Skip to main content
Erschienen in: Respiratory Research 1/2017

Open Access 01.12.2017 | Research

Combination of glycopyrronium and indacaterol inhibits carbachol-induced ERK5 signal in fibrotic processes

verfasst von: Yukiko Namba, Shinsaku Togo, Miniwan Tulafu, Kotaro Kadoya, Kumi Yoneda Nagahama, Hikari Taka, Naoko Kaga, Akira Orimo, Xiangde Liu, Kazuhisa Takahashi

Erschienen in: Respiratory Research | Ausgabe 1/2017

Abstract

Background

Airway fibrosis is one of the pathological features of chronic obstructive pulmonary disease (COPD), and recent studies revealed that acetylcholine plays an important role in the development of airway remodeling by stimulating proliferation and collagen synthesis of lung fibroblasts. This study was designed to examine the effects of a long-acting muscarinic receptor antagonist (LAMA) glycopyrronium and a long-acting β2 adrenergic receptor agonist (LABA) indacaterol on acetylcholine-mediated fibrotic responses in lung fibroblasts.

Methods

After carbachol (CCh) or transforming growth factor-β1 (TGF-β1) exposure, the response to glycopyrronium and indacaterol was determined in vitro in fibroblasts isolated from mild-to-moderate COPD lung tissue. The ability of fibroblasts to mediate the contraction of collagen gels was assessed. The expression of α-smooth muscle actin (α-SMA) and the phosphorylation of extracellular-signal-regulated kinase 5 (ERK5) were determined by immunoblot. TGF-β1 was quantified by ELISA and acetylcholine was quantified by liquid chromatography tandem-mass spectrometry.

Results

CCh stimulated fibroblast-mediated collagen gel contraction and α-SMA expression and TGF-β1 release by fibroblasts. Blockade of autocrine TGF-β1 attenuated CCh-mediated fibrotic responses, while TGF-β1 did not stimulate acetylcholine release. Glycopyrronium plus indacaterol significantly attenuated CCh- and TGF-β1-mediated fibrotic responses through inhibition of ERK5 phosphorylation. Notably, the magnitudes of CCh- and TGF-β1-stimulated gel contraction, CCh-induced TGF-β1 release, and ERK5 phosphorylation were greater in fibroblasts isolated from COPD subjects than in those from non-smokers.

Conclusions

CCh induced TGF-β1 self-sustaining signaling loops by potentiating ERK5 signaling and promoted myofibroblast activity. This autocrine signaling mechanism may be an attractive therapeutic target to block the fibrotic response, which was modulated by the combination of glycopyrronium and indacaterol.
Abkürzungen
ALK5
Activin receptor–like kinase 5
CCh
Carbachol
ChAT
Choline-acetyltransferase
COPD
Chronic obstructive pulmonary disease
ELISA
Enzyme-linked immunosorbent assay
ERK1/2
Extracellular signal-regulated kinase 1/2
ERK5
Extracellular-signal-regulated kinase 5
FEV1
Forced expiratory volume in one second
FVC
Forced vital capacity
GLY
Glycopyrronium
IND
Indacaterol
LABA
Long-acting β2 adrenergic receptor agonist
LAMA
Long-acting muscarinic receptor antagonist
LC-MS/MS
Liquid chromatography tandem-mass spectrometry
MAPK
Mitogen-activated protein kinase
TGF-β1
Transforming growth factor-β1
α-SMA
α-smooth muscle actin

Background

Persistent long-term airway inflammation contributes to airway remodeling and permanent alterations of the airway wall structure. Airway remodeling, including fibrosis, arises during the early stages of asthma and chronic obstructive pulmonary disease (COPD), and small airway fibrosis in COPD is associated with a greater decline in forced expiratory volume in 1 s (FEV1) than emphysema [13].
Resident lung fibroblasts are activated under chronic airway inflammation and differentiated into myofibroblasts, accompanied by an increased expression of α-smooth muscle actin (α-SMA) and augmented a persistence of contractile activity (3), contributing to irreversible airflow limitation [4]. Transforming growth factor (TGF)-β is a well-known fibrogenic mediator involved in COPD [5, 6], but recent evidence indicates the potential role of non-neuronal acetylcholine as a paracrine/autocrine mediator in the regulation of airway fibrosis [7]. A clinical study also showed that methacholine inhalations induced sub-epithelial collagen deposition and TGF-β immunoreactivity within the airway epithelium, which are suppressed by a short acting β2-agonist [8].
Acetylcholine is classically known as a neurotransmitter involved in airway muscle contraction and mucus secretion via the activation of muscarinic receptors, and the increased cholinergic tone of smooth muscle is a major therapeutic target of airflow limitation in COPD [9]. Notably, acetylcholine may not only be released from parasympathetic nerve, but also from airway epithelial cells and inflammatory cells [10, 11], and cholinergic stimuli increase the proliferation and collagen synthesis of human lung fibroblasts via extracellular signal-regulated kinase 1/2 (ERK1/2) [1214] and enhance α-SMA expression and TGF-β1 production through ERK1/2 and Rho kinase [15]. Furthermore, fibroblasts express choline-acetyltransferase (ChAT), an enzyme regulating acetylcholine synthesis, and TGF-β1 stimulates ChAT expression [15, 16]. However, the acetylcholine release from fibroblasts has not yet been directly demonstrated. Additionally, extracellular-signal-regulated kinase 5 (ERK5), a member of the mitogen-activated protein kinase (MAPK) family, has recently garnered the attention as the positive regulator of TGF-β signaling, and inhibition of ERK5 ameliorated bleomycin-induced pulmonary fibrosis in mice [17], but the roles of ERK5 in airway fibrosis remain unknown.
Long-acting muscarinic antagonists (LAMAs) and long-acting β2 adrenergic receptor agonists (LABAs) are widely used as bronchodilators for maintenance therapy in COPD and asthma [18, 19]. QVA149, a fixed-dose combination of the LAMA, glycopyrronium (GLY), and the LABA, indacaterol (IND), is a novel, once-daily dual bronchodilator that demonstrated rapid and sustained bronchodilation in patients with moderate-to-severe COPD [20]. However, whether the combination of LAMA and LABA modifies airway fibrosis remains unclear, particularly during early/mild COPD.
In this study, we investigated the effect of GLY and IND in regulating carbachol (CCh) and TGF-β1-induced ERK5 activation during the fibrotic processes in lung fibroblasts isolated from patients with COPD. In addition, we explored a possible link between CCh and TGF-β1 signaling.

