10
CALL FOR PAPERS Biomarkers in Lung Diseaes: from Pathogenesis to Prediction to New Therapies A-kinase-anchoring proteins coordinate inflammatory responses to cigarette smoke in airway smooth muscle X Wilfred J. Poppinga, 1,2,3 Irene H. Heijink, 2,5 Laura J. Holtzer, 1,3 Philipp Skroblin, 3 Enno Klussmann, 3 Andrew J. Halayko, 4 Wim Timens, 2,5 Harm Maarsingh, 1,2,6 and Martina Schmidt 1,2 1 University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands; 2 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands; 3 Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany; 4 University of Manitoba, Departments of Physiology and Pathophysiology, and Internal Medicine, Winnipeg, Manitoba, Canada; 5 University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands; 6 Palm Beach Atlantic University, Lloyd L. Gregory School of Pharmacy, Department of Pharmaceutical Sciences, West Palm Beach, Florida Submitted 16 October 2014; accepted in final form 29 January 2015 Poppinga WJ, Heijink IH, Holtzer LJ, Skroblin P, Klussmann E, Halayko AJ, Timens W, Maarsingh H, Schmidt M. A-kinase- anchoring proteins coordinate inflammatory responses to cigarette smoke in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 308: L766 –L775, 2015. First published January 30, 2015; doi:10.1152/ajplung.00301.2014.—2 -Agonist inhibitors can relieve chronic obstructive pulmonary disease (COPD) symptoms by stimu- lating cyclic AMP (cAMP) signaling. A-kinase-anchoring proteins (AKAPs) compartmentalize cAMP signaling by establishing protein complexes. We previously reported that the 2 -agonist fenoterol, direct activation of protein kinase A (PKA), and exchange factor directly activated by cAMP decrease cigarette smoke extract (CSE)- induced release of neutrophil attractant interleukin-8 (IL-8) from human airway smooth muscle (ASM) cells. In the present study, we tested the role of AKAPs in CSE-induced IL-8 release from ASM cells and assessed the effect of CSE on the expression levels of different AKAPs. We also studied mRNA and protein expression of AKAPs in lung tissue from patients with COPD. Our data show that CSE exposure of ASM cells decreases AKAP5 and AKAP12, both capable of interacting with 2 -adrenoceptors. In lung tissue of patients with COPD, mRNA levels of AKAP5 and AKAP12 were decreased compared with lung tissue from controls. Using immunohistochem- istry, we detected less AKAP5 protein in ASM of patients with COPD Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage II compared with control subjects. St-Ht31, which disrupts AKAP- PKA interactions, augmented CSE-induced IL-8 release from ASM cells and diminished its suppression by fenoterol, an effect mediated by disturbed ERK signaling. The modulatory role of AKAP-PKA interactions in the anti-inflammatory effects of fenoterol in ASM cells and the decrease in expression of AKAP5 and AKAP12 in response to cigarette smoke and in lungs of patients with COPD suggest that cigarette smoke-induced changes in AKAP5 and AKAP12 in patients with COPD may affect efficacy of pharmacotherapy. compartmentalization; A-kinase-anchoring protein; inflammation; cy- clic AMP; chronic obstructive pulmonary disease CHRONIC OBSTRUCTIVE PULMONARY DISEASE (COPD) is character- ized by airway inflammation, neutrophils, mucus hypersecre- tion, airway remodeling, and parenchymal tissue destruction (emphysema), all contributing to irreversible airflow limitation and accelerated lung function decline (1). Cigarette smoking is the main risk factor for the development of COPD (7). Smok- ing cessation is the most effective strategy to slow down the accelerated decline in lung function observed in the disease (2, 23), but it does not reverse the structural changes in the lungs. Pharmacological COPD management is mainly focused on the treatment of symptoms, including airway obstruction and inflammation. In COPD, mainstays for eliciting bronchodila- tion include inhaled anticholinergics or 2 -agonists (23, 30). 2 -Agonists primarily act on airway smooth muscle (ASM) cells, increasing the intracellular production of the second messenger cyclic AMP (cAMP) to induce ASM relaxation and bronchodilation (19). In addition to their role in regulating airway diameter, ASM cells also possess secretory and proin- flammatory functions and through this contribute to local chronic airway inflammation in COPD (3, 6, 28, 29). Although anti-inflammatory effects of 2 -agonists in vitro are established (13, 20), the evidence for this in vivo is still under debate (34). cAMP induces its biological actions by activating various effectors, including protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac) (36, 40). CAMP and its effectors are under tight spatiotemporal control by a family of scaffolding proteins with over 50 members called A-kinase- anchoring proteins (AKAPs) (32, 37, 38). AKAPs regulate diverse cellular processes, including release of inflammatory cytokines from cardiomyocytes and alveolar macrophages (8, 21). For example, in alveolar macrophages, AKAP10 is re- quired for the potentiation of LPS-induced IL-6 and IL-10 production by the cAMP-elevating agonist prostaglandin E 2 (PGE 2 ) (21). Although less pronounced, AKAP11 also plays a modulatory role in the above-mentioned processes (21). Human ASM cells are known to express AKAP1-AKAP3, AKAP5, AKAP9-AKAP13, MAP2B, and Ezrin (15, 24). In- hibition of AKAP-PKA interactions using the PKA-anchoring Address for reprint requests and other correspondence: A. Deusinglaan, 1, XB10, 9713 AV Groningen, The Netherlands (e-mail: [email protected]). Am J Physiol Lung Cell Mol Physiol 308: L766 –L775, 2015. First published January 30, 2015; doi:10.1152/ajplung.00301.2014. 1040-0605/15 Copyright © 2015 the American Physiological Society http://www.ajplung.org L766 by 10.220.33.4 on June 15, 2017 http://ajplung.physiology.org/ Downloaded from

A-kinase-anchoring proteins coordinate inflammatory ... · A-kinase-anchoring proteins coordinate inflammatory ... stage II compared with control ... different healthy donors during

Embed Size (px)

Citation preview

CALL FOR PAPERS Biomarkers in Lung Diseaes: from Pathogenesis to Prediction

to New Therapies

A-kinase-anchoring proteins coordinate inflammatory responses to cigarettesmoke in airway smooth muscle

X Wilfred J. Poppinga,1,2,3 Irene H. Heijink,2,5 Laura J. Holtzer,1,3 Philipp Skroblin,3 Enno Klussmann,3

Andrew J. Halayko,4 Wim Timens,2,5 Harm Maarsingh,1,2,6 and Martina Schmidt1,2

1University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands; 2University of Groningen,University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, TheNetherlands; 3Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany; 4University of Manitoba, Departments ofPhysiology and Pathophysiology, and Internal Medicine, Winnipeg, Manitoba, Canada; 5University of Groningen, UniversityMedical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands; 6Palm BeachAtlantic University, Lloyd L. Gregory School of Pharmacy, Department of Pharmaceutical Sciences, West Palm Beach,Florida

