You are on page 1of 6

A2B Adenosine Receptors Induce IL-19 from Bronchial

Epithelial Cells, Resulting in TNF-␣ Increase


Hongyan Zhong, Yuzhi Wu, Luiz Belardinelli, and Dewan Zeng
Department of Drug Research and Pharmacological Sciences, CV Therapeutics, Inc., Palo Alto, California

Adenosine is a signaling nucleoside that has been proposed to to the pathogenesis of asthma and chronic obstructive pulmonary
contribute to the pathogenesis of asthma and chronic obstructive disease (COPD) (3). This hypothesis is based on the findings
pulmonary disease. Previous studies suggest that adenosine might that the interstitial concentration of adenosine is elevated in the
play an important role in modulating levels of inflammatory media- lungs of individuals with asthma (4) and inhaled adenosine
tors in the lung. Because airway epithelium is an important cellular causes bronchoconstriction in patients with asthma (5). The
source of inflammatory mediators, the objective of the present bronchoconstrictive effect of adenosine has been attributed to
study was to determine whether adenosine affects the expression
the activation of lung mast cells (6, 7). In addition to the mast
and release of inflammatory cytokines from human bronchial
cells (8, 9), adenosine has been found to modulate the functions
epithelial cells (HBECs). Among the four subtypes of adenosine
of other inflammatory cells such as lymphocytes (10), eosinophils
receptors, the A2B receptor was expressed at the highest level.
5ⴕ-(N-ethylcarboxamido)-adenosine (NECA), a stable analog of
(11, 12), neutrophils (13), and macrophages (14), and lung cells
adenosine, increased the release of IL-19 by 4.6- ⴞ 1.1-fold. A selec- such as bronchial smooth muscle cells (15, 16) and fibroblasts (17).
tive antagonist of the A2B receptor, CVT-6694, attenuated this effect The physiologic effects of adenosine on lung epithelial cells have
of NECA. The amount of IL-19 released from HBEC was sufficient also been investigated. Adenosine is able to modulate the activity
to activate a human monocytic cell line (THP-1) and increase the of ion channels (18–20), and induce fibronectin (21) and mucin
release of TNF-␣. Furthermore, TNF-␣ was found to upregulate A2B gene expression (22). However, it is unknown whether and how
receptor expression in HBECs by 3.1- ⴞ 0.3-fold. Hence, these data adenosine affects the release of inflammatory cytokines from epi-
indicate that NECA increases the release of IL-19 from HBECs via thelial cells and epithelial cell–inflammatory cell communication.
activation of A2B receptors, and IL-19 in turn activates human mono- In this study, we used the primary cultured human bronchial
cytes to release TNF-␣, which upregulates A2B receptor expression epithelial cells (HBECs) as the model system. The objectives of
in HBECs. The results of this study suggest that there is a novel this study were to determine (1 ) whether adenosine affects the
pathway whereby adenosine can initiate and amplify an inflamma- release of inflammatory cytokines from bronchial epithelial cells,
tory response which might be important in pathogenesis of inflam- (2 ) what the potential effects of these cytokines on lung inflam-
matory lung diseases. mation might be, and (3 ) which adenosine receptor subtype is
Keywords: adenosine; TNF-␣; bronchial epithelial cells; IL-19 responsible for the effect of adenosine.

