Next Article in Journal
Inhibition of CDK1 by RO-3306 Exhibits Anti-Tumorigenic Effects in Ovarian Cancer Cells and a Transgenic Mouse Model of Ovarian Cancer
Previous Article in Journal
Insights into the Mechanism of Action of Antipsychotic Drugs Derived from Animal Models: Standard of Care versus Novel Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Ivy Leaf Dry Extract EA 575® Has an Inhibitory Effect on the Signalling Cascade of Adenosine Receptor A2B

Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(15), 12373; https://doi.org/10.3390/ijms241512373
Submission received: 7 July 2023 / Revised: 26 July 2023 / Accepted: 26 July 2023 / Published: 3 August 2023
(This article belongs to the Section Molecular Plant Sciences)

Abstract

:
Ivy leaf dry extract EA 575® is used to improve complaints of chronic inflammatory bronchial diseases and acute inflammation of the respiratory tract accompanied by coughing. Its mechanism of action has so far been explained by influencing β2-adrenergic signal transduction. In the present study, we investigated a possible influence on adenosine receptor A2B (A2BAR) signalling, as it has been described to play a significant and detrimental role in chronic inflammatory airway diseases. The influence of EA 575® on A2BAR signalling was assessed with measurements of dynamic mass redistribution. Subsequently, the effects on A2BAR-mediated second messenger cAMP levels, β-arrestin 2 recruitment, and cAMP response element (CRE) activation were examined using luciferase-based HEK293 reporter cell lines. Lastly, the impact on A2BAR-mediated IL-6 release in Calu-3 epithelial lung cells was investigated via the Lumit™ Immunoassay. Additionally, the adenosine receptor subtype mediating these effects was specified, and A2BAR was found to be responsible. The present study demonstrates an inhibitory influence of EA 575® on A2BAR-mediated general cellular response, cAMP levels, β-arrestin 2 recruitment, CRE activation, and IL-6 release. Since these EA 575®-mediated effects occur within a time frame of several hours of incubation, its mode of action can be described as indirect. The present data are the first to describe an inhibitory effect of EA 575® on A2BAR signalling. This may offer an explanation for the beneficial clinical effects of the extract in adjuvant asthma therapy.

1. Introduction

The use of medicinal products containing ivy leaf dry extract EA 575® is recommended for the treatment of chronic inflammatory airway diseases and acute respiratory tract inflammation accompanied by coughing [1,2,3,4]. Until now, the main mechanism of action has been explained by influencing the β2-adrenergic receptor (β2-AR). It has been shown that α-hederin identified in EA 575® indirectly inhibits the GRK2-mediated phosphorylation of β2-AR [5], which is the reason for the decrease in the recruitment of β-arrestin 2 by EA 575® [6]. This leads to the inhibition of β2-AR internalisation [7,8,9], which, in turn, results in increased β2-adrenergic responsiveness, as evidenced by a corresponding increase in receptor binding and cAMP formation [6,8]. This biased signalling by EA 575® provides an explanation for the bronchospasmolytic and secretolytic effects and a reduction in β-arrestin-mediated negative adverse effects [6]. The anti-inflammatory effects of EA 575® are substantiated by the β-arrestin-independent inhibition of NFκB, presumably by the inhibition of IĸBα phosphorylation, leading to decreased IL-6 release [6,10,11].
Since additional receptor classes are involved in the pathogenesis of these respiratory diseases, we investigated other signalling pathways that may be affected by EA 575® and might explain its beneficial effects in the treatment of inflammatory airway diseases. Many authors have described the harmful influence of adenosine via its adenosine receptor A2B (A2BAR) in chronic inflammatory airway diseases [12,13,14,15] based on the following findings.
Elevated adenosine concentrations were found in the bronchoalveolar lavage fluid (BALF) and exhaled breath condensate (EBC) of patients with chronic respiratory diseases such as asthma and chronic obstructive pulmonary disease (COPD), indicating increased adenosine levels in the lungs [16,17,18,19]. These elevations of adenosine were correlated to decreased forced expiratory volumes in 1 s (FEV1) and higher Global Initiative for Chronic Obstructive Lung Disease (GOLD) stages [19,20]. Inhalation of adenosine or adenosine monophosphate (AMP) causes bronchoconstriction in patients with asthma and COPD but not in healthy subjects [21,22,23]. Remarkably, elevated transcript levels of A2BAR were recovered from the lung tissues of patients with severe COPD, pulmonary fibrosis, and pulmonary arterial hypertension (PAH) [24,25]. Further elevation of transcript and A2BAR protein levels was determined in the lung tissues of patients with COPD when accompanied by pulmonary hypertension, indicating a correlation with disease severity [26].
Several experiments in mouse models provide additional evidence for the detrimental impact of adenosine and A2BAR on airway diseases such as asthma, COPD, and pulmonary fibrosis. Genetically altered adenosine deaminase (ADA)-deficient mice developed severe pulmonary inflammation and airway remodelling as observed in these disorders [27,28]. Lowering adenosine concentrations via ADA enzyme therapy ameliorates lung injury, indicating a correlation between adenosine levels and inflammation as well as fibrosis in the lung [29,30]. Attenuation of pulmonary inflammation, fibrosis, and airway enlargement can also be achieved by administration of specific A2BAR antagonists [26,31]. Transcript levels of A2BAR were also increased in this model and could be decreased by specific antagonism of A2BAR [28,31,32].
Similar observations were made in another mouse model. Mice treated with bleomycin exhibited elevated levels of adenosine and transcripts of A2BAR in their lungs and developed pulmonary fibrosis and inflammation. This disease progression could be counteracted with specific A2BAR antagonists [31,33].
The proinflammatory and profibrotic cytokine interleukin-6 (IL-6) is associated with the signalling of adenosine via A2BAR and also plays a pivotal role in these pulmonary disorders [13,34]. Patients with asthma exhibit elevated levels of IL-6 in their sputum and BALF compared to healthy subjects [35,36,37] and compared to asymptomatic asthmatics [38,39], who already have higher serum IL-6 concentrations [40]. A further IL-6 increase in serum and BALF occur after allergen inhalation and during allergic attacks [40,41]. Most remarkably, the sputum IL-6 levels of asthmatics inversely correlate with FEV1 and peak expiratory flow [37,42,43].
Elevations of IL-6 have also been found in the sputum and serum of patients with COPD compared to healthy controls [44,45], further increasing during exacerbations [46]. As in asthmatics, higher IL-6 levels inversely correlate with lung function [45,46]. Furthermore, correlations with disease severity [47], GOLD stage and BODE index [44], and even mortality [48] have been described.
Correspondingly, genetic IL-6-knockout or anti-IL-6 antibody treatment led to reduced pulmonary inflammation and fibrosis in both of the aforementioned mouse models [49,50]. Further evidence for the deleterious interaction of IL-6 and A2BAR has been provided by experiments using genetic A2BAR-knockouts. While the adenosine receptor agonist NECA caused increased IL-6 secretion in mouse macrophages, reduced IL-6 levels were found in A2BAR-knockout mice [51,52]. Bleomycin-treated mice also exhibited elevated IL-6 levels, which could be decreased by A2BAR-knockout, resulting in improved lung function as well as attenuated pulmonary fibrosis and hypertension [25,33,53]. Moreover, ADA enzyme therapy in ADA-deficient mice was able to reduce IL-6 levels [50].
The present study demonstrates the influence of the ivy leaf dry extract EA 575® on adenosine receptor A2B signalling and subsequent IL-6 release. These processes have been described to play a significant and detrimental role in chronic inflammatory pulmonary diseases. The findings may provide an explanation for the positive clinical effects of EA 575® in adjuvant asthma therapy by inhibiting A2BAR signalling.

2. Results

2.1. Dynamic Mass Redistribution Measurements

To investigate the influence of the ivy leaf dry extract, EA 575®, on the cellular reaction evoked by the stimulation of A2BAR, dynamic mass redistribution (DMR) assays were performed. The cellular reaction of HEK cells to 0.5 µM of the A2BAR agonist BAY 60-6583 resulted in a positive wavelength shift mediated by the dynamic mass redistribution of intracellular particles, which was significantly and dose-dependently inhibited up to 45.95 ± 12.36% by pre-incubation with 160–240 µg/mL EA 575® for 16 h (Figure 1A).
In order to examine whether the observed effect results from direct inhibition of the receptor, pre-incubation of HEK cells with EA 575® was conducted for 1 and 16 h. A reduction in wavelength shifts was exclusively observed with 16 h of pre-incubation time, while a shorter incubation period of 1 h did not alter BAY 60-6583-mediated wavelength shifts (Figure 1B).

2.2. cAMP Measurements

The influence of EA 575® on the intracellular cAMP level induced by the stimulation of A2BAR was determined using HEK cells stably expressing luciferase fused with a cAMP binding domain (GloSensor™, Promega, Mannheim, Germany). Elevation of the second messenger cAMP was elicited by simultaneous stimulation with 1 µM BAY 60-6583 and 1 µM forskolin for one hour. Pre-incubation with 160–240 µg/mL EA 575® for 16 h led to a significant and dose-dependent reduction in cAMP levels by up to 39.02 ± 9.92%, compared to stimulated control cells not pre-incubated with EA 575® (Figure 2A).
In order to determine if the observed effect was mediated by direct inhibition of the receptor and after what time it occurred, different pre-incubation periods were tested. A pre-incubation time of at least 8 h was required to observe a significant decrease in cAMP levels (Figure 2B).

2.3. Measurements of β-Arrestin 2 Recruitment

To investigate another possible process involved in the inhibition of A2BAR by EA 575®, measurements of β-arrestin 2 recruitment were performed. HEK cells transiently expressing A2BAR-LgBiT and SmBiT-β-arrestin 2 generated a luminescence signal after stimulation with the unspecific adenosine receptor agonist NECA or BAY 60-6583, indicating β-arrestin 2 recruitment to A2BAR. Pre-incubation with 160 µg/mL EA 575® for 16 h led to significant inhibition of both NECA- and BAY 60-6583-mediated β-arrestin 2 recruitment by 16.90 ± 4.69% and 9.31 ± 4.82%, respectively (Figure 3).