Methods

Materials

Dulbecco’s Modified Eagle’s Medium (DMEM) was purchased from Wako (Osaka, Japan) and FCS from Sigma-Aldrich (St. Louis, MO, USA). Novartis Pharma AG (Basel, Switzerland) provided GLY and IND. CCh was from Sigma-Aldrich and TGF-β1 from R&D Systems (Minneapolis, MN, USA). ERK5 inhibitor, BIX02189, and activin receptor–like kinase 5 (ALK5) inhibitor, SB431542, were from Selleckchem (Houston, TX, USA) and ERK1/2 inhibitor, PD98059, from Calbiochem (La Jolla, CA, USA). Rho-associated coiled-coil forming kinase/Rho binding kinase inhibitor, Y-27632, was from Wako. The primary antibodies were anti-ERK5 (Cell Signaling Technology, Danvers, MA, USA: Catalogue no. #3372), anti–α-smooth muscle actin (α-SMA) (Sigma-Aldrich: Catalogue no. A2547), and anti- ChAT antibody (Merck KGaA, Darmstadt, Germany: Catalogue no. AB144P).

Isolation and primary culture of lung fibroblasts

Human primary lung parenchymal fibroblasts from patients undergoing lung resection for localized tumor were cultured as described previously [21]. Briefly, the portion of lung parenchymal tissue that was as distal from any tumors as possible and free of the pleural surface was minced and cultured in DMEM supplemented with 10% FCS, 100 μg/ml penicillin, 250 μg/ml streptomycin, and 1 μg/ml amphotericin B in a humidified atmosphere of 5% CO2. Cells were assessed in passage 2 after primary culture and displayed the typical fibroblast morphology and were positive for vimentin and negative for cytokeratin staining. Lung fibroblasts from 22 patients: 11 non-smokers without clinical airway symptom or lung functional abnormalities, 11 smokers with mild-to-moderate COPD according to the Global Initiative for Chronic Obstructive Lung Disease classification (GOLD 1 and 2) were obtained. Patients with a history of asthma and marked eosinophil infiltration in airways of resected lungs were excluded. The Institutional Review Board at the Juntendo University School of Medicine approved the procedures. All patients provided written, informed consent (No. 2013051).

Cell culture

Human fetal lung fibroblasts (HFL1; catalogue number CCL-153) and A549 (catalogue number CCL-185) were purchased from the American Type Culture Collection (Manassas, VA, USA). Lung fibroblasts and A549 cells were cultured in DMEM supplemented with 10% FCS, 100 μg/ml penicillin, 250 μg/ml streptomycin, and 1 μg/ml amphotericin B in a humidified atmosphere of 5% CO2. Sub-confluent cells were removed from the dishes by 0.05% trypsin-EDTA (Wako, Osaka, Japan). For three-dimensional collagen gel contraction and ELISA, primary lung fibroblasts were used at the fourth to sixth passages after isolation to exclude the effect of differences in passage number and culturing conditions.

Type I collagen preparation

Type I collagen (rat tail tendon collagen, RTTC) was extracted from rat tail tendons as previously described [22].

Three-dimensional collagen gel contraction assay

The effect of CCh and TGF-β1 on fibroblast-mediated collagen gel contraction was assessed in the presence/absence of GLY and IND, BIX02189, and SB431542, using a modification of a previously described method [23]. Briefly, sub-confluent fibroblasts were deprived of serum for 24 h, detached with 0.05% trypsin-EDTA, and resuspended in serum free DMEM (SF-DMEM). The cell number was then counted with a Coulter Counter (Beckman Coulter, Inc., Fullerton, CA, USA). Collagen gels were prepared by mixing the appropriate amount of RTTC, distilled water, 4× concentrated DMEM, and cell suspension. The final mixture was 0.75 mg/ml of collagen, 3 × 105 fibroblasts/ml gel, and a physiologic ionic strength of 1× DMEM, and a pH of 7.4. A portion of the gel solution (500 μl) was then casted into each well of a 24-well tissue culture plate with a 1.9 cm2 growth area (Corning, Costar, NY, USA). Gelation occurred in 20 min at room temperature, after which the gels were released from the surface of the culture well using a sterile spatula. They were then transferred into 60 mm tissue culture dishes (three gels in each dish) containing 5 ml of SF-DMEM with or without designated reagents and incubated at 37 °C, 5% CO2 for 3 days. The ability of fibroblasts to contract the floating gels was determined by quantifying the area of the gels daily using an LAS4000 image analyzer (GE Healthcare Bio-Science AB, Uppsala, Sweden). Data are expressed as the percentage of gel area compared with the original gel size. Contraction is then expressed as a decrease in gel area.

Measurement of TGF-β1 release

TGF-β1 release in the monolayer culture media was measured by ELISA. Sub-confluent lung fibroblasts on a 6-well plate were deprived of serum for 24 h and stimulated with CCh in the presence or absence of GLY or/and IND. Supernatants were harvested after 24 h, frozen, and stored at − 80 °C until assay. TGF-β1 production from lung fibroblasts was determined by human TGF-β1 Immunoassay (R&D Systems; Minneapolis, MN, USA) according to the manufacturer’s instructions.

Determination of acetylcholine by LC–MS/MS

Acetylcholine (ACh) in the culture supernatants was analyzed by liquid chromatography tandem-mass spectrometry (LC-MS/MS). Sub-confluent lung fibroblasts on a 6-well plate were deprived of serum for 24 h and stimulated with TGF-β1. After 24 h, supernatants were transferred to 1.5 ml tubes in the presence of 1 μM rivastigmine (Sigma-Aldrich) to prevent ACh degradation. For ACh measurement, 5 μl of the aliquots were injected directly into the LC-MS/MS system. LC-MS/MS analysis were performed on Gilson HPLC system (Gilson, Villiers-le-Bel, France) connected to TSQ Quantum Ultra AM mass spectrometer (Thermo Fisher Scientific). ACh was analyzed in the positive mode and selected reaction monitoring (SRM) analysis was carried out. MS conditions were as follows: The collision energy was 15 eV and the Argon gas as collision gas was set at 1mTorr. The spray voltage was 3.5 kV; the capillary temperature was set at 300 °C. The nitrogen flow rate was 25 units for sheath gas and 5 units for auxiliary gas. Discovery HS F5-3 2.1 mm i.d × 150 mm, 3 μm column (Supelco, Bellefonte, PA, USA) was used for LC separation. The column temperature was set at 40 °C. The mobile phase A consisted of 0.1% formic acid in water and the mobile phase B consisted of 0.1% formic acid in 80% acetonitrile (v/v). The elution program started at 0% B and increased to 40% for 20 min. The flow rate was set to 0.2 ml/min. To obtain the internal standard report, acetylcholine (Wako, Osaka, Japan) was added to the SF-DMEM as control.

