Next Article in Journal
Integration of Ligand-Based Drug Screening with Structure-Based Drug Screening by Combining Maximum Volume Overlapping Score with Ligand Docking
Previous Article in Journal
Synthesis and COX-2 Inhibitory Activity of 4-[(E)-2-(4-Oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethenyl]benzene-1-sulfonamide and Its Analogs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multiple Facets of cAMP Signalling and Physiological Impact: cAMP Compartmentalization in the Lung

1
Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands
2
Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2012, 5(12), 1291-1331; https://doi.org/10.3390/ph5121291
Submission received: 18 September 2012 / Revised: 15 November 2012 / Accepted: 20 November 2012 / Published: 30 November 2012

Abstract

:
Therapies involving elevation of the endogenous suppressor cyclic AMP (cAMP) are currently used in the treatment of several chronic inflammatory disorders, including chronic obstructive pulmonary disease (COPD). Characteristics of COPD are airway obstruction, airway inflammation and airway remodelling, processes encompassed by increased airway smooth muscle mass, epithelial changes, goblet cell and submucosal gland hyperplasia. In addition to inflammatory cells, airway smooth muscle cells and (myo)fibroblasts, epithelial cells underpin a variety of key responses in the airways such as inflammatory cytokine release, airway remodelling, mucus hypersecretion and airway barrier function. Cigarette smoke, being next to environmental pollution the main cause of COPD, is believed to cause epithelial hyperpermeability by disrupting the barrier function. Here we will focus on the most recent progress on compartmentalized signalling by cAMP. In addition to G protein-coupled receptors, adenylyl cyclases, cAMP-specific phospho-diesterases (PDEs) maintain compartmentalized cAMP signalling. Intriguingly, spatially discrete cAMP-sensing signalling complexes seem also to involve distinct members of the A-kinase anchoring (AKAP) superfamily and IQ motif containing GTPase activating protein (IQGAPs). In this review, we will highlight the interaction between cAMP and the epithelial barrier to retain proper lung function and to alleviate COPD symptoms and focus on the possible molecular mechanisms involved in this process. Future studies should include the development of cAMP-sensing multiprotein complex specific disruptors and/or stabilizers to orchestrate cellular functions. Compartmentalized cAMP signalling regulates important cellular processes in the lung and may serve as a therapeutic target.

1. Introduction

Cyclic adenosine monophosphate (cAMP), the most common and universal secondary messenger, regulates physiological processes as diverse as calcium handling, secretion, ion channel conductance, learning and memory, metabolic events, cardiac and smooth muscle contraction, cell growth and differentiation, apoptosis, inflammation, and barrier functioning [1,2]. The impact and complexity of research into the molecular architecture of cAMP signalling is not only reflected by five Nobel awards since the discovery of cAMP in 1957 by Sutherland and colleagues [1], but also by a unique interplay of signalling components that tightly control the cellular content of cAMP. Next to G protein-coupled receptors, adenylyl cyclases (ACs) and cAMP-specific phosphodiesterases (PDEs) maintain the spatio-temporal nature of cAMP signalling by shaping a cAMP gradient throughout the cell [3,4,5]. Subcellular membrane clustering of receptors, ACs and PDEs to lipid rafts and caveolae [6,7,8], and cell compartment-specific (co)localization to distinct cAMP effectors [9,10,11,12,13] further support the maintenance of spatio-temporal compartmentalized cAMP signalling. Moreover, A-kinase anchoring proteins (AKAPs) facilitate subcellular cAMP spatio-temporal compartmentalization by generating spatially discrete signalling complexes that create local gradients of cAMP, and thereby permit and control specific cellular responses (Figure 1) [14,15,16,17]. Dysfunctions of cAMP-sensing AKAP complexes seem to contribute to the progression of a wide variety of diseases, including chronic heart failure, cardiac arrhythmia, Alzheimer’s dementia, HIV infection, diabetes mellitus and cancer [17,18,19,20,21], hence, current research intends to target the spatio-temporal cAMP-responsive complexes to provide novel therapeutical interventions [5,11,12,17,22].
In this review we will discuss the recent progress on spatio-temporal compartmentalized cAMP signalling from the receptors coupled to the cAMP pathway up to the subtle interplay between the distinct cAMP-sensitive effectors that maintain cAMP-sensing multiprotein complexes. In particular, we will focus on the impact of perturbation of cAMP-sensing signalling complexes in the development and progression of chronic obstructive pulmonary disease (COPD), a chronic inflammatory lung disease characterized by airway obstruction, emphysema and airway remodelling. Remodelling processes encompass increased airway smooth muscle mass, epithelial changes, goblet cell and submucosal gland hyperplasia—leading to mucus hypersecretion [23,24,25,26,27,28,29,30]. In addition to inflammatory cells, airway smooth muscle cells and (myo)fibroblasts, epithelial cells underpin a variety of key responses in the airways such as inflammatory cytokine release, airway remodelling, mucus hypersecretion and the barrier function [23,24,25,26,27,28,29,30]. Cigarette smoke—together with environmental pollution—is the main risk factor for COPD and induces inflammatory processes, alveolar destruction (emphysema), fibrosis and epithelial hyperpermeability by disrupting the barrier function, releasing proteases and inducing multiple inflammatory genes [27,28,29,31]. Disruption of the epithelial barrier is associated with epithelial remodelling that also accounts for goblet cell metaplasia and mucus gland hypertrophy in COPD [32,33]. Mucus hypersecretion contributes to the morbidity and mortality of COPD, particularly in those patients with more severe disease [27,28,34]. In the treatment of obstructive lung diseases, including COPD, cAMP elevating drugs are widely used. Already in the early eighties it has been reported that cAMP elevating agents, such as β2-agonists, prostanoids and the direct AC activator forskolin (Figure 1), temper oedema in whole animal, isolated lung, and clinical studies of lung injury, phenomena which could be linked to an increase in barrier function in pulmonary endothelial cells [2].
Figure 1. Overview of compartmentalization of cAMP signalling. Gs-protein coupled receptors are stimulated by their appropriate ligands such as β2-agonists and prostanoids. Subsequently, activation of adenylyl cyclase (AC) will lead to the production of the second messenger cyclic AMP (cAMP), whereas cAMP-specific phosphodiesterases (PDEs) will shape the cAMP gradient throughout the cell. Alternatively, AC can be directly activated by the cell membrane-permeable diterpene forskolin from the Indian plant Coleus forskolhlii. Elevation of cellular cAMP will simultaneously induce the activation of protein kinase A (PKA) and of the exchange protein directly activated by cAMP (Epac). Members of the A-kinase anchoring protein (AKAP) family will support the maintenance of cAMP compartmentalization upon binding to the cAMP-producing receptors, the cAMP effectors PKA and/or Epac as well as PDEs. The generation of cAMP-sensing multiprotein complexes by AKAPs is of tremendous importance to maintain spatio-temporal cAMP signalling at specific and discrete locations within the cell to regulate specific cellular responses upon signalling to several distinct effector proteins including vasodilator-stimulated phosphoprotein (VASP), a subset of small GTPases, and phospholipase C-ε (PLC-ε). Shown are tools being used to study the functioning of the cAMP-sensing multiprotein complexes: st-Ht31, the PKA binding blocking peptide known to act as a generic AKAP inhibitor [14,15,16]; 8-pCPT-2'-O-Me-cAMP and/or Sp-8-pCPT-2'-O-Me-cAMP, activator of Epac; 6-Bnz-cAMP, activator of PKA; Rp-8-CPT-cAMP, Rp-cAMPs, Rp-8-Bromo-cAMPs inhibitors of PKA.
Figure 1. Overview of compartmentalization of cAMP signalling. Gs-protein coupled receptors are stimulated by their appropriate ligands such as β2-agonists and prostanoids. Subsequently, activation of adenylyl cyclase (AC) will lead to the production of the second messenger cyclic AMP (cAMP), whereas cAMP-specific phosphodiesterases (PDEs) will shape the cAMP gradient throughout the cell. Alternatively, AC can be directly activated by the cell membrane-permeable diterpene forskolin from the Indian plant Coleus forskolhlii. Elevation of cellular cAMP will simultaneously induce the activation of protein kinase A (PKA) and of the exchange protein directly activated by cAMP (Epac). Members of the A-kinase anchoring protein (AKAP) family will support the maintenance of cAMP compartmentalization upon binding to the cAMP-producing receptors, the cAMP effectors PKA and/or Epac as well as PDEs. The generation of cAMP-sensing multiprotein complexes by AKAPs is of tremendous importance to maintain spatio-temporal cAMP signalling at specific and discrete locations within the cell to regulate specific cellular responses upon signalling to several distinct effector proteins including vasodilator-stimulated phosphoprotein (VASP), a subset of small GTPases, and phospholipase C-ε (PLC-ε). Shown are tools being used to study the functioning of the cAMP-sensing multiprotein complexes: st-Ht31, the PKA binding blocking peptide known to act as a generic AKAP inhibitor [14,15,16]; 8-pCPT-2'-O-Me-cAMP and/or Sp-8-pCPT-2'-O-Me-cAMP, activator of Epac; 6-Bnz-cAMP, activator of PKA; Rp-8-CPT-cAMP, Rp-cAMPs, Rp-8-Bromo-cAMPs inhibitors of PKA.
Pharmaceuticals 05 01291 g001
Our current knowledge, however, about the molecular mechanisms underlying proper epithelial barrier functioning in the airways is mainly based on studies with focus on the endothelial barrier in the vasculature [2]. For the purpose of this review, we will outline our current knowledge about compartmentalized cAMP signalling. We will highlight the role of the epithelial barrier to maintain proper lung functioning and to alleviate COPD symptoms. The regulation of the endothelial barrier will serve as a starting point, and whenever appropriate, we will focus on the epithelial barrier function.

2. Spatio-Temporal Nature of Compartmentalized cAMP Signalling: Paradigm Shifts

The formation of cAMP is initiated by the stimulation of Gs-protein-coupled receptors, such as the β2-adrenoceptor and distinct prostanoid receptor subtypes. As members of the largest superfamily of cell surface signalling molecules, cAMP-elevating Gs-protein-coupled receptors represent the most prominent family of validated pharmacological targets in biomedicine [1,35,36]. In obstructive airways diseases short- and long-acting β2-agonists, such as salbutamol/albuterol, fenoterol, formoterol and indaceterol, are clinically widely used and act via stimulation of Gs-protein-coupled receptors [37,38,39,40]. In addition, recent studies emphasize also substantial progress to pharmacologically target the prostanoid PGE2-receptors to alleviate symptoms of obstructive lung diseases [41,42,43,44,45]. Over the last years substantial progress has been made to decipher the distinct signalling properties of cAMP. Initially, elevation of cellular cAMP by β2-agonists and prostanoids were expected to simultaneously stimulate both protein kinase A (PKA) and the exchange protein directly activated by cAMP (Epac) [1,46,47]. Meanwhile, it is generally accepted that spatio-temporal compartmentalization of cAMP maintained by cAMP-sensing AKAP-bearing multiprotein complexes and PDEs is of utmost importance to gain signalling specificity of cAMP [4,9,10,11,12,13,14,15,16,17].
Generally, G protein coupled receptors are considered as cell surface recognition sites sensing ions, hormones, neurotransmitters, autocoids and extracellular matrix components [36,38,48,49,50]. More recent studies showed that also internalized G protein-coupled receptors—until now believed to act as a ‘loss-of-function’ receptor signal—maintain signalling properties [18,51,52,53]. Using fluorescence resonance energy transfer to track intracellular cAMP fluctuations following activation of typically Gs-protein-coupled receptors [51,52], it has been reported that AC signalling is not necessarily restricted to the plasma membrane, but could be also detected in the endosome compartment. Indeed, endosomes, in which internalized receptors may end up, are now recognized as essential sites of cellular signalling [54,55], In addition, actin-stabilized endosomal microdomains profoundly affect the endosomal recycling and thereby the signalling properties of the β2-adrenoceptor [56]. Strikingly, Nikolaev and colleagues demonstrated that the β2-adrenoceptor its redistributed in heart failure, thereby compartmentalizing cAMP, a process proposed to contribute to the failing myocardial phenotype [18]. While the novel concept of cAMP signalling by internalized G protein-coupled receptors has recently been adapted to the signalling properties of the β2-adrenoceptor in human small airways [37], evidence that such mechanisms are operational in distinct structural airway cell subtypes, including bronchial epithelial cells, still has to be provided.
Intriguingly, ligand-directed signalling or biased agonism, referring to Gs-induced cAMP- versus β-arrestin-mediated signalling in response to different agonists [48,49,57,58,59], adds another level of complexity of Gs-protein-coupled receptor signalling, and has recently been reviewed within the context of obstructive lung diseases and the β2-adrenoceptor [38,40,60,61,62]. In mice, genetic ablation of either β-arrestin-1 or -2 prevented against bleomycin-induced pulmonary fibrosis and fibroblast invasion, suggesting a role for β-arrestin in fibrosis [63]. In support, β-arrestin-2 expression in increased in cell models of cystic fibrosis as well as in nasal tissue from patients [64]. In human bronchial epithelial cells, β-arrestin is necessary for the transcription of matrix metalloproteinases (MMPs) by diesel exhaust particles, a risk factor for COPD [65]. In addition to modulation of remodelling processes, β-arrestin is involved in agonist-induced desensitisation of the β2-receptor by inducing the internalization of this receptor [48]. Lefkowitz and colleagues reported that β-arrestin-mediated signalling exerts an even higher degree of regulation that relies on distinct phosphorylation sites of seven transmembrane receptors [66,67,68]. Likewise, β-arrestin-dependent signalling and trafficking of the β2-adrenoceptor also involve an unique deubiquitinase-ligase interplay [69,70]. Although recent studies indicate that ligand-directed signalling contributes to the functional responses of airway smooth muscle cells and lung fibroblasts [71,72], comparable studies in airway epithelial cells are still lacking. Further regulation of Gs signalling is mediated by the AKAP family members AKAP5 (aka AKAP79/150) and AKAP12 (aka AKAP250/Gravin), which regulate the de- and resensitization of the β2-adrenoceptor, respectively, and interact next to cAMP signalling proteins also with β-arrestin [14,15,16,17,73,74,75]. Thus, it is tempting to speculate that biased agonism might also profoundly alter the functional responses of AKAP-bearing multiprotein complexes. Moreover, receptors that ‘typically’ signal via Gs, including the β2-adrenoceptors, have also been shown to couple to other G-proteins, including Gi and G12/13, adding another layer of complexity to the regulatory pathways [76].
Recent studies indicate that, next to PDEs and ACs [77], members of the AKAP superfamily are of tremendous importance to maintain compartmentalized cAMP signalling and to prevent the progression of several diseases, such as chronic heart failure, Alzheimer’s dementia and cancer [17,18,19,77]. AKAPs exhibit a distinct (sub)cellular expression pattern and linkage to a diverse subset of target proteins including Gs-protein coupled receptors and ACs, cAMP effector proteins like PKA and Epac as well as cAMP-degrading PDEs (Figure 1). Cooper and colleagues reported recently that AKAP5 is target of palmitylation-dependent localization to lipid rafts and that the lipid modification of AKAP5 promotes its regulation of the calcium-sensitive AC subtype 8, adding an additional regulatory and targeting option for AKAP members [78]. Based on their binding specificity, cellular expression profiles and cellular localization, AKAPs integrate differential coupling of cAMP to specific cellular responses, including smooth muscle tone, cell proliferation and differentiation, learning and memory, inflammation, fibrosis, and barrier functioning (Figure 1) [14,15,16,17]. Intriguingly, the first cAMP-responsive multiprotein complexes identified in the heart and neurons possess a rather distinct composition: i) the cardiac-specific cAMP-responsive complex is maintained by the nuclear envelope-associated mAKAP, PKA, PDE4D3 and Epac1 [79], whereas ii) the neuronal cAMP-sensing complex is maintained by the plasma membrane-associated AKAP5, PKA, Epac2 and phosphoinositide 3-kinase-dependent protein kinase B (PKB/Akt) [80]. Generation of distinct cAMP-sensitive multiprotein complexes maintained by AKAP family members might turn out to be the key to explain that even though Epac and PKA can act independently, most cAMP-dependent processes are interconnectively regulated by Epac and PKA. Classically, most cAMP effects were assigned to PKA [81,82,83].The identification of Epac as an cAMP-regulated guanine nucleotide exchange factor (GEF) that favours GDP/GTP exchange and thereby activation of small Ras-like GTPases, profoundly changed the classical cAMP-PKA dogma [84,85]. The cAMP mediators Epac1 (aka cAMP-GEF-I) and Epac2 (aka cAMP-GEF-II) function as molecular links between members of the Ras superfamily such as Rho, Rac and Ras [86,87,88,89,90]. Members of the Ras superfamily belong to the GTP-ases which switch between a GDP-bound (inactive) state and a GTP-bound (active) state. Guanine exchange factors (GEFs) such as Epac, will exchange GDP for GTP, activating the effector. GTPase-activating proteins (GAPs) will reduce GTP-ase activity due to GTP hydrolyzation. GTP-binding of Rho, Rac or Ras will induce a diversity of cellular processes (see Figure 1).
Upon activation of distinct subset of small GTPases, Epac1 and Epac2 signal to phospholipase C-ε [91,92], phospholipase D [90,93], extracellular signal-regulated kinases (ERK1/2) [94,95,96,97,98,99], PKB/Akt [80,99,100,101,102,103,104] and NF-κB [105,106,107], and thereby control distinct cellular responses, including calcium handling, smooth muscle tone, cell proliferation and differentiation, migration, fibrogenic and inflammatory responses as well as barrier functioning [2,46,107,108,109,110]. We would like to refer the reader to excellent recent reviews with focus on the molecular signalling properties of Epac [2,46,47,108,109,110,111,112].
Recent studies reported on the contribution of cAMP-sensing AKAP-bearing multiprotein complexes to functional responses of different cell types of the airways. Human airway smooth muscle express seven of the nine membrane-bound AC subtypes [113]. The AC subtypes 5/6 differentially respond to β2-adrenoceptor and prostanoid receptor agonists, and thereby represent key molecules to generate cAMP [113,114,115], a process predominantly tuned by PDE4D5 [3,4,61]. As the expression of PDE4D5 is up-regulated by cAMP on the level of gene expression, protein expression and activity [116], cAMP seems to provide a feed-backward signal to diminish its own signalling properties. As PDE4D5 forms a complex with AKAP5 [74], up-regulation of PDE4D5 may alter the delicate spatio-temporal cAMP compartmentalization in human airway smooth muscle and thereby contribute to the progression of airway obstruction. Penn and colleagues reported very recently on the expression of a distinct subset of AKAPs, particularly AKAP12 and ezrin and its impact on compartmentalized cAMP signalling in human airway smooth muscle [117].
Human lung fibroblasts have been reported to express six of the nine membrane-bound AC subtypes [118] (Table 1). As in human airway smooth muscle, the AC subtypes 5/6 generate cAMP [118], whereas PDE4 subtypes hydrolyse cAMP in human lung fibroblasts [119,120,121]. A recent study by Peters-Golden and colleagues showed that the AKAP family member AKAP9 (aka AKAP450) represents a key protein for cAMP compartmentalization in fibrotic lung fibroblasts [122]. They reported that collagen deposition is controlled by a prostaglandin E2 (PGE2)-sensing AKAP9-PKA-protein phosphatase 2A multiprotein complex [122]. Activation of fibroblasts and subsequent collagen synthesis is inhibited by extracellular anti-fibrotic plasmin by restoring the PGE2-sensitivity of the fibroblast-specific AKAP9 (splice variant AKAP450) complex [122]. Intriguingly, AKAP9 (splice variant AKAP450) was found to generate a complex with PDE4 [123]. As selective inhibitors of PDE4 including rolipram, cilomilast and roflumilast are studied in clinical trials or licensed for use in COPD [34,124,125,126], targeting AKAP9-PDE4 complexes might be of benefit for COPD patients. The smallest splice variant of AKAP9, Yotiao can associate with ACs subtypes 1, 2, 3 and 9, leading to more phosphorylation of effectors by PKA [127].
Table 1. Expression of elements of cAMP signalling in cells and tissues involved in the pathogenesis of lung diseases.
Table 1. Expression of elements of cAMP signalling in cells and tissues involved in the pathogenesis of lung diseases.
EpacPKAAKAPPDEACsmall GTPases
Bronchial epitheliumEpac1 [128]
Epac 1 & 2 [129]
PKA [29]AKAP9 [130]++ PDE4, PDE1 [131,132]
+- PDE3, PDE5 [131,132]
PDE4D [133]
PDE3A [134]
PDE7A1&2 [135]
AC9 [136,137]
AC1, 4, 7, 8 [138]
sAC [139]
Rap [140,141,142]
Rac [29,141,142,143,144,145]
Rap1 [129,143]
Rap2
Vascular endotheliumEpac1 [146,147,148,149]PKA [147]AKAP9 [149]
Gravin [150]
PDE4D [22,147,151]
PDE4 [135]
PDE3 [135]
Membrane bound [2]
Soluble AC [2]
AC2, 3, 5, 6 [138]
Rap [143,146,147,149,152]
Rac [146,147,152,153,154,155,156,157]
Rac1 [158]
RRas [147,159]
Airway smooth muscle cellsEpac1 [95,107,129]
Epac2 [95,107,129]
PKA [107]AKAP5, 9, 12 [160]
Gravin, ezrin [117,150]
PDE1C, 3, 5A, 7 [135,161]
PDE7A1&2 [135]
7 membrane bound subtypes [118]
1, 3-7, 9 [115]
2, 6, 7, 9 [113]
RhoA [87]
Rac1 [87]
Rap1 [95]
Rap2 [95]
Vascular smooth muscle cellsEpac1 [162]
Epac2 [162]
PKA [162]AKAP12 [163]PDE1(C), 3(A), 5 [135]
PDE7A1&2 [135]
1A, 1C, 2A, 3A, 3B, 4A, 4B, 4C, 4D, 5A, 7A, 7B, 8A, 9A, 9B, 10A and 11A [164]
AC1, 2, 3, 4, 6, 7, 9 [165]
2, 3, 5, 6, 7, 8 [138]
Rap1 [166]
Rac1 [167]
RhoA [168, 169]
Pulmonary fibroblastsEpac [72]
Epac1 [170]
Epac2* [170]
*only mRNA not protein
PKA [122]AKAP9 [122]PDE4A, B, D [119]
3A&B, 4A5, 4B2, 4C1, 4D3, 7 [171]
6 membrane bound subtypes [118]Rho A [172]
Rac1 [172]
Rac2 [172]
Rap1 [173]
Inflammatory cellsEpac1 [109]PKA [109]Ezrin [174]
AKAP9 [175]
PDE4B2 [176]
PDE1B, 3A, 7A1, 2, 3 [135]
7A1&7A2 [177]
AC [178]
1, 2, 6, 9 [179]
4, 5, 6, 7, 9, sAC [180]
sAC [181]
Rap [182]
Ras [182]
Rac1 [183]
Rho [183]
Since airway smooth muscle cells and lung fibroblast both differentially contribute to chronic inflammation, airway obstruction and airway remodelling in COPD there might be a distinct underlying molecular mechanism for these symptoms in airway smooth muscle cells and lung fibroblast. The diverse complex profile of PDE4 subtypes to AKAP5 in airway smooth muscle compared to AKAP9 in lung fibroblasts may be responsible for this.
In contrast to the substantial progress of our current knowledge on the spatio-temporal compartmentalization of cAMP signalling in human airway smooth muscle and lung fibroblasts including subcellular clustering of receptors, ACs and PDEs, neither expression profiling of AC and PDE subtypes nor identification of cAMP-sensing AKAP-bearing multiprotein complexes in human bronchial and/or alveolar epithelial cells have yet been studied in detail (for an overview see Table 1). Whole lung tissue has been reported to express eight of the nine membrane bound AC subtypes [6,184,185]. In particular, the AC subtype 9 seems to represent the key molecule to generate cAMP and thereby alleviate symptoms of obstructive pulmonary disorders [136,137]. Interestingly, a recent report indicated that production of cAMP by the soluble AC subtype contributes to ciliary beat frequency in fully differentiated ciliated airway epithelial cells [186]. Different PDE isoforms are expressed in different epithelial cells. Whereas whole lung tissue primarily expresses PDE4 [187], primary alveolar A549 cells highly express PDE4 compared to PDE1, PDE3 and PDE5 and primary human bronchial epithelial (HBE) cells equally express PDE4 and PDE1, but express lower levels of PDE3 and PDE5 [131,132]. In contrast to studies in endothelial cells, cAMP-sensing multiprotein complexes maintained by AKAPs have not been studied yet in airway epithelial cells, even though PDE4 is complexed with AKAP9 [123]. In human umbilical vein endothelial cells (HUVECs), Epac1-dependent Rap1 activation and subsequent elevations in cortical actin and VE-cadherin increased barrier function. Parallel to this pathway Epac1-AKAP9 complex is required for microtubule growth, integrin adhesion at cell-cell borders and the endothelial barrier function [149]. In line with the previous finding that AKAP9 controls spatio-temporal cAMP dynamics of PDE4 [123], Maurice and colleagues demonstrated that PDE4D-dependent binding of Epac1 to a VE-cadherin-based signalling complex controls vascular permeability [147]. Cell-cell adhesion and integrin-extracellular matrix interactions also seem to rely on Epac, particularly Epac1 [22,151]. Interestingly, in polarized human Calu-3 airway epithelial cells it has been reported that a functional cAMP diffusion barrier maintained by PDE4D determines the activity of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel, indicating that functional responses to cAMP in airway epithelial cells rely on cAMP microdomains even though the existence of cAMP-sensing AKAP-bearing multiprotein complexes have not been studied yet [133]. However, Zaccolo and colleagues demonstrated recently that CFTR regulation in human airway epithelial cells required next to the subcortical cytoskeleton compartmentalized cAMP signalling. High cAMP levels localized at the plasma membrane are needed for PKA-dependent activation of CFTR. Compartmentalization is accomplished by a multi-protein complex with Na/H exchange regulatory factor-1 and ezrin. Next to PKA, Epac has also been shown to activate CFTR in response via activation of Rap2 [188] In models of cystic fibrosis, a disease associated with mutations in the CFTR gene, overexpression of wildtype CFTR resulted in an organised cAMP compartmentalization and restored fluid homeostasis [189]. In bronchial epithelial cells, compartmentalized cAMP signaling at the CFTR is regulated by PDE3A in a macrocomplex. PDE3A decreases cAMP at the membrane, thereby inactivating CFTR [134]. Compartmentalization of cAMP at the CFTR is further maintained by binding of the cAMP efflux transporter multidrug resistance protein-4 to CFTR, which reduces cAMP levels near the plasma membrane and reduces CFTR function [190].
In addition, multiprotein complexes supporting compartmentalized cAMP signalling are involved in secretory and proliferative functions in airway smooth muscle cells and are maintained by AKAP5 (aka AKAP79/150) and AKAP12 (aka AKAP250). Cigarette smoke-induced perturbation of airway smooth muscle compartmentalization of cAMP might contribute to the development and progression of COPD [160,191]. To summarize, distinct composition of cAMP-responsive AKAP-bearing multiprotein complexes seem to generate and to maintain local cAMP gradients, and thereby to regulate cell-type specific functions in structural airway cells.
The functional role of cAMP-signalling complexes can be studied using pharmacological tools. Perturbation of AKAP-bearing complexes can be achieved with the PKA-binding blocking peptides such as st-Ht31 [14,16,17]. Novel developed AKAP complex disrupters seem to effectively modulate compartmentalized cardiac cAMP signalling [192,193]. Direct activation of cAMP-generating AC subtypes can be achieved by the cell membrane-permeable diterpene forskolin from the Indian plant Coleus forskolhlii [6]. Novel N6-derivatives of cAMP, such as 6-Bnz-cAMP, directly and selectively activate PKA [194,195], whereas inhibition of PKA is achieved by H89 and the more selective inhibitors Rp-8-CPT-cAMPS, Rp-cAMPS and Rp-8-Bromo-cAMPS [195,196,197]. Direct activation of Epac (both Epac1 and Epac2) can be achieved by 8-pCPT-2’-O-Me-cAMP [194,195,198,199] and the PDE-resistant and cell membrane-permeable Sp-8-pCPT-2’-O-Me-cAMP, which exhibits an even increased specificity towards Epac [195,200]. Although pharmacological inhibitors of Epac proteins are not available, in vitro down-regulation of Epac expression by silencing RNA provided the first insight into Epac-specific functions [95,107,170,173] (Figure 1). The recently developed Epac1 and Epac2 knock-out mice should be supportive to specifically assign cellular functions to Epac1 and Epac2 [201,202]. These tools allowed studies on the novel aspects of the spatio-temporal nature of (compartmentalized) cAMP signalling in COPD.