2.4. Measurements of CRE Activation

The influence of EA 575® on cAMP response elements (CRE) was investigated using HEK cells transiently expressing NanoLuc®-PEST under the control of a promoter with cAMP response elements.
The impact of EA 575® on adenosine-mediated CRE activation was examined first. Compared to stimulated control cells not pre-incubated with EA 575®, pre-incubation with 80–240 µg/mL EA 575® for 16 h led to a significant reduction by a maximum of 24.23 ± 9.38% of the CRE activation mediated by stimulation with 100 µM adenosine (Figure 4A). Pre-incubation with EA 575® also specifically inhibited A2BAR-mediated CRE activation by 10 µM BAY 60-6583 in a dose-dependent manner up to 20.18 ± 2.67%, being significant at 80–240 µg/mL (Figure 4B).
Since adenosine is a non-specific agonist binding to all adenosine receptor subtypes, identification of the receptor accountable for the observed effect was addressed next. For this purpose, cells were pre-incubated with the A2BAR-specific antagonist PSB-603 or the A2AAR-specific antagonist SCH 442416 for one hour prior to stimulation with 100 µM adenosine. PSB-603 was able to significantly inhibit the adenosine-mediated CRE activation by 33.37 ± 6.63% at a concentration of 1 µM, while SCH 442416 significantly reduced the CRE activation by 54.11 ± 7.90% at a concentration of 10 µM. Lower concentrations of both antagonists did not show a significant reduction in the signal, whereas 0.1 µM SCH 442416 significantly increased CRE activation by 34.64 ± 12.25% (Figure 4C).
CRE activation could also be mediated by stimulation with 10 µM of the A2AAR agonist CGS 21680, resulting in a significant 3.10 ± 0.24-fold increase in the luminescence signal compared to completely untreated control cells. Nevertheless, this A2AAR stimulation elicited a smaller effect than 10 µM BAY 60-6583, which showed a 5.14 ± 0.93-fold elevation, and 100 µM adenosine, for which the signal was highest with a 7.06 ± 1.09-fold increase (Figure 4D).

2.5. IL-6 Measurements

IL-6 release of Calu-3 cells was provoked by stimulation with 100 µM adenosine and measured as luminescence using the Lumit™ IL-6 (Human) Immunoassay by Promega.
The influence of EA 575® on this non-specifically mediated IL-6 release was investigated. Compared to stimulated control cells not pre-incubated with EA 575®, pre-incubation with 80–240 µg/mL EA 575® for 16 h led to a significant and dose-dependent reduction by up to 33.73 ± 10.76% of the adenosine-mediated IL-6 release (Figure 5A). Also, when pre-incubating with 40–240 µg/mL EA 575®, BAY 60-6583-mediated IL-6 release was significantly inhibited up to 36.36 ± 2.16% in a dose-dependent manner (Figure 5B).
Receptor-specific antagonists PSB-603 and SCH 442416 were employed to identify the specific receptor subtype involved in the induction of adenosine-mediated IL-6 release. PSB-603 was able to significantly inhibit the adenosine-mediated IL-6 release by 22.56 ± 8.19% at a concentration of 1 µM, whereas SCH 442416 increased the concentration of IL-6 even further by up to 24.99 ± 9.57% (Figure 5C).
In another approach, stimulation was performed with 10 µM BAY 60-6583, resulting in a 2.91 ± 0.35-fold increase in IL-6 release, while A2AAR agonist CGS 21680 had no significant effect, showing a maximal 1.33 ± 0.20-fold IL-6 increase, compared to completely untreated control cells (Figure 5D).

2.6. Measurements of NFκB Transcriptional Activity

In order to evaluate whether the observed effect of an IL-6 release via adenosine receptors in Calu-3 cells was NFκB-dependent, the influence of adenosine receptor agonists on NFκB transcriptional activity was investigated using an NFκB reporter gene cell line. Calu-3 cells stably expressing a secreted NanoLuc® under the control of an NFκB-binding sequence reacted with a 1.62 ± 0.11-fold increase in the luminescence signal to stimulation with 25 ng/mL TNFα for 3 h. However, neither 10–100 µM adenosine, nor 1–10 µM NECA, nor 1–10 µM BAY 60-6583 showed any effect on the NFκB transcriptional activity (Figure 6).

3. Discussion

In the present study, we further explored the mechanism of action of the ivy leaf dry extract EA 575® to explain its positive effects on inflammatory airway diseases [2,3,4]. Since adenosine receptor A2B (A2BAR) plays an essential role in the pathogenesis of chronic inflammatory airway diseases such as asthma, COPD, and pulmonary fibrosis [12,13,14,15], we investigated a possible effect of EA 575® on this receptor.
We first examined a possible impact of EA 575® on the cellular response of HEK cells to stimulation of the A2BAR using DMR measurements. This label-free technology is well-suited for an initial investigation of the general influence of ligands on GPCR signalling due to its capability to provide a holistic overview of the complex cellular response [54]. We observed the dose-dependent inhibition of the cellular reaction to A2BAR stimulation by EA 575®. This effect was observed after 16 h of pre-incubation, while no impact was seen after a short incubation time of 1 h, indicating indirect inhibition of A2BAR.
To further specify the influenced cellular response observed in the DMR experiments, second messenger cAMP was assessed. In alignment with the DMR results, a corresponding decrease in cAMP levels was observed after pre-treatment with EA 575® under A2BAR stimulatory conditions. A similar effect was shown for the highly selective A2BAR antagonist PSB-603 using the same cAMP biosensor technology [55]. Next, we challenged the required minimum incubation time period and found that at least 8 h of EA 575® pre-incubation was necessary to mediate a reduction in cAMP levels. Such long incubation periods are unlikely to be in accordance with a direct mode of action, as these ligands typically compete for receptor binding sites within minutes. PSB-603, for example, requires a pre-incubation period of only 30 min in this assay [55]. These findings confirm that EA 575® is able to inhibit the A2BAR signalling pathway and that this can be explained by an indirect mechanism of action. Since adenosine receptors are ubiquitously expressed in humans, this could be advantageous over orthosteric antagonism in terms of adverse effects [56]. To our knowledge, the present study is the first to show the influence of an ivy extract on adenosine receptor A2B.
To further investigate the mode of action of EA 575® on A2BAR and to determine whether it also influences other downstream signalling pathways, the recruitment of β-arrestin 2 to A2BAR was examined. For this purpose, a slightly modified assay system recently described by Saecker et al. was used [57]. In this system, A2BAR is tagged with LgBiT and β-arrestin 2 with SmBiT. When β-arrestin 2 recruitment occurs specifically to A2BAR, luminescence is generated by reversible complementation of a nanoluciferase. Stimulation was performed using the unspecific adenosine receptor agonist NECA, as well as the specific A2BAR agonist BAY 60-6583. Since A2BAR is the only receptor tagged with LgBiT in this system, the effects detected after stimulation with NECA can also be considered specific for this receptor in this case. EA 575® caused a reduction in β-arrestin 2 recruitment to A2BAR and thus affected the receptor signalling in an inhibitory manner in several ways. This is remarkable as under β2-AR stimulatory conditions, EA 575® inhibits β-arrestin 2 recruitment while enhancing G protein/cAMP signalling [6].
In the downstream signalling cascade of Gs protein-coupled receptors such as A2BAR, cAMP causes phosphorylation of cAMP response element-binding protein via PKA, PKC, and ERK, and can thereby activate cAMP response elements (CRE) located in promoter regions, thus affecting the transcriptional activity of genes [58,59,60,61]. Since EA 575® reduced cAMP levels under A2BAR stimulatory conditions, subsequent inhibition of CRE was expected. Indeed, EA 575® inhibited CRE activation mediated both non-specifically by adenosine as well as A2BAR-specifically by BAY 60-6583. A similar result was found for a specific A2BAR antagonist, which reduced the NECA-mediated phosphorylation of CRE binding protein [62].
One of the genes regulated by CRE, encoding IL-6, plays a crucial role in several inflammatory and airway diseases, such as asthma and COPD [34]. The IL-6 promoter region contains several elements that activate IL-6 expression, one of which is a CRE [63,64,65,66,67]. The release of IL-6 and other inflammatory and chemotactic mediators, in turn, can be mediated by adenosine via the A2BAR signalling pathway, and CRE might be at least one important factor in this signalling cascade [68,69]. Since EA 575® inhibits both A2BAR signalling and CRE activation, an effect on IL-6 release seemed plausible. Therefore, we tested the potential effect of EA 575® on the adenosine-mediated IL-6 release in Calu-3 cells. It was found that EA 575®, in fact, reduces the adenosine-mediated release of IL-6, indicating a possible reduction in IL-6-mediated airway inflammation and fibrosis. This inhibition of A2BAR-mediated IL-6 release could also be reproduced with the specific agonist BAY 60-6583.
This is a new finding that complements the previously published decrease in IL-6 release by EA 575® via inhibition of NFκB. In our experiments, we were able to demonstrate that neither adenosine, NECA, nor BAY 60-6583 influenced NFκB transcriptional activity in Calu-3 cells. Although the IL-6 promoter region contains an NFκB binding element [70,71], the release of IL-6 via adenosine receptors is not mediated by this promoter element. This is in line with the results published by Sitaraman et al., who showed that the NFκB binding site, in contrast to the CRE binding site, is not important for adenosine-mediated IL-6 release [69]. Similarly, Zhong et al. found that NECA does not affect NFκB-mediated transcription but rather affects CRE-mediated transcription [68]. Therefore, in this case, EA 575® affects IL-6 release via another mechanism, which could be the inhibition of CRE.
Additionally, we wanted to specify which adenosine receptor subtype is responsible for the observed effects in the IL-6 and CRE activation assays. Therefore, we investigated the inhibition of the adenosine-mediated CRE activation and IL-6 release with the A2AAR antagonist SCH 442416 or the A2BAR antagonist PSB-603, as these are the predominantly expressed adenosine receptor subtypes in the cell types we used [72,73,74]. Only PSB-603 reduced both CRE activation and IL-6 release after stimulation with adenosine. In contrast, SCH 442416 caused the inhibition of CRE activation but simultaneously led to a slight increase in IL-6 release. In addition, CGS 21680, a specific A2AAR agonist, was shown to increase CRE activation in a dose-dependent manner but had no effect on IL-6 release. These data suggest that A2AAR signalling activates CRE but does not result in an increase in IL-6. This might be because IL-6 is regulated not only by the cAMP response element but also by several other elements, as mentioned above. These findings suggest that adenosine mediates IL-6 release through CRE activation via A2BAR. Furthermore, adenosine mediates IL-6 release only in concentrations as high as 10–100 µM [61,68,69,75]. Considering the affinities of adenosine to the different receptor subtypes (EC50: A1 = 0.31 µM, A2A = 0.73 µM, A2B = 23.5 µM, A3 = 0.29 µM) [76], this also suggests mediation via A2BAR. Taken together, published receptor-affinity data and our results indicate that adenosine-induced IL-6 release is mediated through the A2BAR signalling pathway. These findings disagree with those of Sun et al., who stated that A2AAR, but not A2BAR, is responsible for adenosine-mediated IL-6 release [61], but are consistent with data from Sitaraman et al. and Zhong et al., who both identified A2BAR as being responsible for adenosine-mediated IL-6 release [68,69,75]. Although not particularly distinguishing between the two receptors, several other studies have provided additional evidence for the A2BAR-mediated inhibition of IL-6 release. Elevated IL-6 levels in ADA-deficient and bleomycin-treated mice were reduced by the administration of a specific A2BAR antagonist [31,33]. Additionally, a NECA-mediated increase in IL-6 was reduced by antagonists of A2BAR in these models [33,50]. A reduction in NECA-mediated IL-6 elevation by A2BAR antagonists was also shown in macrophages [24,52]. Secretion of IL-6 after treating pulmonary arterial smooth muscle cells of PAH patients with BAY 60-6583 under hypoxic conditions was also reduced by a specific antagonist of A2BAR [25].
Moreover, A2AAR signalling is described as anti-inflammatory and lung protective, which basically matches our results, suggesting that this receptor does not contribute to IL-6 release but rather attenuates it [12,13,14]. Nevertheless, our findings differ from others in terms of the influence of A2AAR on IL-6. There have been reports of both increases [77] as well as reductions [78,79,80,81] by the stimulation of A2AAR with CGS 21680, whereas other research, similar to our study, found no effect [82,83]. Furthermore, the genetic knockout of A2AAR in mice led to higher expressions of IL-6 in one study [84] but did not affect IL-6 in another study examining an ADA/A2AAR double knockout [85]. However, in other studies, antagonism of A2AAR caused the elevation of IL-6 levels, matching our findings [79,80,82].
Several factors may account for these controversial findings. First, the effect of adenosine may be dependent on its concentration and the stage of the disease. At low levels, adenosine activates high-affinity receptors such as A2AAR, triggering anti-inflammatory pathways. In highly inflammatory environments and chronic disease states such as asthma or COPD, higher levels of adenosine are released. Adenosine concentrations have been estimated to reach about 200 µM in the lungs of asthmatics [16]. At such high concentrations, low-affinity A2BAR is activated, which may lead to further exacerbation of airway inflammation [15]. Inhibition of A2BAR is therefore considered beneficial in chronic inflammatory airway diseases [12,13,14]. In a guinea pig model of asthma, antagonism of A2BAR ameliorated the changes provoked by an allergen challenge, whereas A2AAR antagonism deteriorated them [86]. A2BAR antagonism also proved beneficial in mouse models of chronic lung diseases as it attenuated bronchoconstriction, airway inflammation, pulmonary fibrosis, and airspace enlargement [26,31,33,87].
In conclusion, this offers a possible explanation for the positive clinical effects of the extract in adjuvant asthma therapy by means of a possible reduction in adenosine-mediated inflammation and bronchoconstriction [88]. Still, further research is necessary to fully understand the mechanisms of action of this versatile extract. It is still unclear which constituents play a role in the observed effects. It has been shown that a fraction of an ivy leaf extract enriched in phenolics and flavonoids elicits anti-inflammatory properties [89,90]. Since some of the enriched substances have also been identified in EA 575®, future investigations should be performed with the pure compounds. However, the results presented here provide a rationale for further studies in animals or human subjects to prove further effects regarding chronic airway diseases such as asthma or COPD.