Immunoblotting

The expression of α-SMA and ChAT as well as ERK5 phosphorylation were evaluated by immunoblotting analysis of cell lysates by using anti-α-SMA antibody (1: 5000), anti-ChAT antibody (1: 1000), and anti-ERK5 antibody (1: 1000). ERK5 phosphorylation can be detected by the presence of a band with a slower electrophoretic mobility shift [24]. Lung fibroblasts were seeded on 60 mm dish at a density of 5 × 104/ml and cultured for 24 h. The medium was then changed to SF-DMEM for 24 h, following which cells were stimulated with CCh and TGF-β1 in the presence or absence of inhibitors for 48 h. After treatment, the cells were washed with ice-cold PBS and lysed with radioimmunoprecipitation assay lysis buffer (Wako, Osaka, Japan) supplemented with protease inhibitor (Thermo Fisher Scientific) and phosphatase inhibitor cocktail (Thermo Fisher Scientific). The lysates were briefly sonicated and then centrifuged at 12,000 rpm for 10 min. The protein concentration in the supernatants was measured by BCA Protein Assay Kit (Thermo Fisher Scientific). Lysates were solubilized in 4× Laemmli Sample Buffer (Bio-RAD). Equal amounts of protein were loaded into each lane, separated in 7.5–10% SDS-PAGE and transferred to polyvinylidene difluoride membrane. Membranes were blocked for 1 h or overnight in blocking reagent. The membrane was probed with antibodies to ERK5, ChAT, α-SMA, and β-actin. Bound antibodies were visualized using peroxidase-conjugated second antibodies and enhanced chemiluminescence with LAS4000 image analyzer (GE, Healthcare Bio-Science AB) and luminescent images were analyzed with an ImageQuant TL (GE Healthcare Bio-Science AB).

Statistical analysis

Results are expressed as the means ± SEM. Grouped data were evaluated by one-way analysis of variance (ANOVA), corrected by the Bonferroni’s test. Samples that appeared different within a series were further assessed by Student’s t-test. Comparisons between non-smokers and patients with COPD were performed using the unpaired, two-tailed Mann–Whitney test. For evaluation of experimental studies within a group where paired samples were available, the Wilcoxon test was used. For these comparisons, each subject was considered as an individual point. P values less than 0.05 were considered significant. All data were analyzed using GraphPad Prism 6 (GraphPad Software, San Diego, CA, USA).

Results

Clinical and demographic features

The clinical and demographic features of the patients are shown in Table 1. The two groups were similar in age and sex. Patients with COPD were classified according to the GOLD consensus report criteria as GOLD 1 (n = 3) or GOLD 2 (n = 8). The lung function differed significantly between the two groups. As expected, patients with COPD had lower FEV1/FVC and FEV1% predicted value.
Table 1
Clinical and demographic features of patients
 
No. of Non-smokers Patients
Mean ± SD
No. of Patients with COPD
Mean ± SD
Age, y
11
64 ± 8
 
11
66 ± 9
Sex, no. male/female
5/6
  
8/3
 
Smoking status
 Never smoked
11
  
0
 
 Former/Current
0/0
  
4/7
 
Pack-years
11
0
 
11
52.2 ± 32
GOLD 0/1/2/3/4
11/0/0/0/0
  
0/3/8/0/0
 
FEV1/FVC, %
11
77.2 ± 5
 
11
60.4 ± 9*
FEV1, % predicted value
11
107.2 ± 10
 
11
73.2 ± 15*
VC, L
11
3.5 ± 1
 
11
3.6 ± 1
COPD chronic obstructive pulmonary disease, GOLD global initiative for chronic obstructive lung disease
*p < 0.001 compared with control

Effect of GLY and/or IND on CCh or TGF-β1-induced collagen gel contraction and α-SMA expression

To investigate the effect of acetylcholine on tissue fibrotic response, fibroblast-mediated collagen gel contraction assay was used as an in vitro model of tissue remodeling. α-SMA expression was determined as one of contractile stress fibers, leading to gel contraction. Acetylcholine is easily metabolized by acetylcholinesterase. Therefore, we selected CCh as the long acting cholinergic agonist to stimulate muscarinic receptors. The expression of muscarinic receptor (M1-3) and β2 adrenergic receptor in HFL1 cells were confirmed using western blot analysis (data not shown). CCh significantly augmented HFL1-mediated collagen gel contraction and α-SMA expression in a time and concentration-dependent manner (P < 0.05 at 10−6 M, Fig. 1a-d). CCh were widely used as the concentration of 10−5M in laboratory setting, but previous study reported human normal bronchi contained 2–3 nmol/g of acetylcholine [25], suggesting the possibility of lower concentration than 10−5M acetylcholine. Therefore, we decided to select 10−6 M CCh in our experiments.
We examined the effect of GLY or IND on HFL1-mediated collagen gel contraction. IND inhibited gel contraction in a dose-dependent manner (P < 0.05 at concentrations of 10−8 to 10−6 M IND compared to control, Fig. 1f). However, GLY had no inhibitory effect (Fig. 1e). GLY or IND inhibited CCh-augmented gel contraction in a dose-dependent manner (P < 0.05 at concentrations of 10−8 to 10−6 M GLY and of 10−9 to 10−6 M IND compared with control, Fig. 2a and b). IND inhibited gel contraction in the presence of TGF-β1, which is known as a key mediator in the development of fibrosis and strongly stimulates collagen gel contraction and α-SMA expression in fibroblasts [26, 27] (P < 0.05 at concentrations of 10−9 to 10−6 M IND compared to TGF-β1 alone, Fig. 2d). Nevertheless, GLY had no effect on TGF-β1-mediated gel contraction (Fig 2c). In clinical settings, the maximum concentration in plasma of GLY 50 μg and IND 150 μg which were administered by inhalation once daily for 14 days were 216 pg/ml (0.5 nM) and 438.6 pg/ml (1.1 nM), respectively [28, 29]. However, there are no direct evidence for lung concentration after inhalation, we decided to select 10−8 M GLY and 10−9 M IND to access the pharmacological interaction between GLY and IND. Next, we investigated the effect of a combination of GLY and IND on CCh- or TGF-β1-mediated gel contraction in HFL1 cells. The combination of GLY 10−8 M and IND 10−9 M significantly suppressed CCh-augmented collagen gel contraction (CCh; 67.0 ± 1.3% vs. CCh + GLY + IND; 76.0 ± 1.0%: P < 0.01) as well as TGF-β1-augmented gel contraction (TGF-β1; 66.1 ± 1.1% vs. TGF-β1 + GLY + IND; 71.2 ± 0.7%: P < 0.01) (Fig. 2e and g). The combination of 10−8 M GLY and 10−9 M IND significantly suppressed CCh- or TGF-β1-induced α-SMA expression (P < 0.01 and P < 0.05, respectively) (Fig. 2f and h).