Airway epithelium is known to play an important role in airway MATERIALS AND METHODS
defense mechanism via a mucociliary system and as a mechanical
barrier. Recent studies further indicate that the epithelium is Materials
not only a barrier but can also actively generate a range of A selective antagonist to the A2B receptor (CVT-6694) was synthesized
molecules, including lipid mediators, growth factors, and a vari- by the Department of Bio-Organic Chemistry at CV Therapeutics Inc.
ety of cytokines/chemokines, that are important in the inflam- (Palo Alto, CA), and was described in our previous publication (15). All
matory and remodeling responses that occur in the lungs (1). other reagents, such as rolipram, forskolin, 5⬘-(N-ethylcarboxamido)-
adenosine (NECA), and adenosine deaminase (ADA), were purchased
In asthma, the epithelium seems more sensitive and responds
from Sigma (St. Louis, MO) unless otherwise stated.
abnormally to various stimuli (2). In addition, airway epithelial
cells are able to interact with immune and inflammatory cells Cell Culture
via direct adhesion as well as by releasing mediators including
Primary cultured normal HBECs were obtained from Clonetics (San
cytokines (1). Thus, the epithelium is actively involved as regula-
Diego, CA) and cultured using bronchial epithelial cell growth medium
tor of airway inflammatory responses important in the pathogen- (Clonetics). HBECs were routinely grown in a humidified incubator with
esis of airway disorders. 5% CO2 at 37⬚C. Cells from three different donors and from passages 2–3
Adenosine is a nucleoside that can elicit many physiologic were used in the following studies. THP-1 cells were purchased from
and pathophysiologic responses by activating one or more of its ATCC (Manassas, VA) and cultured according to ATCC’s instructions.
four subtypes of G protein–coupled receptors (A1, A2A, A2B, and
A3) on target cells. Adenosine has been proposed to contribute Stimulation of HBECs
HBECs were seeded into 12-well tissue culture plates at a density of
1 ⫻ 105 cells/well and allowed to adhere overnight and reach ⵑ 90%
confluence. Cells were washed twice in HEPES-buffered saline, and
(Received in original form December 21, 2005 and in final form June 1, 2006 )
cultured in bronchial epithelial cell basal medium (Clonetics) containing
various agonists or antagonists of AdoRs for 1 or 24 h.
An abstract (less than 250 words) related to this study has been published
for the American Thoracic Society International Conference in 2005: Zhong H,
RNA Extraction and Real-Time RT-PCR
Wu Y, Belardinelli L, Zeng D. Adenosine indirectly activates monocytes by releasing
IL-19 from human bronchial epithelial cells. Proc Am Thorac Soc 2005;2:A110. Total RNA was extracted from HBECs using the Stratagene Absolutely
Correspondence and requests for reprints should be addressed to Hongyan Zhong, RNA RT-PCR Miniprep Kit (Stratagene Corp., La Jolla, CA) followed
Ph.D., CV Therapeutics, Inc., 3172 Porter Drive, Palo Alto, CA 94304. E-mail: by DNase treatment to eliminate potential genomic DNA contamina-
hongyan.zhong@cvt.com tion. Real-time RT-RCR for adenosine receptors was performed as
Am J Respir Cell Mol Biol Vol 35. pp 587–592, 2006
previously described (15). Specific primers for IL-19 (forward: 5⬘-AAA
Originally Published in Press as DOI: 10.1165/rcmb.2005-0476OC on June 15, 2006 CAATCTCCCCAAGGTGGAT-3⬘; reverse: 5⬘-AGGAAATGCTGT
Internet address: www.atsjournals.org CAAGGTTTGC-3⬘), GRO-␤ (forward: 5⬘-TTTCTTCGTGATGACA
588 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 35 2006

TATCACATGT-3⬘; reverse: 5⬘-TCCTCAGCCTCTATCACAGTGG-


3⬘), and GRO-␥ (forward: 5⬘-TGCTTGTAGGGCATAATGCCT-3⬘;
reverse: 5⬘-GGGAAAGAGAAACGCTGCAG-3⬘) were designed using
Primer Express 2.0 (Applied Biosystems, Foster City, CA) following
the recommended guidelines based on sequences from GenBank. At
the end of the PCR cycle, a dissociation curve was generated to ensure
the amplification of a single product, and the threshold cycle times (Ct
values) for each gene were determined. Relative mRNA levels were
calculated based on the Ct values, normalized to ␤-actin in the same
sample, and presented as percentages of ␤-actin mRNA.

Measurement of cAMP Accumulation Figure 1. The mRNA levels of AdoR subtypes in HBECs. Total RNA iso-
Cells were harvested using 0.0025% trypsin and 2 mM EDTA in PBS, lated from HBECs was subjected to real-time RT-PCR analysis. The rela-
washed and resuspended in phenol-free DMEM to a concentration of tive levels of the AdoR transcripts are presented as percentages of the
5 ⫻ 105 cells/ml, and then incubated with 1 U/ml of ADA for 30 min ␤-actin transcript. Data shown are averages ⫾ SEM (n ⫽ 6). nd denotes
at room temperature. Cells were then treated with AdoR agonists, not detected.
antagonists, and forskolin in the presence of 50 ␮M of the phosphodies-
terase IV inhibitor, rolipram. After incubating for 15 min at 37⬚C, cells
were lysed and cAMP concentrations were determined using cAMP-
Screen Direct System (Applied Biosystems) according to the manufac- mined. NECA is a stable analog of adenosine, and it activates
turer’s instructions. all four AdoR subtypes including A2B receptors. As shown in
Figure 2A, NECA increased cellular cAMP accumulation in a
cDNA Array Analysis concentration-dependent manner, with potency (EC50 value) of
Expression of inflammatory cytokines was determined using a cDNA 8.8 ⫾ 1.3 ␮M. In contrast, the A2A-selective agonist CGS-21680
array kit (Cat. No. HS-033; SuperArray, Frederick, MD). The assay (⭐ 10 ␮M) did not cause a significant increase in cellular cAMP
was performed according to manufacturer’s instructions. Biotinylated concentration. In addition, the A1-selective agonist, CPA (1 ␮M),
cDNA probes were generated from 1–2 ␮g of total RNA using GEArray
and the A3-selective agonist, IB-MECA (1 ␮M), failed to inhibit
RT-labeling enzyme kit (SuperArray). The labeled cDNA probes were
the cellular cAMP accumulation caused by forskolin (10 ␮M,
then hybridized with gene-specific cDNA fragments spotted on nylon
membrane. The signals were detected with the chemiluminescent detec- Figure 2B). Because there is no selective agonist for A2B re-
tion reagents. The relative expression level of each gene was analyzed ceptors, the effect of a selective antagonist to A2B receptors,
using GEArray analysis software (SuperArray). CVT-6694, on NECA-induced cellular cAMP accumulation was