4. Materials and Methods

4.1. Chemicals

Ivy leaf dry extract EA 575® (DER 5-7.5:1, 30% ethanol; batch number 14B0310) was received from Engelhard Arzneimittel (Niederdorfelden, Germany) and is well characterised by 17 ingredients from the natural product classes of flavonoids, saponins, and dicaffeoylquinic acids identified via LC-MS analysis [7]. All reagents for luciferase assays were received from Promega (Mannheim, Germany) if not stated otherwise. Coelenterazine h was obtained from Prolume (Pinetop-Lakeside, AZ, USA). Adenosine, BAY 60-6583, forskolin, 5′-N-Ethylcarboxamidoadenosine (NECA), and SCH 442416 were obtained from Sigma-Aldrich (Crailsheim, Germany). TNFα was received from Merck (Darmstadt, Germany). PSB-603 and CGS 21680 were obtained from Biomol (Hamburg, Germany).

4.2. Cell Culture

Human embryonic kidney cells (HEK293), subsequently called HEK cells, were obtained from DSMZ (No. ACC 305; Braunschweig, Germany). HEK cells and all constructed clones were cultivated at 37 °C with 5% CO2 in DMEM supplemented with 100 units/mL penicillin, 100 µg/mL streptomycin, and 10% fetal bovine serum (FBS) (all obtained from Thermo Fisher Scientific, Waltham, MA, USA). Cells were subcultured 1:10 every 3–4 days in 10 cm cell culture dishes.
Calu-3 cells were obtained from ATCC (HTB-55; Manassas, VA, USA). Calu-3 cells and all constructed clones were cultivated at 37 °C with 5% CO2 in DMEM/F-12 supplemented with GlutaMAX™, 100 units/mL penicillin, 100 µg/mL streptomycin, and 15% FBS (all obtained from Thermo Fisher Scientific). Cells were subcultured 1:5 every 5–7 days in 10 cm cell culture dishes.

4.3. Dynamic Mass Redistribution Measurements

Dynamic mass redistribution (DMR) measurements were performed using the Corning Epic® biosensor. HEK GloSensor™ cells were seeded at a density of 3000 cells per well in a 384-well plate by Corning (#5042; Corning, NY, USA) and allowed to grow for at least 24 h in full growth medium. Pre-incubation was performed with 40, 80, 160, or 240 µg/mL EA 575® for up to 16 h. After pre-incubation, the medium was replaced by HBSS supplemented with 20 mM HEPES and the cells were allowed to equilibrate at 37 °C for one hour. A baseline of 10 measurement points was recorded before stimulation was performed using a CyBi®-SELMA semi-automatic pipetting system (Analytik Jena AG, Jena, Germany) at 37 °C. Subsequently, the wavelength shift mediated by the dynamic mass redistribution of intracellular particles was measured for another 70 min.

4.4. cAMP Measurements

The establishment of HEK cells expressing a cAMP sensor and the measurement of cAMP were performed as described by Bussmann et al. [8]. Briefly, pre-incubation was carried out with 40, 80, 160, or 240 µg/mL EA 575® for up to 16 h in full growth medium. After pre-incubation, the medium was changed to a substrate solution containing 4% GloSensor™ cAMP reagent stock solution in HEPES-buffered DMEM. Cells were incubated at 37 °C for one hour and subsequently equilibrated at room temperature in the plate reader (Tecan Infinite® 200 PRO, Tecan, Männedorf, Switzerland) for another hour. Stimulation was performed with 1 μM BAY 60-6583 and 1 μM forskolin simultaneously, and cAMP increase was measured as luminescence for one hour after stimulation.

4.5. Measurements of β-Arrestin 2 Recruitment

The plasmid coding for human adenosine receptor A2B (A2BAR) fused to the N-terminus of Large BiT (LgBiT) was generated by initially removing the region coding for YFP of the vector pEYFP-N1-A2BR, which was a gift from Robert Tarran (Addgene plasmid # 37202; http://n2t.net/addgene:37202 (accessed 13 November 2019); RRID:Addgene_37202) [91], using BamHI/NotI. The open reading frame coding for LgBiT was amplified via PCR (forward primer: 5′-GATCGGATCCAAGTGGTAGCGGGGTCTTTACCCTG-3′; reverse primer: 5′-GATCGCGGCCGCTAGCTACCACCGCATCC-3′). The PCR product was cut with BamHI/NotI and inserted into the vector via ligation.
For expression of rat β-arrestin 2 with an N-terminal Small BiT (SmBiT), the coding sequence was taken from pECFP-N1-rβ-arrestin-2 (a gift from M. Bouvier, Montreal, QC, Canada) by restriction with NheI/SalI. The fragment was introduced into pcDNA™3.1/Zeo(+) Mammalian Expression Vector (Invitrogen, Waltham, MA, USA) containing the information for the SmBit via NheI and XhoI sites.
HEK cells were co-transfected to transiently express A2BAR-LgBiT and SmBiT-β-arrestin 2 using branched polyethylenimine (PEI) (Sigma-Aldrich). For this purpose, cells were seeded in a 6-well plate at a density of 350,000 cells per well and allowed to grow for one day. For each DNA, 1.5 µg was diluted into 200 µL of 150 mM NaCl, then 7.5 µL of a 1 mg/mL PEI solution was added, and the mixture was vortexed immediately for 10 s. After 10 min at RT, the DNA/PEI mixture was added to the cells and incubated for 24 h. The transiently transfected cells were seeded in a 96-well plate at a density of 15,000 cells per well and allowed to grow for one day in full growth medium. Pre-incubation was carried out with 160 µg/mL EA 575® for 16 h.
In general, measurements of recruitment of β-arrestin 2 to A2BAR were performed as recently described by Saecker et al. for A1AR [57]. Briefly, pre-incubation medium was replaced by a solution of 2.5 µM coelenterazine h in HBSS supplemented with 20 mM HEPES. A baseline of 3 measurement points was recorded before stimulation was performed. Subsequently, luminescence corresponding to the recruitment of β-arrestin 2 was measured for another 27 min using a Spark® plate reader by Tecan.