Phenotypic differences in fibroblasts from non-smokers and patients with COPD on CCh- or TGF -β1-induced collagen gel contraction

We next examined the phenotypic features of fibroblasts from non-smokers and patients with COPD in response to CCh and TGF-β1 stimulation. CCh significantly augmented collagen gel contraction mediated by the fibroblasts isolated from non-smokers (control; 82.0 ± 2.7% vs. CCh; 80.0 ± 3.2%; P < 0.05) or patients with COPD (control; 79.0 ± 2.8% vs. CCh; 73.0 ± 2.9%; P < 0.01) (Fig. 3a). Similarly, TGF-β1 also augmented collagen gel contraction mediated by the fibroblasts isolated from non-smokers (control; 82.0 ± 2.7% vs. TGF-β1; 78.8 ± 2.7%; P < 0.01) or patients with COPD (control; 79.0 ± 2.8% vs. TGF-β1; 70.1 ± 4.0%, P < 0.01) (Fig. 3b). It is noteworthy that the magnitudes of collagen gel contraction in response to CCh or TGF-β1 stimulation were greater in the fibroblasts isolated from patients with COPD than in those from non-smokers. Combination of GLY 10−8 M and IND 10−9 M significantly suppressed CCh- or TGF-β1-augmented collagen gel contraction in the fibroblasts isolated from both non-smokers and COPD subjects (P < 0.01) (Fig. 3c and d).

Effect of CCh on the release of endogenous TGF-β1

We assessed the effects of CCh on the release of endogenous TGF-β1 and the effect of GLY and IND on the release of endogenous TGF-β1 by fibroblasts. CCh stimulated TGF-β1 release by HFL1 cells. The effect was significantly suppressed by a combination of GLY 10−8 M and IND 10−9 M (Fig. 4a). CCh-stimulated TGF-β1 release was significantly higher in the primary lung fibroblasts from patients with COPD than in those from non-smokers (P < 0.05). The suppression of CCh-stimulated TGF-β1 release by GLY 10−8 M plus IND 10−9 M was significantly stronger in the fibroblasts isolated from patients with COPD than in those from non-smokers (P < 0.05, Fig. 4b).
This result suggested that CCh-induced collagen gel contraction and α-SMA expression might be mediated through a TGF-β1 autocrine release/paracrine mechanism. Moreover, CCh-induced collagen gel contraction and α-SMA expression were partially suppressed by SB431542, a selective inhibitor of the TGF-β1 receptor, ALK5 (Fig. 4c and d).

Quantification of acetylcholine release from lung fibroblasts

Acetylcholine is mainly synthesized from choline and acetyl-CoA by ChAT. Therefore, ChAT protein level in HFL1 and A549 cells was semi-quantitatively assessed by immunoblot. A549 cells were shown to release acetylcholine [30]. ChAT was detected in both HFL1 and A549 cells. It was slightly, but significantly, stimulated by TGF-β1 in HFL1 cells (Fig. 5a, P < 0.05). Interestingly, ChAT protein was significantly higher in A549 cells under basal conditions compared to that in HFL1 cells and TGF-β1 did not alter ChAT protein level in A549 cells (Fig. 5a).
Next, we measured acetylcholine in the supernatants of cultured fibroblasts by LC-MS/MS. Acetylcholine was not detectable in the culture medium of HFL1 cells or primary lung fibroblasts from non-smokers and patients with COPD (Fig. 5b-d), although 2 × 10−9 M acetylcholine could be detected as the positive control (Fig. 5e). Furthermore, TGF-β1 did not stimulate acetylcholine release in fibroblasts (Fig. 5b-d). This result indicated that the release of acetylcholine from fibroblasts might be only a modest level.

Effect of GLY and IND on CCh or TGF-β1 induced ERK5 phosphorylation

We explored the role of ERK5 in mediating CCh and TGF-β1 signaling and the effect of GLY and IND on ERK5 phosphorylation. CCh and TGF-β1 induced ERK5 phosphorylation (P < 0.05, Fig. 6a-d). GLY plus IND and BIX02189, a specific MEK5-ERK5 inhibitor, significantly suppressed CCh- or TGF-β1-induced ERK5 phosphorylation (P < 0.05, Fig. 6c and d). Moreover, CCh-induced ERK5 phosphorylation was suppressed by SB431542 (Fig. 6c), suggesting that TGF-β1 autocrine release was involved in CCh signaling. Notably, the basal level of ERK5 expression was significantly higher in fibroblasts from patients with COPD than in those from non-smokers (P < 0.05, Fig. 6e), although ERK5 phosphorylation did not differ between the two groups (Fig. 6f). However, ERK5 phosphorylation in response to CCh stimulation was higher in fibroblasts from patients with COPD than in those from non-smokers (P < 0.05, Fig. 6g). In contrast, in response to TGF-β1 stimulation, ERK5 phosphorylation was similar in fibroblasts from patients with COPD and non-smokers (Fig. 6h). GLY plus IND significantly suppressed CCh- or TGF-β1-induced ERK5 phosphorylation in fibroblasts regardless of their origin (P < 0.05, Fig. 6g and h).

Blockade of ERK5 phosphorylation resulted in the suppression of collagen gel contraction and α-SMA expression

We assessed whether ERK5 blockade altered CCh- or TGF-β1-augmented collagen gel contraction and α-SMA expression in fibroblasts. Pretreatment with BIX02189 or Y-27632, a Rho kinase inhibitor, resulted in the significant reduction of CCh- or TGF-β1-augmented collagen gel contraction and α-SMA expression in HFL1 cells. However, PD98059, an ERK1/2 inhibitor did not significantly attenuate CCh- or TGF-β1-augmented collagen gel contraction and α-SMA expression (Fig. 7a and b). CCh-augmented collagen gel contraction by fibroblasts from patients with COPD or non-smokers was significantly inhibited by BIX02189 (P < 0.01, Fig. 7c). However, BIX02189 suppression of TGF-β1-augmented collagen gel contraction was more significant in fibroblasts from patients with COPD than in those from non-smokers (P < 0.01 and P < 0.05, respectively) (Fig. 7d).