Measurement of IL-19 and TNF-␣


ELISA for IL-19 was developed according to manufacturer’s protocol
(R&D Systems, Minneapolis, MN). Plates (96-well) were pre-coated
with 50 ng/well anti-human IL-19 Ab (R&D Systems). IL-19 was de-
tected using 100 ng/ml of biotinylated polyclonal IL-19 Ab (R&D sys-
tems), streptavidin-HRP, and TMB (tetramethylbenzidine) chromogen
(Biosource, Camarillo, CA). Serial dilutions of recombinant human IL-
19 (rhIL-19; PeproTech, Rocky Hill, NJ) was used as standard. The
detection limit of rhIL-19 was 0.2 ng/ml. The concentrations of TNF-␣
in the cell medium were determined using ELISA kits obtained from
Biosource according to the manufacturer’s instructions. The minimal
detection level of TNF-␣ with these kits was 1.7 pg/ml.

Statistical Analysis
Data are presented as mean ⫾ SEM of at least three separate experi-
ments. The statistical analysis was performed using ANOVA followed
by Bonferroni test. A P value of ⬍ 0.05 was considered significant.

RESULTS
Expression of AdoR Subtypes in HBECs
Real-time RT-PCR was performed to quantify the levels of
transcripts for AdoRs. Among the four subtypes, the A2B recep-
tor had the highest transcript level (0.49 ⫾ 0.04% of ␤-actin
expression) (Figure 1). Lower levels of A1 and A2A receptor
transcripts were also detected (0.0012 ⫾ 0.0002% and 0.0058 ⫾
0.0003% of ␤-actin, respectively), whereas the transcript for A3
receptors was below the detection level. Hence, the rank order
of AdoR mRNA levels was A2B ⬎ ⬎ A2A ⬎ A1 ⬎ ⬎ A3.
Figure 2. Effects of AdoR agonists and antagonist on cellular cAMP
In many cell types, activation of A2A or A2B receptors leads accumulation in HBECs. (A ) Concentration–response curves of CGS-
to increases in cellular cAMP accumulation, whereas activation 21680 (CGS, circles) and NECA in the absence (squares) or presence
of A1 or A3 receptors decreases cellular cAMP accumulation (triangles) of the A2B receptor antagonist CVT-6694 (1 ␮M). (B ) Lack of
caused by the adenylate cyclase activator, forskolin. To identify effect of CPA (1 ␮M) and IB-MECA (IM, 1 ␮M) on forskolin (Fsk,
the AdoR subtype(s) that are functionally expressed in HBECs, 10 ␮M)-induced cellular cAMP accumulation. Data shown are averages ⫾
the effects of a nonselective agonist NECA and several other SEM (n ⫽ 3). *P ⬍ 0.05, compared with control; #P ⬍ 0.05, compared
selective agonists on cellular cAMP accumulation were deter- with NECA-treated cells in A.
Zhong, Wu, Belardinelli, et al.: Adenosine Induces IL-19 and Activates Monocytes 589