4.6. Measurements of CRE Activation

The plasmid expressing NanoLuc®-PEST (NlucP) from a promoter with cAMP response elements (CRE) was received by Promega. HEK cells were transfected to transiently express this construct using PEI, as described above. Transiently transfected HEK cells were seeded in a 96-well plate at a density of 20,000 cells per well and allowed to grow for one day in fully supplemented medium. Pre-incubation was conducted with 40, 80, 160, or 240 µg/mL EA 575® in full growth medium for 16 h. Pre-incubation with antagonists was carried out for only two hours, simultaneously with the substrate incubation. Nano-Glo® Vivazine™ Live Cell Substrate (Promega) was prepared according to the manufacturer’s instructions using HEPES-buffered medium, and cells were incubated with the substrate for 2 h at 37 °C in a Tecan Spark® plate reader already measuring luminescence. Stimulation was then performed by adding adenosine, BAY 60-6583, or CGS 21680, and measurement was performed for another 22 h.

4.7. IL-6 Measurements

Measurement of IL-6 release was performed using the Lumit™ IL-6 (Human) Immunoassay by Promega. Calu-3 cells were seeded in a 96-well plate at a density of 20,000 cells per well and allowed to grow for at least two days to a confluency of 80–90% in full growth medium. Pre-incubation was carried out with 40, 80, 160, or 240 µg/mL EA 575® in DMEM/F-12 without phenol red supplemented with GlutaMAX™ and 5% FBS for 16 h. Pre-incubation with antagonists was conducted for only one hour. IL-6 release was then provoked by adding adenosine, BAY 60-6583, or CGS 21680 for the following 24 h. Subsequently, the measurement of IL-6 was performed according to the manufacturer’s instructions using a Tecan Spark® plate reader.

4.8. Measurements of NFκB Transcriptional Activity

For the generation of a secreted NanoLuc® expression construct under the control of an NFκB binding sequence, the pNFkB-d2EGFP vector (Clontech, Mountain View, CA, USA) was used as a first template. Destabilised GFP was removed from the vector by PCR (forward primer: 5′-TCGGATATCTCGAGCCGGAATTCGGGGAAGCTTC-3′; reverse primer: 5′-GTTCAGGGGGAGGTGTG-3′) and restriction with BamHI/XhoI. The open reading frame coding for the secreted NanoLuc® was cut from pNL1.3[secNluc] vector (Promega) using BamHI/XhoI and introduced into the vector via ligation.
In a second step, this secreted NanoLuc® expression construct, under the control of an NFκB binding sequence, was cloned into the pcDNA™3.1(+) Mammalian Expression Vector (Invitrogen). Therefore, the CMV promoter was removed from the vector by restriction with BamHI/BglII and re-ligation. Then, the vector was re-cut with NotI/XhoI. The insert was isolated out of the plasmid generated in the first step using NotI/SalI and introduced into the vector via ligation.
Calu-3 cells were transfected using Metafectene® Pro (Biontex, Munich, Germany) according to the manufacturer’s instructions. For the selection of successfully transfected cells, the medium was changed to fully supplemented DMEM/F-12 containing 600 µg/mL G418 (Thermo Fisher Scientific).
Cells were seeded in a 96-well plate at a density of 25,000 cells per well and allowed to grow for at least two days to a confluency of 80–90% in full growth medium. Before stimulating with TNFα, adenosine, NECA, or BAY 60-6583 for 3 h, cells were starved overnight. Measurement of NFκB transcriptional activity was performed in a Tecan Spark® plate reader using the Nano-Glo® Luciferase Assay System (Promega) according to the manufacturer’s instructions.

4.9. Statistical Analysis

For statistical analysis, one-way analysis of variance (ANOVA) with Dunnett’s multiple comparisons test was performed using Prism software version 6.01 (GraphPad Software, San Diego, CA, USA). Results were considered to be significant for p-values of <0.05.