3. Cellular Diversity in cAMP Responses: Compartmentalization?

COPD is a chronic inflammatory lung disease mainly caused by cigarette smoking and environmental pollution, and is expected to be the fifth cause of death worldwide by 2020, based on World Health Organization estimates [203]. COPD is characterized by a very slow progressive onset and by respiratory symptoms such as wheezing, cough, chest tightness and dyspnea [26,34,204,205,206]. COPD mainly afflicts middle-aged and elderly people, who usually bear a history of heavy smoking [207,208]. Long-term exposure to smoke (especially cigarette smoke) represents the main risk factor to develop COPD, although less than 25% of smokers develops COPD and at least 15% of COPD-related mortality occurs in never-smokers, suggesting that other factors may be important as well [207,208,209].Cigarette smoke consist of several distinct components such as tar, nicotine, carbon monoxide, ammonia carbonyls, volatiles, semi-volatiles, phenols, aromatic amines and N-nitrosamines [210], and in particular the high level of reactive organic radicals (particle size < 0.5 μm) seems to profoundly disturb functional responses of airway-related cells in the small airways [24,26,211,212]. Cigarette smoke-induced damage of the airway epithelium initiates a chronic cycle of injury and repair that involves the innate immune response, and the recruitment of macrophages as well of neutrophils [213,214]. Such processes contribute to an increase in the neutrophil attractant interleukin-8 and mucus hypersecretion [27,28,30,34,215,216,217] both of which are known to be associated with a higher risk of bacterial or viral infections [28]. Up to 25% of all exacerbations in COPD patients contain the bacteria strains Haemophilius influenzae and Moraxella catarrhalis, and especially the acquisition of new bacterial strains seems to be important for the onset of exacerbations [34,217]. Mucus hypersecretion and inflammatory mediators not only promote bacterial and/or viral infections, but also enhance inflammatory responses and thereby decrease mucociliarly clearance [34,217]. Thus, cigarette smoke-induced inflammation and mucus hypersecretion most likely promote the development and progression of typical COPD features.
The accelerated, not fully reversible decline in lung function in COPD is characterized by infiltration and activation of inflammatory cells, particularly macrophages, lymphocytes and neutrophils, which promote the release of proteases and inflammatory cytokines, including interleukin-8 and tumor necrosis factor [24,25,26,204]. Small airways and lung parenchyma are predominantly affected upon inflammation in COPD patients and contribute to the airway obstruction and progressive loss of lung function [34,218]. In addition to inflammatory cells, structural cells, including airway smooth muscle and epithelial cells, underpin a variety of key responses in the airways, such as smooth muscle contraction, airway remodelling, inflammatory cytokine release and mucus hypersecretion, features known to underpin airway obstruction in COPD [24,26,34]. Although the occurrence of airway hyperresponsiveness in COPD is debated, a considerable amount of COPD patients have been shown to exhibit higher responsiveness to contractile stimuli and the severity of airway hyperresponsiveness appears to be a good predictor of the rapid decline in lung function in patients with COPD [219,220,221]. Airway smooth muscle mass increases significantly in the small airways in COPD [222,223,224,225], and this increase is believed to be a main contributor to airway hyperresponsiveness [226].
Airway remodelling typically appears later in adult life [227,228], and predominantly affects small airways and lung parenchyma [227,228]. Airway remodelling in COPD is inextricably linked to the inflammatory cell infiltration into lung tissue and encompasses emphysema, increased mucous, squamous cell metaplasia and increased airway smooth muscle mass, enlargement of the bronchial mucus glands, increased mucus content in the airway lumen, airway fibrosis, and increased epithelial cell proliferation [24]. Vascular remodelling due to inflammatory infiltration of the vessels, is also a characteristic feature of COPD and may generate pulmonary hypertension [229]. An imbalance between proteases (including MMPs) and endogenous antiproteases is likely to be involved in the development of emphysema [230,231,232]. Fibrosis around the small airways is also believed to play a major role in the irreversible airway narrowing in COPD [233]. Airway fibrosis is considered to be the result of an abnormal wound repair mechanism, involving the recruitment and activation of (myo)fibroblasts, which may be derived from resident mesenchymal cells, circulating fibrocytes and epithelial-to-mesenchymal transition, a process in which epithelial cells transdifferentiate into fibroblasts [234,235,236]. Activated fibroblasts produce huge amounts of extracellular matrix proteins like collagens, proteoglycans and glycoproteins like fibronectin and laminins, thus contributing to fibrotic responses in COPD [204,235,237]. In surgically resected lung tissues, increased accumulation of inflammatory exudates with mucus in the small airways was noted to correlate with the severity of disease [23]. In COPD, structural changes include increased and altered extracellular matrix deposition and increased airway smooth mass, increased microvasculature, thickening of the reticular basement membrane, goblet cell metaplasia and epithelial changes [204,238,239,240,241]. Epithelial remodelling driven by cigarette smoke induces epithelial hyperpermeability and the disruption of the epithelial barrier function [27,29], processes known to correlate with goblet cell metaplasia and mucus gland hypertrophy [32,33]. In COPD patients with more severe disease, mucus hypersecretion account for morbidity and mortality [27,34].
Currently, no preventive or curative pharmacological treatment exists for COPD. Airflow obstruction in COPD is predominantly treated with anticholinergics and β2-agonists [242,243,244], the latter known to induce bronchodilation by elevating cAMP. β2-Agonists effectively reduce airflow obstruction, but have only a poor effect on airway inflammation [204]. Although airway inflammation in asthma can be well controlled by treatment with inhaled glucocorticosteroids in most patients, COPD patients are often characterized by a relative glucocorticosteroid insensitivity [245]. However, β2-agonists can augment the anti-inflammatory effect of glucocorticosteroids [246]. Although β2-agonists have been shown to inhibit cytokine release in vitro [247,248], evidence for their anti-inflammatory properties in vivo is still lacking [73]. This discrepancy might be explained by the development of β2-adrenoceptor desensitization, particularly in inflammatory cells [249]. Hence, in inflammatory cells inhibitors of phosphodiesterase (PDE) maintain the beneficial effects of β2-agonists without the risk of receptor desensitization due to their capacity to elevate cAMP by preventing its breakdown, and control inflammation in the airways and thereby possibly the frequency of exacerbations [125]. Selective inhibitors of PDE4, such as rolipram and roflumilast, have undergone clinical trials to determine their usefulness/efficacy in the treatment of COPD [124]. In contrast to β2-agonists, these PDE inhibitors only marginally reduce airflow obstruction [34,124,250,251,252,253]. Notably, though both β2-agonists and phosphodiesterase (PDE) inhibitors elevate cAMP, they modulate distinct cellular functions. Compartmentalization of cAMP and its effectors could explain these distinct cellular responses to cAMP by different cAMP elevating drugs.