Discussion

In the current study, we demonstrated for the first time that GLY and IND additively attenuated lung fibroblast-mediated contraction of collagen gels in response to CCh or TGF-β1. Interestingly, compared to lung fibroblasts isolated from non-smokers, the response of lung fibroblasts from patients with mild to moderate COPD to CCh- or TGF-β1-induced collagen gel contraction as well as to CCh-stimulated TGF-β1 release was higher. CCh or TGF-β1 induced ERK5 phosphorylation and inhibition of ERK5 phosphorylation by GLY plus IND resulted in the reduction of CCh- or TGF-β1-augmented collagen gel contraction and α-SMA expression in lung fibroblasts (Fig. 8). This functional involvement of a non-neuronal cholinergic system provide new insights into the pathogenesis of COPD and treatment strategy combining inhaled IND and GLY to patients with developing airway fibrosis in early stage of COPD.
Accumulating evidence indicates that acetylcholine in the airway may not only be released from parasympathetic nerve, but also from bronchial epithelial [10] and inflammatory cell [16] and may contribute in airway remodeling as a paracrine and autocrine mediator. Previous studies reported that normal lung fibroblasts express muscarinic receptors and directly respond to cholinergic stimulation, including proliferation and collagen synthesis through ERK1/2 activation [1214]. The cholinergic agonist, relying on Rho kinase and autocrine TGF-β1 release, also induced fibroblast-to-myofibroblast transition and α-SMA expression [15]. An in vivo study revealed that anticholinergics inhibit lipopolysaccharide-induced small airway fibrosis in guinea pigs [31].
Collagen gel contraction assay is an in vitro fibrosis model developed to assess the regulation of contractile stress fiber, generally indicated as α-SMA, production from activated myofibroblasts. When differentiated myofibroblasts are cultured in such a collagen gel, the gels are contracted by the traction force, suggesting the contribution of fibrosis and abnormal tissue architecture [32]. In this study, we demonstrated that the cholinergic agent, CCh, augmented fibroblast-mediated collagen gel contraction. Compared to lung fibroblasts from non-smokers, parenchymal lung fibroblasts obtained from patients with mild to moderate COPD showed higher sensitivity to CCh or TGF-β1 in the collagen gel contraction assay. Moreover, fibroblasts obtained from patients with COPD showed higher response to CCh stimulating TGF-β1 release than those from non-smokers. These findings suggest that acetylcholine and the subsequent TGF-β1 autocrine loop play an important role in the regulation of small airway fibrosis in patients with COPD. A similar study reported that acetylcholine-induced cell proliferation was enhanced in lung fibroblasts from patients with COPD compared to cells from healthy non-smokers and healthy smokers, and muscarinic receptors were highly expressed in fibroblasts from patients with COPD compared to that in fibroblasts from controls [33]. However, we could not demonstrate different expression levels of muscarinic receptors in mild to moderate COPD fibroblasts compared to non-smoker fibroblasts (data not shown), suggesting different mechanisms of action.
ChAT, the synthase for acetylcholine, is expressed in airway epithelial cells, airway smooth muscle cells, and fibroblasts as well as in inflammatory cells such as macrophages, lymphocytes, and granulocytes [16]. Furthermore, epithelial and inflammatory cells release acetylcholine [16], but that from the other cell types remains to be determined. Thus, we measured the level of acetylcholine in the medium of cultured fibroblasts. However, acetylcholine was not detectable in the culture supernatant of fibroblasts from patients with COPD or non-smokers as well as of HFL1 cells. GLY alone did not show any effect on fibroblasts-mediated gel contraction and the cholinesterase inhibitor also could not potentiate collagen gel contraction by fibroblasts (data not shown), indicating that endogenous acetylcholine may not be involved in this mechanism. Consistently, Profita et al. reported that acetylcholine autocrine mechanism generated by ChAT was not involved in lung fibroblast proliferation [33]. In addition, ChAT expression and acetylcholine release were identified from large airways, but not from lung parenchyma, in ovalbumin-sensitized guinea pigs [9]. These findings indicated that lung parenchymal fibroblasts do not synthesize detectable amounts of acetylcholine although they do express ChAT.
ERK5, an atypical member of the MAPK family, plays an important role in hypertrophic remodeling of cardiomyocytes by enhancing cell viability and in chronic glomerulonephritis by increasing ECM deposition [34]. BIX02189, a pharmacological inhibitor of MEK5/ERK5, improved both lung fibrosis and survival rate in the bleomycin-induced lung fibrosis model [17]. García-Hoz et al. revealed that CCh induces the formation of Gαq/protein kinase C (PKC) ζ and MEK5 protein complexes to trigger ERK5 activation in mouse embryonic fibroblasts [35]. Badshah et al. reported that TGF-β1 mediates ERK5 activation in human podocytes without the involvement of the small GTPase Ras [36]. In the present study, CCh and TGF-β1 induced ERK5 phosphorylation in lung fibroblasts, and BIX02189 attenuated CCh- or TGF-β1-induced gel contraction and α-SMA expression. Furthermore, levels of basal ERK5 expression and CCh-induced ERK5 phosphorylation were higher in fibroblasts obtained from patients with COPD than in fibroblasts obtained from non-smokers, suggesting that ERK5 signaling was involved in developing airway fibrosis in COPD.
In clinical studies, inhalation of a combination of GLY and IND showed additive effects in terms of FEV1 between 5 min and 3 h post-inhalation in patients with moderate-to-sever COPD, with a synergistic effect at 15 min post-inhalation compared to the administration of either drug alone [37]. Previous studies using isolated human airway demonstrated that aclidinium and formoterol showed synergistic inhibition of airway smooth muscle tone induced by acetylcholine from 0 to 6 h when administered at low concentrations [38]. In the current study, we report for the first time that the GLY and IND combination has an additive inhibitory effect on fibroblast-mediated gel contraction in response to CCh or TGF-β1. We set the concentrations of GLY and IND in this experiment using plasma pharmacokinetics from a clinical trial; however, the actual concentration of these compounds in the airway is unclear. To verify the desired effectiveness and safety of GLY, IND, and BIX02189 in preventing airway fibrosis for clinical use, computational simulation techniques, such as fluidic dynamics, are needed to calculate the drug concentration in the peripheral airway [39].

Conclusion

Our results demonstrate that GLY and IND combination inhibits lung fibroblast-mediated collagen gel contraction and α-SMA expression in response to CCh or TGF-β1. CCh induced TGF-β1 self-sustaining signaling loops by potentiating ERK5 signaling, which was suppressed by GLY and IND. Notably, the magnitudes of CCh- and TGF-β1-induced fibrotic activities were greater in fibroblasts isolated from mild to moderate COPD subjects than in those from non-smokers, suggesting the substantial roles of CCh- and TGF-β1 in pathogenesis of airway fibrosis. Although the present study was performed under controlled experimental settings, ERK5 might be a novel therapeutic target for airway fibrosis and the inhaled GLY and IND combination therapy may be useful for early intervention to prevent the development of airway fibrosis in COPD.

Acknowledgements

We thank I. Taki, L. Ning, N. Okamoto, T. Ito, R. Mineki, and E. Kobayashi for excellent assistance and advice.

Funding

This study was supported by Novartis Pharma K.K. Provision of glycopyrronium and indacaterol from Novartis Pharma AG.

Availability of data and materials

Not applicable.