Submitted 16 October 2014; accepted in final form 29 January 2015

Poppinga WJ, Heijink IH, Holtzer LJ, Skroblin P, KlussmannE, Halayko AJ, Timens W, Maarsingh H, Schmidt M. A-kinase-anchoring proteins coordinate inflammatory responses to cigarettesmoke in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol308: L766 –L775, 2015. First published January 30, 2015;doi:10.1152/ajplung.00301.2014.—�2-Agonist inhibitors can relievechronic obstructive pulmonary disease (COPD) symptoms by stimu-lating cyclic AMP (cAMP) signaling. A-kinase-anchoring proteins(AKAPs) compartmentalize cAMP signaling by establishing proteincomplexes. We previously reported that the �2-agonist fenoterol,direct activation of protein kinase A (PKA), and exchange factordirectly activated by cAMP decrease cigarette smoke extract (CSE)-induced release of neutrophil attractant interleukin-8 (IL-8) fromhuman airway smooth muscle (ASM) cells. In the present study, wetested the role of AKAPs in CSE-induced IL-8 release from ASMcells and assessed the effect of CSE on the expression levels ofdifferent AKAPs. We also studied mRNA and protein expression ofAKAPs in lung tissue from patients with COPD. Our data show thatCSE exposure of ASM cells decreases AKAP5 and AKAP12, bothcapable of interacting with �2-adrenoceptors. In lung tissue of patientswith COPD, mRNA levels of AKAP5 and AKAP12 were decreasedcompared with lung tissue from controls. Using immunohistochem-istry, we detected less AKAP5 protein in ASM of patients with COPDGlobal Initiative for Chronic Obstructive Lung Disease (GOLD) stageII compared with control subjects. St-Ht31, which disrupts AKAP-PKA interactions, augmented CSE-induced IL-8 release from ASMcells and diminished its suppression by fenoterol, an effect mediatedby disturbed ERK signaling. The modulatory role of AKAP-PKAinteractions in the anti-inflammatory effects of fenoterol in ASM cellsand the decrease in expression of AKAP5 and AKAP12 in response tocigarette smoke and in lungs of patients with COPD suggest thatcigarette smoke-induced changes in AKAP5 and AKAP12 in patientswith COPD may affect efficacy of pharmacotherapy.

compartmentalization; A-kinase-anchoring protein; inflammation; cy-clic AMP; chronic obstructive pulmonary disease

CHRONIC OBSTRUCTIVE PULMONARY DISEASE (COPD) is character-ized by airway inflammation, neutrophils, mucus hypersecre-tion, airway remodeling, and parenchymal tissue destruction(emphysema), all contributing to irreversible airflow limitationand accelerated lung function decline (1). Cigarette smoking isthe main risk factor for the development of COPD (7). Smok-ing cessation is the most effective strategy to slow down theaccelerated decline in lung function observed in the disease (2,23), but it does not reverse the structural changes in the lungs.

Pharmacological COPD management is mainly focused onthe treatment of symptoms, including airway obstruction andinflammation. In COPD, mainstays for eliciting bronchodila-tion include inhaled anticholinergics or �2-agonists (23, 30).�2-Agonists primarily act on airway smooth muscle (ASM)cells, increasing the intracellular production of the secondmessenger cyclic AMP (cAMP) to induce ASM relaxation andbronchodilation (19). In addition to their role in regulatingairway diameter, ASM cells also possess secretory and proin-flammatory functions and through this contribute to localchronic airway inflammation in COPD (3, 6, 28, 29). Althoughanti-inflammatory effects of �2-agonists in vitro are established(13, 20), the evidence for this in vivo is still under debate (34).

cAMP induces its biological actions by activating variouseffectors, including protein kinase A (PKA) and exchangeprotein directly activated by cAMP (Epac) (36, 40). CAMP andits effectors are under tight spatiotemporal control by a familyof scaffolding proteins with over 50 members called A-kinase-anchoring proteins (AKAPs) (32, 37, 38). AKAPs regulatediverse cellular processes, including release of inflammatorycytokines from cardiomyocytes and alveolar macrophages (8,21). For example, in alveolar macrophages, AKAP10 is re-quired for the potentiation of LPS-induced IL-6 and IL-10production by the cAMP-elevating agonist prostaglandin E2

(PGE2) (21). Although less pronounced, AKAP11 also plays amodulatory role in the above-mentioned processes (21).

Human ASM cells are known to express AKAP1-AKAP3,AKAP5, AKAP9-AKAP13, MAP2B, and Ezrin (15, 24). In-hibition of AKAP-PKA interactions using the PKA-anchoring

Address for reprint requests and other correspondence: A. Deusinglaan, 1,XB10, 9713 AV Groningen, The Netherlands (e-mail: [email protected]).

Am J Physiol Lung Cell Mol Physiol 308: L766–L775, 2015.First published January 30, 2015; doi:10.1152/ajplung.00301.2014.

1040-0605/15 Copyright © 2015 the American Physiological Society http://www.ajplung.orgL766

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

disruptor peptides, Ht31 and AKAP-IS, in human ASM cellsextended the duration of the �2-agonist-induced cAMP signalat the plasma membrane (15). The functional role of AKAP-mediated compartmentalization in the lung, the effects ofcigarette smoke exposure on AKAP expression, and the ex-pression profile of AKAPs in lung diseases such as COPD areunknown.

AKAP5 and AKAP12 directly interact with the �2-adreno-ceptor (5, 11, 16, 32, 39). Thus we hypothesized that altera-tions in AKAP5 and AKAP12 expression may affect theanti-inflammatory effects of �2-agonists on cigarette smoke-induced inflammatory responses. In the current study, weinvestigated AKAP expression in human ASM cells afterexposure to cigarette smoke extract (CSE) as well as in lungtissue from patients with COPD patients. We observed changesin AKAP5 and AKAP12 expression in COPD lungs as well asin cultured, CSE-exposed human ASM cells. Importantly,disruption of AKAP-PKA interactions prevented the anti-in-flammatory properties of the �2-agonist fenoterol via distur-bance of ERK phosphorylation.

MATERIALS AND METHODS

Cell culture. Human bronchial ASM cells obtained from threedifferent healthy donors during lung transplant procedures and im-mortalized by stable ectopic expression of human telomerase reversetranscriptase (hTERT) as described previously (12) were used atpassages 20–29. Primary human ASM cells obtained as describedpreviously (35) were used at passages 1–5. The cells were maintainedin high-glucose Dulbecco’s modified Eagle’s medium containing 10%heat-inactivated fetal calf serum supplemented with HEPES (25 mM),L-glutamine (2 mM), amphotericin B (1.5 �g/ml), and penicillin (100U/ml)/streptomycin (100 �g/ml) in a humidified atmosphere at 37°Cin air-CO2 (95:5% vol/vol). For primary ASM cells, the medium wassupplemented with sodium pyruvate (1 mM), Gibco MEM nonessen-tial amino acids, and gentamicin (45 �g/ml).

Human lung tissue. To study possible differences in AKAP expres-sion between patients with moderate and severe COPD, we used tissuefrom COPD Global Initiative for Chronic Obstructive Lung Disease(GOLD) stage II and stage IV, respectively. Lung tissue was collectedfrom current and ex-smoking patients, using asymptomatic currentand ex-smokers as a control group (for patient characteristics seeTables 1 and 2). Tissue from the control group and patients with

COPD GOLD stage II was derived from nondiseased lung tissue ofpatients undergoing resective surgery for pulmonary carcinoma, incontrols without airway obstruction or chronic airway symptoms, suchas cough and sputum production. Classification of COPD severity wasbased on the GOLD criteria (33). Tissue from patients with GOLD IVwas obtained from subjects undergoing surgery for lung transplanta-tion. All tissue was collected according to the Research Code of andapproved by the University Medical Center Groningen (https://www.umcg.nl/EN/Research/Researchers/General/ResearchCode/Pages/default.aspx), and it conformed to national ethical and professionalguidelines (“Code of conduct; Dutch federation of biomedical scientificsocieties”; http://www.federa.org).