determined. CVT-6694 has a high affinity for the A2B receptor Figure 4. Effect of NECA
(Ki ⫽ 7 nM) and very low affinity for three other AdoR subtypes on the release of IL-19 by
(Ki values are ⬎ 5 ␮M for A1, A2A, and A3 receptors) (15, 17). As HBECs. Cells were treated
shown in Figure 2A, CVT-6694 (1 ␮M) significantly attenuated with vehicle, NECA in the
NECA-induced cellular cAMP accumulation. Thus, using cellu- absence or presence of
CVT-6694 for 24 h. Media
lar cAMP concentration as readout for the functional expression
from treated cells were
of AdoRs, the results indicate that A2B receptors are functionally
collected, and the con-
expressed in HBECs, whereas A1, A2A, or A3 receptors are not. centrations of IL-19 were
determined using ELISA.
Effects of NECA on Expression of Inflammatory Cytokines
CVT-6994 alone did not
The effect of NECA on the gene expression of the inflammatory change the release of IL-19 by HBECs (data not shown). Data shown
cytokines was determined using a cDNA array containing 85 are the averages ⫾ SEM (n ⫽ 3). *P ⬍ 0.05, compared with control;
inflammatory genes involved in asthma. Among the 85 genes on #
P ⬍ 0.05, compared with NECA-treated cells.
the microarray, GRO-␤, GRO-␥, and IL-19 genes were increased
above 2-fold by NECA (data not shown).
To confirm and quantify NECA-induced expression of
GRO-␤, GRO-␥, and IL-19, gene-specific real-time RT-PCR or NECA (10 ␮M) alone had no significant effect on TNF-␣
was performed on HBECs treated with NECA (10 ␮M) for release from THP-1 cells (Figure 5A). These data demonstrate
1 h. As shown in Figure 3, NECA increased mRNA expression that IL-19, but not NECA or GRO-␤/␥, was able to activate
of IL-19, GRO-␤, and GRO-␥ up to 4.3- ⫾ 0.9-, 5.6- ⫾ 0.6-, and THP-1 cells. Furthermore, the conditional media collected from
4.3- ⫾ 0.6-fold above control levels, respectively. HBECs treated with vehicle (control-M) or NECA (NECA-M)
for 24 h were used to incubate THP-1 cells for an additional
Activation of the A2B Receptor Increased the Release of 24 h. NECA-M caused a 4.6- ⫾ 1.0-fold increase in TNF-␣ release
IL-19 from HBECs from THP-1 cells compared with control-M. This effect was
Recent publications suggesting a potential role of IL-19 in lung completely abolished by an IL-19–neutralizing Ab (Figure 5B).
inflammation prompted us to determine the effect of NECA on These results suggest that IL-19 released from NECA-stimulated
IL-19 release. IL-19 concentrations in the culture media from HBECs is able to and sufficient to activate THP-1 cells.
cells treated with NECA were measured using ELISA. The
levels of IL-19 in the media from vehicle- and NECA-treated Effects of TNF-␣ on Expression of AdoRs in HBECs
cells for 24 h were 19.2 ⫾ 6.3 ng/ml and 89.2 ⫾ 20.2 ng/ml, To further explore the interaction between epithelial cells and
respectively (Figure 4). Hence, NECA (10 ␮M) caused 4.6- ⫾ monocytes, the effect of TNF-␣ on AdoR expression in HBECs
1.1-fold increase of IL-19 release compared with vehicle-treated was determined using real-time RT-PCR. TNF-␣ caused a sig-
cells. To determine the role of A2B receptors in NECA-induced nificant time-dependent increase in mRNA expression of A2B
IL-19 production, cells were incubated with CVT-6694 (1 ␮M)
together with NECA. The A2B receptor antagonist CVT-6694
(1 ␮M) reduced the NECA-increased IL-19 release by 88.9 ⫾
0.5%. These results confirmed that NECA-induced IL-19 release
is mediated by the A2B receptor subtype.

Effects of IL-19 Released from HBEC on Activation of a


Human Monocytic Cell Line (THP-1)
To determine whether the amount of IL-19 released from HBEC
can activate inflammatory cells, the effect of IL-19 alone or the
conditional medium from NECA-treated HBEC on the release
of TNF-␣ from a monocytic cell line (THP-1) was determined.
The basal level of TNF-␣ in the media from vehicle-treated
(24 h) THP-1 cells was 7.5 ⫾ 0.5 pg/ml. IL-19 (100 ng/ml) increased
the concentrations of TNF-␣ in the media by 4.3- ⫾ 0.4-fold
(Figure 5A). In contrast, GRO-␤ (100 ng/ml), GRO-␥ (100 ng/ml),

Figure 3. Effects of NECA on


the mRNA levels of GRO-␤,
GRO-␥, and IL-19 determined
using real-time RT-PCR.
HBECs were incubated with Figure 5. Effects of cytokines and conditional medium from HBECs on
NECA (10 ␮M) for 1 h. Cells TNF-␣ release from THP-1 cells. (A ) THP-1 cells were incubated with
incubated with vehicle were vehicle (control), GRO-␤ (100 ng/ml), GRO-␥ (100 ng/ml), IL-19
used as control. The expres- (100 ng/ml), or NECA (10 ␮M) for 24 h. (B ) Conditional medium col-
sion levels of the cytokines lected from HBECs that were treated with vehicle (control-M) or NECA
were normalized to that of ␤-actin and are presented as the ratios of (NECA-M) for 24 h was used to incubate THP-1 cells in the absence or
the expression level in NECA-treated cells versus that in vehicle-treated presence of polyclonal IL-19 Ab (1 ␮g/ml) for an additional 24 h. The
cells. The expression levels of GRO-␤, GRO-␥, and IL-19 in control cells concentration of TNF-␣ in media was measured using ELISA. Data shown
are 0.020 ⫾ 0.003%, 0.008 ⫾ 0.002%, and 0.168 ⫾ 0.025% of are the averages ⫾ SEM (n ⫽ 4). *P ⬍ 0.05, compared with control
␤-actin, respectively. Data shown are averages ⫾ SEM (n ⫽ 3). (A ) or control-M (B ); #P ⬍ 0.05, compared with NECA-M in B.
590 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 35 2006