4. Regulation of Epithelial Barrier Function: Tight Junctions versus Adherens Junctions

The following sections will discuss the maintenance of the epithelial barrier in health and disease to maintain proper lung functioning and to alleviate COPD symptoms. Prior to that, we will summarize central structural features of tight and adherens junctions that underpin the maintenance of a proper barrier. Herein, we will mainly focus on the epithelial barrier, and we would like to refer the reader to excellent reviews with focus on the molecular components of the endothelial barrier [254,255,256,257,258,259,260].
In healthy subjects, the epithelium forms a continuous lining to the airways and to the environment, and play a unique role as a barrier against external deleterious agents by elaboration of a series of defence mechanims developed to protect the airways from insults [212,240,261,262]. The airway epithelium defence system is comprised of several different functions, including structural features of the epithelium that maintain the epithelial barrier integrity, effective mucociliary clearance upon tight regulation of ciliary beating leading to an effective mucociliary clearance, and coordinated regulation of epithelial secretory properties to release molecules with antibacterial, antioxidant, and antiprotease activities [211,236,240,261]. Exposure to cigarette smoke severely alters airway epithelium morphology and function, and subsequently initiates a chronic cycle of injury and repair [29,213,214]. The different types of epithelial cells encompass rather distinct functions within the epithelial cell layer (Figure 2).
Figure 2. Epithelial barrier functions. Shown are key features of the epithelial barrier under healthy conditions and in the presence of toxic particles and/or infectious agents. Intriguingly, the different cell types composing the epithelial barrier including ciliated (C) cells, basal (B) cells, clara (C) cells and goblet (G) cells exhibit rather diverse functions within the barrier. Ciliated cells are responsible for the mucociliary clearance of infectious agents. Goblet cells produce the mucus needed for the clearance process. Clara cells contain granules filled with antiproteases known to be released by these cells upon their activation. Basement membrane is composed of basal cells known to exhibit next to their structural role a variety of distinct functions within the epithelium (see text for further details). Intercellular cell-cell contact between epithelial cells is achieved by tight junctions and adherens junctions. Adherens junctions ensure a tight adhesion of cells, whereas tight junctions act as a size selective barrier for certain ions and molecules. Exposure of epithelial cells to toxic Pharmaceuticals 05 01291 i001 such as cigarette smoke Pharmaceuticals 05 01291 i002 a (persistent) damage of the epithelial barrier, a process being compensated by cell spreading and production of extracellular matrix (ECM) by myofibroblasts to gain cell replacement and differentiation of distinct cells within the epithelium. In addition, toxic particles such as viruses and/or bacteria within the epithelial barrier will induce the recruitment of inflammatory cells and the production of cytokines to diminish the entrance of the devastating particles. For further details see text.
Figure 2. Epithelial barrier functions. Shown are key features of the epithelial barrier under healthy conditions and in the presence of toxic particles and/or infectious agents. Intriguingly, the different cell types composing the epithelial barrier including ciliated (C) cells, basal (B) cells, clara (C) cells and goblet (G) cells exhibit rather diverse functions within the barrier. Ciliated cells are responsible for the mucociliary clearance of infectious agents. Goblet cells produce the mucus needed for the clearance process. Clara cells contain granules filled with antiproteases known to be released by these cells upon their activation. Basement membrane is composed of basal cells known to exhibit next to their structural role a variety of distinct functions within the epithelium (see text for further details). Intercellular cell-cell contact between epithelial cells is achieved by tight junctions and adherens junctions. Adherens junctions ensure a tight adhesion of cells, whereas tight junctions act as a size selective barrier for certain ions and molecules. Exposure of epithelial cells to toxic Pharmaceuticals 05 01291 i001 such as cigarette smoke Pharmaceuticals 05 01291 i002 a (persistent) damage of the epithelial barrier, a process being compensated by cell spreading and production of extracellular matrix (ECM) by myofibroblasts to gain cell replacement and differentiation of distinct cells within the epithelium. In addition, toxic particles such as viruses and/or bacteria within the epithelial barrier will induce the recruitment of inflammatory cells and the production of cytokines to diminish the entrance of the devastating particles. For further details see text.
Pharmaceuticals 05 01291 g002
Essentially, the main cell types within the epithelium are divided between secretory and ciliated cells. Epithelial ciliated cells are engaged to transport the mucus out of the airways and thereby to remove the pathogens and toxic particles trapped in the mucus. The functionally close interplay between ciliated and secretory epithelial cells is illustrated by the fact that mucus being utilized by ciliated cells to maintain their proper transport function is produced and secreted by goblet cells, a secretory cell type of the epithelium, whereas a distinct subset of secretory cells release antimicrobial substances as a defence against unwanted pathogens [212,240,261]. The epithelium also consists of basal cells [263,264,265], a separate layer of cells covering most of the airway basal lamina. Due to their central position within the epithelium, basal cells interact with the columnar epithelium (a single cell layer of epithelium cells lining the respiratory tract), neurons, the basement membrane as well as underlying mesenchymal cells, and represent a key component of the epithelial-mesenchymal trophic unit of larger airways [240,264]. Basal cells execute diverse functions within the epithelial cell layer such as the inflammatory response, transepithelial water movement, oxidant defence of the tissue, the formation of the lateral intercellular space and progenitor cell functions for epithelium-associated cells, in particular during the development of the epithelium [263,264,265].
The epithelium is composed of continuous intercellular barriers such as tight junctions known to account for a size-selective barrier of molecules into the epithelium [266]. Tight junctions are located between the apical and lateral cell surface and thereby maintain cell polarity. Tight junctions are characterized by a unique expression profile of proteins such as claudins, occludins, zona occludens and junction adhesion molecules (JAMs) [266,267,268] (Figure 2). Currently, three members of the JAM family of transmembrane proteins have been identified: JAM-A, JAM-B and JAM-C. In particular, JAM-A is highly expressed in the tight junctions of the epithelial cells. In addition, JAM-A controls neutrophil transmigration, primarily across endothelial cells [267,269,270]. As neutrophil numbers seem to be indicative for COPD severity and exacerbation frequency [34,271], it is tempting to speculate that dysfunctions on the level of JAM-A might be also of importance for typical COPD disease features. Interestingly, the coxsackievirus and adenovirus receptor (CAR) represents another member of the JAM family [272,273], and has been reported to regulate the barrier function of tight junctions in epithelial cells [267,274].
Zona occludens (ZO) proteins represent other components of the tight junctions [262,275], that regulate junction formation and the interaction with the actin cytoskeleton [276,277,278,279]. Of the three members of the ZO protein family, ZO-1, ZO-2 and ZO-3, ZO-1 exhibits a rather abundant expression profile and binds to myosin, F-actin, ZO-2 and ZO-3 [262,275,280,281] (Figure 2). Studies in Madin-Darby canine kidney cells demonstrated that downregulation of ZO-1 using stable expression of a ZO-1 short hairpin silencing RNA profoundly altered junctional morphology and the organization of the actin cytoskeleton. Decrease of ZO-1 largely reduced the amount of cell-cell contacts and resulted in an intracellular accumulation of actin [282]. Importantly, transepithelial electrical resistance measurements demonstrated that the reduction of ZO-1 expression was paralleled by a reduction of the epithelial barrier as shown by a size selective increase in the movement of molecules < 4 Å through the disrupted barrier [282]. Taken together, ZO-1 controls paracellular permeability by coupling to components of the junctional actin cytoskeleton. Next to the tight junctions, the epithelium consists of an additional intercellular barrier, namely the adherens junctions predominantly expressed at the more basal side of the epithelial cells. Adherens junctions are characterized by the expression of cadherin family members such as E-cadherin and catenin [141,262,275,283]. E-cadherin interacts with α-catenin and/or β-catenin to form adherens junctions [262,275], a process strengthened by connection of adherens junctions to actin filaments present at intracellular sites of the cell-cell contacts [141]. Actin present in epithelial cells will form a circumferential belt which is bound to the adherens junction. Myosin, as part of this belt and bounded to actin, can control the shape of the cell via this belt [240,264,284] (Figure 2). Based on this, myosin fulfils an important function in the molecular architecture of both tight junctions and adherens junctions. As the predominantly expressed protein in the muscle, myosin binds to actin and thereby enables actomyosin-mediated muscle contraction, a process being under control of Rho-Rho-kinase [262,268,285,286]. The phosphorylation of the Rho-Rho-kinase target myosin light chain is decreased due to a reduction in the RhoA/Rac1 ratio via cAMP-driven Epac activation. Dephosphorylation of the myosin light chain relaxes the smooth muscle [87,88]. Myosin light chain, not only found in smooth muscle cells, but also in epithelial cells can be phosphorylated by Rho-Rho-kinase, increasing the epithelial barrier.
From the multiple myosin isoforms, myosin IIA and IIB are primarily expressed at cell-cell contacts [287]. Importantly, is has been reported recently that myosin IIA and myosin IIB engage rather distinct signalling cascades to regulate cadherin junctions in MCF7 breast epithelial cells [142]. Junctional localization of myosin IIA requires next to E-cadherin adhesion, Rho-Rho-kinase and myosin light chain-kinase activation, and thereby subsequently increase the contractile force of the circumferential belt and tight junction integrity. Myosin IIB, via Rap1A, supports myosin IIA-Rho-Rho-kinase signalling to E-cadherin and to myosin light chain kinase, and thereby also subsequently induce the stabilisation of the apical ring structure and enhancement of the junctional integrity [142]. Thus, both myosin IIA and myosin IIB might modulate the epithelial barrier function upon enhancement of the (tight) junction integrity through signalling via a rather distinct subset of small GTPases, namely RhoA and Rap1A. Several recent reports indicate that Rap1 regulates cell-cell junction formation through signalling to E-cadherin-catenin and integrin-extracellular matrix complexes [22,151,288,289]. Thus, it is tempting to assume that Epac, by activation of Rap1, importantly regulates the barrier function. As it has been also shown that E-cadherin internalization induces GTP-loading, thus, activation of Rap1 [140], these findings might indicate that the activity state of Rap1 is not only controlled by guanine nucleotide exchange factors but also by structural components of the cell-cell barrier.
The next layer of the epithelial barrier, the basement membrane belongs to the basement membrane zone and is a central component of the epithelial mesenchymal trophic unit, the latter known to consist of opposing layers of epithelial and mesenchymal cells separated by the basement membrane [240,263,264,265]. The basement membrane executes several distinct functions such as epithelium-extracellular matrix attachment, barrier functioning, cell-cell/cell-matrix communication and binding of growth factor, including the epidermal growth factor and the transforming growth factor-β (TGF-β) [240,263,264,265] (Figure 2). Although thickening of the epithelial reticular basement membrane is a typical feature of asthma [228], structural alterations on the level of the basement membrane and its potential impact on typical COPD features is still a matter of debate. The extracellular matrix underneath the basement membrane consists of several distinct components such as collagens, proteoglycans and glycoproteins like fibronectin and laminins [212,261,262,263,264]. In COPD patients a decrease in proteoglycans, such as decorin and biglycan, is observed. Collagen and fibronectin are increased in patients with emphysema leading to airway wall fibrosis [204,238,240,264,284,290].
Activated fibroblasts produce huge amounts of the extracellular matrix components, but also regulate the production of MMPs and endogenous antiproteinases [291,292,293]. Elevation of TGF-β promotes cell proliferation and extracellular matrix deposition by fibroblasts, whereas MMP-2 and MMP-9 (gelatinase / collagenase) and MMP-12 (elastase), which are increased in sputum of COPD patients [204,294] promote pro-fibrotic responses as well as destruction of the alveolar parenchyma. Thus, an imbalance between matrix metalloproteinase and their appropriate antiproteinase may contribute to fibrosis and emphysema in COPD [204,294,295,296,297].
Cigarette smoke belongs to the major risk factors of COPD [32,298], and is known to induce a vicious cycle of injury and repair in the airway epithelium upon adaptation on the level of tight junction and adherens junction morphology and function [31,212,213,214], a process which may eventually end up in transcriptional reprogramming of the airway epithelium [29]. Unfortunately, cigarette smoke-induced repair mechanism may also worsen airway obstruction of COPD patients through mucus hypersecretion by goblet cell hyperplasia, down-regulation of epithelial ciliated cells and hypertrophy of the submucosal gland [24,204,212,213,261]. Importantly, a recent study of Crystal and colleagues demonstrated that cigarette smoke exposure of airway epithelium induced a profound down-regulation of the majority of the typical tight junction and adherens junctions components including claudins, ZO proteins, E-cadherin and catenin [29]. In contrast, cigarette smoke exposure induced a profound up-regulation of molecular pathways known to be critical for epithelial differentiation including the phosphatase and tensin homolog (PTEN), phosphoinositide 3-kinase-dependent PKB/Akt, the cAMP effector PKA and Rac [29]. As several recent studies indicate that a proper signal balance between RhoA versus Rac1 seems to be of utmost importance for the regulation of both the endothelial and epithelial barrier [2,112,153,299,300,301], a subtle interplay between the cAMP-effector Rap1 and Rac1 might be envisioned as a key event to preserve the structure and function of tight and adherens junctions (Figure 3). Particularly, the impact of Rac1 on the endothelial barrier seems to be rather puzzling as both stabilization and destabilization of the endothelial barrier integrity have been observed [147,152,153,154,155,156,157] (Figure 2 and Figure 3). Intriguingly, these opposing findings might be partly explained by the distinct barrier protection properties of Rac1 in micro-vascular versus macro-vascular endothelial cells. Future studies should intend to unravel the precise underlying molecular signalling pathways and thereby to develop novel therapeutical interventions to restore and to maintain proper epithelial barrier in patients with COPD.
In epithelial cells, cAMP elevation will activate Rap and Rac via Epac. Rac in its active state will reduce the binding of IQGAP1 to β-catenin, resulting in a decrease of the epithelial barrier [141]. Rap is also activated by myosin IIB and will thereby enhance the barrier via E-cadherin. Myosin IIB will also activate Rho-Rho-kinase which via myosin light chain phosphorylation will increase the barrier properties [142].

5. Novel Aspects of Barrier Functioning

Insights into the molecular mechanisms being implicated in the maintenance or even restoration of the epithelial barrier function are still rather limited, therefore we will outline the molecular mechanisms that maintain the endothelial barrier function in the vasculature, we will translate these findings onto the regulation of the epithelial barrier function in COPD.
Figure 3. cAMP signalling in endothelial cells vs. epithelial cells. β2-agonists and prostanoids which activate their appropriate G-protein coupled receptor, and forskolin which activates adenylcyclase (AC) will increase cAMP production in both endothelial and epithelial cells resulting in cell type specific responses in both cells. In endothelial cells, cAMP increase will cause activation of both PKA and Epac. Activation of Epac will enhance microtubule growth which is AKAP9-dependent [148]. Epac activation will result in binding to PDE4D (phosphodiesterase 4D) which binds to the E-cadherin complex causing improvement of the barrier [147]. Next to this, Epac activation will reduce the phosphorylation of the myosin light chain, causing relaxation and improvement of the barrier [87,88]. The other effector of cAMP, PKA, anchored to AKAP (A-kinase anchoring protein) will activate both Rap and Rac. Active Rap, stabilized by IQGAP1, can activate KRIT which stabilizes cell-cell contacts. IQGAP2, bound to AKAP220, mediates calcium-dependent Rac activation which can alter actin dynamics [143,144,145,302,303]. In epithelial cells, cAMP elevation will activate Rap and Rac via Epac. Rac in its active state will reduce the binding of IQGAP1 to β-catenin, resulting in a decrease of the epithelial barrier [141]. Rap is also activated by myosin IIB and will thereby enhance the barrier via E-cadherin. Myosin IIB will also activate Rho-Rho-kinase which via myosin light chain phosphorylation will increase the barrier properties [142].
Figure 3. cAMP signalling in endothelial cells vs. epithelial cells. β2-agonists and prostanoids which activate their appropriate G-protein coupled receptor, and forskolin which activates adenylcyclase (AC) will increase cAMP production in both endothelial and epithelial cells resulting in cell type specific responses in both cells. In endothelial cells, cAMP increase will cause activation of both PKA and Epac. Activation of Epac will enhance microtubule growth which is AKAP9-dependent [148]. Epac activation will result in binding to PDE4D (phosphodiesterase 4D) which binds to the E-cadherin complex causing improvement of the barrier [147]. Next to this, Epac activation will reduce the phosphorylation of the myosin light chain, causing relaxation and improvement of the barrier [87,88]. The other effector of cAMP, PKA, anchored to AKAP (A-kinase anchoring protein) will activate both Rap and Rac. Active Rap, stabilized by IQGAP1, can activate KRIT which stabilizes cell-cell contacts. IQGAP2, bound to AKAP220, mediates calcium-dependent Rac activation which can alter actin dynamics [143,144,145,302,303]. In epithelial cells, cAMP elevation will activate Rap and Rac via Epac. Rac in its active state will reduce the binding of IQGAP1 to β-catenin, resulting in a decrease of the epithelial barrier [141]. Rap is also activated by myosin IIB and will thereby enhance the barrier via E-cadherin. Myosin IIB will also activate Rho-Rho-kinase which via myosin light chain phosphorylation will increase the barrier properties [142].
Pharmaceuticals 05 01291 g003
It is generally accepted that several endothelial barrier disrupting agents, such as tumor necrosis factor-, thrombin and the bacterial endotoxin lipopolysaccharide, profoundly alter the molecular architecture as well as the dynamics of the actin-microtubule network known to comprise the barrier tight and adherens junctions [258,259,260,261,262]. Importantly, cAMP elevating agents such as β2-agonists, prostanoids (prostacyclin, prostaglandin, PGE2) and the direct AC activator forskolin effectively reduce the leakage of the endothelial barrier in whole animals, isolated lungs and in clinical studies under both resting conditions and exposure to inflammatory mediators [2,304,305,306]. Several conclusive studies by Waschke and colleagues [153,156,157,307] demonstrated that at least part of the cAMP-dependent enhancement of the human dermal microvascular endothelial barrier (measured by transelectrical resistance (TER)) is mediated via the activation of Rac1 by both vasodilator-stimulated phosphoprotein (VASP) and AKAP-anchored PKA (Figure 3). Activation of Rac1 by AKAP-anchored PKA was sensitive to the PKA-AKAP-binding blocking peptides st-Ht31 [158]. As shown in human lung microvascular endothelial cells [308], PKA-dependent phosphorylation of VASP might also contribute to the barrier protection.
Although the members of the AKAP superfamily involved in the barrier function of human dermal microvascular endothelial cells still have to be identified, these results indicate that compartmentalized cAMP signalling by AKAPs contribute to the endothelial barrier function. The magnitude of the cAMP-dependent endothelial barrier maintenance, a process being paralleled by the subcellular localization of endothelial adherens junction marker VE-cadherin [153,156,157], profoundly differs between the β2-agonist epinephrine, the direct AC activator forskolin and the PDE4 inhibitor rolipram, the latter being non-effective on its own and acquiring biological effectiveness only in the presence of forskolin. These findings indicate that forskolin and rolipram are functionally different and/or require a distinct assembly of cAMP signalling pools. Indeed, studies from Maurice and colleagues demonstrated that PDE4D-bearing VE-cadherin-based multiprotein complexes control the vascular permeability in HUVECs [147,155]. Next to PDE4D and VE-cadherin, cAMP-dependent Epac1 was shown to be vital for the regulation of the endothelial barrier in HUVECs [147]. Indeed, subsequent studies by Waschke and colleagues indicated that the direct Epac activator 8-pCPT-2'-O-Me-cAMP mimicked the effect of epinephrine and forskolin on the barrier function and subcellular distribution of VE-cadherin in human microvascular endothelial cells [153,157]. As shown before for VASP and AKAP-anchored PKA, direct activation of Epac induced also the GTP-loading of Rac1 [153,157]. In addition, Hordijk and colleagues reported that Rac1 induced the production of reactive oxygen species and subsequently induced subcellular redistribution of VE-cadherin-catenin complexes in HUVECs [309]. Such mechanisms might worsen the endothelial barrier function under inflammatory disease conditions (Figure 3).
Interestingly, protection of the endothelial barrier upon activation of Rac1 was restricted to microvascular endothelial cells [157]. It is tempting to speculate that a distinct subset of cAMP-sensing AKAP-bearing multiprotein complexes might be expressed in micro-vascular versus macro-vascular endothelial cells. In support, recent studies point to the existence of a rather heterogeneous composition of such multiprotein complexes in cardiomyocytes [79], neurons [19], human lung fibroblasts [82,122] and airway smooth muscle [117]. Such diversity may also -at least in part- explain the opposing effects on the endothelial barrier – both protection and disruption - observed upon β2-adrenegic receptor stimulation [153,156].
Birukova and colleagues characterized the molecular mechanisms leading to Rac1 activation by Epac1 in human pulmonary artery endothelial cells in vitro and in ventilator-induced lung injury in vivo [146,310,311,312,313]. Elevation of cellular cAMP content, e.g., by prostaglandin E2 and prostacyclin I2, induced GTP-loading of Rac1 via Epac1-dependent Rap1 activation and the engagement of the Rac-specific GEFs Tiam1 and Vav2, processes being supported by PKA and the inhibition of p115 Rho-GEF-dependent activation of RhoA. Altogether, these mechanisms contribute to the barrier protection observed in human pulmonary artery endothelial cells in vitro and to the attenuation of ventilator-induced lung injury in vivo [146,310,311,312,313]. As reported for the molecular mechanisms leading to the relaxation of smooth muscle [87,88], Epac1 most likely protects the endothelial barrier by decreasing the phosphorylation of the Rho-Rho-kinase target myosin light chain by skewing the balance of RhoA/Rac1 activation towards Rac1. These recent findings confirmed initial studies by the research groups of Mayadas [148,314] and Mochizuki [22,314], reporting on the first molecular link of Epac and the actin-microtubule, and its impact on the regulation of the barrier function in HUVECs and human pulmonary aortic endothelial cells. Recently, Ginsberg and colleagues demonstrated that Krit1 (Krev1 interaction trapped gene) is required for the stabilization of β-catenin-bearing cell-cell contacts by the Epac effector Rap1, and that the Epac-Rap1 effector Krit1 is required for the maintenance of the endothelial barrier [315,316]. Loss of Krit1, known to account for the loss of endothelial junctions in cerebral cavernous malformations [316], induces destabilization of the endothelial barrier by increasing the phosphorylation of the Rho-Rho-kinase target myosin light chain. Although the involvement of Epac has not been studied by Ginsberg and colleagues [316], these findings might indicate that Epac-bearing multiprotein complexes are of utmost importance to maintain the endothelial barrier properties (Figure 3).
Intriguingly, Mayadas and colleagues reported recently that AKAP9 induced Epac1-dependent-microtubule growth resulting in stabilization of the barrier function in HUVECs and human dermal microvascular endothelial cells [149]. As AKAP9 has been reported to bind to PDE4 [123], PDE4D-dependent Epac1 binding to VE-cadherin-based signalling complexes might contribute to the maintenance of the micro-vascular endothelial permeability [147]. This process which might be supported by an Epac1 dependent enhancement of cell-cell adhesion and integrin-extracellular matrix interactions [22,151]. As Waschke and colleagues reported recently on the activation of Rac1 by AKAP-anchored PKA [158], it is tempting to speculate that the AKAP9 mediates the Epac-dependent Rac1 activation in human micro-vascular endothelial cells. In addition, it has been reported that a GTPase-deficient mutant of IQGAP1 induced GTP-loading of Rac1 and inhibited IQGAP1 sequestration of β-catenin, and thereby subsequently stabilized E-cadherin-dependent barrier function of MCF7 breast epithelial cells which show epithelium characteristics [143,144,145]. As IQGAP1 binds also to the Epac-effector Rap1 [143], Epac-dependent compartmentalized cAMP signalling in human micro-vascular endothelial cells might require next to AKAP superfamily members IQGAPs. In line with this assumption, Scott and colleagues demonstrated recently that a calcium-dependent AKAP220-IQGAP2 complex mediated Rac activation and thereby cellular actin remodelling [143,302,303]. The existence of such AKAP-IQGAP complexes in the endothelium and their contribution to the regulation of the endothelial barrier still has to be studied.
Although it is generally accepted that cigarette smoke alters the epithelial functions in COPD remodelling [27,28,29,30,31,32,33], the molecular mechanisms contributing to the regulation of the epithelial barrier are by far less characterized compared to several recent studies with focus on the endothelial barrier. Little information is known on the epithelial barrier in COPD, but studies performed in other types of epithelial cells may indicate a potential role of cAMP signalling pathway in the restoration of the barrier in COPD patients.
It has been reported for podocytes (renal glomerular visceral epithelial cells) that the AC activator forskolin induced a redistribution of ZO-1, E-cadherin, and β-catenin to cell-cell contacts [317]. On the other hand, reduction of cellular cAMP levels upon PDE inhibition by pentoxifylline attenuated tight junctions of immunostimulated Caco-2 human intestinal epithelial cells [318].
Importantly, Menke and colleagues reported recently that transformation of the human pancreatic carcinoma epithelial-like cell line PANC-1 with constitutively active Rac1(V12) profoundly altered the subcellular distribution of E-cadherin- β-catenin complexes and thereby epithelial cell-cell contacts in an IQGAP1-dependent manner, whereas cell transformation with dominant negative Rac1 (N17) had the opposite effect [141]. Together with the finding that in MCF7 breast epithelial cells myosin IIB signals via Rap1A to E-cadherin and to the Rho-Rho-kinase effector myosin light chain and subsequently enhance junctional integrity [142], a diligent balance of the GDP/GTP-loading of the small GTPase superfamily members Rac1 and Rap1 -most likely driven by compartmentalized cAMP signalling by a distinct subset of AKAPs and IQGAPs- seem to be of key importance to maintain a proper barrier function in epithelial cells. Of interest to note is that IQGAP1 and IQGAP2 seem to be differentially involved in the regulation of the cellular barrier [143,144,145] and the actin-microtubule network dynamics [302,303]. Indeed, recent research indicate also that IQGAP1 and IQGAP2 fulfill distinct functions in tumorigenesis, whereas IQGAP1 acts as an oncogene IQGAP2 seems to act as a tumor suppressor (Figure 3) [319]. Preliminary results of our group show that cigarette smoke-mediated disruption of human bronchial epithelial (HBE) barrier correlates with a down-regulation of AKAP9 [130]. As AC9 is the main isoform in bronchial epithelial cells [123,136], it is tempting to speculate that the previous described interaction between AC9 and AKAP9/Yotiao [127] is involved in this process. Next to AKAP9, AKAP5 and AKAP12 are also expressed in human bronchial epithelial cells, but their expression is not sensitive to cigarette smoke exposure. Importantly, the expression of AKAP9 mRNA was also down-regulated in primary epithelial cells of current smokers compared to non/ex-smokers as well as in lung biopsies from COPD patients [320].
Taken together, compartmentalized cAMP signalling maintained by a distinct subset of cAMP-responsive multi-protein complexes seen also to account for the proper functioning of the epithelial barrier, the latter known to be derailed in patients with COPD. Future research should aim to target cell-type specific cAMP-sensing complexes to augment our current therapeutically treatment regimes for chronic inflammatory disorders such as COPD.