CSE preparation. CSE was prepared using 3R4F research ciga-rettes (Reference Cigarette Program, University of Kentucky) (4) asdescribed previously (25, 26). In short, two cigarettes were combustedusing a peristaltic pump (Watson Marlow 323 E/D) in to 25 ml ofserum-free medium; this was designated 100% CSE and was dilutedto 15% CSE using serum-free medium.

Cell stimulation. hTERT ASM cells were grown to confluence, andgrowth was halted by exchange of the complete medium for serum-free medium for 24 h. Cells were pretreated with 50 �M st-Ht31(Promega) to disrupt AKAP-PKA interactions (4, 18), for 20 minbefore stimulation with 15% CSE in serum-free medium. Controlcultures were pretreated with vehicle alone. When used, fenoterol(0.001–0.1000 �M), the PKA activator 6-Bnz-cAMP (6-Bnz; 500�M), or the Epac activator 8-pCPT-2=-O-Me-cAMP (8-pCPT; 100�M) were added 10 min after the addition of st-Ht31. The action ofthese cAMP analogs has previously been characterized in this system(29). Cells were stimulated with 15% CSE for different time points:10 min for vasodilator-stimulated phosphoprotein (VASP) phosphor-ylation, 1 h for ERK phosphorylation, 2 h for NF-�B translocation,and 24 h for AKAP mRNA and protein expression and ELISAmeasurements of IL-8. Serum-free medium (vehicle) served as controlfor all experiments, as st-Ht31P, the commonly used control forst-Ht31, has been reported to be a direct inhibitor of PKA (24). Inaddition, st-Ht31P decreased cell viability in a dose-dependent man-ner (Fig. 6E), rendering st-Ht31P invalid as a control in this setup andin line with previous observations from our group (28).

RII overlay. RII overlay assay was performed as described previ-ously (14, 18). In brief, proteins were subjected to SDS-PAGE,transferred to PVDF membranes, and incubated in RII blocking buffer(5% milk powder, 0.1% BSA, 0.1% sodium azide in PBS) for 3 h atroom temperature. The membranes were incubated with [�32P]-la-beled PKA RII� subunits in fresh RII blocking buffer overnight atroom temperature. Membranes were washed twice with blockingbuffer and twice with PBS (10 min each). Binding of [�32P]-RIIsubunits was visualized by autoradiography using a STORM 830Scanner (Molecular Dynamics).

Table 1. Characteristics of the study objects used for thegene expression

Subject Groups

Control COPD Stage II COPD Stage IV

Number ofsubjects 5 5 5

Age, yr 57 (46–67) 71 (49–80) 58 (55–61)Male/female 1/4 5/0 0/5Ex-smoker/current

smoker 3/2 4/1 5/0Pack yr 32 (10–40)2� 23 (8.5–30)� 33 (30–54)FEV1% predicted 98.3 (86.1–102.6) 59.1† (54.6–61.1) 16.8† (14.0–23.6)FEV1/FVC 80.9 (70.6–85.3) 63.9* (45.7–71.1) 25.7† (19.2–41.8)

All values except number of subjects, sex, and smoking status are expressedas median values with minimum and maximum range in parentheses. COPD,chronic obstructive pulmonary disease; Ex-smoker, nonsmoker for at least 1yr; FEV1% predicted, forced expiratory volume in 1 s as percentage ofpredicted value; FVC: forced vital capacity. *P � 0.01, †P � 0.001 comparedwith control group. �1 data point unknown, 2�2 data points unknown.

Table 2. Characteristics of the control persons and patientswhose lung tissues were used for the immunohistochemistry

Subject Groups

Control COPD Stage II COPD Stage IV

Number of subjects 10 8 9Age, yr 67 (55–74) 71* (58–80) 58‡ (50–66)Male/female 4/6 6/2 3/6Pack yr 15 (1.5–65)2� 36.5 (10–44)� 26 (18–38)�

FEV1% predicted 97.1 (63.7–121)2� 65* (41.7–71)� 20.6†§ (13–36)FEV1/FVC 73.6 (64–86.2)� 63† (45.7–64.1)� 36.8†‡ (25–66)

All values except number of subjects and sex are expressed as median valueswith minimum and maximum range in parentheses. Only ex-smokers wereincluded. *P � 0.05, †P � 0.001 compared with control group, ‡P � 0.05, §P �0.001 compared with COPD stage II (Mann-Whitney U-test). �1 data pointunknown, 2�2 data points unknown.

L767ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

Western blot. Cells were lysed in RIPA lysis buffer [containing 0.5mM phenylmethylsulphonyl fluoride, 0.1 mM sodium orthovanadate,1 mM sodium fluoride, 3.5 �M �-glycerolphosphate, 1 mM EDTA,1% Triton X-100 (vol/vol), 0.1% sodium dodecyl sulphate (wt/vol),50 mM Tris·HCl (pH 7.4), 150 mM NaCl, 2 �g/�l soybean trypsininhibitor, 1.43 �g/�l aprotinin, and 0.8 mM benzamidin] followed bysonication. Samples for detection of AKAP9 (611518; BD Biosci-ences), AKAP12 (ab87067; Abcam), Ezrin (Ab4069; Abcam),AKAP5 (sc-6442; Santa Cruz Biotechnology), AKAP8 (sc-10766;Santa Cruz Biotechnology), (phospho)VASP (3112; Cell SignalingTechnology), or (phospho)ERK (9101 and 9102; Cell Signaling Tech-nology) expression were subjected to SDS-PAGE. Immunoblotting

with primary antibodies (dilution 1:500 for AKAPs antibodies,1:1,000 for the VASP antibody, 1:2,000 for ERK antibodies) wasdone overnight at 4°C. Blots were incubated with secondary antibody(1:2,000 A9044 or A0545; Sigma-Aldrich). Signals were detectedwith chemiluminescence reagents according to the manufacturer’sprotocol. GAPDH (1:2,000; sc-47724; Santa Cruz Biotechnology)served as a control for equal loading. Blots were quantified usingImageJ.