receptors (Figure 6). After 24 h incubation with TNF-␣, mRNA with COPD compared with the control group, whereas the ra-
level of A2B receptors was increased to 3.1 ⫾ 0.3 fold above dioligand affinity for A2B receptors was not altered (29). How-
basal levels (Figure 6). Incubation of TNF-␣ for 5 and 24 h ever, it is not entirely clear which cells contribute to the decrease
also significantly upregulated the mRNA levels of A1 and A2A of A2B receptors in COPD, how the expression of A2B receptor
receptors up to 2.0- ⫾ 0.3- and 1.6- ⫾ 0.1-fold above basal levels, is regulated in allergic diseases versus COPD, and the precise
although the expression of both subtypes of receptors was still role of A2B receptor in these diseases. Interestingly, in the same
very low (below 0.01% of ␤-actin mRNA level). The mRNA study, the expression level of the A2B receptor was found to be
expression of the A3 receptors remained below the detection highest in mast cells and macrophages. Regardless, the authors
level after TNF-␣ treatment (Figure 6). concluded that the A2B receptor might play an important role
in the pathogenesis of COPD. Therefore, future studies to deter-
DISCUSSION mine the effect of A2B antagonist in COPD would be of interest.
Functional expression of the A2B receptor in airway epithelial
The main objective of this study was to determine the effect of cells has been reported. This receptor has been shown to mediate
adenosine on the inflammatory responses mediated by epithelial adenosine-modulated ion transport (18, 19); however, the func-
cells. The main findings of our study are: (1 ) A2B receptors tion of A2B receptor in mediating cytokine release is not known.
are the predominant subtype of AdoRs expressed in primary The results of our study showed that activation of A2B receptors
HBECs; (2 ) activation of A2B receptors on HBECs increases upregulates the expression of proinflammatory cytokines such
the release of IL-19, which is able to and sufficient to induce as IL-19, GRO-␤, and GRO-␥. GRO-␤ (CXCL2) and GRO-␥
TNF-␣ production from human monocytes; and (3 ) TNF-␣ can (CXCL3) are CXC chemokines. The functions of these two
upregulate the expression of A2B receptors in HBECs. These chemokines are similar to IL-8 (CXCL8), which belongs to the
findings support the hypothesis that adenosine can initiate and same chemokine family. GRO-␤ and GRO-␥ chemoattract and
amplify inflammatory responses which might be important in activate neutrophils (30), and promote angiogenesis by activating
pathogenesis of inflammatory lung diseases. the CXCR2 receptor on endothelial cells (31). Because neutro-
Among the four subtypes of AdoRs, the A2B receptor has phils have been implicated in the pathogenesis of inflammatory
the lowest affinity for the natural ligand, adenosine, requiring lung diseases including asthma and COPD, and because CXC
at least micromolar concentrations of adenosine for its activation chemokine–mediated angiogenesis is associated with idiopathic
(23). Hence, under normal physiologic conditions, adenosine pulmonary fibrosis (32), CXCR2 is a potential therapeutic target
levels might be too low to activate the A2B receptor. However, for these diseases. A more important finding of the present study
higher levels of adenosine have been detected during hypoxia, is that NECA increases the release of IL-19 from HBECs.
ischemia, and inflammation (24–26). In addition, results of recent IL-19 belongs to the IL-10 family, which includes IL-10, IL-19,
studies (16, 17, 26, 27) suggest the A2B receptor expression can IL-20, IL-22, IL-24, and IL-26 (33). In contrast to the pleiotropic
markedly increase during inflammation and hypoxia. The molec- role of IL-10, IL-19 has been shown to have mainly proinflam-
ular mechanism of how A2B receptor expression is modulated matory roles. IL-19 stimulates T cells to produce IL-4 and
during inflammation is largely unknown. The results of the cur- IL-13 (34) and alters the balance of Th1/Th2 in favor of Th2.
rent study demonstrate that TNF-␣, a well-known inflammatory IL-19 also induces production of IL-6, TNF-␣, and reactive oxy-
marker and mediator, upregulates the expression of A1, A2A, gen species from mouse monocytes (35). IL-19 level is elevated
and A2B receptors in HBECs. This is consistent with an earlier in the serum of patients with asthma compared with healthy
report suggesting that TNF-␣ regulates the expression of A2A control subjects, and its transcript is also increased in the lung
and A2B receptors in microvascular endothelial cells (28). of an allergen-challenged mouse model (34). Because adenosine
It should be noted that the expression of A2B receptors are is elevated in the asthmatic lung to hundred-micromolar range
upregulated in the lung of ragweed-challenged mice (27) and (4), and at this level it can increase the release of IL-19 from
IL-13–transgenic mice (26). More strikingly, increased expres- HBECs (data not shown), it is plausible to postulate that elevated
sion of A2B receptors in the lung of IL-13–transgenic mice has adenosine in the asthmatic lung may contribute to the elevation
been localized in bronchial epithelium. On the other hand, re- of IL-19. Interestingly, recent studies have shown that elevated
sults of a recent study suggested that the density of A2B receptor, adenosine play a key role in the upregulation of IL-13 and several
determined using saturation binding assays with an antagonist other cytokines in mouse models of pulmonary diseases (26).
radioligand, was significantly decreased in the lungs of patients Consistent with earlier reports on the proinflammatory roles
of IL-19, the results of our study demonstrate that IL-19 released
by bronchial epithelial cells can activate human monocytes. In
the present study, conditional medium of NECA-stimulated
HBECs was able to activate THP-1 cells. This effect is not due
Figure 6. Effect of TNF-␣ to the activation of AdoRs on THP-1 cells, because this effect was
on the mRNA levels of completely abolished by the IL-19–neutralizing Ab and NECA
AdoR subtypes in HBECs. alone did not stimulate TNF-␣ production from THP-1 cells.
HBECs were incubated Furthermore, the results in this study suggest that elevated
with TNF-␣ (5 ng/ml) for TNF-␣ could have a positive feedback on the expression of the
0, 1, 5, and 24 h. The
adenosine receptors on the epithelial cells. Hence, adenosine
relative levels of the
may play an important role in the interaction between epithelial
AdoR transcripts are pre-
sented as percentages of
cells and inflammatory cells in the airway.
the ␤-actin transcript. Activation of the A2B receptor increases cAMP, which is
Data shown are aver- generally believed to elicit anti-inflammatory responses. How-
ages ⫾ SEM (n ⫽ 3). nd ever, proinflammatory roles of this receptor have been reported
denotes not detected. (36). For example, in human mast cells (HMC-1), activation of
*P ⬍ 0.05, compared A2B receptors increase cAMP (37) and also increase the release
with 0 h. of IL-4, IL-8, and IL-13 (8). A possible explanation is that A2B
receptor can couple to other cell signaling pathways, such as
Zhong, Wu, Belardinelli, et al.: Adenosine Induces IL-19 and Activates Monocytes 591