6. Conclusions

G-protein-coupled receptors, adenylyl cyclases and PDEs regulate, in a spatio-temporal manner, the cellular cAMP concentration and subsequent cAMP signalling. Compartimentalization of cAMP signalling through a distinct subset of multi-domain proteins of the AKAP family supports fine-tuning of the net-outcome of cAMP-regulated cellular responses. Novel insights into cAMP compartmentalization upon manipulation of these multi-protein complexes may lead to new therapies in diseases like heart failure, cancer and COPD, known to be characterized by cAMP dysfunction.
In COPD, dysfunction of the epithelial barrier results in progression of disease symptoms. Since cAMP exhibits protection of the barrier function in endothelial cells, targeting the cAMP pathway may also restore the damaged epithelial barrier in COPD. Given the importance of compartmentalized cAMP signalling in regulating cellular barrier functions, alterations in maintenance of the protein-protein communication may lead to the observed barrier dysfunction in COPD. Upon activation of Rap and inhibition of Rho, the cAMP effectors Epac and PKA increase cellular barrier function.
Future studies with focus on cAMP compartmentalization will be required to further unravel the underlying molecular mechanisms. Further understanding of this compartmentalized cAMP signalling will be of benefit for improvement of the current therapeutic arsenal for the treatment of COPD.

Acknowledgements

This work was supported by a grant by the Nederlands Astma Fonds (NAF 2009: projectnr: 3.2.09.034, NAF 2011: projectnr: 3.2.11.015) and a Rosalind Franklin Fellowship from the University of Groningen to M. Schmidt. Harm Maarsingh is supported by MSD (Oss, The Netherlands).

Conflict of interest

The authors declare no conflict of interest.