Real-time PCR. After 24 h, total RNA was isolated using theNucleoSpin RNA II isolation kit (Machery-Nagel), and cDNA wasobtained using the Reverse Transcription System (A3500, Promega)following the manufacturers’ instructions. This cDNA was used as a

Fig. 1. Cigarette smoke extract (CSE) changes theexpression of several A-kinase-anchoring proteins(AKAPs) in human airway smooth muscle (ASM). A:RII-overlay for the detection of AKAPs before and after24 h of CSE treatment (15%). Preincubation with pep-tide AKAP18-L314E (L314E), which binds to regula-tory RII subunits of PKA and thereby blocks the inter-action of the radioactive RII subunits with AKAPs onthe membrane, served as a control to confirm the spec-ificity of the signals. The peptide AKAP18-PP (PP),which does not bind RII subunits, does not interferewith the interaction of the labeled RII subunits andAKAPs on the membrane. B: expression of AKAP5(AKAP79), AKAP12 (AKAP250, Gravin), AKAP8(AKAP95), AKAP9 (AKAP450, AKAP350, Yotiao),and Ezrin (AKAP78) was detected by immunoblotting.C and D: AKAP expression was studied by immuno-blotting and immunofluorescence before and after treat-ment with 15% CSE for 24 h. Results are expressed asmeans SE, n � 3–6 independent experiments. *P �0.05, **P � 0.01 compared with unstimulated controls.Normal distribution was determined using the Shapiro-Wilk test, and, if normal distribution was determined,differences between 2 groups were compared by pairedsampled t-test.

L768 ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

template for quantitative real-time PCR for AKAP5 (forward: 5=-GACGCCCTACGTTGATCT-3=, reverse: 5=- GAAATGCCCAG-TTTCTCTATG-3=) and AKAP12 (forward: 5=-CAAGCACAGGAG-GAGTTACAG-3=; reverse: 5=- CTGGTCTTCCAAACAGACAATG-3=) using the PIXO Real-Time PCR (Helixis). Relative quantificationsof gene expression were normalized against 18S expression (forward:5=-CGCCGCTAGAGGTGAAATTC-3=; reverse: 5=- TTGGCAAAT-GCTTTCGCTC-3=).

Immunohistochemistry. Three-micron sections from paraffin-em-bedded lung tissue from patients with COPD and control subjects(Table 2) were deparaffinized, and antigen retrieval was performed ina Pascal pressure cooker (DakoCytomation) using preheated 0.1 mMTris·HCl buffer for 15 min at 125°C. Endogenous peroxidase wasblocked with 0.3% H2O2 in PBS for 30 min. Mouse monoclonalantibodies against AKAP5 (sc-17772, Santa Cruz Biotechnology;1:25), AKAP12 (ab87067, Abcam; 1:25), or Ezrin [3C12] (ab4069,Abcam; 1:25) in PBS containing 1% BSA were used as a firstantibody and incubated overnight at 4°C. Incubation time of thesecondary antibody (rabbit-�-mouse, 1:100) and third antibody (goat-�-rabbit, 1:100) in 1% human antibody serum in 1% BSA/PBS was 30min. Visualization was performed by 0.1% 3.3=-diaminobenzidinestaining for 10 min. Sections were counterstained with hematoxylin

for 3 min. Semiquantitative evaluation of the intensity of the stainingin ASM or total tissue was performed by two persons by independentidentification of the intensity of the staining in four classes: 0, no; 1,low; 2, medium; and 3, strong.

Measurements of IL-8 and cell viability. IL-8 was measured in cellsupernatants using an ELISA kit (PeliKine compact; Sanquin) accord-ing to the manufacturer’s instructions. After removal of the media, thecells were washed twice with PBS, and viability was determined usingthe AlamarBlue method (36, 44).

Immunofluorescence. Cells were seeded on coverslips, after 24 hput to serum-free conditions overnight, and stimulated for 2 h. Afterstimulation, cells were fixed in 3% paraformaldehyde (PFA) for 15min; subsequently 3% PFA and 0.3% Triton X-100 were added.Blocking was performed (1% BSA and 2% donkey serum inCytoTBS-T for 1 h) followed by incubation in a humidifying chamberwith the primary antibody against p65 (1:20 in blocking solution) at4°C. Nuclei were stained with a Hoechst staining (1:10,000). ProLongGold antifade reagent was added to mount the cells before cells werevisualized. Colocalization of p65 with nuclear staining was quantifiedusing Tissuefaxs (TissueGnostics).

Data processing and statistical analysis. Normal distribution wasdetermined using the Shapiro-Wilk test, and, if normal distribution

Fig. 2. Expression of AKAP5 and AKAP12 mRNA is altered in patients with chronic obstructive pulmonary disease (COPD) and by CSE in vitro. Expressionof AKAP5 (A) and AKAP12 (B) was measured in lung tissue of subjects without COPD, COPD Global Initiative for Chronic Obstructive Lung Disease (GOLD)stages II or IV (n � 5 each group). See Table 1 for characteristics of the subjects. C: gene expression of AKAP5 and AKAP12 was measured in primary ASMcells after exposure to 15% CSE for 24 h; each data point reflects a separate donor. Results are expressed as individual data points with the median of the separateexperiments. *P � 0.05, **P � 0.01 compared with unstimulated control subjects (A and B) or basal controls (C). Normal distribution was determined usingthe Shapiro-Wilk test, and, if normal distribution was determined, differences between 2 groups were compared by paired sampled t-test.

Fig. 3. Expression of AKAP5 in lung tissue frompatients with COPD and control subjects. AKAP5protein expression was analyzed by immunohisto-chemistry. A representative image from a controlpatient’s lung is shown (A). Bars indicate 100 �m.B: semiquantification of expression in the ASM ofCOPD vs. control patients. See Table 2 for details ofthe patients; all individuals were ex-smokers. Whitearrows indicate ASM. Results are expressed as in-dividual data points for each patient with the medianof the separate experiments, *P � 0.05, controlpatients vs. patients with COPD using a Kruskal-Wallis 1-way ANOVA followed by Mann-WhitneyU-test.

L769ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

was determined, differences between two groups were compared bypaired sampled t-test, when looking at cell culture experiments, unlessstated otherwise. A Kruskal-Wallis one-way ANOVA followed byMann-Whitney U-test was used when comparing patient material. Atwo-tailed P value �0.05 was considered statistically significant. Alltests were performed using SPSS 22.0 for Windows.

RESULTS

To investigate AKAP expression in hTERT ASM cells, weinitially carried out RII overlay assays. Protein samples from

the cells were separated by SDS-PAGE, blotted onto nitrocel-lulose, and overlaid with radioactively labeled regulatory RII�subunits of PKA that bind to AKAPs on the nitrocellulosemembranes (14, 17, 18) (Fig. 1A). Preincubation of RII�subunits with the peptide AKAP18-L314E (L314E), which,like the st-Ht31 peptide (17), binds with subnanomolar affinityto the AKAP-binding site of RII subunits to abolish AKAP-RIIinteractions, abrogated binding of RII� to AKAPs (Fig. 1A,right). This confirms that AKAP detection carried out in the

Fig. 4. Expression of AKAP12 in patients withCOPD and control subjects. AKAP12 protein ex-pression was analyzed by immunohistochemistry. Arepresentative picture from a control patient’s lungis shown (A). Bars indicate 100 �m. B: semiquan-tification of expression in the ASM of COPD vs.control patients. See Table 2 for details of thepatients; all individuals were ex-smokers. Whitearrows indicate ASM. Results are expressed as in-dividual data points for each patient with the medianof the separate experiments.

Fig. 5. Expression of Ezrin expression in lung tissueof patients with COPD and control subjects. Ezrinprotein expression was analyzed by immunohisto-chemistry in lung tissue from patients with COPDand control subjects. A: representative images areshown. Bars indicate 200 �m. B: semiquantificationof the signals for patients with COPD vs. controlsubjects was independently performed in duplicateby 2 persons. See Table 2 for details of the patients;all individuals were ex-smokers. Results are ex-pressed as individual data points for each patientwith the median of the separate experiments, *P �0.05, control patients vs. patients with COPD usinga Kruskal-Wallis 1-way ANOVA followed byMann-Whitney U-test.

