Human Airway Inflammation. Sampling Techniques and Analytical Protocols

2001 ◽  
Vol 31 (11) ◽  
pp. 1796-1796
Author(s):  
A. Menzies-Gow
2001 ◽  
Vol 91 (2) ◽  
pp. 995-1003 ◽  
Author(s):  
Patrick Berger ◽  
J. Manuel Tunon-De-Lara ◽  
Jean-Pierre Savineau ◽  
Roger Marthan

Tryptase, the major mast cell product, is considered to play an important role in airway inflammation and hyperresponsiveness. Tryptase produces different, sometimes opposite, effects on airway responsiveness (bronchoprotection and/or airway contraction). This study was designed to examine the effect of human lung tryptase and activation of protease-activated receptor (PAR)-2 by synthetic activated peptide (AP) SLIGKV-NH2 on Ca2+ signaling in human airway smooth muscle (HASM) cells. Immunocytochemistry revealed that PAR-2 was expressed by HASM cells. Tryptase (7.5–30 mU/ml) induced a concentration-dependent transient relative rise in cytoplasmic Ca2+ concentration ([Ca2+]i) that reached 207 ± 32 nM ( n = 10) measured by indo 1 spectrofluorometry. The protease inhibitors leupeptin or benzamidine (100 μM) abolished tryptase-induced [Ca2+]iincrease. Activation of PAR-2 by AP (1–100 μM) also induced a concentration-dependent transient rise in [Ca2+]i, whereas the reverse peptide produced no effect. There was a homologous desensitization of the [Ca2+]i response on repeated stimulation with tryptase or AP. U-73122, a specific phospholipase C (PLC) antagonist, xestospongin, an inositol trisphosphate (IP3)-receptor antagonist, or thapsigargin, a sarcoplamic Ca2+-ATPase inhibitor, abolished tryptase-induced [Ca2+]iresponse, whereas Ca2+ removal, in the additional presence of EGTA, had no effect. Calphostin C, a protein kinase C inhibitor, increased PAR-2 [Ca2+]i response. Our results indicate that tryptase activates a [Ca2+]iresponse, which appears as PAR-2 mediated in HASM cells. Signal transduction implicates the intracellular Ca2+ store via PLC activation and thus via the IP3 pathway. This study provides evidence that tryptase, which is increasingly recognized as an important mediator in airway inflammation and hyperresponsiveness, is also a potent direct agonist at the site of airway smooth muscle.


2016 ◽  
Vol 17 (6) ◽  
pp. 636-645 ◽  
Author(s):  
Suzanne M Bal ◽  
Jochem H Bernink ◽  
Maho Nagasawa ◽  
Jelle Groot ◽  
Medya M Shikhagaie ◽  
...  

Author(s):  
Paola Rogliani ◽  
Beatrice Ludovica Ritondo ◽  
Francesco Facciolo ◽  
Ivan Nikolaev ◽  
Luigino Calzetta

CHEST Journal ◽  
1998 ◽  
Vol 113 (1) ◽  
pp. 182-185 ◽  
Author(s):  
Roelof M. Aleva ◽  
Jan Kraan ◽  
Mieke Smith ◽  
Nick H.T. Ten Hacken ◽  
Dirkje S. Postma ◽  
...  

2004 ◽  
Vol 287 (3) ◽  
pp. L584-L591 ◽  
Author(s):  
Steven G. Kelsen ◽  
Mark O. Aksoy ◽  
Yi Yang ◽  
Syed Shahabuddin ◽  
Judith Litvin ◽  
...  

Activation of the chemokine receptor CXCR3 by its cognate ligands induces several differentiated cellular responses important to the growth and migration of a variety of hematopoietic and structural cells. In the human respiratory tract, human airway epithelial cells (HAEC) release the CXCR3 ligands Mig/CXCL9, IP-10/CXCL10, and I-TAC/CXCL11. Simultaneous expression of CXCR3 by HAEC would have important implications for the processes of airway inflammation and repair. Accordingly, in the present study we sought to determine whether HAEC also express the classic CXCR3 chemokine receptor CXCR3-A and its splice variant CXCR3-B and hence may respond in autocrine fashion to its ligands. We found that cultured HAEC (16-HBE and tracheocytes) constitutively expressed CXCR3 mRNA and protein. CXCR3 mRNA levels assessed by expression array were ∼35% of β-actin expression. In contrast, CCR3, CCR4, CCR5, CCR8, and CX3CR1 were <5% β-actin. Both CXCR3-A and -B were expressed. Furthermore, tracheocytes freshly harvested by bronchoscopy stained positively for CXCR3 by immunofluorescence microscopy, and 68% of cytokeratin-positive tracheocytes (i.e., the epithelial cell population) were positive for CXCR3 by flow cytometry. In 16-HBE cells, CXCR3 receptor density was ∼78,000 receptors/cell when assessed by competitive displacement of 125I-labeled IP-10/CXCL10. Finally, CXCR3 ligands induced chemotactic responses and actin reorganization in 16-HBE cells. These findings indicate constitutive expression by HAEC of a functional CXC chemokine receptor, CXCR3. Our data suggest the possibility that autocrine activation of CXCR3 expressed by HAEC may contribute to airway inflammation and remodeling in obstructive lung disease by regulating HAEC migration.


Sign in / Sign up

Export Citation Format

Share Document