2. Holgate ST. Epithelial damage and response. Clin Exp Allergy 2000;30:
37–41.
3. Holgate ST, Church MK, Polosa R. Adenosine: a positive modulator of
airway inflammation in asthma. Ann N Y Acad Sci 1991;629:227–236.
4. Driver AG, Kukoly CA, Ali S, Mustafa SJ. Adenosine in bronchoalveolar
lavage fluid in asthma. Am Rev Respir Dis 1993;148:91–97.
5. Cushley MJ, Tattersfield AE, Holgate ST. Adenosine-induced broncho-
constriction in asthma: antagonism by inhaled theophylline. Am Rev
Respir Dis 1984;129:380–384.
6. Cushley MJ, Holgate ST. Adenosine-induced bronchoconstriction in
asthma: role of mast cell-mediator release. J Allergy Clin Immunol
1985;75:272–278.
7. Polosa R, Ng WH, Crimi N, Vancheri C, Holgate ST, Church MK,
Mistretta A. Release of mast-cell-derived mediators after endobron-
chial adenosine challenge in asthma. Am J Respir Crit Care Med
1995;151:624–629.
8. Ryzhov S, Goldstein AE, Matafonov A, Zeng D, Biaggioni I, Feoktistov
I. Adenosine-activated mast cells induce IgE synthesis by B lympho-
cytes: an A2B-mediated process involving Th2 cytokines IL-4 and
IL-13 with implications for asthma. J Immunol 2004;172:7726–7733.
9. Zhong H, Shlykov SG, Molina JG, Sanborn BM, Jacobson MA,
Tilley SL, Blackburn MR. Activation of murine lung mast cells by the
adenosine A3 receptor. J Immunol 2003;171:338–345.
10. Huang S, Apasov S, Koshiba M, Sitkovsky M. Role of A2a extracellular
adenosine receptor-mediated signaling in adenosine-mediated inhibi-
tion of T-cell activation and expansion. Blood 1997;90:1600–1610.
11. Walker BA, Jacobson MA, Knight DA, Salvatore CA, Weir T, Zhou D,
Bai TR. Adenosine A3 receptor expression and function in eosino-
phils. Am J Respir Cell Mol Biol 1997;16:531–537.
12. Young HW, Molina JG, Dimina D, Zhong H, Jacobson M, Chan LN,
Chan TS, Lee JJ, Blackburn MR. A3 adenosine receptor signaling
contributes to airway inflammation and mucus production in adenosine
deaminase-deficient mice. J Immunol 2004;173:1380–1389.
Figure 7. Schematic representation of an inflammatory response initi- 13. Cronstein BN. Adenosine regulation of neutrophil function and inhibi-
ated and amplified by adenosine involving HBECs and monocytes. Aden- tion of inflammation via adenosine receptors. In: Jacobson KA, Jarvis
osine, via activation of the A2B receptors, increases the release of IL-19 MF, editors. Purinergic approaches in experimental therapeutics.
from HBECs. IL-19, in turn, stimulates human monocytes to release New York: Wiley-Liss; 1997. pp. 285–299.
TNF-␣. TNF-␣ upregulates A2B receptor expression in HBECs. 14. Hasko G, Szabo C, Nemeth ZH, Kvetan V, Pastores SM, Vizi ES. Adeno-
sine receptor agonists differentially regulate IL-10, TNF-alpha, and
nitric oxide production in RAW 264.7 macrophages and in endotoxe-
mic mice. J Immunol 1996;157:4634–4640.
Ca/PLC, and MAP kinases (38), and these pathways are likely 15. Zhong H, Belardinelli L, Maa T, Feoktistov I, Biaggioni I, Zeng D. A2B
to mediate the proinflammatory effect of adenosine or adenosine adenosine receptors increase cytokine release by bronchial smooth
muscle cells. Am J Respir Cell Mol Biol 2004;30:118–125.
analog via activation of A2B receptors. However, it remains to
16. Feoktistov I, Ryzhov S, Zhong H, Goldstein AE, Matafonov A, Zeng D,
be established which signaling pathway is responsible for the Biaggioni I. Hypoxia modulates adenosine receptors in human endo-