L770 ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

presence of the inactive control peptide, AKAP18-PP, wasspecific (Fig. 1A, left). The molecular weights of the detectedproteins correspond to those of several known AKAPs, andsubsequent Western blotting identified AKAP5, AKAP8,AKAP9, AKAP12, and Ezrin in hTERT ASM cells (Fig. 1B).The RII overlay showed that exposure to CSE altered theAKAP protein pattern with most AKAP signals being reduced(Fig. 1A). Using immunoblotting, the effects of CSE exposureon the protein expression of the identified AKAPs were com-

pared (Fig. 1C). AKAP5, AKAP9, and AKAP12 were de-creased, whereas Ezrin expression was increased, and AKAP8was not affected. Immunofluorescence microscopic analysesrevealed similar effects of CSE on AKAP5, AKAP12, andEzrin levels (Fig. 1D).

To assess whether similar changes in AKAP expression arealso seen upon long-term cigarette smoke exposure in patientswith COPD, we studied mRNA expression of the differentAKAPs in lung tissue. We observed that the mRNA levels of

Fig. 6. AKAPs coordinate CSE-induced IL-8 release from human telomerase reverse transcriptase ASM. A and B: IL-8 release was measured using ELISA after24-h exposure to 15% CSE in the absence or presence of st-Ht31, fenoterol, 6-Bnz-cAMP, or 8-pCPT-2=-O-Me-cAMP as indicated. C and D: cell viability wastested after 24 h using AlamarBlue. E and F: cells were incubated with the peptide st-Ht31P in the indicated concentrations for 24 h (n � 3). The AKAP-PKAinteraction disruptor peptide st-Ht31 (50 �M) was added 20 min before the addition of fenoterol, 8-pCPT, or 6-Bnz; CSE was added 25 min after this (n � 4–15).*P � 0.05, **P � 0.01, ***P � 0.001 compared with unstimulated control or as indicated. Normal distribution was determined using the Shapiro-Wilk test,and, if normal distribution was determined, differences between 2 groups were compared by paired sampled t-test.

L771ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

AKAP5 (Fig. 2A) and AKAP12 (Fig. 2B) were lower inpatients with COPD with both GOLD stages II and IV com-pared with control subjects. No significant differences inAKAP5 or AKAP12 expression were seen between patientswith GOLD stage II and IV. To study whether the reducedmRNA levels of AKAP5 and AKAP12 in lung tissue ofpatients with COPD could be the result of cigarette smokeexposure, we studied the effect of CSE on AKAP5 andAKAP12 mRNA levels in primary ASM cells. Indeed, in allprimary ASM cell lines tested, AKAP5 and AKAP12 mRNAwas reduced upon CSE exposure (Fig. 2C).

We next assessed AKAP protein immunoreactivity in lungsections from patients with stage II and stage IV COPD andcontrol subjects, all of which are ex-smokers (Table 2). Similarto AKAP9 expression (25), AKAP5, AKAP12 (Figs. 3A and4A), and Ezrin (Fig. 5) exhibited prominent immunoreactivityin the epithelium. In the ASM layer, Ezrin was not detected(received a score of 0), but strong immunoreactivity forAKAP5 and AKAP12 was evident (Figs. 3A and 4A, whitearrows). AKAP5 in the ASM showed a decrease in the COPDstage II vs. control tissue, whereas no significant differencewas observed between control specimens and COPD stage IVtissue (P � 0.05 and P � 0.09 respectively, Fig. 4A). AKAP12was not significantly different in the COPD stages II and IV vs.controls (Fig. 4B).

We next examined functional implications for altered AKAPexpression in CSE-exposed ASM cells and ASM from lungs ofsubjects with COPD. In agreement with our previous findings(26), CSE induced IL-8 release from hTERT ASM cells, withthe levels in cell culture supernatant being increased by 77 21% compared with basal levels in untreated cultures (P �0.01, Fig. 6, A and B). Cell viability under these conditions wasunaffected (Fig. 6, C and D). We found that CSE-induced IL-8release was dose dependently attenuated by the �2-agonist,fenoterol, with a maximum reduction of 50% being reachedwith 0.1 �M fenoterol (P � 0.01; Fig. 6A). The PKA activator6-Bnz (500 �M) also reduced CSE-induced IL-8 release byabout 50% (Fig. 6B). Similarly, as we observed previously(26), selective pharmacological activation of Epac with8-pCPT (100 �M) decreased CSE-induced IL-8 release (P �0.07, Fig. 6B).

To determine the role of AKAP-PKA interactions in CSE-induced IL-8 release and in the inhibitory effects of the�2-agonist fenoterol, the PKA activator 6-Bnz, and the Epacactivator 8-pCPT, we next measured the impact of the cell-permeant PKA-anchoring disruptor peptide, st-Ht31. The con-trol peptide st-Ht31P could not be used as a control because itcaused death of the ASM cells (Fig. 6E), which is in line withobservations in human bronchial epithelial cells (25). Treat-ment with st-Ht31 (50 �M) did not affect viability of the

Fig. 7. Disruption of AKAP-PKA interactions does notaffect the NF-�B pathway. With the use of immunoflu-orescence, nuclear translocation of p65 was visualizedafter 2 h of exposure to 15% CSE in the absence orpresence of st-Ht31 using TNF-� as a positive control.Representative pictures of 3 independent experimentsare shown. Bars indicate 10 �m.

L772 ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

hTERT ASM under any condition (Fig. 6, C and D), whereasit caused a small but significant increase of IL-8 release underbasal conditions and markedly augmented IL-8 release fromCSE-exposed ASM cells (P � 0.05 both; Fig. 6, A and B). Inaddition, disrupting AKAP-PKA interactions with st-Ht31 sig-nificantly reversed the inhibitory effects of the �2-agonistfenoterol on CSE-induced IL-8 release by about 50% (Fig. 6A),and the presence of fenoterol could not prevent the augmenta-tion of IL-8 release that was induced by st-Ht31 in CSE-exposed cultures. We also studied the effect of the peptide inthe presence of the PKA activator 6-Bnz, which fully activatesPKA throughout the cell beyond any compartmental restric-tions. 6-Bnz significantly reduced IL-8 release, and disruptionof AKAP-PKA interactions with st-Ht31 did not reverse thesuppressive effects of 6-Bnz on CSE-induced IL-8 release (Fig.6B); this further supports the notion that AKAP-PKA interac-tions are required for the inhibitory effect of cAMP on IL-8. Incontrast, we observed that increased IL-8 release induced byst-Ht31 in CSE-exposed cells was refractory to treatment withthe Epac activator, 8-pCPT (Fig. 6B).