References

  1. Beavo, J.A.; Brunton, L.L. Cyclic nucleotide research -- still expanding after half a century. Nat. Rev. Mol. Cell Biol. 2002, 3, 710–718. [Google Scholar]
  2. Sayner, S.L. Emerging themes of cAMP regulation of the pulmonary endothelial barrier. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 300, L667–L678. [Google Scholar] [CrossRef]
  3. Conti, M.; Beavo, J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu. Rev. Biochem. 2007, 76, 481–511. [Google Scholar] [CrossRef]
  4. Houslay, M.D. Underpinning compartmentalised cAMP signalling through targeted cAMP breakdown. Trends Biochem. Sci. 2010, 35, 91–100. [Google Scholar] [CrossRef]
  5. McCahill, A.C.; Huston, E.; Li, X.; Houslay, M.D. PDE4 associates with different scaffolding proteins: modulating interactions for certain diseases. Handb. Exp. Pharmacol 2008, 186, 125–166. [Google Scholar] [CrossRef]
  6. Hanoune, J.; Defer, N. Regulation and role of adenylyl cyclase isoforms. Annu. Rev. Pharmacol Toxicol. 2001, 41, 145–174. [Google Scholar] [CrossRef]
  7. Patel, H.H.; Murray, F.; Insel, P.A. G-protein-coupled receptor-signaling components in membrane raft and caveolae microdomains. Handb. Exp. Pharmacol 2008, 167–184. [Google Scholar]
  8. Patel, H.H.; Murray, F.; Insel, P.A. Caveolae as organizers of pharmacologically relevant signal transduction molecules. Annu. Rev. Pharmacol Toxicol. 2008, 48, 359–391. [Google Scholar] [CrossRef]
  9. Zaccolo, M.; Pozzan, T. Discrete microdomains with high concentration of cAMP in stimulated rat neonatal cardiac myocytes. Science 2002, 295, 1711–1715. [Google Scholar] [CrossRef]
  10. Di Benedetto, G.; Zoccarato, A.; Lissandron, V.; Terrin, A.; Li, X.; Houslay, M.D.; Baillie, G.S.; Zaccolo, M. Protein kinase A type I and type II define distinct intracellular signaling compartments. Circ. Res. 2008, 103, 836–844. [Google Scholar]
  11. Zaccolo, M. cAMP signal transduction in the heart: understanding spatial control for the development of novel therapeutic strategies. Br. J. Pharmacol. 2009, 158, 50–60. [Google Scholar] [CrossRef]
  12. Zaccolo, M. Spatial control of cAMP signalling in health and disease. Curr. Opin. Pharmacol. 2011, 11, 649–655. [Google Scholar] [CrossRef]
  13. Stangherlin, A.; Zaccolo, M. Phosphodiesterases and subcellular compartmentalized cAMP signaling in the cardiovascular system. Am. J. Physiol Heart Circ. Physiol. 2012, 302, H379–H390. [Google Scholar] [CrossRef]
  14. Beene, D.L.; Scott, J.D. A-kinase anchoring proteins take shape. Curr. Opin. Cell Biol. 2007, 19, 192–198. [Google Scholar] [CrossRef]
  15. Skroblin, P.; Grossmann, S.; Schafer, G.; Rosenthal, W.; Klussmann, E. Mechanisms of protein kinase A anchoring. Int. Rev. Cell Mol. Biol. 2010, 283, 235–330. [Google Scholar] [CrossRef]
  16. Wong, W.; Scott, J.D. AKAP signalling complexes: focal points in space and time. Nat. Rev. Mol. Cell Biol. 2004, 5, 959–970. [Google Scholar] [CrossRef]
  17. Tröger, J.; Moutty, M.C.; Skroblin, P.; Klussmann, E. A-kinase anchoring proteins as potential drug targets. Br. J. Pharmacol. 2012, 166, 420–433. [Google Scholar] [CrossRef]
  18. Nikolaev, V.O.; Moshkov, A.; Lyon, A.R.; Miragoli, M.; Novak, P.; Paur, H.; Lohse, M.J.; Korchev, Y.E.; Harding, S.E.; Gorelik, J. Beta2-adrenergic receptor redistribution in heart failure changes cAMP compartmentation. Science 2010, 327, 1653–1657. [Google Scholar] [CrossRef]
  19. Ostroveanu, A.; van der Zee, E.; Eisel, U.L.; Schmidt, M.; Nijholt, I.M. Exchange protein directly activated by cyclic AMP2 (Epac2) plays a specific and time-limited role in memory retrieval. Hippocampus 2010, 20, 1018–1026. [Google Scholar] [CrossRef]
  20. Aye, T.-T.; Soni, S.; van Veen, T.A.B.; van der Heyden, M.A.G.; Cappadona, S.; Varro, A.; de Weger, R.A.; de Jonge, N.; Vos, M.A.; Heck, A.J.R.; Scholten, A. Reorganized PKA-AKAP associations in the failing human heart. J. Mol. Cell. Cardiol. 2012, 52, 511–518. [Google Scholar] [CrossRef]
  21. Kovanich, D.; van der Heyden, M.A.; Aye, T.T.; van Veen, T.A.; Heck, A.J.; Scholten, A. Sphingosine kinase interacting proteins is an A-kinase anchroing protein specific for type I cAMP-dependent protein kinase. Chembiochem. 2010, 3, 963–971. [Google Scholar]
  22. Noda, K.; Zhang, J.; Fukuhara, S.; Kunimotot, S.; Yoshimura, M.; Mochizuki, N. Vascular endothelial-cadherin stabilizes at cell-cell junctions by anchoring to circumferential actin bundles through a- and b-catenins in cyclic AMP-Epac1-Rap1 signal-activated endothelial cells. Mol. Cell. Biol. 2010, 21, 584–596. [Google Scholar]
  23. Hogg, J.C.; Chu, F.; Utokaparch, S.; Woods, R.; Elliott, W.M.; Buzatu, L.; Cherniack, R.M.; Rogers, R.M.; Sciurba, F.C.; Coxson, H.O.; Pare, P.D. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N. Engl. J. Med. 2004, 350, 2645–2653. [Google Scholar] [CrossRef]
  24. Hogg, J.C.; Timens, W. The pathology of chronic obstructive pulmonary disease. Annu. Rev. Pathol. 2009, 4, 435–459. [Google Scholar] [CrossRef]
  25. Barnes, P.J. Immunology of asthma and chronic obstructive pulmonary disease. Nat. Rev. Immunol. 2008, 8, 183–192. [Google Scholar] [CrossRef]
  26. van den Berge, M.; ten Hacken, N.H.; Cohen, J.; Douma, W.R.; Postma, D.S. Small airway disease in asthma and COPD: clinical implications. Chest 2011, 139, 412–423. [Google Scholar]
  27. Rogers, D.F.; Barnes, P.J. Trends in clinical practice: Treatment of airway mucus hypersecretion. Ann. Med. 2006, 38, 116–125. [Google Scholar] [CrossRef]
  28. Lai, H.; Rogers, D.F. New pharmacotherapy for airway mucus hypersecretion in asthma and COPD: Targeting intracellular signaling pathways. J. Aerosol. Med. Pulm. Drug Deliv. 2010, 23, 219–231. [Google Scholar] [CrossRef]
  29. Shaykhiev, R.; Otaki, F.; Bonsu, P.; Dang, D.T.; Teater, M.; Strulovici-Barel, Y.; Salit, J.; Harvey, B.G.; Crystal, R.G. Cigarette smoking reprograms apical junctional complex molecular architecture in the human airway epithelium in vivo. Cell. Mol. Life Sci. 2011, 68, 877–892. [Google Scholar] [CrossRef]
  30. Lai, H.Y.; Rogers, D.F. Mucus hypersecretion in asthma: intracellular signalling pathways as targets for pharmacotherapy. Curr. Opin. Allergy Clin. Immunol. 2010, 10, 67–76. [Google Scholar] [CrossRef]
  31. Heijink, I.H.; Brandenburg, S.M.; Postma, D.S.; van Oosterhout, A.J.M. Cigarette smoke impairs airway epithelial barrier function and cell-cell contact recovery. Eur. Respir. J. 2012, 39, 419–428. [Google Scholar] [CrossRef]
  32. Lapperre, T.S.; Sont, J.K.; van Schadewijk, A.; Gosman, M.M.E.; Postma, D.S.; Bajema, I.M.; Timens, W.; Maua, T.; Hiemstra, P.S.; the GLUCOLD Study group. Smoking cessation and bronchial epithelial remodelling in COPD: a cross-sectional study. Respir. Res. 2007, 8, 85. [Google Scholar] [CrossRef] [Green Version]
  33. Voynow, J.A.; Rubin, B.K. Mucins, mucus, and sputum. Chest 2009, 135, 505–512. [Google Scholar] [CrossRef]
  34. Hurst, J.R.; Vestbo, J.; Anzueto, A.; Locantore, N.; Mullerova, H.; Tal-Singer, R.; Miller, B.; Lomas, D.A.; Agusti, A.; Macnee, W.; Calverley, P.; Rennard, S.; Wouters, E.F.; Wedzicha, J.A. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N. Engl. J. Med. 2010, 363, 1128–1138. [Google Scholar]
  35. Hopkins, A.L.; Groom, C.R. The druggable genome. Nat. Rev. Drug Discov. 2002, 1, 727–730. [Google Scholar] [CrossRef]
  36. Pierce, K.L.; Premont, R.T.; Lefkowitz, R.J. Seven-transmembrane receptors. Nat. Rev. Mol. Cell Biol. 2002, 3, 639–650. [Google Scholar]
  37. Cooper, P.R.; Kurten, R.C.; Zhang, J.; Nicholls, D.J.; Dainty, I.A.; Panettieri, R.A. Formoterol and salmeterol induce a similar degree of beta2-adrenoceptor tolerance in human small airways but via different mechanisms. Br. J. Pharmacol. 2011, 163, 521–532. [Google Scholar]
  38. Penn, R.B. Embracing emerging paradigms of G protein-coupled receptor agonism and signaling to address airway smooth muscle pathobiology in asthma. Naunyn Schmiedebergs Arch. Pharmacol. 2008, 378, 149–169. [Google Scholar] [CrossRef]
  39. Bateman, E.D.; Hurd, S.S.; Barnes, P.J.; Bousquet, J.; Drazen, J.M.; FitzGerald, M.; Gibson, P.; Ohta, K.; O'Byrne, P.; Pedersen, S.E.; Pizzichini, E.; Sullivan, S.D.; Wenzel, S.E.; Zar, H.J. Global strategy for asthma management and prevention: GINA executive summary. Eur. Respir. J. 2008, 31, 143–178. [Google Scholar] [CrossRef]
  40. Penn, R.B. Agonizing over agonism: should asthmatics turn their beta-receptors on or off? Proc. Natl. Acad. Sci. USA 2009, 106, 2095–2096. [Google Scholar] [CrossRef]
  41. Yan, H.; Deshpande, D.A.; Misior, A.M.; Miles, M.C.; Saxena, H.; Riemer, E.C.; Pascual, R.M.; Panettieri, R.A.; Penn, R.B. Anti-mitogenic effects of {beta}-agonists and PGE2 on airway smooth muscle are PKA dependent. FASEB J. 2011, 25, 389–397. [Google Scholar]
  42. Bauman, K.A.; Wettlaufer, S.H.; Okunishi, K.; Vannella, K.M.; Stoolman, J.S.; Huang, S.K.; Courey, A.J.; White, E.S.; Hogaboam, C.M.; Simon, R.H.; Toews, G.B.; Sisson, T.H.; Moore, B.B.; Peters-Golden, M. The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice. J. Clin. Invest. 2010, 120, 1950–1960. [Google Scholar]
  43. Huang, S.K.; Fisher, A.S.; Scruggs, A.M.; White, E.S.; Hogaboam, C.M.; Richardson, B.C.; Peters-Golden, M. Hypermethylation of PTGER2 confers prostaglandin E2 resistance in fibrotic fibroblasts from humans and mice. Am. J. Pathol. 2010, 177, 2245–2255. [Google Scholar]
  44. Stumm, C.L.; Wettlaufer, S.H.; Jancar, S.; Peters-Golden, M. Airway remodeling in murine asthma correlates with a defect in PGE2 synthesis by lung fibroblasts. Am. J. Physiol Lung Cell Mol. Physiol. 2011, 301, L636–L644. [Google Scholar] [CrossRef]
  45. Sears, M.R. Safe use of long-acting b-agonists: what have we learnt? Expert. Opin. Drug. Saf. 2012, 10, 767–778. [Google Scholar] [CrossRef]
  46. Bos, J.L. Epac: a new cAMP target and new avenues in cAMP research. Nat. Rev. Mol. Cell. Biol. 2003, 4, 733–738. [Google Scholar] [CrossRef]
  47. Gloerich, M.; Bos, J.L. Epac: defining a new mechanism for cAMP action. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 355–375. [Google Scholar] [CrossRef]
  48. Whalen, E.J.; Rajagopal, S.; Lefkowitz, R.J. Therapeutic potential of beta-arrestin- and G protein-biased agonists. Trends Mol. Med. 2011, 17, 126–139. [Google Scholar]
  49. Rajagopal, S.; Rajagopal, K.; Lefkowitz, R.J. Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat Rev. Drug Discov. 2010, 9, 373–386. [Google Scholar]
  50. Reiter, E.; Ahn, S.; Shukla, A.K.; Lefkowitz, R.J. Molecular mechanisms of b-arrestin-biased agonism at seven-transmembrane receptors. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 179–197. [Google Scholar] [CrossRef]
  51. Calebiro, D.; Nikolaev, V.O.; Gagliani, M.C.; de, F.T.; Dees, C.; Tacchetti, C.; Persani, L.; Lohse, M.J. Persistent cAMP-signals triggered by internalized G-protein-coupled receptors. PLoS. Biol. 2009, 7, 1–8. [Google Scholar]
  52. Ferrandon, S.; Feinstein, T.N.; Castro, M.; Wang, B.; Bouley, R.; Potts, J.T.; Gardella, T.J.; Vilardaga, J.-P. Sustained cyclic AMP production by parathroid hormone receptor endocytosis. Nat. Chem. Biol. 2009, 5, 734–742. [Google Scholar] [CrossRef]
  53. Calebiro, D.; Nikolaev, V.O.; Lohse, M.J. Imaging of persistent cAMP signaling by internalized G protein-coupled receptors. J. Mol. Endocrinol. 2010, 45, 1–8. [Google Scholar]
  54. Murphy, J.E.; Padilla, B.E.; Hasdemir, B.; Cottrell, G.S.; Bunnett, N.W. Endosomes: a legitimate platform for the signaling train. Proc. Natl. Acad. Sci. USA 2009, 106, 17615–17622. [Google Scholar]
  55. Platta, H.W.; Stenmark, H. Endocytosis and signaling. Curr. Opin. Cell Biol. 2011, 23, 393–403. [Google Scholar] [CrossRef]
  56. Puthenveedu, M.A.; Lauffer, B.; Temkin, P.; Vistein, R.; Carlton, P.; Thorn, K.; Taunton, J.; Weiner, O.D.; Parton, R.G.; von Zastrow, M. Sequence-dependent sorting of recycling proteins by actin-stabilized endosomal microdomains. Cell 2010, 143, 761–773. [Google Scholar] [CrossRef]
  57. Kendall, R.T.; Strungs, E.G.; Rachidi, S.M.; Lee, M.-H.; El-Shewy, H.M.; Luttrell, D.K.; Janech, M.G.; Luttrell, L.M. The b-arrestin pathway-selective type 1A angiotensin receptor (AT1a) agonist [Sar1, IL4, Ile8] angiotensin II regulates a robust G protein-independent signaling network. J. Biol. Chem. 2011, 286, 19880–19891. [Google Scholar]
  58. Walters, R.W.; Shukla, A.K.; Kovacs, J.J.; Violin, J.D.; DeWire, S.M.; Lam, C.M.; Chen, J.R.; Muehlbauer, M.J.; Whalen, E.J.; Lefkowitz, R.J. b-arrestin 1 mediates nicotinic acid-induced flushing, but not its antilipolytic effects, in mice. J. Clin. Invest. 2009, 119, 1312–1321. [Google Scholar]
  59. Rajagopal, S.; Ahn, S.; Gowen-MacDonald, W.; Lam, C.M.; DeWire, S.M.; Violin, J.D.; Lefkowitz, R.J. Quantifying ligand bias at seven-transmembrane receptors. Mol. Pharmacol. 2011, 80, 367–377. [Google Scholar] [CrossRef]
  60. Nguyen, L.P.; Lin, R.; Parra, S.; Omoluabi, O.; Hanania, N.A.; Tuvim, M.J.; Knoll, B.J.; Dickey, B.F.; Bond, R.A. Beta2-adrenoceptor signaling is required for the development of an asthma phenotype in a murine model. Proc. Natl. Acad. Sci. USA 2009, 106, 2435–2440. [Google Scholar]
  61. Billington, C.K.; Hall, I.P. Novel cyclic AMP Signalling Paradigms: Therapeutic Implications for Airway Disease. Br. J. Pharmacol. 2011, 166, 401–410. [Google Scholar] [CrossRef]
  62. Walker, J.K.; Penn, R.B.; Hanania, N.A.; Dickey, B.F.; Bond, R.A. New perspectives regarding beta(2) -adrenoceptor ligands in the treatment of asthma. Br. J. Pharmacol. 2011, 163, 18–28. [Google Scholar] [CrossRef]
  63. Lovgren, A.K.; Kovacs, J.J.; Xie, T.; Potts, E.N.; Li, Y.; Foster, W.M.; Liang, J.; Meltzer, E.B.; Jiang, D.; Lefkowitz, R.J.; Noble, P.W. beta-arrestin deficiency protects against pulmonary fibrosis in mice and prevents fibroblast invasion of extracellular matrix. Sci. Transl. Med. 2011, 3, 74–ra23. [Google Scholar] [CrossRef]
  64. Manson, M.E.; Corey, D.A.; White, N.M.; Kelley, T.J. cAMP-mediated regulation of cholesterol accumulation in cystic fibrosis and Niemann-Pick type C cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2008, 295, L809–L819. [Google Scholar] [CrossRef]
  65. Li, J.; Ghio, A.J.; Cho, S.H.; Brinckerhoff, C.E.; Simon, S.A.; Liedtke, W. Diesel exhaust particles activate the matrix-metalloproteinase-1 gene in human bronchial epithelia in a beta-arrestin-dependent manner via activation of RAS. Environ. Health Perspect. 2009, 117, 400–409. [Google Scholar]
  66. Nobles, K.N.; Xiao, K.; Ahn, S.; Shukla, A.K.; Lam, C.M.; Rajagopal, S.; Strachan, R.T.; Huang, T.Y.; Bressler, E.A.; Hara, M.R.; Shenoy, S.K.; Gygi, S.P.; Lefkowitz, R.J. Distinct phosphorylation sites on the b(2)-adrenergic receptor establish a barcode that encodes differential functions of b-arrestin. Sci. Signal. 2011, 4, ra51. [Google Scholar] [CrossRef]
  67. Khasai, A.W.; Xiao, K.; Rajagopal, S.; Ahn, S.; Shukla, A.K.; Sun, J.; Oas, T.G.; Lefkowitz, R.J. Multiple ligand-specific conformations of the b2-adrenergic receptor. Nat. Chem. Biol. 2011, 7, 692–700. [Google Scholar] [CrossRef]
  68. Xiao, K.; Sun, J.; Kim, J.; Rajagopal, S.; Zhai, B.; Villen, J.; Haas, W.; Kovacs, J.J.; Shukla, A.K.; Hara, M.R.; Hernandez, M.; Lachmann, A.; Zhao, S.; Lin, Y.; Cheng, Y.; Mizuno, K.; Maáyan, A.; Gygi, S.P.; Lefkowitz, R.J. Global phosphorylation analysis of b-arrestin-mediated signaling downstream of a seven transmembrane receptor (7TMR). Proc. Natl. Acad. Sci. US A 2010, 107, 15299–15304. [Google Scholar]
  69. Berthouze, M.; Venkataramanan, V.; Li, Y.; Shenoy, S.K. The deubiquitinases USP33 and USP20 coordinate b2 adrenergic receptor recycling and resensitization. EMBO J. 2009, 28, 1684–1696. [Google Scholar] [CrossRef]
  70. Shenoy, S.K.; Modi, A.S.; Shukla, A.K.; Xiao, K.; Berthouze, M.; Ahn, S.; Wilkinson, K.D.; Miller, W.E.; Lefkowitz, R.J. b-Arrestin-dependent signaling and trafficking of 7-transmembrane receptor is reciprocally regulated by the deubititinase USP33 and the E3 ligase Mdm2. Proc. Natl. Acad. Sci. USA 2009, 106, 6650–6655. [Google Scholar]
  71. Billington, C.K.; Hall, I.P. Real time analysis of beta(2)-adrenoceptor-mediated signaling kinetics in human primary airway smooth muscle cells reveals both ligand and dose dependent differences. Respir. Res. 2011, 12, 89. [Google Scholar] [CrossRef]
  72. Lamyel, F.; Warnken-Uhlich, M.; Seemann, W.K.; Mohr, K.; Kostenis, E.; Ahmedat, A.S.; Smit, M.; Gosens, R.; Meurs, H.; Miller-Larsson, A.; Racke, K. The beta2-subtype of adrenoceptors mediates inhibition of pro-fibrotic events in human lung fibroblasts. Naunyn Schmiedebergs Arch. Pharmacol. 2011, 384, 133–145. [Google Scholar] [CrossRef]
  73. Giembycz, M.A.; Newton, R. Beyond the dogma: novel beta2-adrenoceptor signalling in the airways. Eur. Respir. J. 2006, 27, 1286–1306. [Google Scholar] [CrossRef]
  74. Lynch, M.J.; Baillie, G.S.; Mohamed, A.; Li, X.; Maisonneuve, C.; Klussmann, E.; Van, H.G.; Houslay, M.D. RNA silencing identifies PDE4D5 as the functionally relevant cAMP phosphodiesterase interacting with beta arrestin to control the protein kinase A/AKAP79-mediated switching of the beta2-adrenergic receptor to activation of ERK in HEK293B2 cells. J. Biol. Chem. 2005, 280, 33178–33189. [Google Scholar]
  75. Lin, F.; Wang, H.; Malbon, C.C. Gravin-mediated formation of signaling complexes in beta2-adrenergic receptor desensitization and resensitization. J. Biol. Chem. 2000, 275, 19025–19034. [Google Scholar]
  76. Anderson, G.P. Current issues with beta2-adrenoceptor agonists: pharmacology and molecular and cellular mechanisms. Clin. Rev. Allergy Immunol. 2006, 31, 119–130. [Google Scholar] [CrossRef]
  77. Cooper, D.M. Compartmentalization of adenylate cyclase and cAMP signalling. Biochem. Soc Trans. 2005, 33, 1319–1322. [Google Scholar] [CrossRef]
  78. Delint-Ramirez, I.; Willoughby, D.; Hammond, G.V.R.; Ayling, L.J.; Cooper, D.M.F. Palmitylation targets AKAP79 protein to lipid rafts and promotes its regulation of calcium-sensitive adenylyl cyclase type 8. J. Biol. Chem. 2011, 286, 32962–32975. [Google Scholar]
  79. Dodge-Kafka, K.L.; Soughayer, J.; Pare, G.C.; Carlisle Michel, J.J.; Langeberg, L.K.; Kapiloff, M.S.; Scott, J.D. The protein kinase A anchoring protein mAKAP coordinates two integrated cAMP effector pathways. Nature 2005, 437, 574–578. [Google Scholar] [CrossRef]
  80. Nijholt, I.M.; Dolga, A.M.; Ostroveanu, A.; Luiten, P.G.; Schmidt, M.; Eisel, U.L. Neuronal AKAP150 coordinates PKA and Epac-mediated PKB/Akt phosphorylation. Cell Signal. 2008, 20, 1715–1724. [Google Scholar] [CrossRef]
  81. Cohen, P. Protein kinases--the major drug targets of the twenty-first century? Nat. Rev. Drug Discov. 2002, 1, 309–315. [Google Scholar] [CrossRef]
  82. Tasken, K.; Aandahl, E.M. Localized effects of cAMP mediated by distinct routes of protein kinase A. Physiol Rev. 2004, 84, 137–167. [Google Scholar] [CrossRef]
  83. Zambon, A.C.; Zhang, L.; Minovitsky, S.; Kanter, J.R.; Prabhakar, S.; Salomonis, N.; Vranizan, K.; Dubchak, I.; Conklin, B.R.; Insel, P.A. Gene expression patterns define key transcriptional events in cell-cycle regulation by cAMP and protein kinase A. Proc. Natl. Acad. Sci. USA 2005, 102, 8561–8566. [Google Scholar]