A2B-mediated upregulation of IL-19. It should be noted that anti- thelial and smooth muscle cells toward an A2B angiogenic phenotype.
inflammatory roles of the A2B receptor have also been reported Hypertension 2004;44:649–654.
in other in vitro studies (39–41). Therefore, further studies using 17. Zhong H, Belardinelli L, Maa T, Zeng D. Synergy between A2B adeno-
animal models or patients will be helpful to determine the role sine receptors and hypoxia in activating human lung fibroblasts. Am
of A2B receptors in the lung. J Respir Cell Mol Biol 2005;32:2–8.
18. Huang P, Lazarowski ER, Tarran R, Milgram SL, Boucher RC, Stutts MJ.
In summary, adenosine, IL-19, and TNF-␣ are important pro-
From the Cover: Compartmentalized autocrine signaling to cystic fi-
inflammatory mediators in lung inflammation. These mediators brosis transmembrane conductance regulator at the apical membrane
may interact with one another to cause the alleviation or exacer- of airway epithelial cells. Proc Natl Acad Sci USA 2001;98:14120–
bation of the disease. The results of the present study demon- 14125.
strated that the A2B receptor subtype is the predominant AdoR 19. Kornerup KN, Page CP, Moffatt JD. Pharmacological characterisation
expressed in HBECs. Activation of this AdoR subtype increased of the adenosine receptor mediating increased ion transport in the
the release of IL-19. IL-19 released from these cells, in turn, mouse isolated trachea and the effect of allergen challenge. Br J
stimulated the human monocytes to release TNF-␣. TNF-␣ is Pharmacol 2005;144:1011–1016.
20. Szkotak AJ, Ng AM, Sawicka J, Baldwin SA, Man SF, Cass CE, Young JD,
able to upregulate the expression of A2B receptors in HBECs Duszyk M. Regulation of K(⫹) current in human airway epithelial
(Figure 7). Thus, our findings provide a novel mechanism that cells by exogenous and autocrine adenosine. Am J Physiol Cell Physiol
could explain the interactions among adenosine, IL-19, and 2001;281:C1991–C2002.
TNF-␣ in the initiation, maintenance, and amplification of in- 21. Roman J, Rivera HN, Roser-Page S, Sitaraman SV, Ritzenthaler JD.
flammatory responses. In addition, our findings suggest that the Adenosine induces fibronectin expression in lung epithelial cells: impli-
A2B receptor might be a novel therapeutic target for inflamma- cations for airway remodeling. Am J Physiol Lung Cell Mol Physiol
tory lung diseases. 2006;290:L317–L325.
22. McNamara N, Gallup M, Khong A, Sucher A, Maltseva I, Fahy JV,
Conflict of Interest Statement : H.Z., Y.W., L.B., and D.Z. are employees of CV Basbaum C. Adenosine up-regulation of the mucin gene, MU2, in
Therapeutics, Inc. (CVT) and own stock and stock options in this company. CVT asthma. FASEB J 2004;18:1770–1772.
has patented numerous A2B antagonists, and one such antagonist is currently 23. Fredholm BB, Irenius E, Kull B, Schulte G. Comparison of the potency
being developed for the treatment of asthma.
of adenosine as an agonist at human adenosine receptors expressed
in Chinese hamster ovary cells. Biochem Pharmacol 2001;61:443–448.
References 24. Mentzer RM Jr, Rubio R, Berne RM. Release of adenosine by hypoxic
1. Takizawa H. Airway epithelial cells as regulators of airway inflammation canine lung tissue and its possible role in pulmonary circulation. Am
[review]. Int J Mol Med 1998;1:367–378. J Physiol 1975;229:1625–1631.
592 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 35 2006