Authors’ contributions

Conception and design: YN, ST, XL, KT; Acquisition of data: MT, KK, KYN, HT, NK, AO; Analysis and interpretation of data: YN, ST, AO, XL, KT; Drafting the manuscript for important intellectual content: YN, ST, MT, KK, KYN, HT, NK; Revising the manuscript for important intellectual content: YN, ST, AO, XL, KT; Final approval of the manuscript: YN, ST, MT, KK, KYN, HT, NK, AO, XL, KT.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
The Institutional Review Board at the Juntendo University School of Medicine approved the procedures. All patients provided written, informed consent (No. 2013051).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Berankova K, Uhlik J, Honkova L, Pohunek P. Structural changes in the bronchial mucosa of young children at risk of developing asthma. Pediatr Allergy Immunol. 2014;25:136–42.CrossRefPubMed Berankova K, Uhlik J, Honkova L, Pohunek P. Structural changes in the bronchial mucosa of young children at risk of developing asthma. Pediatr Allergy Immunol. 2014;25:136–42.CrossRefPubMed
2.
Zurück zum Zitat Bhatt SP, Soler X, Wang X, Murray S, Anzueto AR, Beaty TH, Boriek AM, Casaburi R, Criner GJ, Diaz AA, et al. Association Between Functional Small Airways Disease and FEV Decline in COPD. Am J Respir Crit Care Med. 2016. Bhatt SP, Soler X, Wang X, Murray S, Anzueto AR, Beaty TH, Boriek AM, Casaburi R, Criner GJ, Diaz AA, et al. Association Between Functional Small Airways Disease and FEV Decline in COPD. Am J Respir Crit Care Med. 2016.
3.
Zurück zum Zitat Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–53.CrossRefPubMed Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–53.CrossRefPubMed
4.
Zurück zum Zitat Rennard SI, Wachenfeldt K. Rationale and emerging approaches for targeting lung repair and regeneration in the treatment of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2011;8:368–75.CrossRefPubMed Rennard SI, Wachenfeldt K. Rationale and emerging approaches for targeting lung repair and regeneration in the treatment of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2011;8:368–75.CrossRefPubMed
5.
Zurück zum Zitat Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, Okada Y, Yamasawa F, Nakahara K, Umeda A. Increased expression of transforming growth factor-beta1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD). Am J Respir Crit Care Med. 2001;163:1476–83.CrossRefPubMed Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, Okada Y, Yamasawa F, Nakahara K, Umeda A. Increased expression of transforming growth factor-beta1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD). Am J Respir Crit Care Med. 2001;163:1476–83.CrossRefPubMed
6.
Zurück zum Zitat Gohy ST, Detry BR, Lecocq M, Bouzin C, Weynand BA, Amatngalim GD, Sibille YM, Pilette C. Polymeric immunoglobulin receptor down-regulation in chronic obstructive pulmonary disease. Persistence in the cultured epithelium and role of transforming growth factor-beta. Am J Respir Crit Care Med. 2014;190:509–21.CrossRefPubMed Gohy ST, Detry BR, Lecocq M, Bouzin C, Weynand BA, Amatngalim GD, Sibille YM, Pilette C. Polymeric immunoglobulin receptor down-regulation in chronic obstructive pulmonary disease. Persistence in the cultured epithelium and role of transforming growth factor-beta. Am J Respir Crit Care Med. 2014;190:509–21.CrossRefPubMed
7.
Zurück zum Zitat Kistemaker LE, Oenema TA, Meurs H, Gosens R. Regulation of airway inflammation and remodeling by muscarinic receptors: perspectives on anticholinergic therapy in asthma and COPD. Life Sci. 2012;91:1126–33.CrossRefPubMed Kistemaker LE, Oenema TA, Meurs H, Gosens R. Regulation of airway inflammation and remodeling by muscarinic receptors: perspectives on anticholinergic therapy in asthma and COPD. Life Sci. 2012;91:1126–33.CrossRefPubMed
8.
Zurück zum Zitat Grainge CL, Lau LC, Ward JA, Dulay V, Lahiff G, Wilson S, Holgate S, Davies DE, Howarth PH. Effect of bronchoconstriction on airway remodeling in asthma. N Engl J Med. 2011;364:2006–15.CrossRefPubMed Grainge CL, Lau LC, Ward JA, Dulay V, Lahiff G, Wilson S, Holgate S, Davies DE, Howarth PH. Effect of bronchoconstriction on airway remodeling in asthma. N Engl J Med. 2011;364:2006–15.CrossRefPubMed
9.
Zurück zum Zitat Wessler I, Kilbinger H, Bittinger F, Unger R, Kirkpatrick CJ. The non-neuronal cholinergic system in humans: expression, function and pathophysiology. Life Sci. 2003;72:2055–61.CrossRefPubMed Wessler I, Kilbinger H, Bittinger F, Unger R, Kirkpatrick CJ. The non-neuronal cholinergic system in humans: expression, function and pathophysiology. Life Sci. 2003;72:2055–61.CrossRefPubMed
10.
Zurück zum Zitat Klapproth H, Reinheimer T, Metzen J, Munch M, Bittinger F, Kirkpatrick CJ, Hohle KD, Schemann M, Racke K, Wessler I. Non-neuronal acetylcholine, a signalling molecule synthezised by surface cells of rat and man. Naunyn Schmiedebergs Arch Pharmacol. 1997;355:515–23.CrossRefPubMed Klapproth H, Reinheimer T, Metzen J, Munch M, Bittinger F, Kirkpatrick CJ, Hohle KD, Schemann M, Racke K, Wessler I. Non-neuronal acetylcholine, a signalling molecule synthezised by surface cells of rat and man. Naunyn Schmiedebergs Arch Pharmacol. 1997;355:515–23.CrossRefPubMed
11.
Zurück zum Zitat Kummer W, Wiegand S, Akinci S, Schinkel AH, Wess J, Koepsell H, Haberberger RV, Lips KS. Role of acetylcholine and muscarinic receptors in serotonin-induced bronchoconstriction in the mouse. J Mol Neurosci. 2006;30:67–8.CrossRefPubMed Kummer W, Wiegand S, Akinci S, Schinkel AH, Wess J, Koepsell H, Haberberger RV, Lips KS. Role of acetylcholine and muscarinic receptors in serotonin-induced bronchoconstriction in the mouse. J Mol Neurosci. 2006;30:67–8.CrossRefPubMed
12.
Zurück zum Zitat Matthiesen S, Bahulayan A, Holz O, Racke K. MAPK pathway mediates muscarinic receptor-induced human lung fibroblast proliferation. Life Sci. 2007;80:2259–62.CrossRefPubMed Matthiesen S, Bahulayan A, Holz O, Racke K. MAPK pathway mediates muscarinic receptor-induced human lung fibroblast proliferation. Life Sci. 2007;80:2259–62.CrossRefPubMed
13.