  84. de Rooij, J.; Zwartkruis, F.J.; Verheijen, M.H.; Cool, R.H.; Nijman, S.M.; Wittinghofer, A.; Bos, J.L. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature 1998, 396, 474–477. [Google Scholar] [CrossRef]
  85. Kawasaki, H.; Springett, G.M.; Mochizuki, N.; Toki, S.; Nakaya, M.; Matsuda, M.; Housman, D.E.; Graybiel, A.M. A family of cAMP-binding proteins that directly activate Rap1. Science 1998, 282, 2275–2279. [Google Scholar] [CrossRef]
  86. Krugmann, S.; Williams, R.; Stephens, L.; Hawkins, P.T. ARAP3 is a PI3K- and RAP-regulated GAP for RhoA. Curr. Biol. 2004, 14, 1380–1384. [Google Scholar] [CrossRef]
  87. Roscioni, S.S.; Maarsingh, H.; Elzinga, C.R.; Schuur, J.; Menzen, M.; Halayko, A.J.; Meurs, H.; Schmidt, M. Epac as a novel effector of airway smooth muscle relaxation. J. Cell Mol. Med. 2011, 15, 1551–1562. [Google Scholar] [CrossRef]
  88. Zieba, B.J.; Artamonov, M.V.; Jin, L.; Momotani, K.; Ho, R.; Franke, A.S.; Neppl, R.L.; Stevenson, A.S.; Khromov, A.S.; Chrzanowska-Wodnicka, M.; Somlyo, A.V. The cAMP-responsive Rap1 guanine nucleotide exchange factor, Epac, induces smooth muscle relaxation by down-regulation of RhoA activity. J. Biol. Chem. 2011, 286, 16681–16692. [Google Scholar]
  89. Li, Y.; Asuri, S.; Rebhun, J.F.; Castro, A.F.; Paranavitana, N.C.; Quilliam, L.A. The RAP1 guanine nucleotide exchange factor Epac2 couples cyclic AMP and Ras signals at the plasma membrane. J. Biol. Chem. 2006, 281, 2506–2514. [Google Scholar]
  90. Lopez De Jesus, M.; Stope, M.B.; Oude Weernink, P.A.; Mahlke, Y.; Borgermann, C.; Ananaba, V.N.; Rimmbach, C.; Rosskopf, D.; Michel, M.C.; Jakobs, K.H.; Schmidt, M. Cyclic AMP-dependent and Epac-mediated activation of R-Ras by G protein-coupled receptors leads to phospholipase D stimulation. J. Biol. Chem. 2006, 281, 21837–21847. [Google Scholar]
  91. Oestreich, E.A.; Wang, H.; Malik, S.; Kaproth-Joslin, K.A.; Blaxall, B.C.; Kelley, G.G.; Dirksen, R.T.; Smrcka, A.V. Epac-mediated activation of phospholipase C(epsilon) plays a critical role in beta-adrenergic receptor-dependent enhancement of Ca2+ mobilization in cardiac myocytes. J. Biol. Chem. 2007, 282, 5488–5495. [Google Scholar]
  92. Schmidt, M.; Evellin, S.; Weernink, P.A.; von, D.F.; Rehmann, H.; Lomasney, J.W.; Jakobs, K.H. A new phospholipase-C-calcium signalling pathway mediated by cyclic AMP and a Rap GTPase. Nat. Cell Biol. 2001, 3, 1020–1024. [Google Scholar] [CrossRef]
  93. Han, L.; Stope, M.B.; de Jesus, M.L.; Oude Weernink, P.A.; Urban, M.; Wieland, T.; Rosskopf, D.; Mizuno, K.; Jakobs, K.H.; Schmidt, M. Direct stimulation of receptor-controlled phospholipase D1 by phospho-cofilin. EMBO J. 2007, 26, 4189–4202. [Google Scholar] [CrossRef]
  94. Wang, C.; Gu, Y.; Li, G.W.; Huang, L.Y. A critical role of the cAMP sensor Epac in switching protein kinase signalling in prostaglandin E2-induced potentiation of P2X3 receptor currents in inflamed rats. J. Physiol. 2007, 584, 191–203. [Google Scholar] [CrossRef]
  95. Roscioni, S.S.; Kistemaker, L.E.; Menzen, M.H.; Elzinga, C.R.; Gosens, R.; Halayko, A.J.; Meurs, H.; Schmidt, M. PKA and Epac cooperate to augment bradykinin-induced interleukin-8 release from human airway smooth muscle cells. Respir. Res. 2009, 10, 88. [Google Scholar] [CrossRef]
  96. Roscioni, S.S.; Dekkers, B.G.; Prins, A.G.; Menzen, M.H.; Meurs, H.; Schmidt, M.; Maarsingh, H. cAMP inhibits modulation of airway smooth muscle phenotype via the exchange protein activated by cAMP (Epac) and protein kinase A. Br. J. Pharmacol. 2011, 162, 193–209. [Google Scholar] [CrossRef]
  97. Keiper, M.; Stope, M.B.; Szatkowski, D.; Bohm, A.; Tysack, K.; Vom, D.F.; Saur, O.; Oude Weernink, P.A.; Evellin, S.; Jakobs, K.H.; Schmidt, M. Epac- and Ca2+ -controlled activation of Ras and extracellular signal-regulated kinases by Gs-coupled receptors. J. Biol. Chem. 2004, 279, 46497–46508. [Google Scholar]
  98. Kiermayer, S.; Biondi, R.M.; Imig, J.; Plotz, G.; Haupenthal, J.; Zeuzem, S.; Piiper, A. Epac activation converts cAMP from a proliferative into a differentiation signal in PC12 cells. Mol. Biol. Cell 2005, 16, 5639–5648. [Google Scholar] [CrossRef]
  99. Grandoch, M.; López de Jesús, M.; Oude Weernink, P.A.; Weber, A.-A.; Jakobs, K.H.; Schmidt, M. B cell receptor-induced growth arrest and apoptosis in WEHI-231 immature B lymphoma cells involve cyclic AMP and Epac proteins. Cell Signal. 2009, 4, 609–621. [Google Scholar]
  100. Kwak, H.J.; Park, K.M.; Choi, H.E.; Chung, K.S.; Lim, H.J.; Park, H.Y. PDE4 inhibitor, roflumilast protects cardiomyocytes against NO-induced apoptosis via activation of PKA and Epac dual pathways. Cell Signal. 2008, 20, 803–814. [Google Scholar] [CrossRef]
  101. Mei, F.C.; Qiao, J.; Tsygankova, O.M.; Meinkoth, J.L.; Quilliam, L.A.; Cheng, X. Differential signaling of cyclic AMP: opposing effects of exchange protein directly activated by cyclic AMP and cAMP-dependent protein kinase on protein kinase B activation. J. Biol. Chem. 2002, 277, 11497–11504. [Google Scholar]
  102. Misra, U.K.; Pizzo, S.V. Coordinate regulation of forskolin-induced cellular proliferation in macrophages by protein kinase A/cAMP-response element-binding protein (CREB) and Epac1-Rap1 signaling: effects of silencing CREB gene expression on Akt activation. J. Biol. Chem. 2005, 280, 38276–38289. [Google Scholar] [CrossRef]
  103. Misra, U.K.; Kaczowka, S.; Pizzo, S.V. The cAMP-activated GTP exchange factor, Epac1 upregulates plasma membrane and nuclear Akt kinase activities in 8-CPT-2-O-Me-cAMP-stimulated macrophages: Gene silencing of the cAMP-activated GTP exchange Epac1 prevents 8-CPT-2-O-Me-cAMP activation of Akt activity in macrophages. Cell Signal. 2008, 20, 1459–1470. [Google Scholar] [CrossRef]
  104. Hewer, R.C.; Sala-Newby, G.B.; Wu, Y.-J.; Newby, A.C.; Bond, M. PKA and Epac synergistically inhibit smooth muscle cell proliferation. J. Mol. Cell. Cardiol. 2011, 50, 87–98. [Google Scholar] [CrossRef]
  105. Fuld, S.; Borland, G.; Yarwood, S.J. Elevation of cyclic AMP in Jurkat T-cells provokes distinct transcriptional responses through the protein kinase A (PKA) and exchange protein activated by cyclic AMP (EPAC) pathways. Exp. Cell Res. 2005, 309, 161–173. [Google Scholar] [CrossRef]
  106. Scheibner, K.A.; Boodoo, S.; Collins, S.; Black, K.E.; Chan-Li, Y.; Zarek, P.; Powell, J.D.; Horton, M.R. The adenosine a2a receptor inhibits matrix-induced inflammation in a novel fashion. Am. J. Respir. Cell Mol. Biol. 2009, 40, 251–259. [Google Scholar]
  107. Oldenburger, A.; Roscioni, S.S.; Jansen, E.; Menzen, M.H.; Halayko, A.J.; Timens, W.; Meurs, H.; Maarsingh, H.; Schmidt, M. Anti-inflammatory role of the cAMP effectors Epac and PKA: implications in chronic obstructive pulmonary disease. PLoS One 2012, e31574. [Google Scholar]
  108. Borland, G.; Smith, B.O.; Yarwood, S.J. EPAC proteins transduce diverse cellular actions of cAMP. Br. J. Pharmacol. 2009, 158, 70–86. [Google Scholar] [CrossRef]
  109. Grandoch, M.; Roscioni, S.S.; Schmidt, M. The role of Epac proteins, novel cAMP mediators, in the regulation of immune, lung and neuronal functions. Br. J. Pharmacol. 2010, 159, 265–284. [Google Scholar] [CrossRef]
  110. Roscioni, S.S.; Elzinga, C.R.; Schmidt, M. Epac: effectors and biological functions. Naunyn Schmiedebergs Arch. Pharmacol. 2008, 377, 345–357. [Google Scholar] [CrossRef]
  111. Schmidt, M.; Sand, C.; Jakobs, K.H.; Michel, M.C.; Oude Weernink, P.A. Epac and the cardiovascular system. Curr. Opin. Pharmacol. 2007, 7, 193–200. [Google Scholar] [CrossRef]
  112. Lorenowicz, M.J.; Fernandez-Borja, M.; Hordijk, P.L. cAMP signaling in leukocyte transendothelial migration. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 1014–1022. [Google Scholar] [CrossRef]
  113. Billington, C.K.; Hall, I.P.; Mundell, S.J.; Parent, J.L.; Panettieri, R.A., Jr.; Benovic, J.L.; Penn, R.B. Inflammatory and contractile agents sensitize specific adenylyl cyclase isoforms in human airway smooth muscle. Am. J. Respir. Cell Mol. Biol. 1999, 21, 597–606. [Google Scholar]
  114. Bogard, A.S.; Xu, C.; Ostrom, R.S. Human bronchial smooth muscle cells express adenylyl cyclase isoforms 2, 4, and 6 in distinct membrane microdomains. J. Pharmacol Exp. Ther. 2011, 337, 209–217. [Google Scholar] [CrossRef]
  115. Xu, D.; Isaacs, C.; Hall, I.P.; Emala, C.W. Human airway smooth muscle expresses 7 isoforms of adenylyl cyclase: a dominant role for isoform V. Am. J. Physiol Lung Cell Mol. Physiol 2001, 281, L832–L843. [Google Scholar]
  116. Le Jeune, I.; Shepherd, M.; Van, H.G.; Houslay, M.D.; Hall, I.P. Cyclic AMP-dependent transcriptional up-regulation of phosphodiesterase 4D5 in human airway smooth muscle cells. Identification and characterization of a novel PDE4D5 promoter. J. Biol. Chem. 2002, 277, 35980–35989. [Google Scholar]
  117. Horvat, S.J.; Deshpande, D.A.; Yan, H.; Panettieri, R.A.; Codina, J.; DuBose, T.D.; Xin, W.; Rich, T.C.; Penn, R.B. A-kinase anchoring proteins regulate compartmenalized cAMP signaling in airway smooth muscle. FASEB J. 2012, 9, 3670–3679. [Google Scholar]
  118. Liu, X.; Ostrom, R.S.; Insel, P.A. cAMP-elevating agents and adenylyl cyclase overexpression promote an antifibrotic phenotype in pulmonary fibroblasts. Am. J. Physiol Cell Physiol. 2004, 286, C1089–C1099. [Google Scholar]
  119. Selige, J.; Hatzelmann, A.; Dunkern, T. The differential impact of PDE4 subtypes in human lung fibroblasts on cytokine-induced proliferation and myofibroblast conversion. J. Cell Physiol. 2011, 226, 1970–1980. [Google Scholar] [CrossRef]
  120. Selige, J.; Tenor, H.; Hatzelmann, A.; Dunkern, T. Cytokine-dependent balance of mitogenic effects in primary human lung fibroblasts related to cyclic AMP signaling and phosphodiesterase 4 inhibition. J. Cell Physiol. 2010, 223, 317–326. [Google Scholar]
  121. Togo, S.; Liu, X.; Wang, X.; Sugiura, H.; Kamio, K.; Kawasaki, S.; Kobayashi, T.; Ertl, R.F.; Ahn, Y.; Holz, O.; Magnussen, H.; Fredriksson, K.; Skold, C.M.; Rennard, S.I. PDE4 inhibitors roflumilast and rolipram augment PGE2 inhibition of TGF-{beta}1-stimulated fibroblasts. Am. J. Physiol. Lung Cell Mol. Physiol. 2009, 296, L959–L969. [Google Scholar] [CrossRef]
  122. Okunishi, K.; Sisson, T.H.; Huang, S.K.; Hogaboam, C.M.; Simon, R.H.; Peters-Golden, M. Plasmin overcomes resistance to prostaglandin E2 in fibrotic lung fibroblasts by reorganizing protein kinase A signaling. J. Biol. Chem. 2011, 286, 32231–32243. [Google Scholar]
  123. Tasken, K.A.; Collas, P.; Kemmner, W.A.; Witczak, O.; Conti, M.; Tasken, K. Phosphodiesterase 4D and protein kinase a type II constitute a signaling unit in the centrosomal area. J. Biol. Chem. 2001, 276, 21999–22002. [Google Scholar]
  124. Calverley, P.M.; Rabe, K.F.; Goehring, U.M.; Kristiansen, S.; Fabbri, L.M.; Martinez, F.J. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet 2009, 374, 685–694. [Google Scholar] [CrossRef]
  125. Spina, D. PDE4 inhibitors: current status. Br. J. Pharmacol. 2008, 155, 308–315. [Google Scholar] [CrossRef]
  126. Michalski, J.M.; Golden, G.; Ikari, J.; Rennard, S.I. PDE4: A novel target in the treatment of chronic obstructive pulmonary disease. Clin. Pharmacol. Ther. 2011. [Google Scholar] [CrossRef]
  127. Li, Y.; Chen, L.; Kass, R.S.; Dessauer, C.W. The A-kinase anchoring protein Yotiao facilitates complex formation between adenylyl cyclase type 9 and the IKs potassium channel in heart. J Biol. Chem. 2012, 287, 29815–29824. [Google Scholar] [CrossRef]
  128. Qin, Y.; Stokman, G.; Yan, K.; Ramaiahgari, S.; Verbeek, F.; de Graauw, M.; van de Water, B.; Price, L.S. Cyclic AMP signalling protects proximal tubular epithelial cells from cisplatin-induced apoptosis via activation of Epac. Br. J. Pharmacol. 2011. [Google Scholar] [CrossRef]
  129. Roscioni, S.S. Epac as a novel regulator of airway smooth muscle phenotype and function. Potential implications in asthma and COPD. Ph.D. Degree, University of Groningen, The Netherland, 2010. [Google Scholar]
  130. Oldenburger, A.; Rijks, W.; Poppinga, W.; Roscioni, S.S.; Heijink, I.H.; Maarsingh, H.; Schmidt, M. Interaction between cigarette smoke and cyclic AMP signaling in human bronchial epithelial function. FASEB J. 2011, 25, 659.13. [Google Scholar]
  131. Dent, G.; White, S.R.; Tenor, H.; Bodtke, K.; Schudt, C.; Leff, A.R.; Magnussen, H.; Rabe, K.F. Cyclic nucleotide phosphodiesterase in human bronchial epithelial cells: characterization of isoenyzmes and functional effects of PDE inhibitors. Pulm. Pharmacol. Ther. 1998, 11, 47–56. [Google Scholar] [CrossRef]
  132. Mata, M.; Sarria, B.; Buenestado, A.; Cortijo, J.; Cerda, M.; Morcillo, E.J. Phosphodiesterase 4 inhibition decreases MUC5AC expression induced by epidermal growth factor in human airway epithelial cells. Thorax 2005, 60, 144–152. [Google Scholar] [CrossRef]
  133. Barnes, A.P.; Livera, G.; Huang, P.; Sun, C.; O'Neal, W.K.; Conti, M.; Stutts, M.J.; Milgram, S.L. Phosphodiesterase 4D forms a cAMP diffusion barrier at the apical membrane of the airway epithelium. J. Biol. Chem. 2005, 280, 7997–8003. [Google Scholar]
  134. Penmatsa, H.; Zhang, W.; Yarlagadda, S.; Li, C.; Conoley, V.G.; Yue, J.; Bahouth, S.W.; Buddington, R.K.; Zhang, G.; Nelson, D.J.; Sonecha, M.D.; Manganiello, V.; Wine, J.J.; Naren, A.P. Compartmentalized cyclic adenosine 3', 5'-monophosphate at the plasma membrane clusters PDE3A and cystic fibrosis transmembrane conductance regulator into microdomains. Mol. Biol Cell 2010, 21, 1097–1110. [Google Scholar]
  135. Halpin, D.M. ABCD of the phosphodiesterase family: interaction and differential activity in COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 2008, 3, 543–561. [Google Scholar]
  136. Small, K.M.; Brown, K.M.; Theiss, C.T.; Seman, C.A.; Weiss, S.T.; Liggett, S.B. An Ile to Met polmorphism in the catalytic domain of adenylyl cycalse type 9 confers reduced beta2-adrenergic receptor stimulation. Pharmacogenetics 2003, 13, 535–541. [Google Scholar] [CrossRef]
  137. Tantisira, K.G.; Small, K.M.; Litonjua, A.A.; Weiss, S.T.; Liggett, S.B. Molecular properties and pharmacogenetics of a polymorphism of adenylyl cyclase type 9 in asthma: interaction between b-agonist and corticosteroid pathways. Hum. Mol. Genet. 2005, 14, 1671–1677. [Google Scholar] [CrossRef]
  138. Jourdan, K.B.; Mason, N.A.; Long, L.; Philips, P.G.; Wilkins, M.R.; Morrell, N.W. Characterization of adenylyl cyclase isoforms in rat peripheral pulmonary arteries. Am. J. Physiol Lung Cell Mol. Physiol. 2001, 280, L1359–L1369. [Google Scholar]
  139. Wang, Y.; Lam, C.S.; Wu, F.; Wang, W.; Duan, Y.; Huang, P. Regulation of CFTR channels by HCO(3)--sensitive soluble adenylyl cyclase in human airway epithelial cells. Am. J. Physiol. Cell. Physiol. 2005, 289, C1145–C1151. [Google Scholar] [CrossRef]
  140. Blazac, F.; Avolio, M.; Degani, S.; Kaverina, I.; Torti, M.; Silengo, L.; Small, J.V.; Retta, S.F. E-cadherin endocytosis regulates the activity of Rap1: a traffic light GTPase at the croosraods between cadherin and integrin function. J. Cell Sci. 2005, 118, 4765–4783. [Google Scholar] [CrossRef]
  141. Hage, B.; Meinel, K.; Baum, I.; Giehl, K.; Menke, A. Rac1 activation inhibits E-cadherin-mediated adherens junctions via binding to IQGAP1 in pancreatic carcinoma cells. Cell Commun. Signal. 2009, 7, 23. [Google Scholar] [CrossRef]
  142. Smutny, M.; Cox, L.H.; Leerberg, J.M.; Kovacs, E.M.; Conti, M.A.; Ferguson, C.; Hamilton, N.A.; Parton, R.G.; Adelstein, R.S.; Yap, A.S. Myosin II isoforms identify distinct functional modules that support integrity of the epithelial zonula adherens. Nat. Cell Biol. 2010, 12, 696–702. [Google Scholar] [CrossRef]
  143. Jeong, H.-W.; Li, Z.; Brown, M.D.; Sacks, D.B. IQGAP1 binds Rap1 and modulates its activity. J. Biol. Chem. 2007, 282, 20752–20762. [Google Scholar] [CrossRef]
  144. Kuroda, S.; Fukata, M.; Nakagawa, M.; Fuji, K.; Nakmura, T.; Ookubo, T.; Izawa, I.; Nagase, T.; Nomura, N.; Tani, H.; Shoji, I.; Matsuura, Y.; Ynoehara, S.; Kaibuchi, K. Role of IQGAP1, a target of the small GTPases Cdc42 and Rac1, in regulation of E-cadherin mediated cell-cell adhesion. Science 1998, 281, 832–835. [Google Scholar] [CrossRef]
  145. Noritake, J.; Watanbe, T.; Sato, K.; Wang, S.; Kaibuchi, K. IQGAP1: a key regulator of adhesion and migration. J. Cell Sci. 2005, 118, 2085–2092. [Google Scholar] [CrossRef]
  146. Birukova, A.A.; Burdette, D.; Moldobaeva, N.; Xing, J.; Fu, P.; Birukov, K.G. Rac GTPase is a hub for protein kinase A and epac signaling in endothelial barrier protection by cAMP. Microvascul. Res. 2010, 79, 128–138. [Google Scholar] [CrossRef]
  147. Rampersad, S.N.; Ovens, J.D.; Huston, E.; Umana, M.B.; Wilson, L.S.; Netherton, S.J.; Lynch, M.J.; Baillie, G.S.; Houslay, M.D.; Maurice, D.H. Cyclic AMP phosphodieseterase 4D (PDE4D) tethers EPAC1 in a vascular endothelial cadherin (VE-Cad)-based signaling complex and controls cAMP-mediated vascular permeabilbity. J. Biol. Chem. 2010, 285, 33614–33622. [Google Scholar]
  148. Sehrawat, S.; Cullere, X.; Patel, S.; Italiano, J.; Mayadas, T.N. Role of Epac1, an exchange factor for rap GTPases, in endothelial microtubule dynamics and barrier function. Mol. Biol. Cell 2008, 19, 1261–1270. [Google Scholar]
  149. Sehrawat, S.; Ernandez, T.; Cullere, X.; Takahashi, M.; Ono, Y.; Komarova, Y.; Mayadas, T.N. AKAP9 regulation of microtubule dynamics promotes Epac1-induced endothelial barrier properties. Blood 2011, 117, 708–718. [Google Scholar] [CrossRef]
  150. Diviani, D.; Soderling, J.; Scott, J.D. AKAP-Lbc anchors protein kinase A and nucleates Galpha 12-selective Rho-mediated stress fiber formation. J. Biol. Chem. 2001, 276, 44247–44257. [Google Scholar] [CrossRef]
  151. Suh, H.N.; Han, H.J. Laminin regulates mouse embryonic stem cell migration: involvement of Epac1/Rap1 and Ra1/cdc42. Am. J. Physiol. Cell Physiol. 2010, 298, C1159–C1169. [Google Scholar] [CrossRef]
  152. Netherton, S.J.; Sutton, J.A.; Wilson, L.S.; Carter, R.L.; Maurice, D.H. Both protein kinase A and exchange protein directly activated by cAMP coordinate adhesion of human vascular endothelial cells. Circ. Res. 2007, 101, 768–776. [Google Scholar] [CrossRef]
  153. Baumer, Y.; Spindler, V.; Werthmann, R.C.; Bünnemann, M.; Waschke, J. Role of Rac1 and cAMP in endothelial barrier stabilization and thrombin-induced barrier breakdown. J. Cell. Physiol. 2008, 220, 716–726. [Google Scholar]
  154. Pullar, C.E.; Grahn, J.C.; Liu, W.; Isseroff, R.R. b2-Adrenergic receptor activation delays wound healing. FASEB J. 2004, 20, 76–86. [Google Scholar]
  155. Raymond, D.R.; Wilson, L.S.; Carter, R.L.; Maurice, D.H. Numerous distinct PKA-, or Epac-based, signalling complexes allow selective phosphodiesterase 3 and phosphodiesterase 4 coordination of cell adhesion. Cell Signal. 2007, 19, 2507–2512. [Google Scholar] [CrossRef]
  156. Spindler, V.; Waschke, J. Beta-adrenergic stimulation contributes to maintenance of endothelial barrier functions under baseline conditions. Microcirculation 2011, 18, 118–127. [Google Scholar] [CrossRef]
  157. Spindler, V.; Peter, D.; Harms, G.S.; Asan, E.; Waschke, J. Ultrastructural analysis reveals cAMP-dependent enhancement of microvascular endothelial barrier functions via Rac1-mediated reorganization of intercellular junctions. Am. J. Pathol. 2011, 178, 2424–2436. [Google Scholar] [CrossRef]