IL-8 release induced by CSE involves nuclear translocationof the NF-�B subunit, p65, which is regulated by Epac (26). Ina manner similar to TNF-� (used as positive control), CSEcaused nuclear translocation of p65 (Fig. 7). As seen for IL-8release, CSE-induced p65 nuclear translocation was unaffectedby the addition of st-Ht31 (Fig. 7, quantifications not shown).PKA activity, as measured by VASP phosphorylation (26), wasnot affected by st-Ht31, suggesting that disruption of AKAP-PKA complexes by st-Ht31 does not profoundly alter basal orCSE-induced total cellular PKA activity (Fig. 8A). Interest-ingly, CSE alone did induce a significant increase in VASPphosphorylation (Fig. 8A). There is evidence that direct acti-vation of PKA attenuated CSE-induced IL-8 release by inhib-iting ERK1/2 phosphorylation (26). In line with the effect ofst-Ht31 to increase basal IL-8 release, we also observed thatst-Ht31 increased basal phosphorylation of ERK1/2 (Fig. 8B).CSE-induced phosphorylation of ERK1/2 was not statisticallysignificantly affected by st-Ht31. However, the peptide didgive a trend toward preventing the inhibitory effect of fenoterolon CSE-induced ERK1/2 phosphorylation (Fig. 8B, P �0.078). This was similar to the effect of st-Ht31 without CSEstimulation (Fig. 8B). Collectively, these data suggest an im-portant role for AKAP-PKA interactions in the regulation ofASM-mediated inflammatory responses.

DISCUSSION

In the present study, we describe the expression of a subsetof AKAPs that are differently affected by CSE exposure inASM cells. In particular, the expression level of AKAP5,which is an important regulator of �2-AR sensitivity (5, 11, 16,32), was significantly reduced, suggesting a mechanism thatcould link cigarette smoking and COPD pathogenesis (39). Weshow that mRNA for AKAP5 and AKAP12 is reduced in lungtissue obtained from patients with COPD, and the use ofimmunohistochemistry allowed us to observe a significantdecrease of AKAP5 protein in COPD stage II lung specimens.Disruption of AKAP-PKA interactions with st-Ht31 increasedbasal and CSE-induced IL-8 release from ASM cells andprevented the inhibitory effect of the �2-agonist, fenoterol, onIL-8 secretion, i.e., prevented the anti-inflammatory effect of

fenoterol. This indicates that the inhibitory effect of fenoterolat least in part depends on AKAP-PKA interactions. Inhibitionof AKAP-PKA interactions is associated with a disruption ofthe inhibitory action of fenoterol on ERK phosphorylation. Thedata indicate that CSE-induced downregulation of AKAPs maypromote airway inflammation and may reduce the regulatoryeffect of �2-agonists on CSE-induced airway inflammation.Our study is the first to link AKAPs to cigarette smoke-evokedinflammatory responses in ASM cells.

Previously, we had demonstrated that CSE-induced IL-8release from human ASM cells is prevented by treatment withfenoterol as well as by selective activation of the cAMPeffectors PKA and Epac (26). Epac activation had less pro-nounced effects on CSE-induced IL-8 release compared withPKA activation. The more limited effectiveness of Epac acti-vation with 8-pCPT is most likely attributable to a reduction of

Fig. 8. Disruption of AKAP-PKA interactions inhibits fenoterol-induced inhi-bition of ERK phosphorylation without affecting PKA activation. A: PKAactivity was determined using the phosphorylation of vasodilator-stimulatedphosphoprotein (VASP) at Ser157 after 10 min of exposure to 15% CSE in theabsence or presence of st-Ht31. B: phosphorylation of ERK1/2 was visualizedafter 1 h of 15% CSE exposure in the absence or presence of st-Ht31. Proteinphosphorylation was corrected for total protein loaded in each lane either bytaking into account the ratio of the density of the phosphorylated-proteinagainst the sum of the phosphorylated and nonphosphorylated protein (A) orthe ratio of the phosphorylated protein against total protein (B). *P � 0.05,**P � 0.01, ***P � 0.001 statistically significant differences, compared withunstimulated control. Normal distribution was determined using the Shapiro-Wilk test, and, if normal distribution was determined, differences between 2groups were compared by paired sampled t-test.

L773ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

Epac1 expression that can be induced by CSE (26, 27). Wereported earlier that activation of PKA inhibits CSE-inducedIL-8 release by suppressing ERK1/2 phosphorylation (26). Toanalyze the potential involvement of AKAPs in the reductionof CSE-induced IL-8 release by fenoterol/PKA, we used theAKAP-PKA interaction inhibitor st-Ht31, demonstrating thatAKAPs are required for spatial coordination of PKA activitythat suppresses ERK1/2 via �2-adrenoceptors. CSE-inducedphosphorylation of ERK1/2 was not affected by st-Ht31, pos-sibly because the phospho-ERK1/2 level was already at itsmaximum. With the observation that, in contrast to ERK1/2phosphorylation, CSE-induced IL-8 release is further enhancedwith the addition of st-Ht31, it is possible that another AKAP-PKA-sensitive pathway is involved in IL-8 release. For exam-ple, other studies have identified that p38- and JNK-inducedsignaling pathways can regulate the release of IL-8 (42) andthat these kinases could also be affected by PKA.

A role for AKAPs in coordinating the duration of cAMPproduction after stimulation of the �2-adrenoceptor has beenshown in several cell types, including ASM cells transfectedwith a cyclic nucleotide gated ion channel-based reporter (15).In that study, disruptors of AKAP-PKA interactions, Ht31 andAKAP-IS, did not cause a change in whole cell cAMP accu-mulation after stimulation with the �2-agonist isoprenaline orforskolin, a direct activator of cAMP-producing adenylyl cy-clases (31). However, the duration of the transient local cAMPsignal measured at the plasma membrane was significantlysustained by AKAP-PKA interaction disruptors (15). It wasshown that AKAP12 is responsible for resensitization andrecycling of the �2-adrenoceptor after treatment with isopro-terenol, whereas AKAP5 is responsible for PKA-mediated�2-adrenoceptor phosphorylation and subsequent switchingfrom the cAMP pathway to �-arrestin-ERK1/2 signaling (5,11, 22, 32, 39). Therefore, the altered expression of AKAP5after exposure to CSE and in COPD might have importantconsequences for ERK1/2 in IL-8 signaling.

We observed that CSE causes a downregulation of AKAP5and AKAP12 mRNA and protein in both immortalized andprimary ASM cells, suggesting a novel mechanism for CSEinfluences on �2-adrenoceptor-directed cell functions. Becausest-Ht31 reduces the anti-inflammatory effects of fenoterol,CSE-induced alterations in AKAP expression may contributeto increased IL-8 release from ASM. We also observed thedownregulation of AKAP5 and AKAP12 mRNA in lung tissuefrom patients with COPD, suggesting roles of these AKAPs in�2-adrenoceptor responses. Previously, it was observed inBeWo trophoblast cells that a common pathway may regulateexpression of AKAP5 and AKAP12 protein and mRNA, whichis different from that of AKAP8 mRNA regulation (9). Alongthese lines, in the present study, we observed that CSE reducedAKAP5 and AKAP12 mRNA expression, whereas AKAP8was not affected.