Zurück zum Zitat Matthiesen S, Bahulayan A, Kempkens S, Haag S, Fuhrmann M, Stichnote C, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of human lung fibroblast proliferation. Am J Respir Cell Mol Biol. 2006;35:621–7.CrossRefPubMed Matthiesen S, Bahulayan A, Kempkens S, Haag S, Fuhrmann M, Stichnote C, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of human lung fibroblast proliferation. Am J Respir Cell Mol Biol. 2006;35:621–7.CrossRefPubMed
14.
Zurück zum Zitat Haag S, Matthiesen S, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of collagen synthesis in human lung fibroblasts. Eur Respir J. 2008;32:555–62.CrossRefPubMed Haag S, Matthiesen S, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of collagen synthesis in human lung fibroblasts. Eur Respir J. 2008;32:555–62.CrossRefPubMed
15.
Zurück zum Zitat Milara J, Serrano A, Peiro T, Gavalda A, Miralpeix M, Morcillo EJ, Cortijo J. Aclidinium inhibits human lung fibroblast to myofibroblast transition. Thorax. 2012;67:229–37.CrossRefPubMed Milara J, Serrano A, Peiro T, Gavalda A, Miralpeix M, Morcillo EJ, Cortijo J. Aclidinium inhibits human lung fibroblast to myofibroblast transition. Thorax. 2012;67:229–37.CrossRefPubMed
16.
17.
Zurück zum Zitat Kim S, Lim JH, Woo CH. ERK5 inhibition ameliorates pulmonary fibrosis via regulating Smad3 acetylation. Am J Pathol. 2013;183:1758–68.CrossRefPubMed Kim S, Lim JH, Woo CH. ERK5 inhibition ameliorates pulmonary fibrosis via regulating Smad3 acetylation. Am J Pathol. 2013;183:1758–68.CrossRefPubMed
18.
Zurück zum Zitat Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al.: Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. Respirology 2017. Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al.: Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. Respirology 2017.
19.
Zurück zum Zitat Kerstjens HA, Engel M, Dahl R, Paggiaro P, Beck E, Vandewalker M, Sigmund R, Seibold W, Moroni-Zentgraf P, Bateman ED. Tiotropium in asthma poorly controlled with standard combination therapy. N Engl J Med. 2012;367:1198–207.CrossRefPubMed Kerstjens HA, Engel M, Dahl R, Paggiaro P, Beck E, Vandewalker M, Sigmund R, Seibold W, Moroni-Zentgraf P, Bateman ED. Tiotropium in asthma poorly controlled with standard combination therapy. N Engl J Med. 2012;367:1198–207.CrossRefPubMed
20.
Zurück zum Zitat Dahl R, Chapman KR, Rudolf M, Mehta R, Kho P, Alagappan VK, Chen H, Banerji D. Safety and efficacy of dual bronchodilation with QVA149 in COPD patients: the ENLIGHTEN study. Respir Med. 2013;107:1558–67.CrossRefPubMed Dahl R, Chapman KR, Rudolf M, Mehta R, Kho P, Alagappan VK, Chen H, Banerji D. Safety and efficacy of dual bronchodilation with QVA149 in COPD patients: the ENLIGHTEN study. Respir Med. 2013;107:1558–67.CrossRefPubMed
21.
Zurück zum Zitat Holz O, Zuhlke I, Jaksztat E, Muller KC, Welker L, Nakashima M, Diemel KD, Branscheid D, Magnussen H, Jorres RA. Lung fibroblasts from patients with emphysema show a reduced proliferation rate in culture. Eur Respir J. 2004;24:575–9.CrossRefPubMed Holz O, Zuhlke I, Jaksztat E, Muller KC, Welker L, Nakashima M, Diemel KD, Branscheid D, Magnussen H, Jorres RA. Lung fibroblasts from patients with emphysema show a reduced proliferation rate in culture. Eur Respir J. 2004;24:575–9.CrossRefPubMed
23.
Zurück zum Zitat Bell E, Ivarsson B, Merrill C. Production of a tissue-like structure by contraction of collagen lattices by human fibroblasts of different proliferative potential in vitro. Proc Natl Acad Sci U S A. 1979;76:1274–8.CrossRefPubMedPubMedCentral Bell E, Ivarsson B, Merrill C. Production of a tissue-like structure by contraction of collagen lattices by human fibroblasts of different proliferative potential in vitro. Proc Natl Acad Sci U S A. 1979;76:1274–8.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Xu BE, Stippec S, Lenertz L, Lee BH, Zhang W, Lee YK, Cobb MH. WNK1 activates ERK5 by an MEKK2/3-dependent mechanism. J Biol Chem. 2004;279:7826–31.CrossRefPubMed Xu BE, Stippec S, Lenertz L, Lee BH, Zhang W, Lee YK, Cobb MH. WNK1 activates ERK5 by an MEKK2/3-dependent mechanism. J Biol Chem. 2004;279:7826–31.CrossRefPubMed
25.
Zurück zum Zitat Reinheimer T, Bernedo P, Klapproth H, Oelert H, Zeiske B, Racke K, Wessler I. Acetylcholine in isolated airways of rat, guinea pig, and human: species differences in role of airway mucosa. Am J Physiol. 1996;270:L722–728.PubMed Reinheimer T, Bernedo P, Klapproth H, Oelert H, Zeiske B, Racke K, Wessler I. Acetylcholine in isolated airways of rat, guinea pig, and human: species differences in role of airway mucosa. Am J Physiol. 1996;270:L722–728.PubMed
26.
Zurück zum Zitat Liu X, Wen FQ, Kobayashi T, Abe S, Fang Q, Piek E, Bottinger EP, Roberts AB, Rennard SI. Smad3 mediates the TGF-beta-induced contraction of type I collagen gels by mouse embryo fibroblasts. Cell Motil Cytoskeleton. 2003;54:248–53.CrossRefPubMed Liu X, Wen FQ, Kobayashi T, Abe S, Fang Q, Piek E, Bottinger EP, Roberts AB, Rennard SI. Smad3 mediates the TGF-beta-induced contraction of type I collagen gels by mouse embryo fibroblasts. Cell Motil Cytoskeleton. 2003;54:248–53.CrossRefPubMed
27.
Zurück zum Zitat Kobayashi T, Liu X, Wen FQ, Kohyama T, Shen L, Wang XQ, Hashimoto M, Mao L, Togo S, Kawasaki S, et al. Smad3 mediates TGF-beta1-induced collagen gel contraction by human lung fibroblasts. Biochem Biophys Res Commun. 2006;339:290–5.CrossRefPubMed Kobayashi T, Liu X, Wen FQ, Kohyama T, Shen L, Wang XQ, Hashimoto M, Mao L, Togo S, Kawasaki S, et al. Smad3 mediates TGF-beta1-induced collagen gel contraction by human lung fibroblasts. Biochem Biophys Res Commun. 2006;339:290–5.CrossRefPubMed
28.