25. Hagberg H, Andersson P, Lacarewicz J, Jacobson I, Butcher S, Sandberg odoulou S, Boucher W, Riccioni G, Conti P, Theoharides TC. Interleu-
M. Extracellular adenosine, inosine, hypoxanthine, and xanthine in kin-19 (IL-19) network revisited. Int J Immunopathol Pharmacol 2003;
relation to tissue nucleotides and purines in rat striatum during 16:95–97.
transient ischemia. J Neurochem 1987;49:227–231. 34. Liao SC, Cheng YC, Wang YC, Wang CW, Yang SM, Yu CK, Shieh CC,
26. Blackburn MR, Lee CG, Young HW, Zhu Z, Chunn JL, Kang MJ, Cheng KC, Lee MF, Chiang SR, et al. IL-19 induced Th2 cytokines
Banerjee SK, Elias JA. Adenosine mediates IL-13-induced inflamma- and was up-regulated in asthma patients. J Immunol 2004;173:6712–
tion and remodeling in the lung and interacts in an IL-13-adenosine 6718.
amplification pathway. J Clin Invest 2003;112:332–344. 35. Liao YC, Liang WG, Chen FW, Hsu JH, Yang JJ, Chang MS. IL-19
27. Fan M, Qin W, Mustafa SJ. Characterization of adenosine receptor(s) induces production of IL-6 and TNF-alpha and results in cell apoptosis
involved in adenosine-induced bronchoconstriction in an allergic through TNF-alpha. J Immunol 2002;169:4288–4297.
mouse model. Am J Physiol Lung Cell Mol Physiol 2003;284:L1012– 36. Caruso M, Holgate ST, Polosa R. Adenosine signalling in airways. Curr
L1019. Opin Pharmacol 2006;6:251–256.
28. Khoa ND, Montesinos MC, Reiss AB, Delano D, Awadallah N, Cronstein 37. Feoktistov I, Biaggioni I. Adenosine A2b receptors evoke interleukin-8
BN. Inflammatory cytokines regulate function and expression of aden- secretion in human mast cells: an enprofylline-sensitive mechanism
osine A(2A) receptors in human monocytic THP-1 cells. J Immunol with implications for asthma. J Clin Invest 1995;96:1979–1986.
2001;167:4026–4032. 38. Fredholm BB, Jzerman API, Jacobson KA, Klotz K-N, Linden J. Interna-
29. Varani K, Caramori G, Vincenzi F, Adcock I, Casolari P, Leung E, tional Union of Pharmacology. XXV. Nomenclature and classification
of adenosine receptors. Pharmacol Rev 2001;53:527–552.
MacLennan S, Gessi S, Morello S, Barnes PJ, et al. Alteration of
39. Kreckler LM, Wan TC, Ge Z-D, Auchampach JA. Adenosine inhibits
Adenosine Receptors in Patients with Chronic Obstructive Pulmonary
tumor necrosis factor-␣ release from mouse peritoneal macrophages
Disease. Am J Respir Crit Care Med 2006;173:398–406.
via A2A and A2B but not the A3 adenosine receptor. J Pharmacol
30. Geiser T, Dewald B, Ehrengruber M, Clark-Lewis I, Baggiolini M. The Exp Ther 2006;317:172–180.
interleukin-8-related chemotactic cytokines GRO alpha, GRO beta, 40. Pelletier S, Dube J, Villeneuve A, Gobeil F Jr, Bernier SG, Battistini B,
and GRO gamma activate human neutrophil and basophil leukocytes. Guillemette G, Sirois P. Adenosine induces cyclic-AMP formation
J Biol Chem 1993;268:15419–15424. and inhibits endothelin-1 production/secretion in guinea-pig tracheal
31. Romagnani P, Lasagni L, Annunziato F, Serio M, Romagnani S. CXC epithelial cells through A2B adenosine receptors. Br J Pharmacol 2000;
chemokines: the regulatory link between inflammation and angiogen- 129:243–250.
esis. Trends Immunol 2004;25:201–209. 41. Xaus J, Valledor AF, Cardo M, Marques L, Beleta J, Palacios JM,
32. Strieter RM, Belperio JA, Keane MP. CXC chemokines in angiogenesis Celada A. Adenosine inhibits macrophage colony-stimulating factor-
related to pulmonary fibrosis. Chest 2002;122:298S–301S. dependent proliferation of macrophages through the induction of
33. Kempuraj D, Frydas S, Kandere K, Madhappan B, Letourneau R, Christ- p27kip-1 expression. J Immunol 1999;163:4140–4149.

You might also like