In addition to ASM, we observed high expression of AKAP5and AKAP12 in airway epithelium. Recently, we showed that(25), in contrast to our present findings in ASM cells, expres-sion of AKAP5 and AKAP12 is not significantly altered byCSE in bronchial epithelial cells in vitro. In bronchial epithelialcells, AKAPs (presumably AKAP9) are involved in maintain-ing cell-cell contacts and the epithelial barrier function byinteraction with the adhesion molecule E-cadherin (25). Theobserved dysregulation of AKAP expression after CSE expo-

sure could contribute to an increased inflammatory response, asseen in COPD (1). In line with this, we report here thatdisturbing AKAP functioning using st-Ht31 increased the IL-8release.

Dysfunction of cAMP compartmentalization and local PKAsignaling occurs in various diseases including cardiac andneurological diseases (10, 32, 41), and here we show that thisalso may play a role in COPD. The observed dysregulation ofthe expression of AKAPs that are important in �2-adrenoceptorregulation may pave the way to novel pharmacological ap-proaches for the treatment of COPD. In summary, the presentstudy demonstrates that AKAPs, in particular their interactionswith PKA, are involved in the regulation of proinflammatoryresponses, specifically IL-8 release by ASM. In addition, weshow that AKAP5 and AKAP12, which regulate �2-adreno-ceptor sensitivity, are dysregulated upon CSE exposure and inpatients with COPD. Therefore, there is a potential for regu-lating inflammatory responses and possibly �2-adrenoceptorfunctioning by pharmacological targeting of AKAPs.

GRANTS

W. J. Poppinga was supported by the Dutch Lung Foundation (Grants3.2.09.034 and 3.2.11.015) and Stichting Astma Bestrijding (Grant 2010/019).E. Klussmann was supported by the Deutsche Forschungsgemeinschaft(KL1415/4-2), the Else Kröner-Fresenius-Stiftung (2013_A145), and the Ger-man-Israeli Foundation (I-1210-286.13/2012). A. J. Halayko is supported bythe Canada Research Chairs Program. M. Schmidt was supported by aRosalind Franklin Fellowship. Part of the work has been performed at theUMCG Imaging and Microscopy Center (UMIC), which is sponsored by NWO-grants 40-00506-98-9021 (TissueFaxs) and 175-010-2009-023 (Zeiss 2p).

DISCLOSURES

No conflicts of interest, financial or otherwise are declared by the author(s).

AUTHOR CONTRIBUTIONS

Author contributions: W.J.P., I.H.H., P.S., E.K., H.M., and M.S. conceptionand design of research; W.J.P. and L.J.H. performed experiments; W.J.P.analyzed data; W.J.P., I.H.H., W.T., H.M., and M.S. interpreted results ofexperiments; W.J.P. prepared figures; W.J.P. and M.S. drafted manuscript;W.J.P., I.H.H., L.J.H., P.S., E.K., A.J.H., W.T., H.M., and M.S. edited andrevised manuscript; W.J.P., I.H.H., L.J.H., P.S., E.K., A.J.H., W.T., H.M., andM.S. approved final version of manuscript.

REFERENCES

1. Barnes PJ. Cellular and molecular mechanisms of chronic obstructivepulmonary disease. Clin Chest Med 35: 71–86, 2014.

2. Barnes PJ. Emerging pharmacotherapies for COPD. Chest 134: 1278–1286, 2008.

3. Beeh KM, Kornmann O, Buhl R, Culpitt SV, Giembycz MA, BarnesPJ. Neutrophil chemotactic activity of sputum from patients with COPD:role of interleukin 8 and leukotriene B4. Chest 123: 1240–1247, 2003.

4. Chen PX, Moldoveanu SC. Mainstream smoke chemical analyses for2R4F Kentucky reference cigarette. Contrib Tobacc Res 20: 448, 2003.

5. Cong M, Perry SJ, Lin FT, Fraser ID, Hu LA, Chen W, Pitcher JA,Scott JD, Lefkowitz RJ. Regulation of membrane targeting of the Gprotein-coupled receptor kinase 2 by protein kinase A and its anchoringprotein AKAP79. J Biol Chem 276: 15192–15199, 2001.

6. Damera G, Tliba O, Panettieri RA Jr. Airway smooth muscle as animmunomodulatory cell. Pulm Pharmacol Ther 22: 353–359, 2009.

7. Decramer M, Janssens W, Miravitlles M. Chronic obstructive pulmo-nary disease. Lancet 379: 1341–1351, 2012.

8. del Vescovo CD, Cotecchia S, Diviani D. A-kinase-anchoring protein-Lbc anchors IkappaB kinase beta to support interleukin-6-mediated car-diomyocyte hypertrophy. Mol Cell Biol 33: 14–27, 2013.

9. Delidaki M, Gu M, Hein A, Vatish M, Grammatopoulos DK. Interplayof cAMP and MAPK pathways in hCG secretion and fusogenic geneexpression in a trophoblast cell line. Mol Cell Endocrinol 332: 213–220,2011.

L774 ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from

10. Esseltine JL, Scott JD. AKAP signaling complexes: pointing towards thenext generation of therapeutic targets? Trends Pharmacol Sci 34: 648–655, 2013.

11. Fraser ID, Cong M, Kim J, Rollins EN, Daaka Y, Lefkowitz RJ, ScottJD. Assembly of an A kinase-anchoring protein-beta(2)-adrenergic recep-tor complex facilitates receptor phosphorylation and signaling. Curr Biol10: 409–412, 2000.

12. Gosens R, Stelmack GL, Dueck G, McNeill KD, Yamasaki A, Gerthof-fer WT, Unruh H, Gounni AS, Zaagsma J, Halayko AJ. Role ofcaveolin-1 in p42/p44 MAP kinase activation and proliferation of humanairway smooth muscle. Am J Physiol Lung Cell Mol Physiol 291: L523–L534, 2006.

13. Hallsworth MP, Twort CH, Lee TH, Hirst SJ. Beta(2)-adrenoceptoragonists inhibit release of eosinophil-activating cytokines from humanairway smooth muscle cells. Br J Pharmacol 132: 729–741, 2001.

14. Henn V, Edemir B, Stefan E, Wiesner B, Lorenz D, Theilig F, SchmittR, Vossebein L, Tamma G, Beyermann M, Krause E, Herberg FW,Valenti G, Bachmann S, Rosenthal W, Klussmann E. Identification ofa novel A-kinase anchoring protein 18 isoform and evidence for its role inthe vasopressin-induced aquaporin-2 shuttle in renal principal cells. J BiolChem 279: 26654–26665, 2004.

15. Horvat SJ, Deshpande DA, Yan H, Panettieri RA, Codina J, DuBoseTD, Jr Xin W, Rich TC, Penn RB. A-kinase anchoring proteins regulatecompartmentalized cAMP signaling in airway smooth muscle. FASEB J26: 3670–3679, 2012.

16. Houslay MD, Baillie GS. beta-Arrestin-recruited phosphodiesterase-4desensitizes the AKAP79/PKA-mediated switching of beta(2)-adrenocep-tor signalling to activation of ERK. Biochem Soc Trans 33: 1333–1336,2005.

17. Hundsrucker C, Krause G, Beyermann M, Prinz A, Zimmermann B,Diekmann O, Lorenz D, Stefan E, Nedvetsky P, Dathe M, Christian F,McSorley T, Krause E, McConnachie G, Herberg FW, Scott JD,Rosenthal W, Klussmann E. High-affinity AKAP7 delta-protein kinaseA interaction yields novel protein kinase A-anchoring disruptor peptides.Biochem J 396: 297–306, 2006.

18. Hundsrucker C, Skroblin P, Christian F, Zenn HM, Popara V, JoshiM, Eichhorst J, Wiesner B, Herberg FW, Reif B, Rosenthal W,Klussmann E. Glycogen synthase kinase 3beta interaction protein func-tions as an A-kinase anchoring protein. J Biol Chem 285: 5507–5521,2010.

19. Johnson M. Beta2-adrenoceptors: mechanisms of action of beta2-ago-nists. Paediatr Respir Rev 2: 57–62, 2001.

20. Kaur M, Holden NS, Wilson SM, Sukkar MB, Chung KF, Barnes PJ,Newton R, Giembycz MA. Effect of �2-adrenoceptor agonists and othercAMP-elevating agents on inflammatory gene expression in human ASMcells: a role for protein kinase A. Am J Physiol Lung Cell Mol Physiol 295:L505–L514, 2008.

21. Kim SH, Serezani CH, Okunishi K, Zaslona Z, Aronoff DM, Peters-Golden M. Distinct protein kinase A anchoring proteins direct prostaglan-din E2 modulation of Toll-like receptor signaling in alveolar macrophages.J Biol Chem 286: 8875–8883, 2011.

22. Lynch MJ, Baillie GS, Mohamed A, Li X, Maisonneuve C, KlussmannE, van Heeke G, Houslay MD. RNA silencing identifies PDE4D5 as thefunctionally relevant cAMP phosphodiesterase interacting with beta arres-tin to control the protein kinase A/AKAP79-mediated switching of thebeta(2)-adrenergic receptor to activation of ERK in HEK293B2 cells. JBiol Chem 280: 33178–33189, 2005.

23. Meurs H, Oenema TA, Kistemaker LE, Gosens R. A new perspectiveon muscarinic receptor antagonism in obstructive airways diseases. CurrOpin Pharmacol 13: 316–323, 2013.

24. Misior AM, Deshpande DA, Loza MJ, Pascual RM, Hipp JD, PennRB. Glucocorticoid- and protein kinase A-dependent transcriptome regu-lation in airway smooth muscle. Am J Respir Cell Mol Biol 41: 24–39,2009.

25. Oldenburger A, Poppinga WJ, Kos F, de Bruin HG, Rijks W, HeijinkI, Timens W, Meurs H, Maarsingh H, Schmidt M. A-kinase anchoring

proteins contribute to loss of E-cadherin and bronchial epithelial barrier bycigarette smoke. Am J Physiol Cell Physiol 306: C585–C597, 2014.

26. Oldenburger A, Roscioni SS, Jansen E, Menzen MH, Halayko AJ,Timens W, Meurs H, Maarsingh H, Schmidt M. Anti-inflammatory roleof the cAMP effectors Epac and PKA: implications in chronic obstructivepulmonary disease. PLoS One 7: e31574, 2012.

27. Oldenburger A, van Basten B, Kooistra W, Meurs H, Maarsingh H,Krenning G, Timens W, Schmidt M. Interaction between Epac1 andmiRNA-7 in airway smooth muscle cells. Naunyn Schmiedebergs ArchPharmacol 387: 795–797, 2014.

28. Panettieri RA, Jr. Airway smooth muscle: immunomodulatory cells thatmodulate airway remodeling? Respir Physiol Neurobiol 137: 277–293,2003.

29. Panettieri RA, Jr. Airway smooth muscle: an immunomodulatory cell. JAllergy Clin Immunol 110, Suppl 6: S269–S274, 2002.

30. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS, ScientificCommittee GOLD. Global strategy for the diagnosis, management, andprevention of chronic obstructive pulmonary disease. NHLBI/WHOGlobal Initiative for Chronic Obstructive Lung Disease (GOLD) Work-shop summary. Am J Respir Crit Care Med 163: 1256–1276, 2001.

31. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP,Buneman OP, Davenport AP, McGrath JC, Peters JA, Southan C,Spedding M, Yu W, Harmar AJ, NC-IUPHAR. The IUPHAR/BPSGuide to PHARMACOLOGY: an expert-driven knowledgebase of drugtargets and their ligands. Nucleic Acids Res 42: D1098–D1106, 2014.

32. Poppinga WJ, Munoz-Llancao P, Gonzalez-Billault C, Schmidt M.A-kinase anchoring proteins: Cyclic AMP compartmentalization in neu-rodegenerative and obstructive pulmonary diseases. Br J Pharmacol 171:5603–5623, 2014.

33. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P,Fukuchi Y, Jenkins C, Rodriguez-Roisin R, van Weel C, Zielinski J,Global Initiative for Chronic Obstructive Lung Disease. Global strat-egy for the diagnosis, management, and prevention of chronic obstructivepulmonary disease: GOLD executive summary. Am J Respir Crit CareMed 176: 532–555, 2007.

34. Remington TL, Digiovine B. Long-acting beta-agonists: anti-inflamma-tory properties and synergy with corticosteroids in asthma. Curr OpinPulm Med 11: 74–78, 2005.

35. Roscioni SS, Prins AG, Elzinga CR, Menzen MH, Dekkers BG,Halayko AJ, Meurs H, Maarsingh H, Schmidt M. Protein kinase A andthe exchange protein directly activated by cAMP (Epac) modulate phe-notype plasticity in human airway smooth muscle. Br J Pharmacol 164:958–969, 2011.

36. Schmidt M, Dekker FJ, Maarsingh H. Exchange protein directly acti-vated by cAMP (Epac): a multidomain cAMP mediator in the regulationof diverse biological functions. Pharmacol Rev 65: 670–709, 2013.

37. Scott JD, Dessauer CW, Tasken K. Creating order from chaos: cellularregulation by kinase anchoring. Annu Rev Pharmacol Toxicol 53: 187–210, 2013.

38. Skroblin P, Grossmann S, Schafer G, Rosenthal W, Klussmann E.Mechanisms of protein kinase a anchoring. Int Rev Cell Mol Biol 283:235–330, 2010.

39. Tao J, Malbon CC. G-protein-coupled receptor-associated A-kinase an-choring proteins AKAP5 and AKAP12: differential signaling to MAPKand GPCR recycling. J Mol Signal 3: 19, 2008.

40. Taylor SS, Zhang P, Steichen JM, Keshwani MM, Kornev AP. PKA:lessons learned after twenty years. Biochim Biophys Acta 1834: 1271–1278, 2013.

41. Troger J, Moutty MC, Skroblin P, Klussmann E. A-kinase anchoringproteins as potential drug targets. Br J Pharmacol 166: 420–433, 2012.

42. Wuyts WA, Vanaudenaerde BM, Dupont LJ, Van Raemdonck DE,Demedts MG, Verleden GM. Interleukin-17-induced interleukin-8 re-lease in human airway smooth muscle cells: role for mitogen-activatedkinases and nuclear factor-kappaB. J Heart Lung Transplant 24: 875–881,2005.

L775ANTI-INFLAMMATORY ROLE OF AKAPs IN AIRWAY SMOOTH MUSCLE

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00301.2014 • www.ajplung.org

by 10.220.33.4 on June 15, 2017http://ajplung.physiology.org/

Dow

nloaded from