Author Contributions

Conceptualisation, H.H. and S.F.; methodology, F.M., H.H. and S.F.; software, F.M.; validation, F.M.; formal analysis, F.M.; investigation, F.M.; resources, H.H. and S.F.; data curation, F.M.; writing—original draft preparation, F.M.; writing—review and editing, H.H. and S.F.; visualisation, F.M.; supervision, H.H. and S.F.; project administration, H.H. and S.F.; funding acquisition, H.H. and S.F. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant from Engelhard Arzneimittel GmbH & Co. KG, Niederdorfelden, Germany, grant number H-061.0037.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All relevant data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Kardos, P.; Dinh, Q.T.; Fuchs, K.H.; Gillissen, A.; Klimek, L.; Koehler, M.; Sitter, H.; Worth, H. Guidelines of the German Respiratory Society for Diagnosis and Treatment of Adults Suffering from Acute, Subacute and Chronic Cough. Pneumologie 2019, 73, 143–180. [Google Scholar] [CrossRef] [PubMed]
  2. Lang, C.; Röttger-Lüer, P.; Staiger, C. A Valuable Option for the Treatment of Respiratory Diseases: Review on the Clinical Evidence of the Ivy Leaves Dry Extract EA 575®. Planta Med. 2015, 81, 968–974. [Google Scholar] [CrossRef] [PubMed]
  3. Schaefer, A.; Ludwig, F.; Giannetti, B.M.; Bulitta, M.; Wacker, A. Efficacy of Two Dosing Schemes of a Liquid Containing Ivy Leaves Dry Extract EA 575 versus Placebo in the Treatment of Acute Bronchitis in Adults. ERJ Open Res. 2019, 5, 52–57. [Google Scholar] [CrossRef] [Green Version]
  4. Schaefer, A.; Kehr, M.S.; Giannetti, B.M.; Bulitta, M.; Staiger, C. A Randomized, Controlled, Double-Blind, Multi-Center Trial to Evaluate the Efficacy and Safety of a Liquid Containing Ivy Leaves Dry Extract (EA 575®) vs. Placebo in the Treatment of Adults with Acute Cough. Pharmazie 2016, 71, 504–509. [Google Scholar] [CrossRef]
  5. Schulte-Michels, J.; Wolf, A.; Aatz, S.; Engelhard, K.; Sieben, A.; Martinez-Osuna, M.; Häberlein, F.; Häberlein, H. α-Hederin Inhibits G Protein-Coupled Receptor Kinase 2-Mediated Phosphorylation of Β2-Adrenergic Receptors. Phytomedicine 2016, 23, 52–57. [Google Scholar] [CrossRef] [Green Version]
  6. Meurer, F.; Schulte-Michels, J.; Häberlein, H.; Franken, S. Ivy Leaves Dry Extract EA 575® Mediates Biased Β2-Adrenergic Receptor Signaling. Phytomedicine 2021, 90, 153645. [Google Scholar] [CrossRef]
  7. Greunke, C.; Hage-Hülsmann, A.; Sorkalla, T.; Keksel, N.; Häberlein, F.; Häberlein, H. A Systematic Study on the Influence of the Main Ingredients of an Ivy Leaves Dry Extract on the Β2-Adrenergic Responsiveness of Human Airway Smooth Muscle Cells. Pulm. Pharmacol. Ther. 2015, 31, 92–98. [Google Scholar] [CrossRef] [Green Version]
  8. Bussmann, H.; Schulte-Michels, J.; Bingel, M.; Meurer, F.; Aatz, S.; Häberlein, F.; Franken, S.; Häberlein, H. A Comparative Study on the Influence of an Ivy Preparation and an Ivy/Thyme Combination on the Β2-Adrenergic Signal Transduction. Heliyon 2020, 6, e03960. [Google Scholar] [CrossRef]
  9. Sieben, A.; Prenner, L.; Sorkalla, T.; Wolf, A.; Jakobs, D.; Runkel, F.; Häberlein, H. α-Hederin, but Not Hederacoside c and Hederagenin from Hedera Helix, Affects the Binding Behavior, Dynamics, and Regulation of Β2- Adrenergic Receptors. Biochemistry 2009, 48, 3477–3482. [Google Scholar] [CrossRef]
  10. Schulte-Michels, J.; Runkel, F.; Gokorsch, S.; Häberlein, H. Ivy Leaves Dry Extract EA 575® Decreases LPS-Induced IL-6 Release from Murine Macrophages. Pharmazie 2016, 71, 158–161. [Google Scholar] [CrossRef]
  11. Schulte-Michels, J.; Keksel, C.; Häberlein, H.; Franken, S. Anti-Inflammatory Effects of Ivy Leaves Dry Extract: Influence on Transcriptional Activity of NFκB. Inflammopharmacology 2019, 27, 339–347. [Google Scholar] [CrossRef] [PubMed]
  12. Li, X.; Berg, N.K.; Mills, T.; Zhang, K.; Eltzschig, H.K.; Yuan, X. Adenosine at the Interphase of Hypoxia and Inflammation in Lung Injury. Front. Immunol. 2021, 11, 604944. [Google Scholar] [CrossRef] [PubMed]
  13. Le, T.T.T.; Berg, N.K.; Harting, M.T.; Li, X.; Eltzschig, H.K.; Yuan, X. Purinergic Signaling in Pulmonary Inflammation. Front. Immunol. 2019, 10, 1633. [Google Scholar] [CrossRef] [PubMed]
  14. Zhou, Y.; Schneider, D.J.; Blackburn, M.R. Adenosine Signaling and the Regulation of Chronic Lung Disease. Pharmacol. Ther. 2009, 123, 105–116. [Google Scholar] [CrossRef] [Green Version]
  15. Blackburn, M.R. Too Much of a Good Thing: Adenosine Overload in Adenosine-Deaminase-Deficient Mice. Trends Pharmacol. Sci. 2003, 24, 66–70. [Google Scholar] [CrossRef]
  16. Driver, A.G.; Kukoly, C.A.; Ali, S.; Mustafa, S.J. Adenosine in Bronchoalveolar Lavage Fluid in Asthma. Am. Rev. Respir. Dis. 1993, 148, 91–97. [Google Scholar] [CrossRef]
  17. Huszár, É.; Vass, G.; Vizi, É.; Csoma, Z.; Barát, E.; Molnár-Világos, G.; Herjavecz, I.; Horváth, I. Adenosine in Exhaled Breath Condensate in Healthy Volunteers and in Patients with Asthma. Eur. Respir. J. 2002, 20, 1393–1398. [Google Scholar] [CrossRef]
  18. Esther, C.R.; Boysen, G.; Olsen, B.M.; Collins, L.B.; Ghio, A.J.; Swenberg, J.W.; Boucher, R.C. Mass Spectrometric Analysis of Biomarkers and Dilution Markers in Exhaled Breath Condensate Reveals Elevated Purines in Asthma and Cystic Fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2009, 296, L987. [Google Scholar] [CrossRef] [Green Version]
  19. Esther, C.R.; Lazaar, A.L.; Bordonali, E.; Qaqish, B.; Boucher, R.C. Elevated Airway Purines in COPD. Chest 2011, 140, 954–960. [Google Scholar] [CrossRef] [Green Version]
  20. Vizi, É.; Huszár, É.; Csoma, Z.; Böszörményi-Nagy, G.; Barát, E.; Horváth, I.; Herjavecz, I.; Kollai, M. Plasma Adenosine Concentration Increases during Exercise: A Possible Contributing Factor in Exercise-Induced Bronchoconstriction in Asthma. J. Allergy Clin. Immunol. 2002, 109, 446–448. [Google Scholar] [CrossRef]
  21. Cushley, M.; Tattersfield, A.; Holgate, S. Inhaled Adenosine and Guanosine on Airway Resistance in Normal and Asthmatic Subjects. Br. J. Clin. Pharmacol. 1983, 15, 161–165. [Google Scholar] [CrossRef] [Green Version]
  22. Mann, J.S.; Holgate, S.T.; Renwick, A.G.; Cushley, M.J. Airway Effects of Purine Nucleosides and Nucleotides and Release with Bronchial Provocation in Asthma. J. Appl. Physiol. 1986, 61, 1667–1676. [Google Scholar] [CrossRef]
  23. Oosterhoff, Y.; De Jong, J.W.; Jansen, M.A.M.; Koeter, G.H.; Postma, D.S. Airway Responsiveness to Adenosine 5′-Monophosphate in Chronic Obstructive Pulmonary Disease Is Determined by Smoking. Am. Rev. Respir. Dis. 1993, 147, 553–558. [Google Scholar] [CrossRef] [PubMed]
  24. Zhou, Y.; Murthy, J.N.; Zeng, D.; Belardinelli, L.; Blackburn, M.R. Alterations in Adenosine Metabolism and Signaling in Patients with Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. PLoS ONE 2010, 5, e9224. [Google Scholar] [CrossRef] [Green Version]
  25. Mertens, T.C.J.; Hanmandlu, A.; Tu, L.; Phan, C.; Collum, S.D.; Chen, N.Y.; Weng, T.; Davies, J.; Liu, C.; Eltzschig, H.K.; et al. Switching-Off Adora2b in Vascular Smooth Muscle Cells Halts the Development of Pulmonary Hypertension. Front. Physiol. 2018, 9, 555. [Google Scholar] [CrossRef]
  26. Karmouty-Quintana, H.; Weng, T.; Garcia-Morales, L.J.; Chen, N.Y.; Pedroza, M.; Zhong, H.; Molina, J.G.; Bunge, R.; Bruckner, B.A.; Xia, Y.; et al. Adenosine A2B Receptor and Hyaluronan Modulate Pulmonary Hypertension Associated with Chronic Obstructive Pulmonary Disease. Am. J. Respir. Cell Mol. Biol. 2013, 49, 1038–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Blackburn, M.R.; Datta, S.K.; Kellems, R.E. Adenosine Deaminase-Deficient Mice Generated Using a Two-Stage Genetic Engineering Strategy Exhibit a Combined Immunodeficiency. J. Biol. Chem. 1998, 273, 5093–5100. [Google Scholar] [CrossRef] [Green Version]
  28. Chunn, J.L.; Young, H.W.J.; Banerjee, S.K.; Colasurdo, G.N.; Blackburn, M.R. Adenosine-Dependent Airway Inflammation and Hyperresponsiveness in Partially Adenosine Deaminase-Deficient Mice. J. Immunol. 2001, 167, 4676–4685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Blackburn, M.R.; Volmer, J.B.; Thrasher, J.L.; Zhong, H.; Crosby, J.R.; Lee, J.J.; Kellems, R.E. Metabolic Consequences of Adenosine Deaminase Deficiency in Mice Are Associated with Defects in Alveogenesis, Pulmonary Inflammation, and Airway Obstruction. J. Exp. Med. 2000, 192, 159–170. [Google Scholar] [CrossRef] [PubMed]
  30. Chunn, J.L.; Molina, J.G.; Mi, T.; Xia, Y.; Kellems, R.E.; Blackburn, M.R. Adenosine-Dependent Pulmonary Fibrosis in Adenosine Deaminase-Deficient Mice. J. Immunol. 2005, 175, 1937–1946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Sun, C.-X.; Zhong, H.; Mohsenin, A.; Morschl, E.; Chunn, J.L.; Molina, J.G.; Belardinelli, L.; Zeng, D.; Blackburn, M.R. Role of A2B Adenosine Receptor Signaling in Adenosine-Dependent Pulmonary Inflammation and Injury. J. Clin. Investig. 2006, 116, 2173–2182. [Google Scholar] [CrossRef] [Green Version]
  32. Chunn, J.L.; Mohsenin, A.; Young, H.W.J.; Lee, C.G.; Elias, J.A.; Kellems, R.E.; Blackburn, M.R. Partially Adenosine Deaminase-Deficient Mice Develop Pulmonary Fibrosis in Association with Adenosine Elevations. Am. J. Physiol. Lung Cell. Mol. Physiol. 2006, 290, L579–L587. [Google Scholar] [CrossRef]
  33. Karmouty-Quintana, H.; Zhong, H.; Acero, L.; Weng, T.; Melicoff, E.; West, J.D.; Hemnes, A.; Grenz, A.; Eltzschig, H.K.; Blackwell, T.S.; et al. The A 2B Adenosine Receptor Modulates Pulmonary Hypertension Associated with Interstitial Lung Disease. FASEB J. 2012, 26, 2546–2557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rincon, M.; Irvin, C.G. Role of IL-6 in Asthma and Other Inflammatory Pulmonary Diseases. Int. J. Biol. Sci. 2012, 8, 1281–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Marini, M.; Vittori, E.; Hollemborg, J.; Mattoli, S. Expression of the Potent Inflammatory Cytokines, Granulocyte-Macrophage-Colony-Stimulating Factor and Interleukin-6 and Interleukin-8, in Bronchial Epithelial Cells of Patients with Asthma. J. Allergy Clin. Immunol. 1992, 89, 1001–1009. [Google Scholar] [CrossRef]
