Cell Tissue Res DOI 10.1007/s00441-013-1755-y

REVIEW

cAMP with other signaling cues converges on Rac1 to stabilize the endothelial barrier— a signaling pathway compromised in inflammation Nicolas Schlegel & Jens Waschke

Received: 29 September 2013 / Accepted: 31 October 2013 # Springer-Verlag Berlin Heidelberg 2013

Abstract cAMP is one of the most potent signaling molecules to stabilize the endothelial barrier, both under resting conditions as well as under challenge of barrier-destabilizing mediators. The two main signaling axes downstream of cAMP are activation of protein kinase A (PKA) as well as engagement of exchange protein directly activated by cAMP (Epac) and its effector GTPase Rap1. Interestingly, both pathways activate GTP exchange factors for Rac1, such as Tiam1 and Vav2 and stabilize the endothelial barrier via Rac1-mediated enforcement of adherens junctions and strengthening of the cortical actin cytoskeleton. On the level of Rac1, cAMP signaling converges with other barrier-enhancing signaling cues induced by sphingosine-1-phosphate (S1P) and angiopoietin-1 (Ang1) rendering Rac1 as an important signaling hub. Moreover, activation of Rap1 and inhibition of RhoA also contribute to barrier stabilization, emphasizing that regulation of small GTPases is a central mechanism in this context. The relevance of cAMP/Rac1-mediated barrier protection under pathophysiologic conditions can be concluded from data showing that inflammatory mediators causing multiorgan failure in systemic inflammation or sepsis interfere with this signaling axis on the level of cAMP or Rac1. This is in line with the well-known efficacy of cAMP to abrogate the barrier breakdown in response to most barrier-compromising stimuli. New is the notion that the tight endothelial barrier

N. Schlegel Department of General-, Visceral, Vascular and Pediatric surgery, University Hospital Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, Germany J. Waschke (*) Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-Universität Munich, Pettenkoferstrasse 11, 80336 Munich, Germany e-mail: [email protected]

under resting conditions is maintained by (1) continuous cAMP formation induced by hormones such as epinephrine or (2) by activation of Rac1 downstream of S1P that is secreted by erythrocytes and activated platelets. Keywords Endothelial barrier . Adherens junctions . Tight junctions . cAMP . Rho-GTPases . Inflammation

Introduction The endothelium provides a selective barrier between blood vessels and the surrounding interstitial tissue (Mehta and Malik 2006; Michel and Curry 1999). Endothelial barrier functions are deranged under various pathological conditions such as hemorrhagic stroke, vascular malformations, atherosclerosis, tumor formation and, especially, in acute inflammation (Dejana et al. 2009). Under inflammatory conditions, breakdown of the endothelial barrier is predominately caused by opening of interendothelial paracellular junctions in postcapillary venules followed by severe subcutaneous and whole body cavity edema (Goldenberg et al. 2011; Lee and Slutsky 2010; Mehta and Malik 2006). The endothelial barrier consists in intercellular tight junctions (TJ) and adherens junctions (AJ), which are also functionally coupled to cell-matrix contacts. TJs directly limit paracellular permeability. They are composed of occludin and claudins such as claudin 5, which is the only claudin expressed in peripheral endothelium and is thought to play a key role in endothelial TJ maintenance by sealing the intercellular cleft between neighboring endothelial cells (Komarova and Malik 2010). The cytoplasmic domain of TJ proteins is associated with zonula occludens protein-1 (ZO-1), which links TJs to α-catenin, spectrin and the actin cytoskeleton. Barrier-stabilizing effects mediated by the second

Cell Tissue Res

messenger cyclic adenosine monophosphate (cAMP) have at least in part been attributed to an augmented number of TJ strands in the intercellular cleft (Adamson et al. 1998). In addition, AJs contribute to barrier stabilization by providing the mechanical strength to cohesion between endothelial cells (Dejana and Orsenigo 2013). The endothelium-specific AJ adhesion molecule vascular-endothelial (VE-) cadherin undergoes homophilic binding by transinteraction of its extracellular domain in Ca2+-dependent manner. The cytoplasmatic domain of VE-cadherin has binding sites for p120 catenin as well as for β- and γ-catenin, which via α-catenin or other proteins tether the cadherin–catenin complex to the actin cytoskeleton. The association of TJs and AJs with the actin cytoskeleton explains why intracellular signaling that affects actin dynamics is critically involved in the regulation of the endothelial barrier. Mediators that strengthen the cortical actin cytoskeleton lead to endothelial barrier stabilization whereas perturbation of actin dynamics strongly alters endothelial barrier functions in vivo (Waschke et al. 2005). Therefore, it is not surprising that cAMP-mediated signaling pathways regulate the endothelial barrier by modulating actin dynamics. cAMP regulates endothelial permeability by two different signaling pathways. The first involves activation of cAMPdependent protein kinase A (PKA) and phosphorylation of PKA substrate proteins such as MLCK, ERK1/2 and RhoA. The second pathway downstream of cAMP is PKAindependent and mediated through its direct binding to exchange protein directly activated by cAMP (Epac), a guanine nucleotide exchange factor (GEF) for the small GTPase Rap1 (Parnell et al. 2012). It is important to note that the different components involved in these pathways form signaling complexes and the selective anchoring of these complexes to specific subcellular domains enables specific actions of a cAMP-mediated signaling event.

For barrier stabilization cAMP generation is confined to the membranous compartment cAMP synthesis occurs predominately at the plasma membrane (Houslay et al. 2007) upon stimulation of adenylyl cyclases (AC) via G-protein-coupled receptors (GPCR) and inhibition of phosphodiesterases (PDEs), respectively. In endothelial cells, most of the cAMP synthesis is provided by the Ca2+-inhibited type 6 adenylyl cyclase (AC6) (Cioffi et al. 2002) and most of the cAMP hydrolysis is attributable to PDE3 and PDE4 (Lugnier and Schini 1990; Netherton and Maurice 2005). Physiological agents such as prostaglandin (PG) D2, E2 (Birukova et al. 2007e, 2013c; Kobayashi et al. 2013) or atrial natriuretic peptide (ANP) (Birukova et al. 2008b) increase endothelial cAMP levels. Similarly, pharmacologic β-receptor agonists such as isoproterenol (Adamson et al. 1998; Schlegel and Waschke 2009a), adenylyl cyclase

activator forskolin and phosphodiesterase 4 inhibitors have been extensively demonstrated to exert barrier-protective effects via formation of cAMP (Adamson et al. 1998; Baumer et al. 2008, 2009; He et al. 2000; Schick et al. 2012; Schlegel et al. 2008, 2012). In this context, compartimentalization of cAMP generation is thought to be crucial. It has been shown that cAMP elevation in endothelial cells can have opposing effects and, depending on the site of cAMP formation, can either lead to barrier stabilization or breakdown (Fischmeister 2006; Sayner et al. 2006). In general, cAMP synthesis in the membranous compartment, for example, by adenylyl cyclase 6, leads to endothelial barrier stabilization. In contrast, soluble adenylyl cyclases that localize outside this membranous compartment synthesize cAMP in a cytosolic pool and have been reported to cause barrier destabilization by as yet undefined mechanisms (Sayner et al. 2006).

cAMP is one of the most potent barrier-stabilizing signaling molecules It is generally accepted that cAMP under physiologic and most pathophysiologic conditions stabilizes the endothelial barrier (Curry and Adamson 2010, 2013; Mehta and Malik 2006; Michel and Curry 1999; Rigor et al. 2012; Shen et al. 2010; Spindler et al. 2010). Increased permeability in response to histamine, bradykinin, platelet-activating factor (PAF), substance P and thrombin (Michel and Curry 1999), as well as to mediators causing multi-organ failure in sepsis such as lipopolysaccharide (LPS) and tumor necrosis factor-α (TNF-α) in vivo (Schick et al. 2012; Schlegel et al. 2009; Schlegel and Waschke 2009b), can be largely blunted by β2-adrenergic agents or phosphodiesterase 4 inhibition. Similarly, when endothelial barrier function is compromised under more complex pathophysiologic conditions, such as in experimental diabetes or after hypoxia-reoxygenation, glucagon-likepeptide-1 (GLP-1) or cAMP analogs restore endothelial barrier properties (Aslam et al. 2013; Wang et al. 2013). Although maybe not as pronounced, elevated cAMP levels in post-capillary venules in vivo are also capable of reducing baseline hydraulic conductivity as a measure for permeability within a time-course of about 20–40 min (Adamson et al. 1998, 2008; Spindler and Waschke 2011). In vitro, cAMP-mediated endothelial barrier stabilization, as revealed by augmented transendothelial resistance (TER), is usually detectable within 10–20 min (Baumer et al. 2008; Birukova et al. 2009; Spindler et al. 2010, 2011). Interestingly, selective stimulation of the Epac1/Rap1 pathway by O-Me-cAMP in vivo prevents increased permeability but, in contrast to cultured endothelium, does not reduce baseline permeability in intact microvessels (Adamson et al. 2008; Baumer et al. 2008; Cullere et al. 2005; Spindler et al. 2011). This discrepancy between cultured endothelium and endothelial cells in intact microvessels indicates

Cell Tissue Res

that cultured cells display a more unstable, inflammatory phenotype (Curry and Adamson 2010, 2013). Enhanced barrier properties were paralleled by increased and linearized immunostaining for VE-cadherin and claudin 5 or occludin, which on the ultrastructural level was reflected by an increased number of TJ strands as well as augmented intercellular cleft length and formation of complex interdigitating junctions with a higher number of intercellular junctions (Adamson et al. 2008; Spindler et al. 2011). This is in line with increased VE-cadherin-mediated binding of endothelial cells after Epac1/Rap1 stimulation (Fukuhara et al. 2005; Kooistra et al. 2005).

cAMP stabilizes the endothelial barrier by activation of Rac1 Increasing evidence indicates that cAMP primarily stabilizes endothelial barrier functions via regulation of small GTPases, first of all by activation of Rac1 (Curry and Adamson 2013; Spindler et al. 2010). We showed that Rac1 is required for maintenance of endothelial barrier properties in vivo and VEcadherin-mediated binding in vitro and that Rac1 activation was effective in blunting the PAF-mediated permeability increase in intact microvessels, similar to that shown previously for cAMP (Waschke et al. 2004a, 2006). Therefore we hypothesize that Rac1 and cAMP signaling converge. Indeed, we found that cAMP was effective in blocking Rac1 glucosylation by lethal toxin (LT) and thereby the LT-induced endothelial barrier breakdown in a PKA-dependent manner in vivo (Waschke et al. 2004b). Some years later, Rac1 activation caused by increased cAMP was demonstrated in macrovascular and microvascular endothelial cells in vitro (Baumer et al. 2008; Birukova et al. 2007e). In this context, Rac1 activation can be mediated by both PKA- as well as Epac1/Rap1-induced activation of the Rac1 GEFs, Tiam1 and Vav2 (Fig. 1), which after phosphorylation are recruited to AJs (Birukova et al. 2007e, 2010; Glading et al. 2007; Lampugnani et al. 2002, 2010). At this site, Rap1 via afadin and Tiam1 directly interact with VEcadherin (Fig. 2) and VE-cadherin transinteraction in a feedback loop enhances Tiam1 and Rac1 activity (Birukova et al. 2011b, 2013b). Further, PKA-mediated activation of Rac1 requires vasodilator-stimulated phosphoprotein (VASP), β1integrin binding as well as proper PKA compartimentalization via A-kinase anchoring peptides (AKAPs) (Fig. 1) (Schlegel et al. 2008; Schlegel and Waschke 2009c; Schlegel et al. 2009), indicating that the actin-binding protein VASP regulates endothelial barrier functions on the level of Rac1 activation similar to that shown recently for cortactin (Schlegel and Waschke 2010; Schnoor et al. 2011). However, under some conditions, PKA- but not Epac-mediated signaling is engaged to stabilize barrier properties downstream of cAMP, as has been shown for the tissue inhibitor of metalloproteinase 2 (TIMP-2), which

activates adenylyl cyclases after binding to integrin α3β1 via the SH2-containing protein tyrosin phosphatase-1 (Shp-1) (Kim et al. 2012). Interestingly, when NSC-23766 was used to interfere with GEF-mediated Rac1 activation, the barrier protective effects of O-Me-cAMP to selectively activate the Epac/Rap1 pathway were completely blocked but not when overall cAMP was increased, which may indicate that Rac1 is the main downstream effector of Rap1 to induce junction remodeling and barrier stabilization, whereas other signaling molecules may exist to mediate the effects of PKA in addition to Rac1 (Spindler et al. 2011). Although in this study Cdc42 was not found to be activated in response to increased cAMP, Cdc42 may also contribute to cAMP-mediated barrier stabilization since it is effectively activated by cAMP, presumably also via Vav2 (Fig. 1) (Baumer et al. 2009; Birukova et al. 2007e, 2008b; Kobayashi et al. 2013). This body of evidence shows that cAMP stabilizes the barrier primarily via activation of Rac1, supported by Rap1 and Cdc42. Not as easy to understand is the role of RhoA downstream of cAMP. RhoA via Rho kinase and regulation of myosin light chain (MLC) phosphorylation is well known to confer a contractile phenotype to endothelial cells (Spindler et al. 2010). In cultured endothelium, this contractile phenotype can be easily induced when barrierdisrupting agents such as the coagulation factor thrombin are used. However, since the endothelium of intact post-capillary venules does not respond to thrombin unless exposed to inflammatory stimuli and inhibitors of Rho kinase or MLCK do not block the permeability increase in response to typical inflammatory mediators (which thrombin is not), these data indicate that RhoA and the contractile phenotype are less important for barrier regulation in vivo (Adamson et al. 2003; Curry et al. 2003). This fits with the observation that cAMP does not reduce RhoA activity under resting conditions, although RhoA bears a PKA consensus site and can be phosphorylated by PKA (Baumer et al. 2008; Qiao et al. 2003). Instead, under resting conditions, RhoA appears to foster cell junctions as can be concluded from studies where a deficiency of the Rho-GEF TEM4, which associates with the cadherin–catenin complex, causes endothelial barrier disruption (Fig. 1) (Ngok et al. 2013). Moreover, we believe that RhoA activation under inflammatory conditions is downstream of cAMP reduction and Rac1 inactivation (Fig. 1). When thrombin is used, cAMP levels decrease within 30 s as revealed by fluorescence resonance energy transfer (FRET) (Baumer et al. 2009). As soon as 5 min after addition of thrombin, the activities of Rac1 and Cdc42 are reduced while RhoA is activated, suggesting that all these GTPases are involved in thrombin-mediated barrier breakdown (Baumer et al. 2008, 2009). However, when LPS or TNF-α were used to increase permeability in vitro, inactivation of Rac1 coincided with the initial barrier breakdown (Schlegel et al. 2009), whereas RhoA activation was delayed. Moreover, parallel activation of Rac1 and RhoA but not

Cell Tissue Res

Fig. 1 Mechanisms underlying barrier regulation by cAMP and small GTPases. Endothelial barrier properties are maintained by adherens junctions (AJ) and tight junctions (TJ). AJs consist in VE-cadherin, which is linked to the intracellular adapter molecules α-, ß-, γ- and p120-catenin and tethered to the cortical actin cytoskeleton. TJs are composed of claudin5 and occludin, which are connected to the actin cytoskeleton via Zonula occludens protein (ZO-1/2/3). cAMP-mediated effects to stabilize the endothelial barrier involve protein kinase A (PKA) and exchange protein directly activated by cAMP (EPAC), which via guanine

nucleotide exchange factors (GEFs) such as Tiam1 or Vav2 activate Rac1 and Cdc42, a process involving vasodilator-stimulated phosphoprotein (VASP) and A-kinase anchoring proteins (AKAPs). Rac1 stabilization of AJs and TJs occurs in part by strengthening of the cortical actin cytoskeleton and by inhibition of RhoA via p190RhoGAP. RhoA is also regulated by the junction-associated RhoGEF TEM4 and also by GEFH1, which is sequestered by microtubules downstream of cAMP and stathmin phosphorylation (p-stmn). cAMP stabilizes microtubules also via AKAP9

inhibition of Rho kinase was capable of blocking TNF-αinduced permeability in vivo, further indicating that RhoA signaling is not crucial to the increase of permeability under these conditions. In this context, Rac1 may regulate RhoA function on the level of GTP exchange. In endothelial cells, Rac1 inhibits RhoA via p190RhoGAP, which is bound to AJs via p120-catenin (Birukova et al. 2011a; Zebda et al. 2013). In addition, Rac1 can interfere with RhoA activation by p115RhoGEF (Birukova et al. 2007a, c). Alternatively, cAMP via PKA can inhibit RhoA function independent of Rac1 by stabilization of microtubules (Fig. 1). cAMP via stathmin phosphorylation was shown to stabilize microtubules to sequester GEF-H1 and thereby to inhibit RhoA (Tian et al. 2012). Similarly, Epac (independently of Rap1) via AKAP9 caused microtubule polymerization and endothelial barrier stabilization (Sehrawat et al. 2011). The mechanisms by which Rac1 regulates endothelial barrier functions are not the subject of this article and are discussed elsewhere (Spindler et al. 2010; review by van NieuwAmerongen in this issue). However, inhibition of myosin-ATPase did not block the permeability increase caused by LT-mediated Rac1 inactivation in vivo but, similar to MLCK, inhibition in cultured endothelium abolished intercellular gap formation and largely reduced increased permeability in these monolayers. Therefore, it can be concluded that Rac1mediated regulation of endothelial contraction (via RhoA)

in vivo is maybe less important than in cultured cells (Waschke et al. 2004c). Rather, Rac1 via p21-activated kinase and cortactin appears to regulate endothelial barrier properties on the level of the cortical actin cytoskeleton and the cadherin– catenin complex (Birukova et al. 2011b; Waschke et al. 2004c).

On the level of Rac1, cAMP signaling converges with other barrier-enhancing signaling cues Mediators that are known to be important for endothelial barrier stabilization in acute inflammation and under resting conditions affect Rac1 activity, suggesting that on this level cAMP signaling converges with other signaling cues. In this context, the angiopoietin/Tie2 system as well as sphingosine 1-phosphate (S1P) are known to be required for endothelial barrier maintenance under resting conditions (Curry and Adamson 2013; David et al. 2013). The origin of angiopoietin-1 (Ang1) are smooth muscle cells, pericytes and monocytes (Moss 2013). Ang1 binds to the endothelium-specific receptor Tie2, which activates downstream effector phosphatidylinositol 3-kinase and subsequently leads to stimulation of Rac1, a process that requires IQ Domain GTPase-activating protein1 (IQGAP1) binding to the active GTPase (Fig. 2) (David et al. 2011). This is followed by activation of p190 RhoGAP and thereby leads to inhibition of

Cell Tissue Res

Fig. 2 Rac1 activity is crucial for mediators involved in endothelial barrier regulation under resting conditions and in inflammation. Endothelial barrier regulation predominately occurs on the level of postcapillary venules. Inflammatory mediators such as Lipopolysaccharide (LPS), tumor necrosis factor-α (TNF-α) or thrombin lead to the opening of intercellular junctions. Various mediators stabilize the endothelial barrier by cAMP-dependent pathways (left). Prostaglandins, atrial natriuretic peptide (ANP) or β-receptor agonists via their receptors increase cAMP levels in endothelial cells. cAMP levels in turn promote Rac1/Cdc42 activation via GEFs Tiam1 and Vav2 downstream of PKA and Epac1/Rap1. Similarly, angiopoietin1 (Ang1),

hepatocyte growth factor (HGF) and sphingosine-1-phosphate (S1P) via their receptors as well as OxPAPC cause Rac1 activation, a process involving Akt and IQGAP1. Thus, on the level of Rac1, cAMP signaling converges with these barrier-enhancing signaling cues. Endothelial barrier disruption by inflammatory mediators such as LPS, TNF-α, or thrombin via their receptors as well as loss of β-adrenergic stimulation is mediated by reduction of cAMP and Rac1 activity (right). Ang2 via blocking Tie2 receptor autophosphorylation or S1P via S1P2-receptor results in Rac1 inactivation. Interestingly, βadrenergic stimulation, S1P1 signaling and Tie2-activation are permanently required to maintain basal Rac1 activity and endothelial barrier function

RhoA (Mammoto et al. 2007). Because the Tie2 receptor is constitutively activated by Ang1, it is believed that maintenance of endothelial barrier properties is Ang1/Tie2-dependent under resting conditions (David et al. 2013; Thurston et al. 2000). Under inflammatory conditions, Ang1 was effective in preventing LPS-induced endothelial barrier breakdown in different in vivo models (David et al. 2013; Mammoto et al. 2007). The mutual antagonist of Ang1 is Ang2, which is stored within endothelial Weibel–Palade bodies and is secreted rapidly following inflammatory stimulation. Ang2 induces vascular leakage by blocking the Ang1-induced autophosphorylation of the Tie2 receptor (Moss 2013). The pathophysiological relevance of the Ang/Tie2 system is highlighted by the fact that patients with severe systemic inflammation or sepsis display reduced levels of Ang1 but increased levels of Ang2 (David et al. 2013; Fiusa et al. 2013). Another substance that stimulates the endothelium to maintain its barrier function is S1P (Garcia et al. 2001; McVerry

and Garcia 2005). It is believed that S1P is continuously released from erythrocytes or platelets and stimulates the S1 P 1 -re ce ptor that, tog eth er w ith Ak t-me diate d transactivation (Singleton et al. 2009), leads to activation of Rac1 and to endothelial barrier stabilization (Fig. 2) (Curry and Adamson 2013). The main body of evidence for this observation was derived from experiments using single microvessels that were perfused with a physiological solution lacking S1P. This condition led to increased microvascular permeability, which was abrogated after addition of S1P (Curry et al. 2012). Moreover, S1P was also found to effectively attenuate increased permeability in response to the inflammatory mediators bradykinin and PAF (Adamson et al. 2012a, b). It is not known whether S1P plasma levels are reduced in systemic inflammation or sepsis but it has been reported that the S1P carrier protein ApoM is reduced in patients under the latter conditions, which could contribute to manifestation of vascular leakage (Kumaraswamy et al. 2012).

Cell Tissue Res

Hepatocyte growth factor (HGF) is also well known to induce endothelial barrier stabilization (Birukova et al. 2009; Ephstein et al. 2013; Liu et al. 2002; Singleton et al. 2007). HGF is usually found in the alveolar fluid and, because its secretion is augmented under conditions of acute inflammation such as in lung injury, pancreatitis, or after surgical trauma after partial liver resection and nephrectomy, HGF is considered as a rescue factor (Ware and Matthay 2002). Accordingly, it was demonstrated that HGF was effective in preventing VEGFinduced and thrombin-induced endothelial barrier breakdown via Rac1 (Fig. 2) (Birukova et al. 2007a, 2009). Recently, it was shown that HGF-induced Rac1 activation and endothelial barrier stabilization is mediated via S1P1 receptor and integrin β4 transactivation (Ephstein et al. 2013). It is well established that inflammation leads to augmented levels of oxidized phospholipids, which derive from an increased generation of reactive oxygen species (Fu and Birukov 2009). Among the multiple biological effects that oxidized phospholipids exert, it is remarkable that oxidized phospholipids such as oxidized 1-palmitoyl-2-arachidonoylsn-glycero-3-phosphorylcholine (OxPAPC) are capable of protecting the endothelial barrier under challenge of various inflammatory stimuli in vivo and in vitro (Birukova et al. 2013a; Fu and Birukov 2009; Nonas et al. 2008; Starosta et al. 2012; Zebda et al. 2013). Detailed investigations have revealed that endothelial barrier protection by OxPAPC involves activation of the GEFs Tiam1 and βPIX to augment Rac1/Cdc42 activity (Fig. 2) (Birukova et al. 2007d). Upstream mechanisms included activation of GPCR, PKA, Src and PKC (Birukova et al. 2007b, 2008a). Taken together, modulation of Rac1 activity represents a crucial signaling hub not only for cAMP-mediated effects but also of signaling cues that are known to critically influence endothelial barrier functions under resting conditions and in inflammation. Given that cAMP-mediated Rac1 activation and endothelial barrier stabilization is capable of interfering with alterations of Ang/Tie2- or S1P signaling, there is an enormous therapeutic potential to stabilize the endothelial barrier by pharmacological targeting this pathway, e.g., by application of phosphodiesterase inhibitors (Schick et al. 2012; Schlegel et al. 2009) or by newly developed agents such as Sac-1004 (Maharjan et al. 2013). In support of this notion, it was shown that systemic application of a phosphodiesterase-4 inhibitor was effective in preventing microvascular leakage and breakdown of the microcirculation, which drastically augmented survival rates in an animal model of systemic inflammation (Schick et al. 2012). Moreover, intraperitoneal application of phosphodiesterase-4 inhibitors or systemic application of cAMP-increasing agents such as dobutamine or norepinephrine were effective for volume expansion under resting conditions and in acute inflammation (De Backer et al. 2006; Lehtonen et al. 2004; Lin et al. 2012; Secchi et al. 2001; Stephens et al. 2011).

In sepsis and in response to inflammatory mediators, the cAMP/Rac1 signaling axis is impaired From a clinical point of view, endothelial barrier breakdown is increasingly considered to play a key role in the pathophysiology of sepsis (David et al. 2013; Goldenberg et al. 2011; Lee and Slutsky 2010; Schick et al. 2012). In systemic inflammation and sepsis, the consecutive loss of intravascular fluid after endothelial barrier disruption leads to breakdown of the microcirculation, metabolic dysbalance and organ failure, which contribute significantly to the high mortality rate of up to 60 % in these patients (Dellinger et al. 2012). Therefore, the option to enable therapeutic intervention is desperately needed. It has been demonstrated that endothelial barrier disruption by inflammatory stimuli such as bacterial LPS, TNF-α, or thrombin is at least in part caused by loss of intracellular cAMP and Rac1 inactivation (Fig. 2) (Schlegel et al. 2009). Similarly, disturbed cAMP signaling was observed in microvessels in an animal model of systemic inflammation (Schick et al. 2012). Since mRNA levels for several adenylyl cyclase isoforms including AC6 were reduced in vivo when LPS was applied, it is possible that cAMP synthesis is reduced by this mechanism (Risoe et al. 2007). Besides direct effects of inflammatory stimuli on endothelial cells, it is also known that the Ang/Tie2-signaling axis is critically deranged in systemic inflammation and sepsis (Fig. 2) (David et al. 2013). Plasma levels of the Tie2 receptor antagonist Ang2 in septic patients are strongly associated with severity of illness, markers of endothelial inflammation and adverse outcome (Fiusa et al. 2013; Kumpers et al. 2008, 2010). Furthermore, it has been shown that sepsis-induced microvascular alterations can be attributed to Ang2-mediated effects (Kumpers et al. 2011; Ziegler et al. 2013). Therefore, it can be assumed that both Ang1 mimetics as well as strategies to deplete or inhibit Ang2 may become successful therapeutic options to treat inflammation-induced endothelial barrier breakdown (David et al. 2013).

The tight endothelial barrier under resting conditions is maintained by mechanisms that continuously serve to increase cAMP or to activate Rac1 An interesting aspect is that the cAMP/Rac1 signaling axis is not only impaired under inflammatory conditions and stimulation of this signaling pathway can be used to prevent endothelial barrier breakdown but apparently cAMP/Rac1 signaling is constitutively activated, which is required for maintenance of tight barrier properties under resting conditions. For example, S1P appears to be constantly released from erythrocytes and activated platelets (Fig. 2) (Hanel et al. 2007; Pyne and Pyne 2000). Thus, when sphingosine kinase 1 that produces S1P is missing in hematopoetic cells, the lung

Cell Tissue Res

endothelial barrier is compromised (Camerer et al. 2009). Similarly, when intact microvessels are perfused in the absence of erythrocytes or S1P, baseline permeability increases (Curry et al. 2012). Apparently, a similar mechanism is stabilizing the endothelial barrier via the β2-adrenergic receptor (Fig. 2) (Spindler et al. 2011). Inhibition of β2-adrenergic receptor signaling increases baseline hydraulic conductivity in vivo and reduces barrier functions in cultured endothelium. Since in these studies no epinephrine, which is the physiologic ligand of this receptor, was present, it is attractive to assume that endothelial cells can generate epinephrine. These data strongly indicate that catecholamines serve to maintain endothelial barrier properties by constant formation of cAMP. In this context, it is possible that, under resting conditions, cAMP sustains the endothelial barrier primarily via PKA-dependent mechanisms, whereas the Epac pathway is more relevant for stimulated barrier protection (Pannekoek et al. 2011).

Conclusion The main conclusion of this article is that the cAMP/Rac1 signaling axis is pivotal in endothelial barrier regulation. Dysfunction of this pathway leading to microvascular endothelial barrier breakdown significantly contributes to the pathogenicity of several severe diseases, such as multi-organ failure in sepsis. Therefore, because stimulation of cAMP and Rac1 signaling in the endothelium is effective in blocking the permeability increase in response to most inflammatory mediators, this pathway should be considered as a therapeutic target. Three aspects are interesting: (1) blood cells (primarily erythrocytes) constantly supplying mediators such as S1P and hormones may be secreted by endothelial cells to maintain endothelial barrier properties; (2) compartimentalization of signaling molecules in this pathway (AKAPs for PKA and GEFs for Rac1 and RhoA, as well as the GTPase Rap1 itself) to endothelial AJs and cytoskeletal components or via modulation of cAMP diffusion coefficients (Feinstein et al. 2012) is increasingly recognized to be important; and (3) the contractile, RhoA-dependent phenotype is less important for cAMPmediated barrier regulation but rather is induced under inflammatory conditions downstream of Rac1 inactivation.

References Adamson RH, Liu B, Fry GN, Rubin LL, Curry FE (1998) Microvascular permeability and number of tight junctions are modulated by cAMP. Am J Physiol 274:H1885–H1894 Adamson RH, Zeng M, Adamson GN, Lenz JF, Curry FE (2003) PAFand bradykinin-induced hyperpermeability of rat venules is independent of actin-myosin contraction. Am J Physiol Heart Circ Physiol 285:H406–H417

Adamson RH, Ly JC, Sarai RK, Lenz JF, Altangerel A, Drenckhahn D, Curry FE (2008) Epac/Rap1 pathway regulates microvascular hyperpermeability induced by PAF in rat mesentery. Am J Physiol Heart Circ Physiol 294:H1188–H1196 Adamson RH, Sarai RK, Altangerel A, Thirkill TL, Clark JF, Curry F-RE (2012a) Sphingosine-1-phosphate modulation of basal permeability and acute inflammatory responses in rat venular microvessels. Cardiovasc Res 88(2):344–351 Adamson RH, Sarai RK, Clark JF, Altangerel A, Thirkill TL, Curry FE (2012b) Attenuation by sphingosine-1-phosphate of rat microvessel acute permeability response to bradykinin is rapidly reversible. Am J Physiol Heart Circ Physiol 302(10):H1929–H1935 Aslam M, Schluter KD, Rohrbach S, Rafiq A, Nazli S, Piper HM, Noll T, Schulz R, Gunduz D (2013) Hypoxia-reoxygenation-induced endothelial barrier failure: role of RhoA, Rac1 and myosin light chain kinase. J Physiol 591:461–473 Baumer Y, Drenckhahn D, Waschke J (2008) cAMP induced Rac 1mediated cytoskeletal reorganization in microvascular endothelium. Histochem Cell Biol 129:765–778 Baumer Y, Spindler V, Werthmann RC, Bünemann M, Waschke J (2009) Role of Rac 1 and cAMP in endothelial barrier stabilization and thrombin-induced barrier breakdown. J Cell Physiol 220(3):716–726 Birukova AA, Alekseeva E, Mikaelyan A, Birukov KG (2007a) HGF attenuates thrombin-induced endothelial permeability by Tiam1mediated activation of the Rac pathway and by Tiam1/Racdependent inhibition of the Rho pathway. FASEB J 21:2776–2786 Birukova AA, Chatchavalvanich S, Oskolkova O, Bochkov VN, Birukov KG (2007b) Signaling pathways involved in OxPAPC-induced pulmonary endothelial barrier protection. Microvasc Res 73:173–181 Birukova AA, Malyukova I, Mikaelyan A, Fu P, Birukov KG (2007c) Tiam1 and betaPIX mediate Rac-dependent endothelial barrier protective response to oxidized phospholipids. J Cell Physiol 211:608–617 Birukova AA, Malyukova I, Poroyko V, Birukov KG (2007d) Paxillinbeta-catenin interactions are involved in Rac/Cdc42-mediated endothelial barrier-protective response to oxidized phospholipids. Am J Physiol Lung Cell Mol Physiol 293(1):L199–L211 Birukova AA, Zagranichnaya T, Fu P, Alekseeva E, Chen W, Jacobson JR, Birukov KG (2007e) Prostaglandins PGE(2) and PGI(2) promote endothelial barrier enhancement via PKA- and Epac1/Rap1dependent Rac activation. Exp Cell Res 313:2504–2520 Birukova AA, Alekseeva E, Cokic I, Turner CE, Birukov KG (2008a) Cross talk between paxillin and Rac is critical for mediation of barrier-protective effects by oxidized phospholipids. Am J Physiol Lung Cell Mol Physiol 295(4):L593–L602 Birukova AA, Zagranichnaya T, Alekseeva E, Bokoch GM, Birukov KG (2008b) Epac/Rap and PKA are novel mechanisms of ANP-induced Rac-mediated pulmonary endothelial barrier protection. J Cell Physiol 215:715–724 Birukova AA, Cokic I, Moldobaeva N, Birukov KG (2009) Paxillin is involved in the differential regulation of endothelial barrier by HGF and VEGF. Am J Respir Cell Mol Biol 40(1):99–107 Birukova AA, Burdette D, Moldobaeva N, Xing J, Fu P, Birukov KG (2010) Rac GTPase is a hub for protein kinase A and Epac signaling in endothelial barrier protection by cAMP. Microvasc Res 79:128–138 Birukova AA, Zebda N, Cokic I, Fu P, Wu T, Dubrovskyi O, Birukov KG (2011a) p190RhoGAP mediates protective effects of oxidized phospholipids in the models of ventilator-induced lung injury. Exp Cell Res 317:859–872 Birukova AA, Zebda N, Fu P, Poroyko V, Cokic I, Birukov KG (2011b) Association between adherens junctions and tight junctions via Rap1 promotes barrier protective effects of oxidized phospholipids. J Cell Physiol 226:2052–2062 Birukova AA, Starosta V, Tian X, Higginbotham K, Koroniak L, Berliner JA, Birukov KG (2013a) Fragmented oxidation products define barrier disruptive endothelial cell response to OxPAPC. Transl Res 161:495–504

Cell Tissue Res Birukova AA, Tian X, Tian Y, Higginbotham K, Birukov KG (2013b) Rap-afadin axis in control of Rho signaling and endothelial barrier recovery. Mol Biol Cell 24:2678–2688 Birukova AA, Wu T, Tian Y, Meliton A, Sarich N, Tian X, Leff A, Birukov KG (2013c) Iloprost improves endothelial barrier function in lipopolysaccharide-induced lung injury. Eur Respir J 41:165–176 Camerer E, Regard JB, Cornelissen I, Srinivasan Y, Duong DN, Palmer D, Pham TH, Wong JS, Pappu R, Coughlin SR (2009) Sphingosine1-phosphate in the plasma compartment regulates basal and inflammation-induced vascular leak in mice. J Clin Invest 119: 1871–1879 Cioffi DL, Moore TM, Schaack J, Creighton JR, Cooper DM, Stevens T (2002) Dominant regulation of interendothelial cell gap formation by calcium-inhibited type 6 adenylyl cyclase. J Cell Biol 157:1267–1278 Cullere X, Shaw SK, Andersson L, Hirahashi J, Luscinskas FW, Mayadas TN (2005) Regulation of vascular endothelial barrier function by Epac, a cAMP-activated exchange factor for Rap GTPase. Blood 105:1950–1955 Curry FR, Adamson RH (2010) Vascular permeability modulation at the cell, microvessel, or whole organ level: towards closing gaps in our knowledge. Cardiovasc Res 87:218–229 Curry FR, Adamson RH (2013) Tonic regulation of vascular permeability. Acta Physiol (Oxf) 207:628–649 Curry FE, Zeng M, Adamson RH (2003) Thrombin increases permeability only in venules exposed to inflammatory conditions. Am J Physiol Heart Circ Physiol 285:H2446–H2453 Curry FE, Clark JF, Adamson RH (2012) Erythrocyte-derived sphingosine-1-phosphate stabilizes basal hydraulic conductivity and solute permeability in rat microvessels. Am J Physiol Heart Circ Physiol 303(7):H825–H834 David S, Ghosh CC, Mukherjee A, Parikh SM (2011) Angiopoietin-1 requires IQ domain GTPase-activating protein 1 to activate Rac1 and promote endothelial barrier defense. Arterioscler Thromb Vasc Biol 31:2643–2652 David S, Kumpers P, van Slyke P, Parikh SM (2013) Mending leaky blood vessels: the angiopoietin-Tie2 pathway in sepsis. J Pharmacol Exp Ther 345(1):2–6 De Backer D, Creteur J, Dubois MJ, Sakr Y, Koch M, Verdant C, Vincent JL (2006) The effects of dobutamine on microcirculatory alterations in patients with septic shock are independent of its systemic effects. Crit Care Med 34:403–408 Dejana E, Orsenigo F (2013) Endothelial adherens junctions at a glance. J Cell Sci 126:2545–2549 Dejana E, Tournier-Lasserve E, Weinstein BM (2009) The control of vascular integrity by endothelial cell junctions: molecular basis and pathological implications. Dev Cell 16:209–221 Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE, Sprung CL, Douglas IS, Jaeschke R, Osborn TM, Nunnally ME, Townsend SR, Reinhart K, Kleinpell RM, Angus DC, Deutschman CS, Machado FR, Rubenfeld GD, Webb SA, Beale RJ, Vincent JL, Moreno R (2012) Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 41:580–637 Ephstein Y, Singleton PA, Chen W, Wang L, Salgia R, Kanteti P, Dudek SM, Garcia JG, Jacobson JR (2013) Critical role of S1PR1 and integrin beta4 in HGF/c-Met-mediated increases in vascular integrity. J Biol Chem 288:2191–2200 Feinstein WP, Zhu B, Leavesley SJ, Sayner SL, Rich TC (2012) Assessment of cellular mechanisms contributing to cAMP compartmentalization in pulmonary microvascular endothelial cells. Am J Physiol Cell Physiol 302:C839–C852 Fischmeister R (2006) Is cAMP good or bad? Depends on where it’s made. Circ Res 98:582–584 Fiusa MM, Costa-Lima C, de Souza GR, Vigorito AC, Aranha FJ, Lorand-Metze I, Annichino-Bizzacchi JM, De Souza CA, De Paula EV (2013) A high angiopoietin-2/angiopoietin-1 ratio is

associated with a high risk of septic shock in patients with febrile neutropenia. Crit Care 17(4):R169 Fu P, Birukov KG (2009) Oxidized phospholipids in control of inflammation and endothelial barrier. Transl Res 153:166–176 Fukuhara S, Sakurai A, Sano H, Yamagishi A, Somekawa S, Takakura N, Saito Y, Kangawa K, Mochizuki N (2005) Cyclic AMP potentiates vascular endothelial cadherin-mediated cell-cell contact to enhance endothelial barrier function through an Epac-Rap1 signaling pathway. Mol Cell Biol 25(1):136–146 Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR, English D (2001) Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest 108:689–701 Glading A, Han J, Stockton RA, Ginsberg MH (2007) KRIT-1/CCM1 is a Rap1 effector that regulates endothelial cell cell junctions. J Cell Biol 179:247–254 Goldenberg NM, Steinberg BE, Slutsky AS, Lee WL (2011) Broken barriers: a new take on sepsis pathogenesis. Sci Transl Med 3:88ps25 Hanel P, Andreani P, Graler MH (2007) Erythrocytes store and release sphingosine 1-phosphate in blood. FASEB J 21:1202–1209 He P, Zeng M, Curry FE (2000) Dominant role of cAMP in regulation of microvessel permeability. Am J Physiol Heart Circ Physiol 278: H1124–H1133 Houslay MD, Baillie GS, Maurice DH (2007) cAMP-Specific phosphodiesterase-4 enzymes in the cardiovascular system: a molecular toolbox for generating compartmentalized cAMP signaling. Circ Res 100:950–966 Kim SH, Cho YR, Kim HJ, Oh JS, Ahn EK, Ko HJ, Hwang BJ, Lee SJ, Cho Y, Kim YK, Stetler-Stevenson WG, Seo DW (2012) Antagonism of VEGF-A-induced increase in vascular permeability by an integrin alpha3beta1-Shp-1-cAMP/PKA pathway. Blood 120: 4892–4902 Kobayashi K, Tsubosaka Y, Hori M, Narumiya S, Ozaki H, Murata T (2013) Prostaglandin D2-DP signaling promotes endothelial barrier function via the cAMP/PKA/Tiam1/Rac1 pathway. Arterioscler Thromb Vasc Biol 33:565–571 Komarova Y, Malik AB (2010) Regulation of endothelial permeability via paracellular and transcellular transport pathways. Annu Rev Physiol 72:463–493 Kooistra MRH, Corada M, Dejana E, Bos JL (2005) Epac1 regulates integrity of endothelial cell junctions through VE-cadherin. FEBS Lett 579:4966–4972 Kumaraswamy SB, Linder A, Akesson P, Dahlback B (2012) Decreased plasma concentrations of apolipoprotein M in sepsis and systemic inflammatory response syndromes. Crit Care 16(2):R60 Kumpers P, Lukasz A, David S, Horn R, Hafer C, Faulhaber-Walter R, Fliser D, Haller H, Kielstein JT (2008) Excess circulating angiopoietin-2 is a strong predictor of mortality in critically ill medical patients. Crit Care 12(6):R147 Kumpers P, Hafer C, David S, Hecker H, Lukasz A, Fliser D, Haller H, Kielstein JT, Faulhaber-Walter R (2010) Angiopoietin-2 in patients requiring renal replacement therapy in the ICU: relation to acute kidney injury, multiple organ dysfunction syndrome and outcome. Intensive Care Med 36:462–470 Kumpers P, Gueler F, David S, Slyke PV, Dumont DJ, Park JK, Bockmeyer CL, Parikh SM, Pavenstadt H, Haller H, Shushakova N (2011) The synthetic tie2 agonist peptide vasculotide protects against vascular leakage and reduces mortality in murine abdominal sepsis. Crit Care 15(5):R261 Lampugnani MG, Zanetti A, Breviario F, Balconi G, Orsenigo F, Corada M, Spagnuolo R, Betson M, Braga V, Dejana E (2002) VE-cadherin regulates endothelial actin activating Rac and increasing membrane association of Tiam. Mol Biol Cell 13:1175–1189 Lampugnani MG, Orsenigo F, Rudini N, Maddaluno L, Boulday G, Chapon F, Dejana E (2010) CCM1 regulates vascular-lumen organization by inducing endothelial polarity. J Cell Sci 123:1073–1080

Cell Tissue Res Lee WL, Slutsky AS (2010) Sepsis and endothelial permeability. N Engl J Med 363(7):689–691 Lehtonen LA, Antila S, Pentikainen PJ (2004) Pharmacokinetics and pharmacodynamics of intravenous inotropic agents. Clin Pharmacokinet 43:187–203 Lin YC, Adamson RH, Clark JF, Reed RK, Curry FR (2012) Phosphodiesterase 4 inhibition attenuates plasma volume loss and transvascular exchange in volume-expanded mice. J Physiol 590: 309–322 Liu F, Schaphorst KL, Verin AD, Jacobs K, Birukova A, Day RM, Bogatcheva N, Bottaro DP, Garcia JGN (2002) Hepatocyte growth factor enhances endothelial cell barrier function and cortical cytoskeletal rearrangement: potential role of glycogen synthase kinase3-beta. FASEB J 16(9):950–962 Lugnier C, Schini VB (1990) Characterization of cyclic nucleotide phosphodiesterases from cultured bovine aortic endothelial cells. Biochem Pharmacol 39:75–84 Maharjan S, Kim K, Agrawal V, Choi HJ, Kim NJ, Kim YM, Suh YG, Kwon YG (2013) Sac-1004, a novel vascular leakage blocker, enhances endothelial barrier through the cAMP/Rac/cortactin pathway. Biochem Biophys Res Commun 435:420–427 Mammoto T, Parikh SM, Mammoto A, Gallagher D, Chan B, Mostoslavsky G, Ingber DE, Sukhatme VP (2007) Angiopoietin-1 requires p190 RhoGAP to protect against vascular leakage in vivo. J Biol Chem 282:23910–23918 McVerry BJ, Garcia JG (2005) In vitro and in vivo modulation of vascular barrier integrity by sphingosine 1-phosphate: mechanistic insights. Cell Signal 17:131–139 Mehta D, Malik AB (2006) Signaling mechanisms regulating endothelial permeability. Physiol Rev 86:279–367 Michel CC, Curry FE (1999) Microvascular permeability. Physiol Rev 79(3):703–761 Moss A (2013) The angiopoietin:Tie 2 interaction: a potential target for future therapies in human vascular disease. Cytokine Growth Factor Rev. doi:10.1016/j.cytogfr.2013.05.009 Netherton SJ, Maurice DH (2005) Vascular endothelial cell cyclic nucleotide phosphodiesterases and regulated cell migration: implications in angiogenesis. Mol Pharmacol 67(1):263–272 Ngok SP, Geyer R, Kourtidis A, Mitin N, Feathers R, Der C, Anastasiadis PZ (2013) TEM4 is a junctional Rho GEF required for cell-cell adhesion, monolayer integrity and barrier function. J Cell Sci 126: 3271–3277 Nonas S, Birukova AA, Fu P, Xing J, Chatchavalvanich S, Bochkov VN, Leitinger N, Garcia JG, Birukov KG (2008) Oxidized phospholipids reduce ventilator-induced vascular leak and inflammation in vivo. Crit Care 12(1):R27 Pannekoek WJ, van Dijk JJ, Chan OY, Huveneers S, Linnemann JR, Spanjaard E, Brouwer PM, van der Meer AJ, Zwartkruis FJ, Rehmann H, de Rooij J, Bos JL (2011) Epac1 and PDZ-GEF cooperate in Rap1 mediated endothelial junction control. Cell Signal 23:2056–2064 Parnell E, Smith BO, Palmer TM, Terrin A, Zaccolo M, Yarwood SJ (2012) Regulation of the inflammatory response of vascular endothelial cells by EPAC1. Br J Pharmacol 166:434–446 Pyne S, Pyne N (2000) Sphingosine 1-phosphate signalling via the endothelial differentiation gene family of G-protein-coupled receptors. Pharmacol Ther 88:115–131 Qiao J, Huang F, Lum H (2003) PKA inhibits RhoA activation: a protection mechanism against endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 284:L972–L980 Rigor RR, Shen Q, Pivetti CD, Wu MH, Yuan SY (2012) Myosin light chain kinase signaling in endothelial barrier dysfunction. Med Res Rev 33:911–933 Risoe PK, Wang Y, Stuestol JF, Aasen AO, Wang JE, Dahle MK (2007) Lipopolysaccharide attenuates mRNA levels of several adenylyl cyclase isoforms in vivo. Biochim Biophys Acta 1772:32–39

Sayner SL, Alexeyev M, Dessauer CW, Stevens T (2006) Soluble adenylyl cyclase reveals the significance of cAMP compartmentation on pulmonary microvascular endothelial cell barrier. Circ Res 98:675–681 Schick MA, Wunder C, Wollborn J, Roewer N, Waschke J, Germer CT, Schlegel N (2012) Phosphodiesterase-4 inhibition as a therapeutic approach to treat capillary leakage in systemic inflammation. J Physiol 590:2693–2708 Schlegel N, Waschke J (2009a) Impaired cAMP and Rac 1 signaling contribute to TNF-alpha-induced endothelial barrier breakdown in microvascular endothelium. Microcirculation 16(6):521–533 Schlegel N, Waschke J (2009b) Impaired integrin-mediated adhesion contributes to reduced barrier properties in VASP-deficient microvascular endothelium. J Cell Physiol 220:357–366 Schlegel N, Waschke J (2009c) VASP is involved in cAMP-mediated Rac 1 activation in microvascular endothelial cells. Am J Physiol Cell Physiol 296(3):C453–C462 Schlegel N, Waschke J (2010) Vasodilator-stimulated phosphoprotein: crucial for activation of Rac1 in endothelial barrier maintenance. Cardiovasc Res 87(1):1–3 Schlegel N, Burger S, Golenhofen N, Walter U, Drenckhahn D, Waschke J (2008) The role of VASP in regulation of cAMP- and Rac 1mediated endothelial barrier stabilization. Am J Physiol Cell Physiol 294:C178–C188 Schlegel N, Baumer Y, Drenckhahn D, Waschke J (2009) Lipopolysaccharide-induced endothelial barrier breakdown is cAMP dependent in vivo and in vitro*. Crit Care Med 37(5): 1735–1743 Schlegel N, Leweke R, Meir M, Germer CT, Waschke J (2012) Role of NF-kappaB activation in LPS-induced endothelial barrier breakdown. Histochem Cell Biol 138:627–641 Schnoor M, Lai FP, Zarbock A, Klaver R, Polaschegg C, Schulte D, Weich HA, Oelkers JM, Rottner K, Vestweber D (2011) Cortactin deficiency is associated with reduced neutrophil recruitment but increased vascular permeability in vivo. J Exp Med 208:1721–1735 Secchi A, Ortanderl JM, Schmidt W, Walther A, Gebhard MM, Martin E, Schmidt H (2001) Effects of dobutamine and dopexamine on hepatic micro- and macrocirculation during experimental endotoxemia: an intravital microscopic study in the rat. Crit Care Med 29:597–600 Sehrawat S, Ernandez T, Cullere X, Takahashi M, Ono Y, Komarova Y, Mayadas TN (2011) AKAP9 regulation of microtubule dynamics promotes Epac1-induced endothelial barrier properties. Blood 117(2):708–718 Shen Q, Rigor RR, Pivetti CD, Wu MH, Yuan SY (2010) Myosin light chain kinase in microvascular endothelial barrier function. Cardiovasc Res 87:272–280 Singleton PA, Salgia R, Moreno-Vinasco L, Moitra J, Sammani S, Mirzapoiazova T, Garcia JGN (2007) CD44 regulates hepatocyte growth factor-mediated vascular integrity: ROLE OF c-Met, Tiam1/ Rac1, DYNAMIN 2, and CORTACTIN. J Biol Chem 282(42): 30643–30657 Singleton PA, Chatchavalvanich S, Fu P, Xing J, Birukova AA, Fortune JA, Klibanov AM, Garcia JG, Birukov KG (2009) Akt-mediated transactivation of the S1P1 receptor in caveolin-enriched microdomains regulates endothelial barrier enhancement by oxidized phospholipids. Circ Res 104:978–986 Spindler V, Waschke J (2011) Beta-adrenergic stimulation contributes to maintenance of endothelial barrier functions under baseline conditions. Microcirculation 18:118–127 Spindler V, Schlegel N, Waschke J (2010) Role of GTPases in control of microvascular permeability. Cardiovasc Res 87(2):243–253 Spindler V, Peter D, Harms GS, Asan E, Waschke J (2011) Ultrastructural analysis reveals cAMP-dependent enhancement of microvascular endothelial barrier functions via Rac1-mediated reorganization of intercellular junctions. Am J Pathol 178:2424–2436

Cell Tissue Res Starosta V, Wu T, Zimman A, Pham D, Tian X, Oskolkova O, Bochkov V, Berliner JA, Birukova AA, Birukov KG (2012) Differential regulation of endothelial cell permeability by high and low doses of oxidized 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphocholine. Am J Respir Cell Mol Biol 46:331–341 Stephens CT, Uwaydah N, Kramer GC, Prough DS, Salter M, Kinsky MP (2011) Vascular and extravascular volume expansion of dobutamine and norepinephrine in normovolemic sheep. Shock 36:303–311 Thurston G, Rudge JS, Ioffe E, Zhou H, Ross L, Croll SD, Glazer N, Holash J, McDonald DM, Yancopoulos GD (2000) Angiopoietin-1 protects the adult vasculature against plasma leakage. Nat Med 6: 460–463 Tian X, Tian Y, Sarich N, Wu T, Birukova AA (2012) Novel role of stathmin in microtubule-dependent control of endothelial permeability. FASEB J 26:3862–3874 Wang D, Luo P, Wang Y, Li W, Wang C, Sun D, Zhang R, Su T, Ma X, Zeng C, Wang H, Ren J, Cao F (2013) Glucagon-like peptide-1 protects against cardiac microvascular injury in diabetes via a cAMP/PKA/Rho-dependent mechanism. Diabetes 62:1697–1708 Ware LB, Matthay MA (2002) Keratinocyte and hepatocyte growth factors in the lung: roles in lung development, inflammation, and repair. Am J Physiol Lung Cell Mol Physiol 282(5):L924–L940 Waschke J, Baumgartner W, Adamson RH, Zeng M, Aktories K, Barth H, Wilde C, Curry FE, Drenckhahn D (2004a) Requirement of Rac

activity for maintenance of capillary endothelial barrier properties. Am J Physiol Heart Circ Physiol 286:H394–H401 Waschke J, Drenckhahn D, Adamson RH, Barth H, Curry FE (2004b) cAMP protects endothelial barrier functions by preventing Rac-1 inhibition. Am J Physiol Heart Circ Physiol 287:H2427–H2433 Waschke J, Drenckhahn D, Adamson RH, Curry FE (2004c) Role of adhesion and contraction in Rac 1-regulated endothelial barrier function in vivo and in vitro. Am J Physiol Heart Circ Physiol 287:H704–H711 Waschke J, Curry FE, Adamson RH, Drenckhahn D (2005) Regulation of actin dynamics is critical for endothelial barrier functions. Am J Physiol Heart Circ Physiol 288:H1296–H1305 Waschke J, Burger S, Curry FR, Drenckhahn D, Adamson RH (2006) Activation of Rac-1 and Cdc42 stabilizes the microvascular endothelial barrier. Histochem Cell Biol 125:397–406 Zebda N, Tian Y, Tian X, Gawlak G, Higginbotham K, Reynolds AB, Birukova AA, Birukov KG (2013) Interaction of p190RhoGAP with C-terminal domain of p120-catenin modulates endothelial cytoskeleton and permeability. J Biol Chem 288(25):18290–18299 Ziegler T, Horstkotte J, Schwab C, Pfetsch V, Weinmann K, Dietzel S, Rohwedder I, Hinkel R, Gross L, Lee S, Hu J, Soehnlein O, Franz WM, Sperandio M, Pohl U, Thomas M, Weber C, Augustin HG, Fassler R, Deutsch U, Kupatt C (2013) Angiopoietin 2 mediates microvascular and hemodynamic alterations in sepsis. J Clin Invest. doi:10.1172/JCI66549

cAMP with other signaling cues converges on Rac1 to stabilize the endothelial barrier- a signaling pathway compromised in inflammation.

cAMP is one of the most potent signaling molecules to stabilize the endothelial barrier, both under resting conditions as well as under challenge of b...
446KB Sizes 0 Downloads 0 Views