  158. Schlegel, N.; Waschke, J. VASP is involved in cAMP-mediated rac 1 activation in microvascular endothelial cells. Am. J. Cell Physiol. 2009, 296, C453–C462. [Google Scholar] [CrossRef]
  159. Cole, A.L.; Subbanagounder, G.; Mukhopadhyay, S.; Berliner, J.A.; Vora, D.K. Oxidized phospholipid-induced endothelial cell/monocyte interaction is mediated by a cAMP-dependent R-Ras/PI3-kinase pathway. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1384–1390. [Google Scholar] [CrossRef]
  160. Poppinga, W.J.; Holtzer, L.J.; Skroblin, P.; Klussmann, E.; Maarsingh, H.; Schmidt, M. A-kinase anchoring proteins (AKAPs) regulate airway smooth muscle sceretory and proliferative functions. Available online: http://www.pA2online.org/abstracts/Vol10Issue1abst004P.pdf accessed on 28 November 2012.
  161. Omori, K.; Kotera, J. Overview of PDEs and their regulation. Circ. Res. 2007, 100, 309–327. [Google Scholar] [CrossRef]
  162. Purves, G.I.; Kamishima, T.; Davies, L.M.; Quayle, J.M.; Dart, C. Exchange protein activated by cAMP (Epac) mediates cAMP-dependent but protein kinase A-insensitive modulation of vascular ATP-sensitive potassium channels. J. Physiol. 2009, 587, 3639–3650. [Google Scholar] [CrossRef]
  163. Streb, J.W.; Long, X.; Lee, T.H.; Sun, Q.; Kitchen, C.M.; Georger, M.A.; Slivano, O.J.; Blaner, W.S.; Carr, D.W.; Gelman, I.H.; Miano, J.M. Retinoid-induced expression and activity of an immediate early tumor suppressor gene in vascular smooth muscle cells. PLos One 2011, 6, e18538. [Google Scholar]
  164. Murray, F.; Patel, H.H.; Suda, R.Y.; Zhang, S.; Thistlethwaite, P.A.; Yuan, J.X.; Insel, P.A. Expression and activity of cAMP phosphodiesterase isoforms in pulmonary artery smooth muscle cells from patients with pulmonary hypertension: role for PDE1. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 292, L294–L303. [Google Scholar]
  165. El-Haroun, H.; Bradbury, D.; Clayton, A.; Knox, A.J. Interleukin-1beta, transforming growth factor-beta1, and bradykinin attenuate cyclic AMP production by human pulmonary artery smooth muscle cells in response to prostacyclin analogues and prostaglandin E2 by cyclooxygenase-2 induction and downregulation of adenylyl cyclase isoforms 1, 2, and 4. Circ. Res. 2004, 94, 353–361. [Google Scholar] [CrossRef]
  166. Yokoyama, U.; Minamisawa, S.; Quan, H.; Akaike, T.; Jin, M.; Otsu, K.; Ulucan, C.; Wang, X.; Baljinnyam, E.; Takaoka, M.; Sata, M.; Ishikawa, Y. Epac1 is upregulated during neointima formation and promotes vascular smooth muscle cell migration. Am. J. Physiol. Heart. Circ. Physiol. 2008, 295, H1547–H1555. [Google Scholar] [CrossRef]
  167. Diebold, I.; Petry, A.; Djordjevic, T.; Belaiba, R.S.; Fineman, J.; Black, S.; Schreiber, C.; Fratz, S.; Hess, J.; Kietzmann, T.; Gorlach, A. Reciprocal regulation of Rac1 and PAK-1 by HIF-1alpha: a positive-feedback loop promoting pulmonary vascular remodeling. Antioxid. Redox. Signal. 2010, 13, 399–412. [Google Scholar] [CrossRef]
  168. Bailly, K.; Ridley, A.J.; Hall, S.M.; Haworth, S.G. RhoA activation by hypoxia in pulmonary arterial smooth muscle cells is age and site specific. Circ. Res. 2004, 94, 1383–1391. [Google Scholar] [CrossRef]
  169. Guilluy, C.; Eddahibi, S.; Agard, C.; Guignabert, C.; Izikki, M.; Tu, L.; Savale, L.; Humbert, M.; Fadel, E.; Adnot, S.; Loirand, G.; Pacaud, P. RhoA and Rho kinase activation in human pulmonary hypertension: role of 5-HT signaling. Am. J. Respir. Crit. Care. Med. 2009, 179, 1151–1158. [Google Scholar] [CrossRef]
  170. Haag, S.; Warnken, M.; Juergens, U.R.; Racke, K. Role of Epac1 in mediating anti-proliferative effects of prostanoid EP(2) receptors and cAMP in human lung fibroblasts. Naunyn Schmiedebergs Arch. Pharmacol. 2008, 378, 617–630. [Google Scholar] [CrossRef]
  171. Kohyama, T.; Liu, X.; Wen, F.Q.; Zhu, Y.K.; Wang, H.; Kim, H.J.; Takizawa, H.; Cieslinski, L.B.; Barnette, M.S.; Rennard, S.I. PDE4 inhibitors attenuate fibroblast chemotaxis and contraction of native collagen gels. Am. J. Respir. Cell Mol. Biol. 2002, 26, 694–701. [Google Scholar]
  172. Tufvesson, E.; Westergren-Thorsson, G. Biglycan and decorin induce morphological and cytoskeletal changes involving signalling by the small GTPases RhoA and Rac1 resulting in lung fibroblast migration. J. Cell Sci. 2003, 116, 4857–4864. [Google Scholar] [CrossRef]
  173. Huang, S.K.; Wettlaufer, S.H.; Chung, J.; Peters-Golden, M. Prostaglandin E2 inhibits specific lung fibroblast functions via selective actions of PKA and Epac-1. Am. J. Respir. Cell Mol. Biol. 2008, 39, 482–489. [Google Scholar] [CrossRef]
  174. Brudvik, K.W.; Tasken, K. Modulation of T cell immune functions by the prostaglandin E(2) - cAMP pathway in chronic inflammatory states. Br. J. Pharmacol. 2012, 166, 411–419. [Google Scholar] [CrossRef]
  175. El Din El Homasany BS; Volkov, Y.; Takahashi, M.; Ono, Y.; Keryer, G.; Delouvee, A.; Looby, E.; Long, A.; Kelleher, D. The scaffolding protein CG-NAP/AKAP450 is a critical integrating component of the LFA-1-induced signaling complex in migratory T cells. J. Immunol. 2005, 175, 7811–7818. [Google Scholar]
  176. Wang, P.; Wu, P.; Ohleth, K.M.; Egan, R.W.; Billah, M.M. Phosphodiesterase 4B2 is the predominant phosphodiesterase species and undergoes differential regulation of gene expression in human monocytes and neutrophils. Mol. Pharmacol. 1999, 56, 170–174. [Google Scholar]
  177. Smith, S.J.; Brookes-Fazakerley, S.; Donnelly, L.E.; Barnes, P.J.; Barnette, M.S.; Giembycz, M.A. Ubiquitous expression of phosphodiesterase 7A in human proinflammatory and immune cells. Am J Physiol Lung Cell Mol. Physiol. 2003, 284, L279–L289. [Google Scholar]
  178. Mosenden, R.; Tasken, K. Cyclic AMP-mediated immune regulation--overview of mechanisms of action in T cells. Cell Signal. 2011, 23, 1009–1016. [Google Scholar] [CrossRef]
  179. Chang, L.C.; Wang, C.J.; Lin, Y.L.; Wang, J.P. Expression of adenylyl cyclase isoforms in neutrophils. Biochim. Biophys. Acta 2003, 1640, 53–60. [Google Scholar] [CrossRef]
  180. Risoe, P.K.; Wang, Y.; Stuestol, J.F.; Aasen, A.O.; Wang, J.E.; Dahle, M.K. Lipopolysaccharide attenuates mRNA levels of several adenylyl cyclase isoforms in vivo. Biochim. Biophys. Acta 2007, 1772, 32–39. [Google Scholar]
  181. Han, H.; Stessin, A.; Roberts, J.; Hess, K.; Gautam, N.; Kamenetsky, M.; Lou, O.; Hyde, E.; Nathan, N.; Muller, W.A.; Buck, J.; Levin, L.R.; Nathan, C. Calcium-sensing soluble adenylyl cyclase mediates TNF signal transduction in human neutrophils. J. Exp. Med. 2005, 202, 353–361. [Google Scholar]
  182. Reedquist, K.A.; Tak, P.P. Signal transduction pathways in chronic inflammatory autoimmune disease: small GTPases. Open. Rheumatol. J. 2012, 6, 259–272. [Google Scholar]
  183. Zhang, J.; Zhu, J.; Bu, X.; Cushion, M.; Kinane, T.B.; Avraham, H.; Koziel, H. Cdc42 and RhoB activation are required for mannose receptor-mediated phagocytosis by human alveolar macrophages. Mol. Biol. Cell 2005, 16, 824–834. [Google Scholar]
  184. Pierre, S.; Eschenhagen, T.; Geisslinger, G.; Scholich, K. Capturing adenylyl cyclases as potential drug targets. Nat Rev. Drug Discov. 2009, 8, 321–335. [Google Scholar]
  185. Defer, N.; Best-Belpomme, M.; Hanoune, J. Tissue specificity and physiological relevance of various isoforms of adenylyl cyclase. Am. J. Renal Physiol. 2000, 279, F400–F416. [Google Scholar]
  186. Schmid, A.; Sutto, Z.; Nlend, M.-C.; Horvath, G.; Schmid, N.; Buck, J.; Levin, L.R.; Conner, G.E.; Fregien, N.; Salathe, M. Soluble adenylyl cyclase is localized to cilia and contributes to ciliary beat frequency regulation via production of cAMP. J. Gen. Physiol. 2007, 130, 99–109. [Google Scholar]
  187. Lopez, E.; Jarreau, P.-H.; Zana, E.; Franco-Montoya, M.-L.; Schmitz, T.; Evain-Brion, D.; Bourbon, J.; Delacourt, C.; Mehats, C. Differential expression of cyclic nucleotide phosphodiesterases 4 in developing rat lung. Develop. Dynam. 2010, 239, 2470–2478. [Google Scholar] [Green Version]
  188. Hoque, K.M.; Woodward, O.M.; van Rossum, D.B.; Zachos, N.C.; Chen, L.; Leung, G.P.; Guggino, W.B.; Guggino, S.E.; Tse, C.M. Epac1 mediates protein kinase A-independent mechanism of forskolin-activated intestinal chloride secretion. J. Gen. Physiol. 2010, 135, 43–58. [Google Scholar]
  189. Monterisi, S.; Favia, M.G.L.; Cardone, R.A.; Marzulli, D.; Reshkin, S.J.; Casavola, V.; Zaccolo, M. CFTR regulation in human airway epithelial cells requires integrity of the actin cytoskeleton and compartmentalized cAMP and PKA activity. J. Cell Sci. 2012, 125, 1106–1117. [Google Scholar]
  190. Li, C.; Krishnamurthy, P.C.; Penmatsa, H.; Marrs, K.L.; Wang, X.Q.; Zaccolo, M.; Jalink, K.; Li, M.; Nelson, D.J.; Schuetz, J.D.; Naren, A.P. Spatiotemporal coupling of cAMP transporter to CFTR chloride channel function in the gut epithelia. Cell 2007, 131, 940–951. [Google Scholar]
  191. Schmidt, M.; Oldenburger, A.; Poppinga, W.; Roscioni, S.S.; Heijink, I.H.; Timens, W.; Skroblin, P.; Klussmann, E.; Maarsingh, H. Cigarette smoke and A-kinase anchoring proteins (AKAPs) in human airway smooth muscle function. FASEB J. 2011, 25, 864.6. [Google Scholar]
  192. Patel, H.H.; Hamuro, L.L.; Chun, B.J.; Kawaraguchi, Y.; Quick, A.; Olson, G.; Insel, P.A.; Giles, W.R.; Taylor, S.S.; Roth, D.M. Disruption of protein kinase A localization using a trans-activator of transcription (TAT)-conjugated A-kinase-anchoring peptide reduces cardiac function. J. Biol. Chem. 2010, 285, 27632–27640. [Google Scholar]
  193. Christian, F.; Sazaszák, M.; Friedl, S.; Drewianka, S.; Lorenz, D.; Goncalves, A.; Furkert, J.; Vargas, C.; Schmieder, P.; Götz, F.; Zühlke, K.; Moutty, M.; Göttert, H.; Gáspàr, R.; Noack, C.; Bergmann, M.; Kass, R.; Hampel, K.; Kashin, D.; Genieser, H.-G.; Herberg, F.W.; Willoughby, D.; Cooper, D.M.F.; Baillie, G.S.; Houslay, M.D.; von Kries, J.P.; Zimmermann, B.; Rosenthal, W.; Klussmann, E. Small molecule AKAP-protein kinase A (PKA) interaction disruptors that activate PKA interfere with compartmentalized cAMP signaling in cardiac myocytes. J. Biol. Chem. 2011, 286, 9079–9096. [Google Scholar]
  194. Christensen, A.E.; Selheim, F.; de, R.J.; Dremier, S.; Schwede, F.; Dao, K.K.; Martinez, A.; Maenhaut, C.; Bos, J.L.; Genieser, H.G.; Doskeland, S.O. cAMP analog mapping of Epac1 and cAMP kinase. Discriminating analogs demonstrate that Epac and cAMP kinase act synergistically to promote PC-12 cell neurite extension. J. Biol. Chem. 2003, 278, 35394–35402. [Google Scholar]
  195. Poppe, H.; Rybalkin, S.D.; Rehmann, H.; Hinds, T.R.; Tang, X.B.; Christensen, A.E.; Schwede, F.; Genieser, H.G.; Bos, J.L.; Doskeland, S.O.; Beavo, J.A.; Butt, E. Cyclic nucleotide analogs as probes of signaling pathways. Nat. Methods 2008, 5, 277–278. [Google Scholar]
  196. Botelho, L.H.; Rothermel, J.D.; Coombs, R.V.; Jastorff, B. cAMP analog antagonists of cAMP action. Methods Enzymol. 1988, 159, 159–172. [Google Scholar]
  197. Gjertsen, B.T.; Mellgren, G.; Otten, A.; Maronde, E.; Genieser, H.G.; Jastorff, B.; Vintermyr, O.K.; McKnight, G.S.; Doskeland, S.O. Novel (Rp)-cAMPS analogs as tools for inhibition of cAMP-kinase in cell culture. Basal cAMP-kinase activity modulates interleukin-1 beta action. J. Biol. Chem. 1995, 270, 20599–20607. [Google Scholar]
  198. Holz, G.G.; Chepurny, O.G.; Schwede, F. Epac-selective cAMP analogs: new tools with which to evaluate the signal transduction properties of cAMP-regulated guanine nucleotide exchange factors. Cell Signal. 2008, 20, 10–20. [Google Scholar]
  199. Rehmann, H.; Schwede, F.; Doskeland, S.O.; Wittinghofer, A.; Bos, J.L. Ligand-mediated activation of the cAMP-responsive guanine nucleotide exchange factor Epac. J. Biol. Chem. 2003, 278, 38548–38556. [Google Scholar]
  200. Laxman, S.; Riechers, A.; Sadilek, M.; Schwede, F.; Beavo, J.A. Hydrolysis products of cAMP analogs cause transformation of Trypanosoma brucei from slender to stumpy-like forms. Proc. Natl. Acad. Sci. USA 2006, 103, 19194–19199. [Google Scholar]
  201. Shibasaki, T.; Takahashi, H.; Miki, T.; Sunaga, Y.; Matsumura, K.; Yamanaka, M.; Zhang, C.; Tamamoto, A.; Satoh, T.; Miyazaki, J.; Seino, S. Essential role of Epac2/Rap1 signaling in regulation of insulin granule dynamics by cAMP. Proc. Natl. Acad. Sci. USA 2007, 104, 19333–19338. [Google Scholar]
  202. Suzuki, S.; Yokoyama, U.; Abe, T.; Kiyonari, H.; Yamashita, N.; Kato, Y.; Kurotani, R.; Sato, M.; Okumura, S.; Ishikawa, Y. Differential roles of Epac in regulating cell death in neuronal and myocardial cells. J. Biol. Chem. 2010, 285, 24248–24259. [Google Scholar]
  203. Global Initiative for Chronic Obstructive Lung Disease. Global Strategy for Diagnosis, Management, and Prevention of COPD. 2010. Available online: http://www.goldcopd.org/ accessed on 28 November 2013.
  204. Postma, D.S.; Timens, W. Remodeling in asthma and chronic obstructive pulmonary disease. Proc. Am. Thorac. Soc. 2006, 3, 434–439. [Google Scholar] [CrossRef]
  205. Briggs, M.W.; Sacks, D.B. IQGAPs proteins are integral components of cytoskeletal regulation. EMBO Reports 2003, 4, 571–574. [Google Scholar] [CrossRef]
  206. Doherty, D.E. The pathophysiology of airway dysfunction. Am. J. Med. 2004, 117 Suppl 12A, 11S–23S. [Google Scholar]
  207. Rabe, K.F.; Hurd, S.S.; Anzueto, A.; Barnes, P.J.; Buist, S.A.; Calverley, P.; Fukuchi, Y.; Jenkins, C.; Rodriguez-Roisin, R.; van Weel, C.; Zielinski, J. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am. J. Respir. Crit Care Med. 2007, 176, 532–555. [Google Scholar] [CrossRef]
  208. Barnes, P.J.; Shapiro, S.D.; Pauwels, R.A. Chronic obstructive pulmonary disease: molecular and cellular mechanisms. Eur. Respir J. 2003, 22, 672–688. [Google Scholar] [CrossRef]
  209. Mannino, D.M. COPD: epidemiology, prevalence, morbidity and mortality, and disease heterogeneity. Chest 2002, 121, 121S–126S. [Google Scholar] [CrossRef]
  210. Shin, H.J.; Sohn, H.O.; Han, J.H.; Park, C.H.; Lee, H.S.; Hwang, K.J.; Hyun, H.C. Effect of cigarette filters on the chemical composition and in vitro biological activity of cigarette mainstream smoke. Food Chem. Toxicol. 2009, 47, 192–197. [Google Scholar] [CrossRef]
  211. Domagala-Kulawik, J. Effects of cigarette smoke on the lung and systemic immunity. J. Physiol. Pharmacol. 2008, 59 Suppl 6, 19–34. [Google Scholar]
  212. Thorley, A.J.; Tetley, T.D. Pulmonary epithelium, cigarette smoke, and chronic obstructive pulmonary disease. Int. J. Chron. Obstruct. Pulmon. Dis. 2007, 2, 409–428. [Google Scholar]
  213. Crosby, L.M.; Waters, C.M. Epithelial repair mechanisms in the lung. Am. J. Physiol Lung Cell Mol. Physiol. 2010, 298, L715–L731. [Google Scholar] [CrossRef]
  214. Kode, A.; Yang, S.R.; Rahman, I. Differential effects of cigarette smoke on oxidative stress and proinflammatory cytokine release in primary human airway epithelial cells and in a variety of transformed alveolar epithelial cells. Respir. Res. 2006, 7, 132. [Google Scholar] [CrossRef]
  215. Oenema, T.A.; Kolahian, S.; Nanninga, J.E.; Rieks, D.; Hiemstra, P.S.; Zuyderduyn, S.; Halayko, A.J.; Meurs, H.; Gosens, R. Pro-inflammatory mechanisms of muscarinic receptor stimulation in airway smooth muscle. Respir. Res. 2010, 11, 130. [Google Scholar] [CrossRef]
  216. Pease, J.E.; Sabroe, I. The role of interleukin-8 and its receptors in inflammatory lung disease: Implications for therapy. Am. J. Respir. Med. 2002, 1, 19–25. [Google Scholar] [CrossRef]
  217. Taylor, J.D. COPD and the response of the lung to tobacco smoke exposure. Pulm. Pharmacol. Ther. 2010, 23, 376–383. [Google Scholar] [CrossRef]
  218. Cornwell, W.D.; Kim, V.; Song, C.; Rogers, T.J. Pathogenesis of inflammation and repair in advanced COPD. Semin. Respir. Crit. Care Med 2010, 31, 257–266. [Google Scholar] [CrossRef]
  219. Postma, D.S.; Kerstjens, H.A. Characteristics of airway hyperresponsiveness in asthma and chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 1998, 158, S187–S192. [Google Scholar]
  220. Tashkin, D.P.; Altose, M.D.; Connett, J.E.; Kanner, R.E.; Lee, W.W.; Wise, R.A. Methacholine reactivity predicts changes in lung function over time in smokers with early chronic obstructive pulmonary disease. The Lung Health Study Research Group. Am. J. Respir. Crit. Care Med. 1996, 153, 1802–1811. [Google Scholar]
  221. Tashkin, D.P.; Altose, M.D.; Bleecker, E.R.; Connett, J.E.; Kanner, R.E.; Lee, W.W.; Wise, R. The lung health study: airway responsiveness to inhaled methacholine in smokers with mild to moderate airflow limitation. The Lung Health Study Research Group. Am. Rev. Respir. Dis. 1992, 145, 301–310. [Google Scholar]
  222. Saetta, M.; Di, S.A.; Turato, G.; Facchini, F.M.; Corbino, L.; Mapp, C.E.; Maestrelli, P.; Ciaccia, A.; Fabbri, L.M. CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am. J. Respir. Crit Care Med. 1998, 157, 822–826. [Google Scholar]
  223. Bosken, C.H.; Wiggs, B.R.; Pare, P.D.; Hogg, J.C. Small airway dimensions in smokers with obstruction to airflow. Am. Rev. Respir. Dis. 1990, 142, 563–570. [Google Scholar] [CrossRef]
  224. Cosio, M.G.; Hale, K.A.; Niewoehner, D.E. Morphologic and morphometric effects of prolonged cigarette smoking on the small airways. Am. Rev. Respir. Dis. 1980, 122, 266–271. [Google Scholar]
  225. Saetta, M.; Turato, G.; Baraldo, S.; Zanin, A.; Braccioni, F.; Mapp, C.E.; Maestrelli, P.; Cavallesco, G.; Papi, A.; Fabbri, L.M. Goblet cell hyperplasia and epithelial inflammation in peripheral airways of smokers with both symptoms of chronic bronchitis and chronic airflow limitation. Am. J. Respir. Crit. Care Med. 2000, 161, 1016–1021. [Google Scholar]
  226. Lambert, R.K.; Wiggs, B.R.; Kuwano, K.; Hogg, J.C.; Pare, P.D. Functional significance of increased airway smooth muscle in asthma and COPD. J. Appl. Physiol. 1993, 74, 2771–2781. [Google Scholar] [CrossRef]
  227. Jeffery, P.K. Remodeling in asthma and chronic obstructive lung disease. Am. J. Respir. Crit Care Med. 2001, 164, S28–S38. [Google Scholar]
  228. Jeffery, P.K. Comparison of the structural and inflammatory features of COPD and asthma. Giles F. Filley Lecture. Chest 2000, 117, 251S–260S. [Google Scholar] [CrossRef]
  229. Barbera, J.A.; Blanco, I. Pulmonary hypertension in patients with chronic obstructive pulmonary disease: advances in pathophysiology and management. Drugs 2009, 69, 1153–1171. [Google Scholar] [CrossRef]
  230. Barnes, P.J. Mechanisms in COPD: differences from asthma. Chest 2000, 117, 10S–14S. [Google Scholar] [CrossRef]
  231. Finlay, G.A.; Russell, K.J.; McMahon, K.J.; D'arcy, E.M.; Masterson, J.B.; FitzGerald, M.X.; O'Connor, C.M. Elevated levels of matrix metalloproteinases in bronchoalveolar lavage fluid of emphysematous patients. Thorax 1997, 52, 502–506. [Google Scholar] [CrossRef]
  232. Ohnishi, K.; Takagi, M.; Kurokawa, Y.; Satomi, S.; Konttinen, Y.T. Matrix metalloproteinase-mediated extracellular matrix protein degradation in human pulmonary emphysema. Lab. Invest. 1998, 78, 1077–1087. [Google Scholar]
  233. Matsuba, K.; Thurlbeck, W.M. The number and dimensions of small airways in emphysematous lungs. Am. J. Pathol. 1972, 67, 265–275. [Google Scholar]
  234. Wynn, T.A. Integrating mechanisms of pulmonary fibrosis. J. Exp. Med. 2011, 208, 1339–1350. [Google Scholar] [CrossRef]
  235. Salazar, L.M.; Herrera, A.M. Fibrotic response of tissue remodeling in COPD. Lung 2011, 189, 101–109. [Google Scholar] [CrossRef]