Zurück zum Zitat Bartels C, Looby M, Sechaud R, Kaiser G. Determination of the pharmacokinetics of glycopyrronium in the lung using a population pharmacokinetic modelling approach. Br J Clin Pharmacol. 2013;76:868–79.CrossRefPubMedPubMedCentral Bartels C, Looby M, Sechaud R, Kaiser G. Determination of the pharmacokinetics of glycopyrronium in the lung using a population pharmacokinetic modelling approach. Br J Clin Pharmacol. 2013;76:868–79.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Khindri S, Sabo R, Harris S, Woessner R, Jennings S, Drollmann AF. Cardiac safety of indacaterol in healthy subjects: a randomized, multidose, placebo- and positive-controlled, parallel-group thorough QT study. BMC Pulm Med. 2011;11:31.CrossRefPubMedPubMedCentral Khindri S, Sabo R, Harris S, Woessner R, Jennings S, Drollmann AF. Cardiac safety of indacaterol in healthy subjects: a randomized, multidose, placebo- and positive-controlled, parallel-group thorough QT study. BMC Pulm Med. 2011;11:31.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Xu ZP, Devillier P, Xu GN, Qi H, Zhu L, Zhou W, Hou LN, Tang YB, Yang K, Yu ZH, et al. TNF-alpha-induced CXCL8 production by A549 cells: involvement of the non-neuronal cholinergic system. Pharmacol Res. 2013;68:16–23.CrossRefPubMed Xu ZP, Devillier P, Xu GN, Qi H, Zhu L, Zhou W, Hou LN, Tang YB, Yang K, Yu ZH, et al. TNF-alpha-induced CXCL8 production by A549 cells: involvement of the non-neuronal cholinergic system. Pharmacol Res. 2013;68:16–23.CrossRefPubMed
31.
Zurück zum Zitat Pera T, Zuidhof A, Valadas J, Smit M, Schoemaker RG, Gosens R, Maarsingh H, Zaagsma J, Meurs H. Tiotropium inhibits pulmonary inflammation and remodelling in a guinea pig model of COPD. Eur Respir J. 2011;38:789–96.CrossRefPubMed Pera T, Zuidhof A, Valadas J, Smit M, Schoemaker RG, Gosens R, Maarsingh H, Zaagsma J, Meurs H. Tiotropium inhibits pulmonary inflammation and remodelling in a guinea pig model of COPD. Eur Respir J. 2011;38:789–96.CrossRefPubMed
32.
Zurück zum Zitat Mio T, Liu X, Toews ML, Adachi Y, Romberger DJ, Spurzem JR, Rennard SI. Bradykinin augments fibroblast-mediated contraction of released collagen gels. Am J Physiol Lung Cell Mol Physiol. 2001;281:L164–171.PubMed Mio T, Liu X, Toews ML, Adachi Y, Romberger DJ, Spurzem JR, Rennard SI. Bradykinin augments fibroblast-mediated contraction of released collagen gels. Am J Physiol Lung Cell Mol Physiol. 2001;281:L164–171.PubMed
33.
Zurück zum Zitat Profita M, Bonanno A, Siena L, Bruno A, Ferraro M, Montalbano AM, Albano GD, Riccobono L, Casarosa P, Pieper MP, Gjomarkaj M. Smoke, choline acetyltransferase, muscarinic receptors, and fibroblast proliferation in chronic obstructive pulmonary disease. J Pharmacol Exp Ther. 2009;329:753–63.CrossRefPubMed Profita M, Bonanno A, Siena L, Bruno A, Ferraro M, Montalbano AM, Albano GD, Riccobono L, Casarosa P, Pieper MP, Gjomarkaj M. Smoke, choline acetyltransferase, muscarinic receptors, and fibroblast proliferation in chronic obstructive pulmonary disease. J Pharmacol Exp Ther. 2009;329:753–63.CrossRefPubMed
34.
Zurück zum Zitat Urushihara M, Takamatsu M, Shimizu M, Kondo S, Kinoshita Y, Suga K, Kitamura A, Matsuura S, Yoshizumi M, Tamaki T, et al. ERK5 activation enhances mesangial cell viability and collagen matrix accumulation in rat progressive glomerulonephritis. Am J Physiol Renal Physiol. 2010;298:F167–176.CrossRefPubMed Urushihara M, Takamatsu M, Shimizu M, Kondo S, Kinoshita Y, Suga K, Kitamura A, Matsuura S, Yoshizumi M, Tamaki T, et al. ERK5 activation enhances mesangial cell viability and collagen matrix accumulation in rat progressive glomerulonephritis. Am J Physiol Renal Physiol. 2010;298:F167–176.CrossRefPubMed
35.
Zurück zum Zitat Garcia-Hoz C, Sanchez-Fernandez G, Diaz-Meco MT, Moscat J, Mayor F, Ribas C. G alpha(q) acts as an adaptor protein in protein kinase C zeta (PKCzeta)-mediated ERK5 activation by G protein-coupled receptors (GPCR). J Biol Chem. 2010;285:13480–9.CrossRefPubMedPubMedCentral Garcia-Hoz C, Sanchez-Fernandez G, Diaz-Meco MT, Moscat J, Mayor F, Ribas C. G alpha(q) acts as an adaptor protein in protein kinase C zeta (PKCzeta)-mediated ERK5 activation by G protein-coupled receptors (GPCR). J Biol Chem. 2010;285:13480–9.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Badshah II, Baines DL, Dockrell ME. Erk5 is a mediator to TGFbeta1-induced loss of phenotype and function in human podocytes. Front Pharmacol. 2014;5:71.CrossRefPubMedPubMedCentral Badshah II, Baines DL, Dockrell ME. Erk5 is a mediator to TGFbeta1-induced loss of phenotype and function in human podocytes. Front Pharmacol. 2014;5:71.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Cazzola M, Calzetta L, Segreti A, Facciolo F, Rogliani P, Matera MG. Translational Study Searching for Synergy between Glycopyrronium and Indacaterol. COPD. 2015;12:175–81.CrossRefPubMed Cazzola M, Calzetta L, Segreti A, Facciolo F, Rogliani P, Matera MG. Translational Study Searching for Synergy between Glycopyrronium and Indacaterol. COPD. 2015;12:175–81.CrossRefPubMed
38.
Zurück zum Zitat Cazzola M, Calzetta L, Page CP, Rogliani P, Facciolo F, Gavalda A, Matera MG. Pharmacological characterization of the interaction between aclidinium bromide and formoterol fumarate on human isolated bronchi. Eur J Pharmacol. 2014;745:135–43.CrossRefPubMed Cazzola M, Calzetta L, Page CP, Rogliani P, Facciolo F, Gavalda A, Matera MG. Pharmacological characterization of the interaction between aclidinium bromide and formoterol fumarate on human isolated bronchi. Eur J Pharmacol. 2014;745:135–43.CrossRefPubMed
39.
Zurück zum Zitat Ravi Kannan R, Przekwas AJ, Singh N, Delvadia R, Tian G, Walenga R. Pharmaceutical aerosols deposition patterns from a Dry Powder Inhaler: Euler Lagrangian prediction and validation. Med Eng Phys. 2016. Ravi Kannan R, Przekwas AJ, Singh N, Delvadia R, Tian G, Walenga R. Pharmaceutical aerosols deposition patterns from a Dry Powder Inhaler: Euler Lagrangian prediction and validation. Med Eng Phys. 2016.
Metadaten
Titel
Combination of glycopyrronium and indacaterol inhibits carbachol-induced ERK5 signal in fibrotic processes
verfasst von
Yukiko Namba
Shinsaku Togo
Miniwan Tulafu
Kotaro Kadoya
Kumi Yoneda Nagahama
Hikari Taka
Naoko Kaga
Akira Orimo
Xiangde Liu
Kazuhisa Takahashi
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2017
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-017-0529-6

Weitere Artikel der Ausgabe 1/2017

Respiratory Research 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.