  36. Mattoli, S.; Mattoso, V.L.; Soloperto, M.; Allegra, L.; Fasoli, A. Cellular and Biochemical Characteristics of Bronchoalveolar Lavage Fluid in Symptomatic Nonallergic Asthma. J. Allergy Clin. Immunol. 1991, 87, 794–802. [Google Scholar] [CrossRef]
  37. Neveu, W.A.; Allard, J.L.; Raymond, D.M.; Bourassa, L.M.; Burns, S.M.; Bunn, J.Y.; Irvin, C.G.; Kaminsky, D.A.; Rincon, M. Elevation of IL-6 in the Allergic Asthmatic Airway Is Independent of Inflammation but Associates with Loss of Central Airway Function. Respir. Res. 2010, 11, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Konno, S.I.; Gonokami, Y.; Kurokawa, M.; Kawazu, K.; Asano, K.; Okamoto, K.I.; Adachi, M. Cytokine Concentrations in Sputum of Asthmatic Patients. Int. Arch. Allergy Immunol. 1996, 109, 73–78. [Google Scholar] [CrossRef] [PubMed]
  39. Broide, D.H.; Lotz, M.; Cuomo, A.J.; Coburn, D.A.; Federman, E.C.; Wasserman, S.I. Cytokines in Symptomatic Asthma Airways. J. Allergy Clin. Immunol. 1992, 89, 958–967. [Google Scholar] [CrossRef]
  40. Yokoyama, A.; Kohno, N.; Fujino, S.; Hamada, H.; Inoue, Y.; Fujioka, S.; Ishida, S.; Hiwada, K. Circulating Interleukin-6 Levels in Patients with Bronchial Asthma. Am. J. Respir. Crit. Care Med. 1995, 151, 1354–1358. [Google Scholar] [CrossRef]
  41. Tillie-Leblond, I.; Pugin, J.; Marquette, C.H.; Lamblin, C.; Saulnier, F.; Brichet, A.; Wallaert, B.; Tonnel, A.B.; Gosset, P. Balance between Proinflammatory Cytokines and Their Inhibitors in Bronchial Lavage from Patients with Status Asthmaticus. Am. J. Respir. Crit. Care Med. 1999, 159, 487–494. [Google Scholar] [CrossRef] [Green Version]
  42. Dixon, A.E.; Raymond, D.M.; Suratt, B.T.; Bourassa, L.M.; Irvin, C.G. Lower Airway Disease in Asthmatics with and without Rhinitis. Lung 2008, 186, 361–368. [Google Scholar] [CrossRef] [Green Version]
  43. Morjaria, J.B.; Babu, K.S.; Vijayanand, P.; Chauhan, A.J.; Davies, D.E.; Holgate, S.T. Sputum IL-6 Concentrations in Severe Asthma and Its Relationship with FEV1. Thorax 2011, 66, 537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Attaran, D.; Lari, S.M.; Towhidi, M.; Marallu, H.G.; Ayatollahi, H.; Khajehdaluee, M.; Ghanei, M.; Basiri, R. Interleukin-6 and Airflow Limitation in Chemical Warfare Patients with Chronic Obstructive Pulmonary Disease. Int. J. Chronic Obstr. Pulm. Dis. 2010, 5, 335–340. [Google Scholar] [CrossRef] [Green Version]
  45. Eickmeier, O.; Huebner, M.; Herrmann, E.; Zissler, U.; Rosewich, M.; Baer, P.C.; Buhl, R.; Schmitt-Grohé, S.; Zielen, S.; Schubert, R. Sputum Biomarker Profiles in Cystic Fibrosis (CF) and Chronic Obstructive Pulmonary Disease (COPD) and Association between Pulmonary Function. Cytokine 2010, 50, 152–157. [Google Scholar] [CrossRef]
  46. Donaldson, G.C.; Seemungal, T.A.R.; Patel, I.S.; Bhowmik, A.; Wilkinon, T.M.A.; Hurst, J.R.; MacCallum, P.K.; Wedzicha, J.A. Airway and Systemic Inflammation and Decline in Lung Function in Patients with COPD. Chest 2005, 128, 1995–2004. [Google Scholar] [CrossRef]
  47. Hacievliyagil, S.S.; Gunen, H.; Mutlu, L.C.; Karabulut, A.B.; Temel, I. Association between Cytokines in Induced Sputum and Severity of Chronic Obstructive Pulmonary Disease. Respir. Med. 2006, 100, 846–854. [Google Scholar] [CrossRef] [Green Version]
  48. Celli, B.R.; Locantore, N.; Yates, J.; Tal-Singer, R.; Miller, B.E.; Bakke, P.; Calverley, P.; Coxson, H.; Crim, C.; Edwards, L.D.; et al. Inflammatory Biomarkers Improve Clinical Prediction of Mortality in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2012, 185, 1065–1072. [Google Scholar] [CrossRef]
  49. Saito, F.; Tasaka, S.; Inoue, K.I.; Miyamoto, K.; Nakano, Y.; Ogawa, Y.; Yamada, W.; Shiraishi, Y.; Hasegawa, N.; Fujishima, S.; et al. Role of Interleukin-6 in Bleomycin-Induced Lung Inflammatory Changes in Mice. Am. J. Respir. Cell Mol. Biol. 2008, 38, 566–571. [Google Scholar] [CrossRef]
  50. Pedroza, M.; Schneider, D.J.; Karmouty-Quintana, H.; Coote, J.; Shaw, S.; Corrigan, R.; Molina, J.G.; Alcorn, J.L.; Galas, D.; Gelinas, R.; et al. Interleukin-6 Contributes to Inflammation and Remodeling in a Model of Adenosine Mediated Lung Injury. PLoS ONE 2011, 6, e22667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Ryzhov, S.; Zaynagetdinov, R.; Goldstein, A.E.; Novitskiy, S.V.; Blackburn, M.R.; Biaggioni, I.; Feoktistov, I. Effect of A2B Adenosine Receptor Gene Ablation on Adenosine-Dependent Regulation of Proinflammatory Cytokines. J. Pharmacol. Exp. Ther. 2008, 324, 694–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Philip, K.; Mills, T.W.; Davies, J.; Chen, N.Y.; Karmouty-Quintana, H.; Luo, F.; Molina, J.G.; Amione-Guerra, J.; Sinha, N.; Guha, A.; et al. HIF1A Up-Regulates the ADORA2B Receptor on Alternatively Activated Macrophages and Contributes to Pulmonary Fibrosis. FASEB J. 2017, 31, 4745–4758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Karmouty-Quintana, H.; Philip, K.; Acero, L.F.; Chen, N.Y.; Weng, T.; Molina, J.G.; Luo, F.; Davies, J.; Le, N.B.; Bunge, I.; et al. Deletion of ADORA2B from Myeloid Cells Dampens Lung Fibrosis and Pulmonary Hypertension. FASEB J. 2015, 29, 50–60. [Google Scholar] [CrossRef] [Green Version]
  54. Schröder, R.; Janssen, N.; Schmidt, J.; Kebig, A.; Merten, N.; Hennen, S.; Müller, A.; Blättermann, S.; Mohr-Andrä, M.; Zahn, S.; et al. Deconvolution of Complex G Protein–Coupled Receptor Signaling in Live Cells Using Dynamic Mass Redistribution Measurements. Nat. Biotechnol. 2010, 28, 943–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Goulding, J.; May, L.T.; Hill, S.J. Characterisation of Endogenous A2A and A2B Receptor-Mediated Cyclic AMP Responses in HEK 293 Cells Using the GloSensorTM Biosensor: Evidence for an Allosteric Mechanism of Action for the A2B-Selective Antagonist PSB 603. Biochem. Pharmacol. 2018, 147, 55–66. [Google Scholar] [CrossRef] [PubMed]
  56. Barresi, E.; Martini, C.; Da Settimo, F.; Greco, G.; Taliani, S.; Giacomelli, C.; Trincavelli, M.L. Allosterism vs. Orthosterism: Recent Findings and Future Perspectives on A2B AR Physio-Pathological Implications. Front. Pharmacol. 2021, 12, 652121. [Google Scholar] [CrossRef]
  57. Saecker, L.; Häberlein, H.; Franken, S. Investigation of Adenosine A1 Receptor-Mediated β-Arrestin 2 Recruitment Using a Split-Luciferase Assay. Front. Pharmacol. 2023, 14, 1172551. [Google Scholar] [CrossRef]
  58. Montminy, M.R.; Bilezikjian, L.M. Binding of a Nuclear Protein to the Cyclic-AMP Response Element of the Somatostatin Gene. Nature 1987, 328, 175–178. [Google Scholar] [CrossRef]
  59. Montminy, M.R.; Sevarino, K.A.; Wagner, J.A.; Mandel, G.; Goodman, R.H. Identification of a Cyclic-AMP-Responsive Element within the Rat Somatostatin Gene. Proc. Natl. Acad. Sci. USA 1986, 83, 6682–6686. [Google Scholar] [CrossRef]
  60. Yamamoto, K.K.; Gonzalez, G.A.; Biggs, W.H.; Montminy, M.R. Phosphorylation-Induced Binding and Transcriptional Efficacy of Nuclear Factor CREB. Nature 1988, 334, 494–498. [Google Scholar] [CrossRef]
  61. Sun, Y.; Wu, F.; Sun, F.; Huang, P. Adenosine Promotes IL-6 Release in Airway Epithelia. J. Immunol. 2008, 180, 4173–4181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Du, X.; Ou, X.; Song, T.; Zhang, W.; Cong, F.; Zhang, S.; Xiong, Y. Adenosine A2B Receptor Stimulates Angiogenesis by Inducing VEGF and ENOS in Human Microvascular Endothelial Cells. Exp. Biol. Med. 2015, 240, 1472–1479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ray, A.; LaForge, K.S.; Sehgal, P.B. On the Mechanism for Efficient Repression of the Interleukin-6 Promoter by Glucocorticoids: Enhancer, TATA Box, and RNA Start Site (Inr Motif) Occlusion. Mol. Cell. Biol. 1990, 10, 5736–5746. [Google Scholar] [CrossRef] [PubMed]
  64. Ray, A.; Sassone-Corsi, P.; Sehgal, P.B. A Multiple Cytokine- and Second Messenger-Responsive Element in the Enhancer of the Human Interleukin-6 Gene: Similarities with c-Fos Gene Regulation. Mol. Cell. Biol. 1989, 9, 5537–5547. [Google Scholar] [CrossRef]
  65. Krueger, J.; Ray, A.; Tamm, I.; Sehgal, P.B. Expression and Function of Interleukin-6 in Epithelial Cells. J. Cell. Biochem. 1991, 45, 327–334. [Google Scholar] [CrossRef]
  66. Ray, A.; Tatter, S.B.; May, L.T.; Sehgal, P.B. Activation of the Human “Beta 2-Interferon/Hepatocyte-Stimulating Factor/Interleukin 6” Promoter by Cytokines, Viruses, and Second Messenger Agonists. Proc. Natl. Acad. Sci. USA 1988, 85, 6701–6705. [Google Scholar] [CrossRef]
  67. Zhang, Y.; Lin, J.X.; Vilcek, J. Synthesis of Interleukin 6 (Interferon-Β2/B Cell Stimulatory Factor 2) in Human Fibroblasts Is Triggered by an Increase in Intracellular Cyclic AMP. J. Biol. Chem. 1988, 263, 6177–6182. [Google Scholar] [CrossRef]
  68. Zhong, H.; Belardinelli, L.; Maa, T.; Feoktistov, I.; Biaggioni, I.; Zeng, D. A2B Adenosine Receptors Increase Cytokine Release by Bronchial Smooth Muscle Cells. Am. J. Respir. Cell Mol. Biol. 2004, 30, 118–125. [Google Scholar] [CrossRef]
  69. Sitaraman, S.V.; Merlin, D.; Wang, L.; Wong, M.; Gewirtz, A.T.; Si-Tahar, M.; Madara, J.L. Neutrophil-Epithelial Crosstalk at the Intestinal Lumenal Surface Mediated by Reciprocal Secretion of Adenosine and IL-6. J. Clin. Investig. 2001, 107, 861–869. [Google Scholar] [CrossRef] [Green Version]
  70. Libermann, T.A.; Baltimore, D. Activation of Interleukin-6 Gene Expression through the NF-Kappa B Transcription Factor. Mol. Cell. Biol. 1990, 10, 2327–2334. [Google Scholar] [CrossRef] [Green Version]
