the ASM cell may alter the ASM response and in some cases recapitulate disease hallmarks of asthma. These phenotypic changes are ultimately determined by multiple stimuli and occur due to alterations in the intricate balance or reversible state that maintains ASM cells in either a contractile or synthetic state, through processes termed maturation or modulation, respectively. To elucidate the role of ASM phenotype in disease states, numerous in vitro studies have suggested a phenotypic switch in ASM primary cell cultures as an explanation for the plethora of responses mediated by ASM cells. Moreover, there is overwhelming evidence suggesting that the immunomodulatory response of ASM is due to the acquisition of a synthetic phenotype; however, whether this degree of plasticity is present in vivo as opposed to cell culture-based models remains speculative. Nonetheless, this review will give an overall scope of ASM phenotypic markers, triggers of ASM phenotype modulation and novel therapeutic approaches to control ASM phenotype plasticity.


Subject(s)
Asthma/physiopathology , Muscle, Smooth/pathology , Myocytes, Smooth Muscle/pathology , Animals , Cell Movement , Cell Proliferation , Humans , Inflammation Mediators/metabolism , Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype
8.
Pulm Pharmacol Ther ; 26(1): 95-104, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22921313

ABSTRACT

In asthma, the airway smooth muscle (ASM) cell plays a central role in disease pathogenesis through cellular changes which may impact on its microenvironment and alter ASM response and function. The answer to the long debated question of what makes a 'healthy' ASM cell become 'asthmatic' still remains speculative. What is known of an 'asthmatic' ASM cell, is its ability to contribute to the hallmarks of asthma such as bronchoconstriction (contractile phenotype), inflammation (synthetic phenotype) and ASM hyperplasia (proliferative phenotype). The phenotype of healthy or diseased ASM cells or tissue for the most part is determined by expression of key phenotypic markers. ASM is commonly accepted to have different phenotypes: the contractile (differentiated) state versus the synthetic (dedifferentiated) state (with the capacity to synthesize mediators, proliferate and migrate). There is now accumulating evidence that the synthetic functions of ASM in culture derived from asthmatic and non-asthmatic donors differ. Some of these differences include an altered profile and increased production of extracellular matrix proteins, pro-inflammatory mediators and adhesion receptors, collectively suggesting that ASM cells from asthmatic subjects have the capacity to alter their environment, actively participate in repair processes and functionally respond to changes in their microenvironment.


Subject(s)
Asthma/physiopathology , Inflammation/pathology , Myocytes, Smooth Muscle/pathology , Animals , Bronchoconstriction , Cellular Microenvironment , Humans , Hyperplasia/pathology , Muscle Contraction , Myocytes, Smooth Muscle/metabolism , Phenotype
9.
Pulm Pharmacol Ther ; 26(1): 112-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22634112

ABSTRACT

Agonists activating ß(2)-adrenoceptors (ß(2)ARs) on airway smooth muscle (ASM) are the drug of choice for rescue from acute bronchoconstriction in patients with both asthma and chronic obstructive pulmonary disease (COPD). Moreover, the use of long-acting ß-agonists combined with inhaled corticosteroids constitutes an important maintenance therapy for these diseases. ß-Agonists are effective bronchodilators due primarily to their ability to antagonize ASM contraction. The presumed cellular mechanism of action involves the generation of intracellular cAMP, which in turn can activate the effector molecules cAMP-dependent protein kinase (PKA) and Epac. Other agents such as prostaglandin E(2) and phosphodiesterase inhibitors that also increase intracellular cAMP levels in ASM, can also antagonize ASM contraction, and inhibit other ASM functions including proliferation and migration. Therefore, ß(2)ARs and cAMP are key players in combating the pathophysiology of airway narrowing and remodeling. However, limitations of ß-agonist therapy due to drug tachyphylaxis related to ß(2)AR desensitization, and recent findings regarding the manner in which ß(2)ARs and cAMP signal, have raised new and interesting questions about these well-studied molecules. In this review we discuss current concepts regarding ß(2)ARs and cAMP in the regulation of ASM cell functions and their therapeutic roles in asthma and COPD.


Subject(s)
Asthma/physiopathology , Cyclic AMP/metabolism , Muscle, Smooth/metabolism , Adrenergic beta-2 Receptor Agonists/administration & dosage , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Asthma/drug therapy , Bronchoconstriction/drug effects , Drug Therapy, Combination , Glucocorticoids/administration & dosage , Glucocorticoids/therapeutic use , Humans , Muscle, Smooth/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Receptors, Adrenergic, beta-2/metabolism
10.
Thorax ; 65(6): 547-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20522856

ABSTRACT

Asthma is characterised by airway hyper-responsiveness and remodelling, and there is mounting evidence that alterations in the phenotype of airway smooth muscle (ASM) play a central role in these processes. Although the concept that dysregulation of ASM Ca(2+) homeostasis may underlie at least part of these alterations has been around for many years, it is only relatively recently that the availability of ASM biopsies from subjects with mild and moderate asthma has allowed it to be properly investigated. In this article, critical components of the pathobiology of asthmatic ASM, including contractile function, proliferation, cell migration and secretion of proinflammatory cytokines and chemokines, are reviewed and related to associated changes in ASM Ca(2+) homeostasis. Based on this evidence, it is proposed that a unifying mechanism for the abnormal asthmatic phenotype is dysregulation of Ca(2+) homeostasis caused at least in part by a downregulation in expression and function of sarcoendoplasmic Ca(2+) ATPases (SERCAs).


Subject(s)
Airway Remodeling/physiology , Asthma/physiopathology , Calcium/metabolism , Muscle, Smooth/physiopathology , Cytokines/metabolism , Homeostasis/physiology , Humans , Muscle Contraction/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology
11.
Proc Natl Acad Sci U S A ; 106(26): 10775-80, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19541629

ABSTRACT

Phenotypic modulation of airway smooth muscle (ASM) is an important feature of airway remodeling in asthma that is characterized by enhanced proliferation and secretion of pro-inflammatory chemokines. These activities are regulated by the concentration of free Ca(2+) in the cytosol ([Ca(2+)](i)). A rise in [Ca(2+)](i) is normalized by rapid reuptake of Ca(2+) into sarcoplasmic reticulum (SR) stores by the sarco/endoplasmic reticulum Ca(2+) (SERCA) pump. We examined whether increased proliferative and secretory responses of ASM from asthmatics result from reduced SERCA expression. ASM cells were cultured from subjects with and without asthma. SERCA expression was evaluated by western blot, immunohistochemistry and real-time PCR. Changes in [Ca(2+)](i), cell spreading, cellular proliferation, and eotaxin-1 release were measured. Compared with control cells from healthy subjects, SERCA2 mRNA and protein expression was reduced in ASM cells from subjects with moderately severe asthma. SERCA2 expression was similarly reduced in ASM in vivo in subjects with moderate/severe asthma. Rises in [Ca(2+)](i) following cell surface receptor-induced SR activation, or inhibition of SERCA-mediated Ca(2+) re-uptake, were attenuated in ASM cells from asthmatics. Likewise, the return to baseline of [Ca](i) after stimulation by bradykinin was delayed by approximately 50% in ASM cells from asthmatics. siRNA-mediated knockdown of SERCA2 in ASM from healthy subjects increased cell spreading, eotaxin-1 release and proliferation. Our findings implicate a deficiency in SERCA2 in ASM in asthma that contributes to its secretory and hyperproliferative phenotype in asthma, and which may play a key role in mechanisms of airway remodeling.


Subject(s)
Asthma/metabolism , Bronchi/metabolism , Sarcoplasmic Reticulum/enzymology , Asthma/pathology , Asthma/physiopathology , Blotting, Western , Bronchi/pathology , Bronchi/physiopathology , Calcium/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CCL11/metabolism , Female , Gene Expression Regulation, Enzymologic , Homeostasis , Humans , Immunohistochemistry , Interleukin-13/pharmacology , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases
SELECTION OF CITATIONS
SEARCH DETAIL
...