  236. Coraux, C.; Roux, J.; Jolly, T.; Birembaut, P. Epithelial cell-extracellular matrix interactions and stem cells in airway epithelial regeneration. Proc. Am. Thorac. Soc. 2008, 5, 689–694. [Google Scholar] [CrossRef]
  237. Fernandes, D.J.; Bonacci, J.V.; Stewart, A.G. Extracellular matrix, integrins, and mesenchymal cell function in the airways. Curr. Drug Targets 2006, 7, 567–577. [Google Scholar] [CrossRef]
  238. Jeffery, P.K. Remodeling and inflammation of bronchi in asthma and chronic obstructive pulmonary disease. Proc. Am. Thorac. Soc. 2004, 1, 176–183. [Google Scholar] [CrossRef]
  239. Mauad, T.; Dolhnikoff, M. Pathologic similarities and differences between asthma and chronic obstructive pulmonary disease. Curr. Opin. Pulm. Med. 2008, 14, 31–38. [Google Scholar] [CrossRef]
  240. Knight, D.A.; Holgate, S.T. The airway epithelium: Structural and functional properties in health and disease. Respirology 2003, 8, 432–446. [Google Scholar] [CrossRef]
  241. Petecchia, L.; Sabatini, F.; Varesio, L.; Camoirano, A.; Usai, C.; Pezzolo, A.; Rossi, G.A. Bronchial airway epithelial cell damage following exposure to cigarette smoke includes disassembly of tight junction components mediated by the extracellular signal-regulated kinase 1/2 pathway. Chest 2009, 135, 1502–1512. [Google Scholar] [CrossRef]
  242. Giembycz, M.A.; Kaur, M.; Leigh, R.; Newton, R. A Holy Grail of asthma management: toward understanding how long-acting beta(2)-adrenoceptor agonists enhance the clinical efficacy of inhaled corticosteroids. Br. J. Pharmacol. 2008, 153, 1090–1104. [Google Scholar]
  243. Barnes, P.J. New therapies for chronic obstructive pulmonary disease. Med. Princ. Pract. 2010, 19, 330–338. [Google Scholar] [CrossRef]
  244. Pauwels, R.A.; Buist, A.S.; Calverley, P.M.; Jenkins, C.R.; Hurd, S.S. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Lung Disease (GOLD) Workshop summary. Am J Respir. Crit. Care Med. 2001, 163, 1256–1276. [Google Scholar]
  245. Keatings, V.M.; Jatakanon, A.; Worsdell, Y.M.; Barnes, P.J. Effects of inhaled and oral glucocorticoids on inflammatory indices in asthma and COPD. Am. J. Respir. Crit Care Med. 1997, 155, 542–548. [Google Scholar]
  246. Barnes, N.C.; Qiu, Y.S.; Pavord, I.D.; Parker, D.; Davis, P.A.; Zhu, J.; Johnson, M.; Thomson, N.C.; Jeffery, P.K. Antiinflammatory effects of salmeterol/fluticasone propionate in chronic obstructive lung disease. Am. J. Respir. Crit Care Med. 2006, 173, 736–743. [Google Scholar] [CrossRef]
  247. Kaur, M.; Holden, N.S.; Wilson, S.M.; Sukkar, M.B.; Chung, K.F.; Barnes, P.J.; Newton, R.; Giembycz, M.A. Effect of beta2-adrenoceptor agonists and other cAMP-elevating agents on inflammatory gene expression in human ASM cells: a role for protein kinase A. Am. J. Physiol Lung Cell Mol. Physiol. 2008, 295, L505–L514. [Google Scholar] [CrossRef]
  248. Hallsworth, M.P.; Twort, C.H.; Lee, T.H.; Hirst, S.J. beta(2)-adrenoceptor agonists inhibit release of eosinophil-activating cytokines from human airway smooth muscle cells. Br. J. Pharmacol. 2001, 132, 729–741. [Google Scholar] [CrossRef]
  249. Shore, S.A.; Moore, P.E. Regulation of beta-adrenergic responses in airway smooth muscle. Respir. Physiol. Neurobiol. 2003, 137, 179–195. [Google Scholar] [CrossRef]
  250. Banner, K.H.; Press, N.J. PDE3/4 inhibitors as therapeutic agents for chronic obstructive pulmonary disease. Br. J. Pharmacol. 2009, 157, 892–906. [Google Scholar] [CrossRef]
  251. Gross, N.J.; Giembycz, M.A.; Rennard, S.I. Treatment of chronic obstructive pulmonary disease with roflumilast, a new phosphodiesterase 4 inhibitor. J. ChronicObstruct. Pulmon. Dis. 2010, 7, 141–153. [Google Scholar] [CrossRef]
  252. Giembycz, M.A.; Field, S.K. Roflumilast: first phosphodiesterase 4 inhibitor approved for treatment of COPD. Drug Desig. Develop. Therap. 2010, 4, 147–158. [Google Scholar]
  253. Press, N.J.; Banner, K.H. PDE4 inhibitors - a review of the current field. Prog. Med. Chem. 2009, 47, 37–74. [Google Scholar] [CrossRef]
  254. Sowa, G. Caveolae, caveolins, cavins, and endothelial cell function: new insights. Front Physiol. 2012, 2, 120. [Google Scholar]
  255. Fish, J.E. A primer on the role of microRNAs in endothelial biology and vascular disease. Semin. Nephrol. 2012, 32, 167–175. [Google Scholar] [CrossRef]
  256. Hirase, T.; Noda, K. Endothelial dysfunction as a cellular mechanism for vascular failure. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H499–H505. [Google Scholar] [CrossRef]
  257. Vestweber, D. Novel insights into leukocyte extravasation. Curr. Opin. Hematol. 2012, 19, 212–217. [Google Scholar] [CrossRef]
  258. Vestweber, D.; Broermann, A.; Schulte, D. Control of endothelial barrier function by regulating vascular endothelial-cadherin. Curr. Opin. Hematol. 2010, 17, 230–236. [Google Scholar] [CrossRef]
  259. Vestweber, D.; Winderlich, M.; Cagna, G.; Nottebaum, A.F. Cell adhesion dynamics at endothelial junctions: VE-cadherin as a major player. Trends Cell Biol. 2009, 19, 8–15. [Google Scholar] [CrossRef]
  260. Vestweber, D. Adhesion and signaling molecules that control endothelial cell contacts. Immunol. Rev. 2007, 218, 178–196. [Google Scholar] [CrossRef]
  261. Davies, D.E. The role of the epithelium in airway remodeling in asthma. Proc. Am. Thorac. Soc. 2009, 6, 678–682. [Google Scholar] [CrossRef]
  262. Niessen, C.M. Tight junctions/adherens junctions: Basic structure and function. J. Investig Dermatol. 2007, 127, 2525–2532. [Google Scholar] [CrossRef]
  263. Evans, M.J.; Fanucchi, M.V.; Plopper, C.G.; Hyde, D.M. Postnatal development of the lamina reticularis in primate airways. Anatom. Rec. 2010, 293, 947–954. [Google Scholar] [CrossRef]
  264. Evans, M.J.; van Winkle, L.S.; Fanucchi, M.V.; Plopper, C.G. Cellular and molecular characteristics of basal cells in airway epithelium. Experiment. Lung Res. 2001, 27, 401–415. [Google Scholar]
  265. Evans, M.J.; van Winkle, L.S.; Fanucchi, M.V.; Toskala, E.; Luck, E.C.; Sannes, P.L.; Plopper, C.G. Three-dimensional organization of the lamina reticularis in the rat tracheal basement membrane. Am. J. Respir. Cell Mol. Biol. 2000, 22, 393–397. [Google Scholar]
  266. Anderson, J.M.; van Italie, C.M. Physiology and function of the tight junction. ColdSpringHarb. Perspect. Biol. 2009, 1, a002584. [Google Scholar]
  267. Chiba, H.; Osanai, M.; Murata, M.; Kojima, T.; Sawada, N. Transmembrane proteins of tight junctions. Biochim. Biophys. Acta 2008, 1778, 588–600. [Google Scholar] [CrossRef]
  268. Terry, S.; Nie, M.; Matter, K.; Balda, M.S. Rho signaling and tight junction functions. Physiology 2010, 25, 16–26. [Google Scholar] [CrossRef]
  269. Corada, M.; Chimeti, S.; Cera, M.R.; Vinci, M.; Salio, M.; Fiordaliso, F.; De Angelis, N.; Villa, A.; Bossi, M.; Staszewsky, L.I.; Vecchi, A.; Parazzoli, D.; Motoike, T.; Latini, R.; Dejana, E. Junctional adhesion molecule-A-deficient polymorphonuclear cells show reduced diapedesis in peritonitis and heart ischemia-reperfusion injury. Proc. Natl. Acad. Sci. USA 2005, 102, 10634–10639. [Google Scholar]
  270. Khandoga, A.; Kessler, J.S.; Meissner, H.; Hanschen, M.; Corada, M.; Motoike, T.; Enders, G.; Dejana, E.; Krombach, F. Junctional adhesion moleculae A deficiency increases hepatic ischmia-reperfusion injury despite reduction of neutrophil transendothelial migration. Blood 2005, 106, 725–733. [Google Scholar] [CrossRef]
  271. Quint, J.K.; Wedzicha, J.A. The neutrophil in chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2007, 119, 1065–1071. [Google Scholar] [CrossRef]
  272. Walters, R.W.; Freimuth, P.; Moninger, T.O.; Ganske, I.; Zabner, J.; Welsh, M.J. Adenovirus fiber disrupts CAR-mediated intercellular adhesion allowing virus escape. Cell 2002, 110, 789–799. [Google Scholar] [CrossRef]
  273. Cohen, C.J.; Shieh, J.T.C.; Pickles, R.J.; Okegawa, T.; Hsieh, J.-T.; Bergelson, J.M. The coxsackievirus and adenovirus receptor is a transmembrane component of the tight junction. Proc. Natl. Acad. Sci. USA 2001, 98, 15191–15196. [Google Scholar]
  274. Zen, K.; Liu, Y.; McCall, I.C.; Wu, T.; Lee, W.; Babbin, B.A.; Nusrat, A.; Parkos, C.A. Neutrophil migration across tight junctions is mediated by adhesive interactions between epithelial coxsackie and adenovirus receptor and a junctional adhesion molecule-like protein on neutrophils. Mol. Biol. Cell 2005, 16, 2694–2703. [Google Scholar] [CrossRef]
  275. Giepsmans, B.N.; Ijzendoorn, S.C. Epithelial cell-cell junctions and plasma membrane domains. Biochim. Biophys. Acta 2009, 1788, 820–831. [Google Scholar] [CrossRef]
  276. Hernandez, S.; Chavez Munguia, B.; Gonzalez-Mariscal, L. ZO-2 silencing in epithelial cells perturbs the gate and fence function of tight junctions and leads to an atypical monolayer architecture. Exp. Cell Res. 2007, 313, 1533–1547. [Google Scholar] [CrossRef]
  277. Fanning, A.S.; Jameson, B.J.; Jesaitis, L.A.; Anderson, J.M. The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton. J. Biol. Chem. 1998, 273, 29745–29753. [Google Scholar]
  278. González-Mariscal, L.; Tapia, R.; Chamorro, D. Crosstalk of tight junctions components with signaling pathways. Biochim. Biophys. Acta 2008, 1778, 729–756. [Google Scholar] [CrossRef]
  279. Guillemot, L.; Paschoud, S.; Pulimeno, P.; Foglia, A.; Citi, S. The cytoplasmic plaque of tight junctions: a scaffolding and signalling center. Biochim. Biophys. Acta 2008, 1778, 601–613. [Google Scholar] [CrossRef]
  280. Rodgers, L.S.; Fanning, A.S. Regulation of epithelial permeability by the actin cytoskeleton. Cytoskeleton 2011, 68, 653–660. [Google Scholar] [CrossRef]
  281. Fanning, A.S.; Anderson, J.M. Zonula occludens-1 and -2 are cytosolic scaffolds that regulate the assembly of cellular junctions. Ann. N. Y. Acad. Sci. 2009, 1165, 113–120. [Google Scholar] [CrossRef]
  282. van Itallie, C.M.; Fanning, A.S.; Bridhges, A.; Anderson, J.M. ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol. Biol. Cell 2009, 20, 3930–3940. [Google Scholar] [CrossRef]
  283. Nawijn, M.C.; Hackett, T.L.; Postma, D.S.; van Oosterhout, A.J.M.; Heijink, I.H. E-cadherin: gatekeeper of airway mucosa and allergic sensitization. Trends Immunol. 2011, 32, 248–255. [Google Scholar] [CrossRef]
  284. Lodish, H.; Berk, A.; Zipursky, S.L. Molecular Cell Biology, 4th ed; W.H. Freeman and Company: New York, NY, USA, 2000. [Google Scholar]
  285. Goeckeler, Z.M.; Wysolmerski, R.B. Myosin phosphatase and cofilin mediate cAMP/cAMP-dependent protein kinase-induced decline in endothelial isometric tension and myosin II regulatory light chain phosphorylation. J. Biol. Chem. 2005, 280, 33083–33095. [Google Scholar] [CrossRef]
  286. Pritchard, C.A.; Hayes, L.; Wojnowski, L.; Zimmer, A.; Marais, R.M.; Norman, J.C. B-Raf acts via ROCKII/LIMK/Cofilin pathway to maintain actin stress fibres in fibroblasts. Mol. Cell. Biol. 2004, 24, 5937–5952. [Google Scholar] [CrossRef]
  287. De La Cruz, E.M.; Ostap, E.M. Relating biochemistry and function in the myosin superfamily. Curr. Opin. Cell Biol. 2004, 16, 61–67. [Google Scholar] [CrossRef]
  288. Pannekoek, W.J.; Kooistra, M.R.; Zwartkruis, F.J.; Bos, J.L. Cell-cell junction formation: The role of Rap1 and Rap1 guanine nucleotide exchange factors. Biochim. Biophys. Acta 2009, 1788, 790–796. [Google Scholar] [CrossRef]
  289. Retta, S.F.; Balzac, F.; Avolio, M. Rap1: A turnabout for the crosstalk between cadherins and integrins. Eur. J. Cell Biol. 2006, 85, 283–293. [Google Scholar] [CrossRef]
  290. van Straaten, J.F.; Coers, W.; Noordhoek, J.A.; Huitema, S.; Flipsen, J.T.; Kauffman, H.F.; Timens, W.; Postma, D.S. Proteoglycan changes in the extracellular matrix of lung tissue from patients with pulmonary emphysema. Mod. Pathol. 1999, 12, 697–705. [Google Scholar]
  291. Greenlee, K.J.; Werb, Z.; Kheradmand, F. Matrix metalloproteinase in lung: Multiple, multifarious, and multifaceted. Physiol. Rev. 2007, 87, 69–98. [Google Scholar] [CrossRef]
  292. Page-McCaw, A.; Ewald, A.J.; Werb, Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat. Rev. Mol. Cell Biol. 2007, 8, 221–233. [Google Scholar] [CrossRef]
  293. Parks, W.C.; Wilson, C.L.; López-Boado, Y.S. Matrix metalloproteinases as modulators of inflammtion and innate immunity. Nat. Rev. Immunol. 2004, 4, 617–629. [Google Scholar] [CrossRef]
  294. Gueders, M.M.; Foidart, J.M.; Noel, A.; Cataldo, D.D. Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the respiratory tract: Potential implications in asthma and other lung diseases. Eur. J. Pharmacol. 2006, 533, 133–144. [Google Scholar] [CrossRef]
  295. Vadenbroucke, R.E.; Dejonckheere, E.; Libert, C. A therapeutic role for matrix metalloproteinase inhibitors in lung diseases? Eur. Respir. J. 2011, 38, 1200–1214. [Google Scholar] [CrossRef]
  296. Demedts, I.K.; Brusselle, G.G.; Bracke, K.R.; Vemaelen, K.Y.; Pauwels, R.A. Matrix metalloproteinases in asthma and COPD. Curr. Opin. Pharmacol. 2005, 5, 257–263. [Google Scholar] [CrossRef]
  297. Oikonomid, S.; Kostikas, K.; Tsilioni, I.; Tanou, K.; Gourgoulianis, K.I.; Kiropoulos, T.S. Matrix metalloproteinases in respiratory diseases: from pathogenesis to potential clinical implications. Curr. Med. Chem. 2009, 16, 1214–1228. [Google Scholar] [CrossRef]
  298. Vial, W.C. Cigarette smoking and lung disease. Am. J. Med. Sci. 1986, 291, 130–142. [Google Scholar] [CrossRef]
  299. Lawrence, D.W.; Comerford, K.M.; Colgan, S.P. Role of VASP in reestablishment of epithelial tight junction assembly after Ca2+ switch. Am. J. Physi. Am. J. Physiol. Cell Physiol. 2002, 282, C1235–C1245. [Google Scholar]
  300. Lorenowicz, M.J.; Fernandez-Borja, M.; van Stalborch, A.M.; van Sterkenburg, M.A.; Hiemstra, P.S.; Hordijk, P.L. Microtubule dynamics and rac-1 signaling independently regulate barrier function in lung epithelial cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 293, L1321–L1331. [Google Scholar] [CrossRef]
  301. Baumer, Y.; Drenckhahn, D.; Waschke, J. cAMP induced Rac1-mediated cytoskeletal reorganization in microvascular endothelium. Histochem. Cell Biol. 2008, 129, 765–778. [Google Scholar]
  302. Logue, J.S.; Whiting, J.L.; Tunquist, B.; Langeberg, L.K.; Scott, J.D. Anchored protein kinase A recruitment of active Rac GTPase. J. Biol. Chem. 2011, 286, 22113–22121. [Google Scholar]
  303. Logue, J.S.; Whiting, J.L.; Scott, J.D. Sequestering Rac with PKA confers cAMP control of cytoskeletal remodeling. SmallGTPases 2011, 3, 173–176. [Google Scholar]
  304. Michel, C.C.; Curry, F.E. Microvascular permeability. Physiol. Rev. 1999, 79, 703–761. [Google Scholar]
  305. Metha, D.; Malik, A.B. Signaling mechanisms regulating endothelial permeability. Physiol. Rev. 2006, 86, 279–367. [Google Scholar] [CrossRef]
  306. Serezani, C.H.; Ballinger, M.N.; Aronoff, D.M.; Peters-Golden, M. Cyclic AMP: master regulator of innate immune cell function. Am. J. Respir. Cell Mol. Biol. 2008, 39, 127–132. [Google Scholar] [CrossRef]
  307. Spindler, V.; Schlegel, N.; Waschke, J. Role of GTPases in control of microvascular permeability. Cardiovasc. Res. 2010, 87, 243–253. [Google Scholar] [CrossRef]
  308. Bogatcheva, N.V.; Zemskova, M.A.; Kovalenkov, Y.; Poirier, C.; Verin, A.D. Molecular mechanisms mediating protective effect of cAMP on lipopolysaccharide (LPS)-induced human lung microvascular endothelial (HLMVEC) hyperpermeability. J. Cell. Physiol. 2009, 221, 750–759. [Google Scholar] [CrossRef]
  309. van Wetering, S.; van Buul, J.D.; Quik, S.; Mul, F.P.J.; Anthony, E.C.; ten Klosster, J.-P.; Collard, J.G.; Hordijk, P.L. Reactive oxygen species mediate Rac-induced loss of cell-cell adhesion in primary human endothelial cells. J. CellSci. 2002, 115, 1837–1846. [Google Scholar]
  310. Birukova, A.A.; Zagranichnaya, T.; Alekseeva, E.; Bokoch, G.M.; Birukov, K.G. Epac/Rap and PKA are novel mechanisms of ANP-induced Rac-mediated pulmonary endothelial barrier protection. J. Cell Physiol. 2008, 215, 715–724. [Google Scholar] [CrossRef]
  311. Birukova, A.A.; Fu, P.; Xing, J.; Birukov, K.G. Rap1 mediates protective effects of iloprost against ventilator-induced lung injury. J. Appl. Physiol. 2009, 107, 1900–1910. [Google Scholar] [CrossRef]
  312. Birukova, A.A.; Zagranichnaya, T.; Fu, P.; Alekseeva, E.; Cheng, X.; Jacobson, J.R.; Birukov, K.G. Prostaglandines PGE2 and PGI2 promote endothelial barrier enhancement via PKA- and Epac1/Rap1-dependent Rac activation. Exp. Cell Res. 2007, 313, 2504–2520. [Google Scholar] [CrossRef]
  313. Xing, J.; Birukova, A.A. ANP attenuates inflammatory signaling and Rho pathway of lung endothelial permeability induced by LPS and TNFa. Microvascul. Res. 2010, 79, 56–62. [Google Scholar] [CrossRef]
  314. Cullere, X.; Shaw, S.K.; Andersson, L.; Hirahashi, J.; Luscinskas, F.W.; Mayadas, T.N. Regulation of vascular endothelial barrier function by Epac, a cAMP-activated exchange factor for Rap GTPase. Blood 2005, 105, 1950–1955. [Google Scholar] [CrossRef]
  315. Glading, A.; Han, J.; Stockton, R.A.; Ginsberg, M.H. KRIT-1/CCM1 is a Rap1 effector that regulates endothelial cell cell junctions. J. Cell Biol. 2007, 179, 247–254. [Google Scholar] [CrossRef]
  316. Stockton, R.A.; Shenkar, R.; Awad, I.A.; Ginsberg, M.H. Cerebral cavernous malformations proteins inhibit Rho kinase to stabilize vascular integrity. J. Exp. Med. 2010, 4, 881–896. [Google Scholar]
  317. Gao, S.; Li, C.; Shimokawa, T.; Terashita, T.; Matsuda, S.; Yaoita, E.; Kobayashi, N. Rho-family small GTPases are involved in forskolin-induced cell-cell contact formation of renal glomerular podocytes in vitro. Cell Tissue Res. 2007, 328, 391–400. [Google Scholar] [CrossRef]
  318. Costantini, T.W.; Deree, J.; Loomis, W.; Putman, J.G.; Choi, S.; Baird, A.; Eliceiri, B.P.; Bansal, V.; Coimbra, R. Phosphodiesterase inhibition attenuates alterations to the tight junction proteins occludin and ZO-1 in immunostimulated caco-2 intestinal monolayers. Life Sci. 2009, 84, 18–22. [Google Scholar] [CrossRef]
  319. White, C.D.; Brown, M.D.; Sacks, D.B. IQGAPs in cancer: A family of scaffold proteins underlying tumorigenesis. FEBS Lett. 2009, 583, 1817–1824. [Google Scholar] [CrossRef]
  320. Oldenburger, A.; Rijks, W.F.; Sewbalaksing, V.D.; Poppinga, W.J.; Heijink, I.H.; Maarsingh, H.; Schmidt, M. A-kinase anchoring proteins (AKAPs) as potential novel therapeutic targets to improve cigarette smoke-induced loss of epithelial barrier function. Available online: http://www.pA2online.org/abstracts/Vol10Issue3abst108P.pdf accessed on 28 November 2012.

Share and Cite

MDPI and ACS Style

Oldenburger, A.; Maarsingh, H.; Schmidt, M. Multiple Facets of cAMP Signalling and Physiological Impact: cAMP Compartmentalization in the Lung. Pharmaceuticals 2012, 5, 1291-1331. https://doi.org/10.3390/ph5121291

AMA Style

Oldenburger A, Maarsingh H, Schmidt M. Multiple Facets of cAMP Signalling and Physiological Impact: cAMP Compartmentalization in the Lung. Pharmaceuticals. 2012; 5(12):1291-1331. https://doi.org/10.3390/ph5121291

Chicago/Turabian Style

Oldenburger, Anouk, Harm Maarsingh, and Martina Schmidt. 2012. "Multiple Facets of cAMP Signalling and Physiological Impact: cAMP Compartmentalization in the Lung" Pharmaceuticals 5, no. 12: 1291-1331. https://doi.org/10.3390/ph5121291

Article Metrics

Back to TopTop