  71. Shimizu, H.; Mitomo, K.; Watanabe, T.; Okamoto, S.; Yamamoto, K. Involvement of a NF-Kappa B-like Transcription Factor in the Activation of the Interleukin-6 Gene by Inflammatory Lymphokines. Mol. Cell. Biol. 1990, 10, 561–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Cobb, B.R.; Ruiz, F.; King, C.M.; Fortenberry, J.; Greer, H.; Kovacs, T.; Sorscher, E.J.; Clancy, J.P. A2 Adenosine Receptors Regulate CFTR through PKA and PLA2. Am. J. Physiol. Lung Cell. Mol. Physiol. 2002, 282, 12–25. [Google Scholar] [CrossRef] [PubMed]
  73. Szkotak, A.J.; Ng, A.M.L.; Man, S.F.P.; Baldwin, S.A.; Cass, C.E.; Young, J.D.; Duszyk, M. Coupling of CFTR-Mediated Anion Secretion to Nucleoside Transporters and Adenosine Homeostasis in Calu-3 Cells. J. Membr. Biol. 2003, 192, 169–179. [Google Scholar] [CrossRef] [PubMed]
  74. Atwood, B.K.; Lopez, J.; Wager-Miller, J.; Mackie, K.; Straiker, A. Expression of G Protein-Coupled Receptors and Related Proteins in HEK293, AtT20, BV2, and N18 Cell Lines as Revealed by Microarray Analysis. BMC Genom. 2011, 12, 14. [Google Scholar] [CrossRef] [Green Version]
  75. Zhong, H.; Belardinelli, L.; Maa, T.; Zeng, D. Synergy between A2B Adenosine Receptors and Hypoxia in Activating Human Lung Fibroblasts. Am. J. Respir. Cell Mol. Biol. 2005, 32, 2–8. [Google Scholar] [CrossRef]
  76. Fredholm, B.B.; IJzerman, A.P.; Jacobson, K.A.; Linden, J.; Müller, C.E. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and Classification of Adenosine Receptors—An Update. Pharmacol. Rev. 2011, 63, 1–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Geraghty, N.J.; Adhikary, S.R.; Watson, D.; Sluyter, R. The A2A Receptor Agonist CGS 21680 Has Beneficial and Adverse Effects on Disease Development in a Humanised Mouse Model of Graft-versus-Host Disease. Int. Immunopharmacol. 2019, 72, 479–486. [Google Scholar] [CrossRef] [PubMed]
  78. Pei, H.; Linden, J. Adenosine Influences Myeloid Cells to Inhibit Aeroallergen Sensitization. Am. J. Physiol. Lung Cell. Mol. Physiol. 2016, 310, L985–L992. [Google Scholar] [CrossRef] [Green Version]
  79. Zheng, X.; Wang, D. The Adenosine A2A Receptor Agonist Accelerates Bone Healing and Adjusts Treg/Th17 Cell Balance through Interleukin 6. Biomed. Res. Int. 2020, 2020, 2603873. [Google Scholar] [CrossRef] [Green Version]
  80. Tang, L.M.; Wang, Y.P.; Wang, K.; Pu, L.Y.; Zhang, F.; Li, X.C.; Kong, L.B.; Sun, B.C.; Li, G.Q.; Wang, X.H. Protective Effect of Adenosine A2A Receptor Activation in Small-for-Size Liver Transplantation. Transpl. Int. 2007, 20, 93–101. [Google Scholar] [CrossRef]
  81. Rogachev, B.; Ziv, N.Y.; Mazar, J.; Nakav, S.; Chaimovitz, C.; Zlotnik, M.; Douvdevani, A. Adenosine Is Upregulated during Peritonitis and Is Involved in Downregulation of Inflammation. Kidney Int. 2006, 70, 675–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Zhang, L.; Franchini, M.; Wehrli Eser, M.; Dip, R. Enhanced IL-6 Transcriptional Response to Adenosine Receptor Ligands in Horses with Lower Airway Inflammation. Equine Vet. J. 2012, 44, 81–87. [Google Scholar] [CrossRef] [PubMed]
  83. Schwaninger, M.; Neher, M.; Viegas, E.; Schneider, A.; Spranger, M. Stimulation of Interleukin-6 Secretion and Gene Transcription in Primary Astrocytes by Adenosine. J. Neurochem. 1997, 69, 1145–1150. [Google Scholar] [CrossRef]
  84. McColl, S.R.; St-Onge, M.; Dussault, A.-A.; Laflamme, C.; Bouchard, L.; Boulanger, J.; Pouliot, M. Immunomodulatory Impact of the A2A Adenosine Receptor on the Profile of Chemokines Produced by Neutrophils. FASEB J. 2006, 20, 187. [Google Scholar] [CrossRef] [Green Version]
  85. Mohsenin, A.; Mi, T.; Xia, Y.; Kellems, R.E.; Chen, J.F.; Blackburn, M.R. Genetic Removal of the A2A Adenosine Receptor Enhances Pulmonary Inflammation, Mucin Production, and Angiogenesis in Adenosine Deaminase-Deficient Mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 293, L753–L761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Pejman, L.; Omrani, H.; Mirzamohammadi, Z.; Shahbazfar, A.A.; Khalili, M.; Keyhanmanesh, R. The Effect of Adenosine A2A and A2B Antagonists on Tracheal Responsiveness, Serum Levels of Cytokines and Lung Inflammation in Guinea Pig Model of Asthma. Adv. Pharm. Bull. 2014, 4, 131–138. [Google Scholar] [CrossRef] [PubMed]
  87. Mustafa, S.J.; Nadeem, A.; Fan, M.; Zhong, H.; Belardinelli, L.; Zeng, D. Effect of a Specific and Selective A2B Adenosine Receptor Antagonist on Adenosine Agonist AMP and Allergen-Induced Airway Responsiveness and Cellular Influx in a Mouse Model of Asthma. J. Pharmacol. Exp. Ther. 2007, 320, 1246–1251. [Google Scholar] [CrossRef] [Green Version]
  88. Zeil, S.; Schwanebeck, U.; Vogelberg, C. Tolerance and Effect of an Add-on Treatment with a Cough Medicine Containing Ivy Leaves Dry Extract on Lung Function in Children with Bronchial Asthma. Phytomedicine 2014, 21, 1216–1220. [Google Scholar] [CrossRef] [Green Version]
  89. Shokry, A.A.; El-Shiekh, R.A.; Kamel, G.; Bakr, A.F.; Ramadan, A. Bioactive Phenolics Fraction of Hedera helix L. (Common Ivy Leaf) Standardized Extract Ameliorates LPS-Induced Acute Lung Injury in the Mouse Model through the Inhibition of Proinflammatory Cytokines and Oxidative Stress. Heliyon 2022, 8, e09477. [Google Scholar] [CrossRef]
  90. Shokry, A.A.; El-Shiekh, R.A.; Kamel, G.; Bakr, A.F.; Sabry, D.; Ramadan, A. Anti-Arthritic Activity of the Flavonoids Fraction of Ivy Leaves (Hedera helix L.) Standardized Extract in Adjuvant Induced Arthritis Model in Rats in Relation to Its Metabolite Profile Using LC/MS. Biomed. Pharmacother. 2022, 145, 112456. [Google Scholar] [CrossRef]
  91. Watson, M.J.; Worthington, E.N.; Clunes, L.A.; Rasmussen, J.E.; Jones, L.; Tarran, R. Defective Adenosine-Stimulated CAMP Production in Cystic Fibrosis Airway Epithelia: A Novel Role for CFTR in Cell Signaling. FASEB J. 2011, 25, 2996–3003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Influence of EA 575® (A) and different incubation periods thereof (B) on the cellular reaction of HEK cells evoked by stimulation of A2BAR. Pre-incubation with different concentrations of EA 575® was performed for 16 h (A) and 1 or 16 h (B) before cells were stimulated with 0.5 µM BAY 60-6583. The positive wavelength shift mediated by the dynamic mass redistribution of intracellular particles was significantly and dose-dependently inhibited by pre-incubation with 160–240 µg/mL EA 575®, compared to stimulated control cells not pre-incubated with EA 575® (SC) (A). With an incubation time of 1 h, no reduction in BAY 60-6583-mediated wavelength shift was measured. A significant inhibition was only achieved by pre-incubation with 240 µg/mL EA 575® for 16 h (B). Data are shown as baseline-corrected (BC) AUC normalised to stimulated control cells not pre-incubated with EA 575® (SC). Results represent the mean and SEM ((A): n = 5 independent experiments performed in triplicate; (B): n = 1 experiment performed in triplicate, * p < 0.05).
Figure 1. Influence of EA 575® (A) and different incubation periods thereof (B) on the cellular reaction of HEK cells evoked by stimulation of A2BAR. Pre-incubation with different concentrations of EA 575® was performed for 16 h (A) and 1 or 16 h (B) before cells were stimulated with 0.5 µM BAY 60-6583. The positive wavelength shift mediated by the dynamic mass redistribution of intracellular particles was significantly and dose-dependently inhibited by pre-incubation with 160–240 µg/mL EA 575®, compared to stimulated control cells not pre-incubated with EA 575® (SC) (A). With an incubation time of 1 h, no reduction in BAY 60-6583-mediated wavelength shift was measured. A significant inhibition was only achieved by pre-incubation with 240 µg/mL EA 575® for 16 h (B). Data are shown as baseline-corrected (BC) AUC normalised to stimulated control cells not pre-incubated with EA 575® (SC). Results represent the mean and SEM ((A): n = 5 independent experiments performed in triplicate; (B): n = 1 experiment performed in triplicate, * p < 0.05).
Ijms 24 12373 g001
Figure 2. Influence of EA 575® (A) and different incubation periods thereof (B) on the intracellular cAMP level in HEK GloSensor™ cells elicited by stimulation of A2BAR. Prior to co-stimulation with 1 µM BAY 60-6583 and 1 µM forskolin, pre-incubation with different concentrations of EA 575® was performed for 16 h (A) and for 1–16 h with 160 µg/mL EA 575® (B). The A2BAR-induced cAMP increase was significantly and dose-dependently inhibited by pre-incubation with 160–240 µg/mL EA 575®, compared to stimulated control cells not pre-incubated with EA 575® (SC) (A). Pre-incubation for at least 8 h was necessary to cause a significant decrease in cAMP levels (B). Data are presented as AUC normalised to stimulated control cells not pre-incubated with EA 575® (SC). Results represent the mean and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Figure 2. Influence of EA 575® (A) and different incubation periods thereof (B) on the intracellular cAMP level in HEK GloSensor™ cells elicited by stimulation of A2BAR. Prior to co-stimulation with 1 µM BAY 60-6583 and 1 µM forskolin, pre-incubation with different concentrations of EA 575® was performed for 16 h (A) and for 1–16 h with 160 µg/mL EA 575® (B). The A2BAR-induced cAMP increase was significantly and dose-dependently inhibited by pre-incubation with 160–240 µg/mL EA 575®, compared to stimulated control cells not pre-incubated with EA 575® (SC) (A). Pre-incubation for at least 8 h was necessary to cause a significant decrease in cAMP levels (B). Data are presented as AUC normalised to stimulated control cells not pre-incubated with EA 575® (SC). Results represent the mean and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Ijms 24 12373 g002
