New insights into acute lung injury

4
Introduction New insights into acute lung injury Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) were rst identied over 40 years ago by Ashbaugh et al. (1967). ALI and ARDS are forms of progressive respiratory failure characterized by an acute onset of dyspnea, decreased arterial oxygen pressure (hypoxemia), bilateral inltrates on chest radiograms, and absence of clinical evidence of primary left heart failure. The American-European Consensus Conference on ARDS has led to a denition of ALI as a PaO 2 /FiO 2 b 300 while ARDS is dened as b 200 for ARDS (Bernard et al., 1994a; Bernard et al., 1994b). Thus, ARDS represents a subset of ALI patients with greater severity of symptoms. In ALI/ARDS, the integrity of the separation between the alveolus and the pulmonary circulation is compromised either by endothelial or epithelial injury or more commonly both. This damage leads to increased vascular permeability, alveolar ooding, and surfactant abnormalities (due to damage of type II pneumocytes). ALI/ARDS can occur in response to a number of insults that either directly or indirectly cause lung injury. The most common indirect insult leading to ALI is the release of lipopolysaccharide (LPS) from the outer cell wall of Gram-negative (G ) bacteria producing sepsis (Hudson et al., 1995). Other common causes include severe trauma with shock, multiple transfusions, burn injury, pneumonia and aspiration of gastric contents. Based on a recent announcement from the National Institute of Health (NIH), ALI and its more severe form, acute respiratory distress syndromes (ARDS) affect approximately 150,000 people in the United States every year. Nearly 28,500 are likely to die. Some survivors recover completely. However, others may have lasting damage to their lungs and additional health problems. Sepsis represents the systemic inammatory response to infection (Jacobi, 2002). Severe sepsis is dened as sepsis complicated by organ dysfunction and hypotension (septic shock). Lungs are among the most frequently affected organs in severe sepsis leading to ALI and ARDS (Martin et al., 2003). The incidence of sepsis has increased by 8.7% from 19792000 (Martin et al., 2003) and mortality ranges from 3050% (Angus et al., 2000; Annane et al., 2000; Rangel-Frausto et al., 1995). Clinical trials targeting inammatory mediators have shown no survival benet(Abraham et al., 1998; Abraham et al., 2001; Dhainaut et al., 1998; Fink, 1998; Fisher et al., 1994; McCloskey et al., 1994) and other strategies have failed to reduce morbidity associated with severe sepsis except for the survival benet that has been identied with low tidal volume mechanical ventilation (Jain & DalNogare, 2006) and the use of recombinant activated protein C (Bernard et al., 2001). With an unacceptably high mortality rate up to 58% (MacCallum & Evans, 2005) it is evident that a clearer understanding of both the mechanisms involved in the pathogenesis of ALI/ARDS and the development of new therapies for the control of the disease are critical. Thus, this special issue has brought together ve studies that are highlight the development of new reagents for the study of the mechanisms involved in ALI, elucidate new signaling pathways that may be involved in the pathophysiology of ALI, as well as evaluating potential new therapeutic agents to prevent the endothelial barrier disruption which is a hallmark of ALI. In this editorial I will briey describe the major ndings of each of the studies and attempt to highlight both the strengths and limitations of these individual studies and the eld of ALI as a whole. The initial site affected during the development of ALI/ARDS is the endothelial cell (EC) layer lining the micro-vessels in the lung. The vascular endothelium is a single-cell layer that acts as a semi-selective barrier between the plasma and interstitial uid. This function is critical for normal vessel wall homeostasis. Endothelial permeability is regulated by the balance between the contractile machinery within the cell and the elements that oppose contraction. The latter include tethering complexes that are responsible for cellcell and cell- substrate contacts and systems granting cell rigidity that prevent cell collapse, such as actin laments, microtubules and intermediate laments (Dudek et al., 2002; Birukov et al., 2002). One of the major limitations in the study of ALI/ARDS is the lack of a reproducible cell culture model that can be used to investigate how EC that line the micro-vessels in the pulmonary system are disrupted by ALI/ARDS. However, a signicant number of published studies utilize pulmonary EC of either bovine or human origin but are isolated from the major vessels such as the pulmonary artery. These vessels are not normally affected by ALI/ARDS. Further, the commercially available EC isolated from human micro-vessels do not appear to maintain the archetypal properties of EC namely their cobblestone appearance. Further, the currently available EC lines require large quantities of agents such as LPS (100 endotoxin units (EU)/ml) for measureable barrier disruption to occur. However, Catravas et al in this issue describe a new method for harvesting and culturing of human lung microvas- cular endothelial cells (HLMVEC). Further, they present convincing data demonstrating the identity of these cells and their response to appropriate stimuli. These HLMVEC appear to be of superior quality to other available cells exhibiting small size, characteristic cobblestone appearance, and a contact-inhibited monolayer. Further, these characteristics are maintained over multiple passages. The HLMVEC exhibited a tight monolayer with excellent transendothelial resis- tance (TER) (1000MΩ) measured using an Applied Biosystems ECIS instrument. Most excitingly, freshly harvested HLMVEC demonstrated excellent sensitivity to a variety of barrier disrupting agents. For example as little as 1 EU/ml elicited a profound decrease in TER. These cells are likely to become the gold-standardfor investigations into the mechanisms underlying the endothelial barrier disruption in ALI/ ARDS. Although extensive investigations have been carried out to delineate the mechanisms underlying the development of ALI/ARDS Vascular Pharmacology 52 (2010) 171174 The Endothelium & ALI 1537-1891/$ see front matter © 2010 Elsevier Inc. All rights reserved. doi:10.1016/j.vph.2010.02.002 Contents lists available at ScienceDirect Vascular Pharmacology journal homepage: www.elsevier.com/locate/vph

Transcript of New insights into acute lung injury

Page 1: New insights into acute lung injury

Vascular Pharmacology 52 (2010) 171–174

TheEndothelium

&ALI

Contents lists available at ScienceDirect

Vascular Pharmacology

j ourna l homepage: www.e lsev ie r.com/ locate /vph

Introduction

New insights into acute lung injury

Acute lung injury (ALI) and acute respiratory distress syndrome(ARDS) were first identified over 40 years ago by Ashbaugh et al.(1967). ALI and ARDS are forms of progressive respiratory failurecharacterized by an acute onset of dyspnea, decreased arterial oxygenpressure (hypoxemia), bilateral infiltrates on chest radiograms, andabsence of clinical evidence of primary left heart failure. TheAmerican-European Consensus Conference on ARDS has led to adefinition of ALI as a PaO2/FiO2b300while ARDS is defined as b200 forARDS (Bernard et al., 1994a; Bernard et al., 1994b). Thus, ARDSrepresents a subset of ALI patients with greater severity of symptoms.In ALI/ARDS, the integrity of the separation between the alveolus andthe pulmonary circulation is compromised either by endothelial orepithelial injury or more commonly both. This damage leads toincreased vascular permeability, alveolar flooding, and surfactantabnormalities (due to damage of type II pneumocytes). ALI/ARDS canoccur in response to a number of insults that either directly orindirectly cause lung injury. The most common indirect insult leadingto ALI is the release of lipopolysaccharide (LPS) from the outer cellwall of Gram-negative (G−) bacteria producing sepsis (Hudson et al.,1995). Other common causes include severe trauma with shock,multiple transfusions, burn injury, pneumonia and aspiration ofgastric contents. Based on a recent announcement from the NationalInstitute of Health (NIH), ALI and its more severe form, acuterespiratory distress syndromes (ARDS) affect approximately 150,000people in the United States every year. Nearly 28,500 are likely to die.Some survivors recover completely. However, othersmay have lastingdamage to their lungs and additional health problems. Sepsisrepresents the systemic inflammatory response to infection (Jacobi,2002). Severe sepsis is defined as sepsis complicated by organdysfunction and hypotension (septic shock). Lungs are among themost frequently affected organs in severe sepsis leading to ALI andARDS (Martin et al., 2003). The incidence of sepsis has increased by8.7% from 1979–2000 (Martin et al., 2003) and mortality ranges from30–50% (Angus et al., 2000; Annane et al., 2000; Rangel-Frausto et al.,1995). Clinical trials targeting inflammatory mediators have shownno survival benefit (Abraham et al., 1998; Abraham et al., 2001;Dhainaut et al., 1998; Fink, 1998; Fisher et al., 1994; McCloskey et al.,1994) and other strategies have failed to reduce morbidity associatedwith severe sepsis except for the survival benefit that has beenidentified with low tidal volume mechanical ventilation (Jain &DalNogare, 2006) and the use of recombinant activated protein C(Bernard et al., 2001). With an unacceptably high mortality rate up to58% (MacCallum & Evans, 2005) it is evident that a clearerunderstanding of both the mechanisms involved in the pathogenesisof ALI/ARDS and the development of new therapies for the control ofthe disease are critical. Thus, this special issue has brought togetherfive studies that are highlight the development of new reagents for

1537-1891/$ – see front matter © 2010 Elsevier Inc. All rights reserved.doi:10.1016/j.vph.2010.02.002

the study of the mechanisms involved in ALI, elucidate new signalingpathways that may be involved in the pathophysiology of ALI, as wellas evaluating potential new therapeutic agents to prevent theendothelial barrier disruption which is a hallmark of ALI. In thiseditorial I will briefly describe themajor findings of each of the studiesand attempt to highlight both the strengths and limitations of theseindividual studies and the field of ALI as a whole.

The initial site affected during the development of ALI/ARDS is theendothelial cell (EC) layer lining the micro-vessels in the lung. Thevascular endothelium is a single-cell layer that acts as a semi-selectivebarrier between the plasma and interstitial fluid. This function iscritical for normal vessel wall homeostasis. Endothelial permeabilityis regulated by the balance between the contractile machinery withinthe cell and the elements that oppose contraction. The latter includetethering complexes that are responsible for cell–cell and cell-substrate contacts and systems granting cell rigidity that preventcell collapse, such as actin filaments, microtubules and intermediatefilaments (Dudek et al., 2002; Birukov et al., 2002). One of the majorlimitations in the study of ALI/ARDS is the lack of a reproducible cellculture model that can be used to investigate how EC that line themicro-vessels in the pulmonary system are disrupted by ALI/ARDS.However, a significant number of published studies utilize pulmonaryEC of either bovine or human origin but are isolated from the majorvessels such as the pulmonary artery. These vessels are not normallyaffected by ALI/ARDS. Further, the commercially available EC isolatedfrom human micro-vessels do not appear to maintain the archetypalproperties of EC namely their cobblestone appearance. Further, thecurrently available EC lines require large quantities of agents such asLPS (∼100 endotoxin units (EU)/ml) for measureable barrierdisruption to occur. However, Catravas et al in this issue describe anew method for harvesting and culturing of human lung microvas-cular endothelial cells (HLMVEC). Further, they present convincingdata demonstrating the identity of these cells and their response toappropriate stimuli. These HLMVEC appear to be of superior quality toother available cells exhibiting small size, characteristic cobblestoneappearance, and a contact-inhibited monolayer. Further, thesecharacteristics are maintained over multiple passages. The HLMVECexhibited a tight monolayer with excellent transendothelial resis-tance (TER) (∼1000MΩ) measured using an Applied Biosystems ECISinstrument. Most excitingly, freshly harvested HLMVEC demonstratedexcellent sensitivity to a variety of barrier disrupting agents. Forexample as little as 1 EU/ml elicited a profound decrease in TER. Thesecells are likely to become the “gold-standard” for investigations intothe mechanisms underlying the endothelial barrier disruption in ALI/ARDS.

Although extensive investigations have been carried out todelineate the mechanisms underlying the development of ALI/ARDS

Page 2: New insights into acute lung injury

TheEndothelium

&ALI

172 Introduction

little of therapeutic value has resulted from these efforts. Thus, newsignaling pathways may need to be elucidated and tested for theirinterventional potential. Thus, the remaining four studies in this serieshave begun to evaluate new mechanisms of endothelial protection inALI/ARDS. In the first of these studies Sharma and colleagues haveexamined the role of protein nitration in the pathophysiology of ALI/ARDS. The nitration of tyrosine residues is mediated by reactivenitrogen species (RNS). Increased RNS production occurs whendysregulated nitric oxide (NO) production reacts with reactiveoxygen species (ROS) such as superoxide. This reaction generatesRNS, including peroxynitrite (ONOO–). ONOO– leads to tyrosinenitration, a covalent modification that adds a nitro group (–NO2) toone ortho carbon of the phenolic ring of tyrosine to form 3-nitrotyrosine (3-NT). This introduces a net negative charge to thetyrosine, altering structural properties and catalytic activity of theprotein. There has been increasing interest in the effects of tyrosinenitration on changes in protein structure in diverse pathologicconditions. The nitration of tyrosine residues to form 3-NT residuesis widely used as a marker of ONOO– formation and Sharma et al havedemonstrated that elevated levels of protein tyrosine nitrationprecedes the endothelial injury associated with the development ofALI in the LPS injected mouse. This increase in ONOO– appears to berelated to eNOS uncoupling mediated via an increase in the levels ofthe endogenous NOS inhibitor asymmetric dimethyargine (ADMA).Further, the increases in ADMA occurs secondary to a post-translational inhibition of the enzyme that degrades ADMA, dimethy-larginine dimethylaminohydrolase (DDAH). Inhibition of DDAHwithout alteration of its gene expression is becoming more widelyappreciated as Lin and colleagues have shown that elevated glucoseraises endothelial ADMA levels by inhibiting DDAH activity via amechanism involving oxidative stress (Lin et al., 2002). Similarly, LPSsignificantly increases the levels of ADMA and decreased DDAHactivity in cultured medium from human endothelial cells (Xin et al.,2007). Of great clinical interest is a recent study indicating that ADMAlevels are elevated in patients with septic shock (O'Dwyer et al., 2006)suggesting that the ADMA-DDAH pathway could be responsive totherapeutic intervention. Further, the studies of Church et alinvestigating the effect of phosphatase and tensin homologue deletedon chromosome 10 (PTEN) signaling on eNOS activity are intriguingwith respect to DDAH activity. PTEN is a lipid phosphatase thatfunctions as a negative regulator of the phosphoinositide-3-kinase(PI3K) pathway. Although most studies on PTEN have focused on itsrole in cancer progressionwhere it is found in the advanced stages of anumber of cancers (Li et al., 1997) it is now becoming apparent thatPTEN can also modulate the blood vessel structure and function(Marsh et al., 1999; Zhong et al., 2000; Tsigkos et al., 2006). Inaddition, some recent studies have shown a role for PTEN in theprogression of ALI as both the pharmacological inhibition of PTEN andits conditional ablation in the epithelia of the lung have been shown toreduce the severity of ALI (Lai et al., 2009; Tiozzo et al., 2009). Thedata indicating that enhancing PTEN signaling decreases NO gener-ation from eNOS through the inhibition of Akt-mediated phosphor-ylation correlates well with the decrease in eNOS-derived NO thatSharma et al have shown to be an early event after I.P. LPS in themouse lung. Although Church et al have focused on changes in seinephosphorylation to explain the decreases in NO induced by PTENover-expression in COS-7 cells, the potential of PTEN to inhibit DDAHactivity is an intriguing but unexplored possibility.

The Sharma study also highlights the limitations of currentinvestigations: the lack of easily available means to identify posttranslational modifications of proteins. This is a major roadblock toour understanding of the potential mechanistic contributions of thesemodifications to disease processes. New methodologies are neededthat go beyond the limitations of current analytical approaches thatfocus on measuring either global changes in protein nitration (as inthe Sharma study) or merely identifying individual proteins that are

susceptible to nitration. Rather priority should be placed on movingtowards identifying both the individual tyrosine residues targeted bynitration and the effect these nitration events have on the structure-function relationship of the nitrated protein.

Recently, attention has been given to the therapeutic potential ofpurinergic agonists in the treatment of cardiovascular and pulmonarydiseases (Burnstock, 2008; Raju et al., 2008; Smyth et al., 2009;Kolosova et al., 2008) and the study in this issue from Umapathy et alhas evaluated the barrier protective properties of adenosine in vitrousing cultured human PAEC. Extracellular purines can function asintercellular signaling molecules when released from differentsources in the body (Burnstock & Williams, 2000) and accumulatingexperimental data suggest that purines could be barrier-protectiveagents against the effects of ALI, as they are present in the ECmicroenvironment in vivo and they decrease permeability in vitro.The dominant pathway modulating the levels of extracellularadenosine is the extracellular catabolism of ATP to adenosine throughthe progressive action of a number of ectonucleotidases (Eltzschig etal., 2004; Thomson et al., 2000). In this study Umapathy et al clearlydemonstrate that the addition of adenosine in physiologically relevantconcentrations (1–5 µM) significantly increases the TER of culturedhuman PAEC. Further, using an elegant siRNA strategy they confirmthat the mechanism of action is mediated via A2A receptors and acAMP-dependent signaling pathway that produces changes in F-actinvia the activation of myosin light chain phosphatase (MLCP). Thisreport also emphasizes an important limitation in our currentknowledge of the role of phosphatases in the pathophysiology ofALI. This is primarily due to the inherent technical difficulties instudying the action of protein phosphatases. MLCP is a type 1 Ser/ThrPPase and the holoenzyme is composed of 3 subunits: a catalyticsubunit (CS1) of 38 kD that was identified as CS1δ isoform (currentlyCS1β) and two non-catalytic subunits of 20–21 and 110–130 kD(Alessi et al., 1992; Shimizu et al., 1994; Shirazi et al., 1994;Hartshorne et al., 1998). The 110–130 kD non-catalytic subunit, calledmyosin PPase targeting subunit 1 (MYPT1), binds to CS1 and thistargets CS1 to MLC and provides substrate specificity (Hubbard &Cohen, 1993; Johnson et al., 1997). Human MYPT1 and its splicevariants are encoded by a single gene on human chromosome 12q15–q21.2 (Takahashi et al., 1997). It is well established that MLC is themajor substrate for MLCP. However, recent data have revealed thatMYPT1 can also bind directly to the ezrin/radixin/moesin (ERM)family of actin-binding proteins (Fukata et al., 1998; Kimura et al.,1998). ERM proteins act as linkers between the actin cytoskeleton andplasma membrane proteins, and as signal transducers in responsesinvolving cytoskeletal remodeling (Bretscher et al., 2002) as Uma-pathy et al have shownwith adenosine. Established a role of the ERMsin the barrier protective effect of adenosine could yield novel potentialtherapeutic targets for ALI. However, it should be noted that a majorlimitation of the Umapathy study is the fact that there was no in vivoconfirmation of the barrier enhancement or protective effects in vivo.Hopefully these important confirmatory studies will be soonforthcoming.

One of the major limitations in our understanding of ALI/ARDS isthat much of the work is carried out using LPS as a model of G− sepsisad studies on G+ infections are less common in the literature. Thus,the final study fromXiong et al is potentially very powerful and timelyin that it has identified a new signaling mechanism and potentialtherapy for the treatment of G+ associated bacterial infections usingthe G+ pore forming toxin, Listeriolysin-O (LLO) isolated from Listeriamonocytogenes, a bacterium that causes a severe food-borne disease inneonates characterized by meningitis and meningo-encephalitis. Thedata presented indicate that the oxidative stress associated with LLOis mediated via the activation of PKCα and that its pharmacologicinhibition using GÖ6976, attenuates the LLO-mediated decreases inTER in the HLMVEC isolated by Catravas et al. Further, and ofsignificant therapeutic interest, the authors have shown that the

Page 3: New insights into acute lung injury

TheEndothelium

&ALI

173Introduction

lectin like domain of TNF-the TIP peptide- prevents the LLO-mediatedincrease in oxidative stress, at least in part, by preventing the up-regulation of Nox-4. However, the mechanism by which the TIPpeptide exerts its barrier protective effect has been only partiallyelucidated as the TIP peptide prevented the LLO-mediated activationof PKCα but did not prevent the influx of intracellular calcium knownto play a key role its activation. Similar to the report by Umapathy etal., this study was also in vitro and did not evaluate the barrierprotective effects of the TIP peptide in vivo in LLO challenged mice.Again it is hoped that these studies will be forthcoming soon.

In conclusion, this special issue focuses on the role of theendothelium in ALI/ARDS and highlights the complexity of the diseaseprocess and the problemswith developing single therapies targeted atalleviating the significant morbidity and mortality associated withALI/ARDS. Several new signaling pathways that could have therapeu-tic potential (DDAH, PTEN, PKCα) are investigated in humanmicrovascular endothelial cells that are isolated from humanpulmonary microvessels and are significantly more relevant to thisetiology of this disease. Finally, new agents are tested for their abilityto prevent endothelial barrier disruption in vitro (adenosine, TIPpeptide) and in vivo (peroxynitrite scavengers) against agentsderived from both G− and G+ bacteria. It is hoped that as thesestudies progress they will begin to elucidate mechanistic similaritiesand differences between the effects of G− and G+ infections and thatthese mechanisms will provide a pathway to new and efficacioustherapeutic strategies that integrates multiple approaches.

Acknowledgements

This work was supported in part by grants HL60190 (to SMB),HL67841, HL72123, HL70061, HL084739, and R21HD057406 all fromthe National Institutes of Health, by a Transatlantic NetworkDevelopment Grant from the Fondation Leducq, and by a Program-matic Development award from the from the CardiovascularDiscovery Institute of the Medical College of Georgia.

References

Ashbaugh, D.G.B.D., Petty, T.L., Levine, B.E., 1967. Acute respiratory distress in adults.Lancet 2 (7511), 319–323.

Bernard, G.R., Artigas, A., Brigham, K.L., Carlet, J., Falke, K., Hudson, L., Lamy, M., Legall,J.R., Morris, A., Spragg, R., 1994. The American-European Consensus Conferenceon ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination.Am. J. Respir. Crit. Care Med. 149 (3 Pt 1), 818–824.

Bernard, G.R., Artigas, A., Brigham, K.L., Carlet, J., Falke, K., Hudson, L., Lamy, M., LeGall, J.R.,Morris, A., Spragg, R., 1994. Report of the American-European Consensus conferenceon acute respiratory distress syndrome: definitions, mechanisms, relevant outcomes,and clinical trial coordination. Consensus Committee. J. Crit. Care 9 (1), 72–81.

Hudson, L.D., Milberg, J.A., Anardi, D., Maunder, R.J., 1995. Clinical risks for developmentof the acute respiratory distress syndrome. Am. J. Respir. Crit. CareMed. 151 (2 Pt 1),293–301.

Jacobi, J., 2002. Pathophysiology of sepsis. Am. J. Health Syst. Pharm. 59 (Suppl 1), S3–S8.Martin, G.S., Mannino, D.M., Eaton, S., Moss, M., 2003. The epidemiology of sepsis in the

United States from 1979 through 2000. N. Engl. J. Med. 348 (16), 1546–1554.Angus, D.C., Birmingham, M.C., Balk, R.A., Scannon, P.J., Collins, D., Kruse, J.A., Graham,

D.R., Dedhia, H.V., Homann, S., MacIntyre, N., 2000. E5 murine monoclonalantiendotoxin antibody in gram-negative sepsis: a randomized controlled trial.E5 Study Investigators. Jama 283 (13), 1723–1730.

Annane, D., Sebille, V., Troche, G., Raphael, J.C., Gajdos, P., Bellissant, E., 2000. A 3-levelprognostic classification in septic shock based on cortisol levels and cortisolresponse to corticotropin. Jama 283 (8), 1038–1045.

Rangel-Frausto, M.S., Pittet, D., Costigan, M., Hwang, T., Davis, C.S., Wenzel, R.P., 1995.The natural history of the systemic inflammatory response syndrome (SIRS). Aprospective study. Jama 273 (2), 117–123.

Abraham, E., Anzueto, A., Gutierrez, G., Tessler, S., San Pedro,G.,Wunderink, R., Dal Nogare,A., Nasraway, S., Berman, S., Cooney, R., Levy, H., Baughman, R., Rumbak,M., Light, R.B.,Poole, L., Allred, R., Constant, J., Pennington, J., Porter, S., 1998. Double-blindrandomised controlled trial of monoclonal antibody to human tumour necrosis factorin treatment of septic shock. NORASEPT II Study Group. Lancet 351 (9107), 929–933.

Abraham, E., Reinhart, K., Svoboda, P., Seibert, A., Olthoff, D., Dal Nogare, A., Postier, R.,Hempelmann, G., Butler, T., Martin, E., Zwingelstein, C., Percell, S., Shu, V., Leighton,A., Creasey, A.A., 2001. Assessment of the safety of recombinant tissue factorpathway inhibitor in patients with severe sepsis: a multicenter, randomized,

placebo-controlled, single-blind, dose escalation study. Crit. Care Med. 29 (11),2081–2089.

Dhainaut, J.F., Tenaillon, A., Hemmer, M., Damas, P., Le Tulzo, Y., Radermacher, P., Schaller,M.D., Sollet, J.P.,Wolff,M., Holzapfel, L., Zeni, F., Vedrinne, J.M., de Vathaire, F., Gourlay,M.L., Guinot, P., Mira, J.P., 1998. Confirmatory platelet-activating factor receptorantagonist trial in patients with severe gram-negative bacterial sepsis: a phase III,randomized, double-blind, placebo-controlled, multicenter trial. BN 52021 SepsisInvestigator Group. Crit. Care Med. 26 (12), 1963–1971.

Fink, M.P., 1998. Therapeutic options directed against platelet activating factor,eicosanoids and bradykinin in sepsis. J. Antimicrob. Chemother. 41 (Suppl A),81–94.

Fisher Jr., C.J., Opal, S.M., Lowry, S.F., Sadoff, J.C., LaBrecque, J.F., Donovan, H.C.,Lookabaugh, J.L., Lemke, J., Pribble, J.P., Stromatt, S.C., et al., 1994. Role ofinterleukin-1 and the therapeutic potential of interleukin-1 receptor antagonistin sepsis. Circ. Shock 44 (1), 1–8.

McCloskey, R.V., Straube, R.C., Sanders, C., Smith, S.M., Smith, C.R., 1994. Treatment ofseptic shock with human monoclonal antibody HA-1A. A randomized, double-blind, placebo-controlled trial. CHESS Trial Study Group. Ann. Intern. Med. 121 (1),1–5.

Jain, R., DalNogare, A., 2006. Pharmacological therapy for acute respiratory distresssyndrome. Mayo Clin. Proc. 81 (2), 205–212.

Bernard, G.R., Vincent, J.L., Laterre, P.F., LaRosa, S.P., Dhainaut, J.F., Lopez-Rodriguez, A.,Steingrub, J.S., Garber, G.E., Helterbrand, J.D., Ely, E.W., Fisher Jr, C.J., 2001. Efficacyand safety of recombinant human activated protein C for severe sepsis. N. Engl. J.Med. 344 (10), 699–709.

MacCallum, N.S., Evans, T.W., 2005. Epidemiology of acute lung injury. Curr. Opin. Crit.Care 11 (1), 43–49.

Dudek, S.M., Birukov, K.G., Zhan, X., Garcia, J.G., 2002. Novel interaction of cortactinwith endothelial cell myosin light chain kinase. Biochem. Biophys. Res. Commun.298 (4), 511–519.

Birukov, K.G., Birukova, A.A., Dudek, S.M., Verin, A.D., Crow, M.T., Zhan, X., DePaola, N.,Garcia, J.G., 2002. Shear stress-mediated cytoskeletal remodeling and cortactintranslocation in pulmonary endothelial cells. Am. J. Respir. Cell Mol. Biol. 26 (4),453–464.

Lin, K.Y., Ito, A., Asagami, T., Tsao, P.S., Adimoolam, S., Kimoto, M., Tsuji, H., Reaven, G.M.,Cooke, J.P., 2002. Impaired nitric oxide synthase pathway in diabetes mellitus: roleof asymmetric dimethylarginine and dimethylarginine dimethylaminohydrolase.Circulation 106 (8), 987–992.

Xin, H.Y., Jiang, D.J., Jia, S.J., Song, K., Wang, G.P., Li, Y.J., Chen, F.P., 2007. Regulation byDDAH/ADMA pathway of lipopolysaccharide-induced tissue factor expression inendothelial cells. Thromb. Haemost. 97 (5), 830–838.

O'Dwyer, M.J., Dempsey, F., Crowley, V., Kelleher, D.P., McManus, R., Ryan, T., 2006.Septic shock is correlated with asymmetrical dimethyl arginine levels, which maybe influenced by a polymorphism in the dimethylarginine dimethylaminohydro-lase II gene: a prospective observational study. Crit. Care 10 (5), R139.

Li, J., Yen, C., Liaw, D., Podsypanina, K., Bose, S., Wang, S.I., Puc, J., Miliaresis, C., Rodgers, L.,McCombie, R., Bigner, S.H., Giovanella, B.C., Ittmann, M., Tycko, B., Hibshoosh, H.,Wigler, M.H., Parsons, R., 1997. PTEN, a putative protein tyrosine phosphatase genemutated in human brain, breast, and prostate cancer. Science 275 (5308), 1943–1947.

Marsh, D.J., Kum, J.B., Lunetta, K.L., Bennett, M.J., Gorlin, R.J., Ahmed, S.F., Bodurtha, J.,Crowe, C., Curtis, M.A., Dasouki, M., Dunn, T., Feit, H., Geraghty, M.T., Graham Jr.,J.M., Hodgson, S.V., Hunter, A., Korf, B.R., Manchester, D., Miesfeldt, S., Murday,V.A., Nathanson, K.L., Parisi, M., Pober, B., Romano, C., Eng, C., et al., 1999. PTENmutation spectrum and genotype–phenotype correlations in Bannayan–Riley–Ruvalcaba syndrome suggest a single entity with Cowden syndrome. Hum. Mol.Genet. 8 (8), 1461–1472.

Zhong, H., Chiles, K., Feldser, D., Laughner, E., Hanrahan, C., Georgescu, M.M., Simons,J.W., Semenza, G.L., 2000. Modulation of hypoxia-inducible factor 1alpha expressionby the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAPpathway in human prostate cancer cells: implications for tumor angiogenesis andtherapeutics. Cancer Res. 60 (6), 1541–1545.

Tsigkos, S., Zhou, Z., Kotanidou, A., Fulton, D., Zakynthinos, S., Roussos, C.,Papapetropoulos, A., 2006. Regulation of Ang2 release by PTEN/PI3-kinase/Akt inlung microvascular endothelial cells. J. Cell. Physiol. 207 (2), 506–511.

Lai, J.P., Bao, S., Davis, I.C., Knoell, D.L., 2009. Inhibition of the phosphatase PTEN protectsmice against oleic acid-induced acute lung injury. Br. J. Pharmacol. 156 (1),189–200.

Tiozzo, C., De Langhe, S., Yu, M., Londhe, V.A., Carraro, G., Li, M., Li, C., Xing, Y., Anderson,S., Borok, Z., Bellusci, S., Minoo, P., 2009. Deletion of Pten expands lung epithelialprogenitor pools and confers resistance to airway injury. Am. J. Respir. Crit. CareMed.

Burnstock, G., 2008. Purinergic receptors as future targets for treatment of functional GIdisorders. Gut 57 (9), 1193–1194.

Raju, N.C., Eikelboom, J.W., Hirsh, J., 2008. Platelet ADP-receptor antagonists forcardiovascular disease: past, present and future. Nat. Clin. Pract. Cardiovasc. Med. 5(12), 766–780.

Smyth, S.S., Woulfe, D.S., Weitz, J.I., Gachet, C., Conley, P.B., Goodman, S.G., Roe, M.T.,Kuliopulos, A., Moliterno, D.J., French, P.A., Steinhubl, S.R., Becker, R.C., 2009. G-protein-coupled receptors as signaling targets for antiplatelet therapy. Arterioscler.Thromb. Vasc. Biol. 29 (4), 449–457.

Burnstock, G., Williams, M., 2000. P2 purinergic receptors: modulation of cell functionand therapeutic potential. J. Pharmacol. Exp. Ther. 295 (3), 862–869.

Eltzschig, H.K., Thompson, L.F., Karhausen, J., Cotta, R.J., Ibla, J.C., Robson, S.C., Colgan,S.P., 2004. Endogenous adenosine produced during hypoxia attenuates neutro-phil accumulation: coordination by extracellular nucleotide metabolism. Blood104 (13), 3986–3992.

Page 4: New insights into acute lung injury

TheEndothelium

&ALI

174 Introduction

Thomson, S., Bao, D., Deng, A., Vallon, V., 2000. Adenosine formed by 5′-nucleotidasemediates tubuloglomerular feedback. J. Clin. Invest. 106 (2), 289–298.

Alessi, D., MacDougall, L.K., Sola, M.M., Ikebe, M., Cohen, P., 1992. The control of proteinphosphatase-1 by targetting subunits. The major myosin phosphatase in aviansmooth muscle is a novel form of protein phosphatase-1. Eur. J. Biochem. / FEBS210 (3), 1023–1035.

Shimizu, H., Ito, M., Miyahara, M., Ichikawa, K., Okubo, S., Konishi, T., Naka, M., Tanaka,T., Hirano, K., Hartshorne, D.J., et al., 1994. Characterization of the myosin-bindingsubunit of smooth muscle myosin phosphatase. J. Biol. Chem. 269 (48),30407–30411.

Shirazi, A., Iizuka, K., Fadden, P., Mosse, C., Somlyo, A.P., Somlyo, A.V., Haystead, T.A.,1994. Purification and characterization of the mammalian myosin light chainphosphatase holoenzyme. The differential effects of the holoenzyme and itssubunits on smooth muscle. J. Biol. Chem. 269 (50), 31598–31606.

Hartshorne, D.J., Ito, M., Erdodi, F., 1998. Myosin light chain phosphatase: subunitcomposition, interactions and regulation. J. Muscle Res. Cell Motil. 19 (4), 325–341.

Hubbard, M.J., Cohen, P., 1993. On target with a new mechanism for the regulation ofprotein phosphorylation. Trends Biochem. Sci. 18 (5), 172–177.

Johnson, D., Cohen, P., Chen, M.X., Chen, Y.H., Cohen, P.T., 1997. Identification of theregions on the M110 subunit of protein phosphatase 1 M that interact with theM21 subunit and with myosin. Eur. J. Biochem. / FEBS 244 (3), 931–939.

Takahashi, N., Ito, M., Tanaka, J., Nakano, T., Kaibuchi, K., Odai, H., Takemura, K., 1997.Localization of the gene coding for myosin phosphatase, target subunit 1 (MYPT1)to human chromosome 12q15–q21. Genomics 44 (1), 150–152.

Fukata, Y., Kimura, K., Oshiro, N., Saya, H., Matsuura, Y., Kaibuchi, K., 1998. Association ofthe myosin-binding subunit of myosin phosphatase and moesin: dual regulation ofmoesin phosphorylation by Rho-associated kinase and myosin phosphatase. J. CellBiol. 141 (2), 409–418.

Kimura, K., Fukata, Y., Matsuoka, Y., Bennett, V., Matsuura, Y., Okawa, K., Iwamatsu, A.,Kaibuchi, K., 1998. Regulation of the association of adducin with actin filamentsby Rho-associated kinase (Rho-kinase) and myosin phosphatase. J. Biol. Chem.273 (10), 5542–5548.

Bretscher, A., Edwards, K., Fehon, R.G., 2002. ERM proteins and merlin: integrators atthe cell cortex. Nat. Rev. 3 (8), 586–599.

Kolosova, I.A., Mirzapoiazova, T., Moreno-Vinasco, L., Sammani, S., Garcia, J.G.N., Verin,A.D., 2008. Protective effect of purinergic agonist ATPγS against acute lung injury.Am. J. Physiol. Lung Cell Mol. Physiol. 294 (2), L319–324.

Stephen M. BlackProgram in Pulmonary Disease, Vascular Biology Center,

1459 Laney Walker Blvd, CB3201B, Medical College of Georgia, Augusta,GA 30912, USA

E-mail address: [email protected].