Figure 3. Influence of EA 575® on the recruitment of β-arrestin 2 to the A2BAR in transiently transfected HEK cells induced by stimulation with NECA or BAY 60-6583. Pre-incubation with 160 µg/mL EA 575® was conducted for 16 h before cells were stimulated with 10 µM NECA or 10 µM BAY 60-6583. EA 575® was able to significantly inhibit the recruitment of β-arrestin 2 to the A2BAR induced by both NECA and BAY 60-6583. Data are shown as AUC of the fold change (FC) after stimulation normalised to equally stimulated control cells not pre-incubated with EA 575® (SC). Results represent the mean and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Figure 3. Influence of EA 575® on the recruitment of β-arrestin 2 to the A2BAR in transiently transfected HEK cells induced by stimulation with NECA or BAY 60-6583. Pre-incubation with 160 µg/mL EA 575® was conducted for 16 h before cells were stimulated with 10 µM NECA or 10 µM BAY 60-6583. EA 575® was able to significantly inhibit the recruitment of β-arrestin 2 to the A2BAR induced by both NECA and BAY 60-6583. Data are shown as AUC of the fold change (FC) after stimulation normalised to equally stimulated control cells not pre-incubated with EA 575® (SC). Results represent the mean and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Ijms 24 12373 g003
Figure 4. Influence of EA 575® (A,B) or the antagonists PSB-603 and SCH 442416 (C) on the CRE activation in transiently transfected HEK cells mediated by stimulation with adenosine (A,C) or BAY 60-6583 (B). Pre-incubation with 40–240 µg/mL EA 575® was conducted for 16 h (A,B) or with 0.1–10 µM of the antagonists for 2 h (C) before cells were stimulated by adding 100 µM adenosine (A,C) or 10 µM BAY 60-6583 (B). The non-specifically mediated CRE activation was significantly inhibited by pre-incubation with 80–240 µg/mL EA 575® (A) or 1 µM PSB-603 (C), compared to stimulated control cells not pre-incubated with EA 575® (SC). SCH 442416 also significantly reduced the CRE activation at a concentration of 10 µM, whereas this effect could not be observed at lower concentrations of both antagonists. Instead, 0.1 µM SCH 442416 slightly increased the CRE activation (C). The inhibition of the specific A2BAR-mediated CRE activation was achieved by pre-incubation with 80–240 µg/mL EA 575® (B). Influence of the A2AAR agonist CGS 21680 and, in comparison, BAY 60-6583 and adenosine on the CRE activation in transiently transfected HEK cells (D). Stimulation was performed with 1–10 µM CGS 21680, 10 µM BAY 60-6583, or 100 µM adenosine. The A2AAR-mediated CRE activation was significantly increased by 10 µM CGS 21680 compared with completely untreated control cells (UTC), but to a lesser extent than that mediated by A2BAR or non-specifically using adenosine (D). Data are shown as AUC of the fold change (FC) after stimulation normalised to stimulated control cells not pre-incubated with EA 575® (SC) (AC) or completely untreated control cells (UTC) (D). Results represent the mean and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Figure 4. Influence of EA 575® (A,B) or the antagonists PSB-603 and SCH 442416 (C) on the CRE activation in transiently transfected HEK cells mediated by stimulation with adenosine (A,C) or BAY 60-6583 (B). Pre-incubation with 40–240 µg/mL EA 575® was conducted for 16 h (A,B) or with 0.1–10 µM of the antagonists for 2 h (C) before cells were stimulated by adding 100 µM adenosine (A,C) or 10 µM BAY 60-6583 (B). The non-specifically mediated CRE activation was significantly inhibited by pre-incubation with 80–240 µg/mL EA 575® (A) or 1 µM PSB-603 (C), compared to stimulated control cells not pre-incubated with EA 575® (SC). SCH 442416 also significantly reduced the CRE activation at a concentration of 10 µM, whereas this effect could not be observed at lower concentrations of both antagonists. Instead, 0.1 µM SCH 442416 slightly increased the CRE activation (C). The inhibition of the specific A2BAR-mediated CRE activation was achieved by pre-incubation with 80–240 µg/mL EA 575® (B). Influence of the A2AAR agonist CGS 21680 and, in comparison, BAY 60-6583 and adenosine on the CRE activation in transiently transfected HEK cells (D). Stimulation was performed with 1–10 µM CGS 21680, 10 µM BAY 60-6583, or 100 µM adenosine. The A2AAR-mediated CRE activation was significantly increased by 10 µM CGS 21680 compared with completely untreated control cells (UTC), but to a lesser extent than that mediated by A2BAR or non-specifically using adenosine (D). Data are shown as AUC of the fold change (FC) after stimulation normalised to stimulated control cells not pre-incubated with EA 575® (SC) (AC) or completely untreated control cells (UTC) (D). Results represent the mean and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Ijms 24 12373 g004
Figure 5. Influence of EA 575® (A,B) or the antagonists PSB-603 and SCH 442416 (C) on the IL-6 release of Calu-3 cells mediated by stimulation with adenosine (A,C) or BAY 60-6583 (B). Pre-incubation with 40–240 µg/mL EA 575® was conducted for 16 h (A,B) or with 0.1–10 µM of the antagonists for 1 h (C) before cells were stimulated by adding 100 µM adenosine (A,C) or 10 µM BAY 60-6583 (B) for another 24 h. The non-specifically mediated IL-6 release was significantly and dose-dependently inhibited by pre-incubation with 80–240 µg/mL EA 575® (A) or 1 µM PSB-603 (C), compared to stimulated control cells not pre-incubated with EA 575® (SC). SCH 442416, however, increased the concentration of IL-6 even further. (C). The inhibition of the specific A2BAR-mediated IL-6 release was achieved by pre-incubation with 40–240 µg/mL EA 575® (B). Influence of the A2AAR agonist CGS 21680 and, in comparison, BAY 60-6583 on the IL-6 release of Calu-3 cells (D). Stimulation was performed with 1–10 µM CGS 21680 or 10 µM BAY 60-6583 for 24 h. The A2AAR-mediated IL-6 release was slightly, but neither significantly nor dose-dependently, increased compared to completely untreated control cells (UTC) (D). Results represent the mean normalised to stimulated control cells not pre-incubated with EA 575® (SC) (AC) or completely untreated control cells (UTC) (D) and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Figure 5. Influence of EA 575® (A,B) or the antagonists PSB-603 and SCH 442416 (C) on the IL-6 release of Calu-3 cells mediated by stimulation with adenosine (A,C) or BAY 60-6583 (B). Pre-incubation with 40–240 µg/mL EA 575® was conducted for 16 h (A,B) or with 0.1–10 µM of the antagonists for 1 h (C) before cells were stimulated by adding 100 µM adenosine (A,C) or 10 µM BAY 60-6583 (B) for another 24 h. The non-specifically mediated IL-6 release was significantly and dose-dependently inhibited by pre-incubation with 80–240 µg/mL EA 575® (A) or 1 µM PSB-603 (C), compared to stimulated control cells not pre-incubated with EA 575® (SC). SCH 442416, however, increased the concentration of IL-6 even further. (C). The inhibition of the specific A2BAR-mediated IL-6 release was achieved by pre-incubation with 40–240 µg/mL EA 575® (B). Influence of the A2AAR agonist CGS 21680 and, in comparison, BAY 60-6583 on the IL-6 release of Calu-3 cells (D). Stimulation was performed with 1–10 µM CGS 21680 or 10 µM BAY 60-6583 for 24 h. The A2AAR-mediated IL-6 release was slightly, but neither significantly nor dose-dependently, increased compared to completely untreated control cells (UTC) (D). Results represent the mean normalised to stimulated control cells not pre-incubated with EA 575® (SC) (AC) or completely untreated control cells (UTC) (D) and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Ijms 24 12373 g005
Figure 6. Influence of different adenosine receptor agonists on NFκB transcriptional activity. Calu-3 cells stably expressing a secreted NanoLuc® under control of an NFκB binding sequence were stimulated for 3 h. An amount of 25 ng/mL TNFα was used as positive control, resulting in a significant increase in the luminescence signal. Amounts of 10–100 µM adenosine, 1–10 µM NECA, or 1–10 µM BAY 60-6583 did not show any effect on the NFκB transcriptional activity. Results represent the mean normalised to completely untreated control cells (UTC) and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Figure 6. Influence of different adenosine receptor agonists on NFκB transcriptional activity. Calu-3 cells stably expressing a secreted NanoLuc® under control of an NFκB binding sequence were stimulated for 3 h. An amount of 25 ng/mL TNFα was used as positive control, resulting in a significant increase in the luminescence signal. Amounts of 10–100 µM adenosine, 1–10 µM NECA, or 1–10 µM BAY 60-6583 did not show any effect on the NFκB transcriptional activity. Results represent the mean normalised to completely untreated control cells (UTC) and SEM (n = 3 independent experiments performed in triplicate, * p < 0.05).
Ijms 24 12373 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Meurer, F.; Häberlein, H.; Franken, S. Ivy Leaf Dry Extract EA 575® Has an Inhibitory Effect on the Signalling Cascade of Adenosine Receptor A2B. Int. J. Mol. Sci. 2023, 24, 12373. https://doi.org/10.3390/ijms241512373

AMA Style

Meurer F, Häberlein H, Franken S. Ivy Leaf Dry Extract EA 575® Has an Inhibitory Effect on the Signalling Cascade of Adenosine Receptor A2B. International Journal of Molecular Sciences. 2023; 24(15):12373. https://doi.org/10.3390/ijms241512373

Chicago/Turabian Style

Meurer, Fabio, Hanns Häberlein, and Sebastian Franken. 2023. "Ivy Leaf Dry Extract EA 575® Has an Inhibitory Effect on the Signalling Cascade of Adenosine Receptor A2B" International Journal of Molecular Sciences 24, no. 15: 12373. https://doi.org/10.3390/ijms241512373

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop