10,760
Views
133
CrossRef citations to date
0
Altmetric
Review Article

Biomarkers of endothelial dysfunction: can they help us deciphering systemic inflammation and sepsis?

, &
Pages S11-S21 | Received 14 Dec 2010, Accepted 10 May 2011, Published online: 28 Jun 2011

Abstract

The endothelial integrity, as mechanical barrier against microorganisms and as natural “anticoagulant”, is crucial for physiologic organ function. Systemic activation of the endothelium upon inflammation, sepsis, and septic shock is always ending in blood–tissue barrier disruption. With increasing dysfunction, uncontrolled clotting activation, capillary microthrombi formation, tissue edema, local hypoxia, and ischemia are initiated. This in turn enhances a vicious circle leading to multiple organ failure and death.

Therefore, biomarkers reflecting this special compartment may help in the early detection of systemic inflammation and its complications. This review provides an overview of the most important endothelial biomarkers and their possible use in sepsis.

Introduction

Sepsis represents the third leading cause of death in the industrialized countries (CitationAnderson 2002; CitationAndreu Ballester et al. 2008; Angus Citation2001a, Citation2001b; CitationDombrovskiy et al. 2007; CitationEngel et al. 2007; CitationMcDonald et al. 2005). The early diagnosis and therapy of sepsis still remain a major challenge, where time plays a crucial role (CitationKumar et al. 2006; CitationLever and Mackenzie 2007; CitationZambon et al. 2008). It is well known that an increase in the number of failing organs during the first 48 h following admission to an intensive care unit (ICU) is a good indicator of mortality in septic patients (CitationFerreira et al. 2001). Therefore, the early diagnosis of sepsis and prevention of organ dysfunction progression to multiorgan failure (MOF) is the primary goal in the treatment of septic patients (CitationSlade et al. 2003). In the daily clinical routine, many markers reflecting i.e. immune status, liver, kidney, or heart failure are commonly used to efficaciously detect the course of sepsis. However, one of the largest and most important organs, the endothelial and vascular system, is not routinely tracked in the daily clinical routine. No recommendations about measuring the extent of endothelial damage are embedded in the latest sepsis guidelines.

As one of the largest “organs”, the vascular and capillary system occupies a central role in the homeostasis of organ functions. It provides a mechanical barrier, which keeps liquids within the vasculature, ensuring transport of nutrients to the different organs. It builds up a natural barrier that prevents microorganisms to invade tissues and it exerts a natural anticoagulant action that prevents from uncontrolled activation of the coagulation system.

Almost every stimulus leading to a systemic inflammatory response i.e. severe infection, trauma, excessive tissue breakdown, solid tumors, leukemia, pregnancy-associated complications, vascular anomalies, liver failure, and toxicological or immunological responses and activation of the coagulation system can be associated with endothelial damage. Systemic inflammation and sepsis are mostly accompanied by an overwhelming activation of the coagulation system, preceded by the endothelial dysfunction and the loss of its antithrombotic properties (CitationKnoebl 2010; CitationLevi 2010; CitationLevi et al. 2002; CitationLevi and van der Poll 2010). Clinically, this pathology first manifests i.e. as a general hemostasis reaction that may vary greatly from a simple drop in platelet counts or subclinical prolongation in clotting times to the full picture of a disseminated intravascular coagulation (DIC) and, finally ends up in a global consumption of clotting factors (CitationLevi 2007; CitationLevi and Ten Cate 1999; CitationTaylor et al. 2001). During sepsis, the release of endotoxin or proinflammatory cytokines initiates an activation cascade in endothelial cells (ECs) leading to impaired homeostasis (CitationInce 2002; CitationTyagi et al. 2009). Due to rapid changes in endothelial function (early contraction followed by a rapid loss of function), microthrombi occur by activation of the clotting system leading to changes in blood rheology and causing organ failure due to misperfusion (CitationLee and Slutsky 2010; CitationLehr et al. 2000). Thus, in septic patients, levels of factors that directly reflect endothelial dysfunction might be changed (CitationFareed et al. 1998; CitationIba et al. 2005).

In sepsis, endothelial activation and dysfunction are critical determinants of the host response and represent an explanation for the complex sepsis pathophysiology (CitationFareed et al. 1998; CitationIba et al. 2005; CitationShapiro et al. 2010). Given this central place of the endothelium, biomarkers reflecting ECs’ state might be useful in the tracking of sepsis (CitationPierrakos and Vincent 2010). However, direct markers reflecting endothelial damage are not commonly used in daily clinical routine.

We have, therefore, reviewed the most common direct endothelial markers, with special view on their physiologic role and changes during systemic inflammation in humans.

von Willebrand factor and ADAMTS13

The acute phase glycoprotein von Willebrand factor (vWF) is a hemostatic cofactor that is physiologically produced in ECs, megakaryocytes, and subendothelial connective tissue. Usually, vWF is able to form large multimers, which circulate in the blood. The multimers undergo proteolysis by a specific plasma metalloprotease known as ADAMTS13 (a disintegrin and metalloprotease with thrombospondin motif). Under physiologic conditions, circulating ultra-large vWF (ULVWF) is barely detectable. Severe hereditary or acquired ADAMTS13 deficiency causes organ dysfunction and systemic inflammatory response syndrome (SIRS). In proinflammatory conditions, ADAMTS13 activity decreases, leading to increased levels of circulating vWF, which in turn causes platelet thrombus formation. Severe sepsis or septic shock results in the accumulation of substantial amounts of plasma ULVWF, which positively correlates with the severity of inflammation and the degree of organ failure (CitationClaus et al. 2010).

The role of vWF as a biologic marker for the diagnosis of sepsis, or the discrimination between individuals at risk for developing acute respiratory distress syndrome (ARDS) or as prognosis marker is very inconsistent. In fact, an increase in plasma levels of vWF can be caused by various septic and non-septic insults (CitationReinhart et al. 2002). The hypothesis that vWF may be increased in patients suffering from ARDS vs. only septic patients could not be fulfilled. The same observation was done, when comparing sepsis vs. non-septic patients with hyperinflammation (CitationRubin et al. 1990). Thus, it seems that vWF is a marker for diagnosing hyperinflammation, without being able to discriminate between specific organ damage (i.e. ARDS) or inflammation accompanied by infection as it occurs in sepsis. Nevertheless, vWF levels could help to differentiate between survivors and non-survivors in sepsis and high levels of vWF were correlated to fewer days organ-failure-free days (CitationMoss et al. 1996; CitationScherpereel et al. 2006). Furthermore, plasma vWF-antigen levels were associated with the development of acute lung injury and 28-day survival. However, these data demonstrated moderate sensitivity (87%) and specificity (77%) (CitationRubin et al. 1990; CitationSadler 1998). CitationMcGill et al. (1998) also demonstrated that vWF might be useful to serve as prognostic marker, predicting survival with a positive predictive value by 80%. Sensitivity was 80% and specificity was 87% (CitationMcGill et al. 1998). However, CitationMoss et al. (1996) could not verify these findings.

Taken together, vWF might only serve as marker to detect systemic endothelial activation upon systemic inflammation. Also, high levels of vWF reflect endothelial damage and correlate with poor survival and fewer organ-failure days. However, it seems that discrimination between infection and specific organ damage is not possible with this marker as data are too inconsistent at current stage.

Concerning the vWF cleaving protease ADAMTS13, data seem to reflect the same tendency as for vWF. Unfortunately, the current literature does not provide any larger studies regarding its role in inflammation. In a small clinical study (50 patients), ADAMTS13 was decreased in septic patients with concomitant DIC, however, with a low predictive value. The incidence of acute renal failure in patients with decreased ADAMTS13 levels was 41.2%. Decrease of the protein was due to a combination of increased cleavage together with a decreased synthesis of new ADAMTS 13 in the liver (CitationFurlan and Lammle 2001; CitationMimuro et al. 2008; CitationOno et al. 2006).

Ang-1 and -2

The angiopoietins (Ang)-1 and -2 belong to the family of growth factors and have been studied mainly in proliferative diseases such as cancer (neoangiogenesis) (CitationDavis et al. 1996; CitationFiedler and Augustin 2006; CitationMaisonpierre et al. 1997; CitationSuri et al. 1996). Ang-1 and -2 are antagonistic factors that trigger endothelial cell activation, involving the most important intracellular pathways (nuclear factor-κB for inflammation, Rho-kinase for interendothelial cell contacts and PI3K/AKT pathway for cell survival). Ang-1 and -2 bind to the endothelial Tie-2 receptor. Activation of the Tie-2 pathway involves processes such as vessel integrity, vascular permeability, and the regulation of inflammation (CitationFiedler et al. 2003; CitationMammoto et al. 2007; CitationPapapetropoulos et al. 2000; CitationWitzenbichler et al. 2005; CitationWong et al. 1997; CitationYuan et al. 2000). The importance of the Ang/Tie system in systemic inflammatory disorders has been demonstrated in many studies. In critically ill patients, the release of Ang-2 directly reflects vascular barrier breakdown. CitationOrfanos et al. (2007) could demonstrate that Ang-2 serves as marker to discriminate between sepsis and severe sepsis (p < 0.05). The authors investigated serum Ang-2 levels and could also show that Ang-2 acts similar to tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, two very important sepsis markers (p < 0.001). The clinical study was performed on a very small group (61 patients, 6 non-SIRS, 8 SIRS, 16 sepsis, 18 severe sepsis, and 13 septic shock patients).

Other small clinical studies demonstrated that circulating Ang-2 levels correlated with the APACHE and SOFA scoring systems as well as with the 28-day mortality reflecting disease severity and prognosis (CitationFiedler et al. 2006; CitationKranidioti et al. 2009; CitationMcCarter et al. 2007; CitationOrfanos et al. 2007; CitationThurston et al. 2000; CitationThurston et al. 1999; CitationWitzenbichler et al. 2005).

Another study with 293 children demonstrated that low Ang-1 and higher Ang-2 concentrations are associated with an unfavorable outcome in children with severe bacterial infection. The authors concluded that alterations of these secreted factors are directly linked to the endothelial damage/dysregulation occurring in severe bacterial infection and that angiopoietins could be used for the early identification of patients at risk of a poor outcome (CitationMankhambo et al. 2010).

Recently, CitationRicciuto et al. (2011) confirmed, in their study with 70 patients, the predictive power of Ang-1 and -2 as prognostic marker in the course of sepsis. Ang-1 plasma levels at admission and during the course of disease, both Ang-1 and -2 highly correlated with 28-day mortality in severe sepsis. Moreover, Ang-2 levels also correlated with disease severity as reflected by markers of organ damage and clinical sepsis scores. With Ang-1 exists a marker that might be of use to predict probability of sepsis mortality already at admission reflecting the critical role of early endothelial dysfunction. Here, low levels correspond with high mortality.

Taken together, Ang-2 and -1 seem to be of interest as prognostic sepsis biomarker. When detected over time, Ang-2 levels show a similar pattern such as TNF-α and IL-6. Ang-2 might present a reliable marker reflecting the direct status of the endothelium that correlates with disease severity (SIRS and sepsis vs. severe sepsis) and outcome (i.e. 28-day survival). Whereas, Ang-1 levels might have a high predictive outcome value, when determined at admission to the ICU. The early detection of severe disease courses and the progression to another pathologic state are extremely important for early decision to therapy escalation; therefore, Ang-1 and -2 levels might be a valuable tool to help taking this decision.

Endocan

Endocan, or originally known as endothelial cell-specific molecule-1, is a proteoglycan that is constitutively expressed in human ECs as well as in the human lung and kidney. It is localized in the vascular network, but also in the corresponding epithelia and in adipocytes (CitationBechard et al. 2000; CitationJanke et al. 2006; CitationWellner et al. 2003). The expression of endocan in ECs is up-regulated in response to TNF-α or IL1-β. Its secreted form circulates in the human bloodstream and can easily be detected (Bechard et al. 2001a, 2001b; CitationLassalle et al. 1996). It is suggested that in patients with septic shock serum levels of endocan may increase dramatically, representing the occurring endothelial damage and correlating to the severity of disease and the outcome (CitationScherpereel et al. 2006).

Scherpereel and colleagues investigated circulating endocan levels in serum samples from septic patients (n = 63). The data were compared to serum levels of healthy volunteers (n = 20) and to serum endocan levels of patients with SIRS (n = 7). The authors only measured endocan levels once, at admission to the ICU. They found that endocan levels significantly varied between patients in septic shock, severe sepsis, and sepsis yielding specificity from about 80% and 100% for SIRS vs. sepsis and a sensitivity of about 82%. Also, the single point detection of endocan at admission was useful to discriminate between 10-day survivors and non-survivors, latter displaying significantly higher levels. Endocan levels concerning 10- and 28-day survival had a high negative predictive power with 92%. Cutoff levels were set at 1.2 for all sepsis vs. SIRS and 3.0 for septic shock vs. all sepsis. Unfortunately, this study lacks kinetic data.

Other studies described endocan as not specific for systemic inflammatory diseases. High levels have also been detected in patients with cancer (CitationAitkenhead et al. 2002; CitationDepontieu et al. 2008; CitationGrigoriu et al. 2006; CitationHuang et al. 2009; CitationScherpereel et al. 2003; CitationZuo et al. 2008). Nonetheless, high levels may be of relevance for the promotion of systemic inflammation, as endocan mediates the recruitment of circulating lymphocytes to inflammatory sites as well as leukocyte adhesion and activation (Bechard et al. 2001b).

Large preclinical investigations underline the previous findings from the clinical studies. Taken together, endocan has a high prognostic value to discriminate between 10- and 28-day survivors. It serves also to distinguish between SIRS and sepsis with a specificity of 100%. However, no data exist for patients undergoing large cancer surgery, as cancer also upregulates endocan circulating levels (CitationSarrazin et al. 2006). Moreover, endocan kinetic data in the course of sepsis are missing. Endocan may be a very robust marker to solely detect endothelial (as organ) damage at early time points, which has been verified in large preclinical trials.

Selectins and other adhesion molecules

Selectins (E-, P-, L-) represent key adhesion molecules during the inflammatory process. They are the key mediators of leukocyte trafficking and hemostasis implicated in many human diseases including systemic inflammatory disorders and sepsis (CitationKansas 1996; Laubli and Borsig 2010; CitationLey 2003; CitationMcEver 1997; CitationWitz 2008). The selective recruitment of subpopulations of circulating leukocytes to the sites of injury or inflammation is a complex process orchestrating the interaction of primary (selectin) and activation-dependent (integrin) adhesion molecules. Selectins are expressed on the surface of activated ECs, platelets, and leukocytes. After activation, selectin receptors are shed from the surface and are measurable in the circulation. Severe injury and subsequent onset of systemic inflammation is resulting upon release of IL-1 β, IL-6, IL-8 (CXCL-8), and TNF-α contributing in turn to the release of adhesion molecules. Therefore, soluble selectins are directly linked to the secretion of proinflammatory mediators and reflect endothelial response upon inflammation (CitationCarlos and Harlan 1994; CitationNewman et al. 1993; CitationSiemiatkowski et al. 2001).

Endothelial leukocyte adhesion molecule (E-selectin or ELAM-1) is exclusively expressed on activated ECs. E-selectin can rapidly be induced (within 2–6 h) in response to IL-1 or TNF-α released by damaged cells or upon disturbances in blood rheology (CitationKansas 1996; CitationMorigi et al. 1995).

During the initiation of inflammation, neutrophils interact with ECs by binding to the endothelial intercellular adhesion molecule-1 (ICAM-1). ICAM-1 instead is constitutively expressed on ECs but is highly inducible upon stimulation (CitationKayal et al. 1998). Together with E-selectin, ICAM-1 plays an important role in the initiation or “rolling” phase of inflammation. E-selectin exists in a bound and a soluble form (CitationCummings et al. 1997; CitationRao et al. 2007).

P-selectin has a similar function, but is constitutively expressed in lung ECs, and therefore correlates with lung endothelial injury (CitationSakamaki et al. 1995). On demand, it can rapidly be released from the Weibel–Palade bodies of ECs or α-granules from thrombocytes via exocytosis (CitationKansas 1996; CitationMcEver 1997) after appropriate activation by histamine, thrombin, complement or reactive oxygen species. Increased soluble (s) P-selectin levels coincide with a raised number of leukocytes “rolling” along the endothelium thereby reflecting their activation (CitationRivera-Chavez et al. 1998). In addition, functionally active plasma-soluble P-selectin may modulate leukocyte adhesion to the P-selectin expressed on ECs (CitationSiemiatkowski et al. 2001).

L-selectin is constitutively expressed on all myeloid cells, on naïve T-cells and some activated memory T-cells. Upon their activation, L-selectin is quickly proteolytically cleaved (CitationKansas 1996; CitationLaubli and Borsig 2010; CitationLey 2003). Soluble L-selectin concentrations are reduced by widespread binding to activated endothelium making it a representative marker reflecting the activation status of the endothelium. Elevated levels of sL-selectin are detected in patients with AIDS, leukemia, and SIRS. Low concentrations can be found in ARDS, brain injury, and severe organ failure (CitationCocks and Chan 1997; CitationDonnelly et al. 1994; CitationKerner et al. 1999; CitationMaekawa et al. 1998; CitationMcKeating et al. 1998; CitationSiemiatkowski et al. 2001).

Similar to the selectins, vascular cell adhesion molecule-1 (VCAM-1) or CD106 participates in the adhesion of leukocytes to the endothelium at the site of inflammation. Besides its mechanical function, it also plays a role in the intercellular signal transduction. Upregulation of VCAM-1 in ECs is transcriptionally activated mainly by cytokines (TNF-α and IL-1). VCAM-1 protein is endothelial specific, and is a ligand for the α4β1 and α4β7 integrins (CitationCook-Mills et al. 2010; CitationRao et al. 2007).

In a cohort of 25 consecutive newborns, the investigation of sICAM-1 revealed that levels increased during the course of sepsis, and being higher in patients with positive vs. negative microbial hemoculture (HC). Soluble VCAM-1 only increased slightly in HC-positive sepsis. The highest sICAM-1 levels could be positively correlated with scores of illness severity. Soluble VCAM-1 levels increased only slightly in HC-positive sepsis, while sL-selectin and sP-selectin did not change (CitationFigueras-Aloy et al. 2007).

In another study including 119 critically ill patients, E-selectin serum levels were assessed and compared to healthy volunteers. The authors could show that sE-selectin levels were higher in serum of patients with microbiologically documented sepsis than in other critically ill medical ICU patients. The E-selectin levels at day 1 of sepsis diagnosis correlated highly with hemodynamic compromise and modestly with subsequent organ dysfunction and survival (CitationCummings et al. 1997).

In 63 septic patients with an infection in one or more major organs, serum sL-selectin could be identified as a predictor of survival in patients with sepsis. Patients with low sL-selectin (<470 ng/mL) were characterized by a high 1-year mortality (CitationSeidelin et al. 2002).

In a pediatric study including 77 children with sepsis and 14 acutely ill children without sepsis, a pronounced and persistent increase in plasma VCAM-1 and ICAM-1 was observed in septic children and persistent MOF. The authors concluded that these changes correlated mainly with the proinflammatory status of the endothelium (CitationWhalen et al. 2000).

Taken together, selectins and adhesion molecules may serve as markers for the detection of general endothelial activation and consequently sepsis. The studies investigating their use as biomarkers revealed that they can be used to monitor i.e. specific organ’s endothelium damage (i.e. L-selectin for ARDS). Also, these molecules seem to be good markers of prognosis for the 1-year survival and for progression of disease in case general endothelial damage has not yet occurred. However, the exact function and biological role of the soluble adhesion factors is so far unclear; some of the factors influencing their plasma levels are yet unknown. These markers, however, act otherwise to secreted factors described above. The soluble adhesion molecules are binding to the activated endothelium and are thus not detectable by tests. Therefore, low levels are often associated with the severity of disease and hence the severity of endothelial activation.

ET-1 and its precursor peptide proET-1

The endothelium-derived 21-residue venous and arterial peptide, endothelin (ET), has a potent and prolonged vasoconstrictor and vasopressor activity. ET has two functions: first, as circulating hormone and second, as paracrine factor mainly involved in the regulation of the vascular tone and systemic blood pressure (CitationWei et al. 1994; CitationYanagisawa et al. 1988). ET production is induced by substances such as epinephrine, IL-1, thrombin, and by conditions such as shear stress and damage to ECs. The ET family consists of three distinct isopeptides, ET-1, ET-2, and ET-3, which are derived from three distinct genes. ET-1 is the most potent vasoconstrictor out of the three. It is synthesized as a much longer polypeptide preproendothelin, being 203 amino acid residues long. Preproendothelin is cleaved by a signal peptidase to proET-1, which in turn is cleaved by ET-converting enzyme (ECE) to the 21 amino acid long ET-1 (CitationBrauner et al. 2000; CitationHemsen et al. 1996; CitationKido et al. 1998; CitationSchuetz et al. 2011; CitationSchweizer et al. 1997; CitationTschaikowsky et al. 2000). Using a sandwich immunoassay, Schuetz and colleagues evaluated proET-1 levels in patients with community-acquired pneumonia (CAP) and found that proET-1 levels at admission were independent predictors of short-term mortality and the need for ICU admission. They concluded that proET-1 levels improved the prognostic accuracy of the commonly used CURB-65 score to predict adverse outcome (CitationSchuetz et al. 2007; CitationSchuetz et al. 2009; CitationSchuetz et al. 2008). Piechota and colleagues could demonstrate in another study including twenty patients with sepsis and severe sepsis that ET-1 levels highly correlate with levels of NT-proBNP, PCT, and CRP, as well as the SOFA score. Mean ET-1 concentrations were 8.39 ± 6.39 pg/mL. Correlation between ET-1 levels and levels of NT-proBNP, PCT, and CRP was 0.3879 (p < 0.001), 0.358 (p < 0.001), and 0.225 (p = 0.011), respectively. Correlation between the ET-1 levels and SOFA score was 0.470 (p < 0.001) (CitationPiechota et al. 2007).

Newborns physiologically present a reduced liver function. This is associated with low CRP levels, as CRP is produced in the liver. Hence, ET-1 might also be a good marker to detect sepsis especially in newborns with the same reliability than CRP or PCT in adults. CitationFigueras-Aloy et al. (2004) demonstrated that plasma ET-1 levels in neonatal sepsis are related to the severity of clinical manifestations, especially oliguria, acidosis, and systemic hypotension. Taken together, proET-1 levels in CAP patients at admission may be independent predictors of short-term mortality and the need for ICU admission, correlating strongly with the CURB-65 score. With disease progression from sepsis to severe sepsis, ET-1 levels strongly correlated with markers of cardiac failure and markers of infectional status, reflecting disease severity. In newborns, where CRP production is physiologically low and thus not reliable, ET-1 might be a valuable marker to monitor disease courses.

VEGF and soluble VEGF-receptor-1 (Flt-1)

Vascular endothelial growth factor (VEGF) is a hypoxia-inducible cell mitogen. It stimulates endothelial cell migration along with vessel permeability (CitationDvorak et al. 1995), and promotes survival of the newly formed blood vessels (CitationFerrara and Davis-Smyth 1997). VEGF is crucial for endothelial cell survival (CitationGerber et al. 1999). Although VEGF is highly specific for ECs, it has become increasingly clear that it also elicits responses in non-ECs types. For example, it is chemotactic for monocytes and can inhibit the maturation of dendritic cells (CitationBarleon et al. 1996; CitationClauss et al. 1990; CitationGabrilovich et al. 1996).

The family of VEGF-related molecules contains five members: VEGF, placenta growth factor, VEGF-B, VEGF-C, and VEGF-D, whereas VEGF-A plays the most important role during sepsis. VEGF-A is 50,000 times more potent than histamine and is a potent inducer of tissue edema (CitationDvorak et al. 1995). VEGFs bind to their receptors, which are mainly expressed on ECs. Currently three types are described: fms-like tyrosine kinase-1 and -4 (Flt-1, -4) and kinase-insert-domain-containing receptor (VEGFR-2 or KDR) (Citationde Vries et al. 1992; CitationTerman et al. 1992). A soluble form of Flt-1 is produced by alternative splicing and acts as a natural occurring inhibitor of the VEGF-signaling. Recent studies have demonstrated the direct role of this receptor in sepsis concerning disease progression, severity, and survival (CitationEbihara et al. 2008; CitationPickkers et al. 2005; Citationvan der Flier et al. 2005; CitationYano et al. 2006).

All studies investigating VEGF-A or its soluble receptor in sepsis were significantly associated with sepsis severity and organ dysfunction (high correlation with organ dysfunction scores). A recent study conducted in a small cohort of 18 patients with severe sepsis and in 40 healthy controls could show that plasma VEGF levels were increased during severe sepsis. Moreover, the authors associated plasma VEGF levels with disease severity and mortality and explained these results by the VEGF-triggered capillary leak (Citationvan der Flier et al. 2005). CitationKarlsson et al. (2008) confirmed the relationship between high VEGF-levels and severe sepsis in a large clinical study including 470 patients. With progression of disease to septic shock, the levels of VEGF decreased, as a reflection of endothelial dysfunction. With this downregulation of VEGF, EC apoptosis rate will in turn increase, reinforcing the vascular disorder and finally leading to death. The authors could also demonstrate this relationship, where low VEGF-levels correlate with survival. These data indicate that VEGF levels increase while sepsis progresses. At the same time, the physiologic counter-regulation via VEGF scavenging by soluble Flt-1 is induced. Therefore, Flt-1 represents an interesting biomarker candidate, recently demonstrated in 101 patients (CitationYang et al. 2011). High plasma sFlt-1 levels significantly correlated with pneumonia-related septic shock and negative 28-day survival with median values of 659 pg/mL.

Taken together, the VEGF and sFlt-1 data seem to be very specific in detecting progression of disease from non-sepsis to severe sepsis and to septic shock. The progression from systemic inflammation, the most common syndrome on ICUs after major surgery, to sepsis needs further evaluation. VEGF and its soluble receptor served also to reliably detect short-term and 28-day survival, where low VEGF and high Flt-1 levels correlated with poor outcome. This may be explained by the massive intrinsic counter-regulation to VEGF-overproduction during sepsis. Compared to well-established daily clinical tests, especially sFlt-1 strongly correlates with Il-6 levels (CitationShapiro et al. 2010). Whether VEGF levels in cancer patients suffering from sepsis following large tumor surgeries are reliable remains to be elucidated as VEGF is also a marker for cancer progression.

PDGF

Platelet-derived growth factor (PDGF) was first described in the 1970s as a serum factor that stimulates proliferation of smooth muscle cells. Its name “platelet-derived,” is misleading due to the fact that the endothelium presents the major source of PDGF during sepsis (CitationBattegay et al. 1994; CitationThommen et al. 1997; CitationYaguchi et al. 2004). The PDGF family compromises four different members, PDGF-A, -B, -C, and -D, forming biological active homodimers (CitationBetsholtz 2003). Lack or loss of PDGF-B leads to vascular dysfunction and vascular leakage (CitationGaengel et al. 2009; CitationReigstad et al. 2005; CitationTallquist and Kazlauskas 2004; CitationTrojanowska 2008).

In a study, involving 46 patients at day 3 of severe sepsis, PDGF-BB levels were measured correlating PDGF-BB levels with the outcome of septic patients and evaluated PDGF-BB as response marker for treatment with recombinant human-activated protein C (rhAPC). The authors proposed PDGF-BB as a useful laboratory marker to predict survival in patients suffering from severe sepsis (CitationBrueckmann et al. 2007).

uPA and PAI-1

Plasminogen activator inhibitor (PAI-1) belongs to the family of serine protease inhibitors (SERPINs). PAI-1 is the most potent inhibitor of the serine proteases tissue plasminogen activator (tPA) and urokinase PA (uPA). The latter are potent activators of plasminogen and hence fibrinolysis. They are described to be involved in the inflammatory process and endothelial cell migration. Moreover, uPA seems to play a critical role in the VEGF-induced vascular permeability change (CitationYang et al. 2011). Under physiological conditions, PAI-1 is released into the circulation and the extracellular space by liver cells, smooth muscle cells, adipocytes, and platelets. This results in plasma levels of only 5–20 ng/mL of active PAI-1, which is sufficient to control fibrinolysis and extracellular proteolysis (CitationBinder et al. 2002). However, during pathological states such as systemic inflammation, large amounts of PAI-1 may be secreted by ECs. Here, mainly lipopolysaccharide derived from gram-negative bacteria, TNF-α, and IL-1 are responsible for the PAI-1 upregulation (CitationBinder et al. 2002; CitationZhang et al. 1997).

A clinical trial with 101 patients (81 patients with pneumonia-related septic shock and 20 with pneumonia without organ dysfunction) demonstrated the direct link between VEGF and uPA. Together with the increase of VEGF receptor, the activity of uPA increased significantly in non-survivors with septic shock. uPA activity was also considered as an independent marker of renal and hematologic dysfunction as well as metabolic acidosis. With increase of the number of failing organs, the uPA activity also increased (CitationYang et al. 2011).

In another large clinical study including 117 patients with sepsis-induced DIC and 1627 patients with non-septic DIC, the potential of PAI-1 as biomarker for sepsis was evaluated. Data showed that in septic DIC patients, plasma PAI-1 levels were significantly higher than in non-septic DIC cases, moreover high PAI-1 levels (>90 ng/mL vs. <30 ng/mL) in septic DIC patients correlated with multiple organ dysfunction scores. The 28-day mortality after DIC diagnosis was higher in patients having high circulating PAI-1 levels in the blood.

Taken together, uPA was described as an independent marker for organ dysfunction, increasing with the number of failing organs. Also, uPA was a marker for survival in septic shock patients, where high levels correlated with poor survival, independent of infection status. More, PAI-1 levels at the time of DIC diagnosis were an independent risk factor for mortality in sepsis-induced and a marker for organ failure progression after DIC onset (CitationMadoiwa et al. 2006). The close link between endothelial dysfunction and coagulation activation is directly reflected by uPA and PAI-1. Their activation shows on the one hand the endothelial homeostasis and on the other hand, the activation of the clotting system.

Fibrin degradation products

During systemic inflammation and sepsis, coagulation is partly activated or increased due to the loss of endothelium’s antithrombotic properties and exposure of highly thrombogenic subendothelial structures (CitationGimbrone et al. 1982 CitationNawroth and Stern 1985; CitationSemeraro and Colucci 1992). Here, massive turnover in coagulation factors and especially fibrinogen are observed. Along with this, the fibrinolysis system is activated, leading to a significant amount of fibrin degradation products (CitationLord 2007; CitationMosesson 2005). Namely, X and Y fragments, D-dimers, D and E fragments, Bβ15–42 and smaller fragments (CitationOlexa et al. 1981; CitationSmith et al. 1990; CitationWalker and Nesheim 1999).

Our understanding of fibrin degradation products until nowadays has been limited by their simple measurement as by-products of fibrinolysis without any biological activity. But, there is emerging evidence that fibrin degradation products also trigger inflammation. So far, only one molecular target has been reported to mediate this effect, namely vascular-endothelial-cadherin, an anchor protein that is part of the tight junctions and thus directly responsible for endothelial leak tightness (CitationPetzelbauer et al. 2005). Even fibrinogen and fibrin monomers are able to bind to ICAM-1 on ECs and thereby promote the attachment of leukocytes and platelets which in turn may then result in vascular occlusion (CitationAltieri et al. 1995) strong vasoconstriction by the stimulation of ET-1 (CitationAnggrahini et al. 2009), or resulting in paracellular hyperpermeability and subsequent fluid and protein leak via RhoA-dependent signaling pathways (CitationGuo et al. 2009).

CitationDeitcher and Eisenberg (1993) observed high concentrations of fibrin degradation products in plasma in 100 patients with a gram-negative infection. The test (measurement of fibrin fragments by enzyme-linked immunosorbent assay) had a high negative predictive value of 100% in the case of gram-negative infection and 91% in the case of general bacteremia. The authors concluded that high levels of fibrin degradation products correlated with a high fibrinolytic activity in patients with gram-negative bacteremia. The fibrin fragment hemagglutinin test, which is—according to the authors—easy to perform (bed-side test that provides results within 3 min and yield a negative predictive value of 96%) could be useful to detect those patients who are less susceptible for gram-negative infections.

Discussion

Sepsis is known for centuries and still remains a current challenge as it is the third leading cause of death in industrialized countries. Novel therapies, and usually very expensive, have emerged in the last decades. However, treatment and its success also require a very specific and reliable diagnostic to be efficacious (CitationDe Waele 2010; CitationRussel 2008).

Progressive organ dysfunction is a hallmark of this disease. One of the largest and most important organs is the endothelial system. Without a functional vascular system, blood rheology, coagulation, and organ perfusion is massively impaired. Endothelial dysfunction is a broad term, which is mainly used to refer to an imbalance of vasoconstriction, vasodilatation and vascular leakage, and edema formation. The endothelial injury, therefore, plays a key role during sepsis pathogenesis and is highly associated with mortality (CitationDuffy et al. 2011). Tissue edema may rapidly impair organ function by elongating the oxygen diffusion distance, favoring the formation of microthrombi with consequent breakdown of adequate organ perfusion (CitationLee and Slutsky 2010). Therefore, biomarkers serving as indicators or as surrogate markers for endothelial cell activation and dysfunction are of particular importance (CitationFilep 2006).

Today, the circulating levels of all these aforementioned factors, cytokines etc. are easily detectable and measurable. However, standardized chemistry tests to evaluate levels of creatinine, LDH, or GOT etc are missing for endothelial failure. Many tests described in this review seem to be robust and reliable tests. It is astonishing, due to the central role of the endothelium and its direct link to coagulation and homeostasis, that these markers have not been followed up until today. Some of the tests described in this review seem to be sensitive and specific enough to track the course of sepsis; however, they are not used broadly in the clinical routine. Economic considerations and physician’s personal interest might surely play an important role in the propagation of such tests. Another aspect might also be the perception of the endothelium as central organ. We would like to encourage the message that the endothelium is far more than just the smallest brick in the vessel wall, but that it keeps upright important gradients in the body. Even, there is no organ damage when the vasculature is working correctly and efficiently.

Conclusion

Loss of endothelial function and subsequent organ damage are hallmarks of systemic inflammatory disorders and sepsis. The central role of the endothelium renders this compartment an attractive target for biomarker research. However, which markers to choose in clinical practice will depend on further validation of the existing markers, the clinical accessibility and feasibility, economic aspects, and the personal experience of the treating physician.

Declaration of interest

P. Paulus and K. Zacharowski are beneficiaries of individual research grants from Bayer Schering Pharma. Besides this, no other financial relationships or conflict of interests has to be disclosed.

References

  • Aitkenhead M, Wang SJ, Nakatsu MN, Mestas J, Heard C, Hughes CC. (2002). Identification of endothelial cell genes expressed in an in vitro model of angiogenesis: induction of ESM-1, (beta)ig-h3, and NrCAM. Microvasc Res 63:159–171.
  • Altieri DC, Duperray A, Plescia J, Thornton GB, Languino LR. (1995). Structural recognition of a novel fibrinogen gamma chain sequence (117-133) by intercellular adhesion molecule-1 mediates leukocyte-endothelium interaction. J Biol Chem 270:696–699.
  • Anderson RN. (2002). Deaths: leading causes for 2000. Natl Vital Stat Rep 50:1–85.
  • Andreu Ballester JC, Ballester F, González Sánchez A, Almela Quilis A, Colomer Rubio E, Peñarroja Otero C. (2008). Epidemiology of sepsis in the Valencian Community (Spain), 1995-2004. Infect Control Hosp Epidemiol 29:630–634.
  • Anggrahini DW, Emoto N, Nakayama K, Widyantoro B, Adiarto S, Iwasa N, Nonaka H, Rikitake Y, Kisanuki YY, Yanagisawa M, Hirata K. (2009). Vascular endothelial cell-derived endothelin-1 mediates vascular inflammation and neointima formation following blood flow cessation. Cardiovasc Res 82:143–151.
  • Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. (2001a). Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 7: 1303–1310.
  • Angus DC, Wax RS. (2001). Epidemiology of sepsis: an update. Crit Care Med 29:S109–S116.
  • Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marmé D. (1996). Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood 87:3336–3343.
  • Battegay EJ, Rupp J, Iruela-Arispe L, Sage EH, Pech M. (1994). PDGF-BB modulates endothelial proliferation and angiogenesis in vitro via PDGF beta-receptors. J Cell Biol 125:917–928.
  • Béchard D, Gentina T, Delehedde M, Scherpereel A, Lyon M, Aumercier M, Vazeux R, Richet C, Degand P, Jude B, Janin A, Fernig DG, Tonnel AB, Lassalle P. (2001). Endocan is a novel chondroitin sulfate/dermatan sulfate proteoglycan that promotes hepatocyte growth factor/scatter factor mitogenic activity. J Biol Chem 276:48341–48349.
  • Bechard D, Meignin V, Scherpereel A, Oudin S, Kervoaze G, Bertheau P, Janin A, Tonnel A, Lassalle P. (2000). Characterization of the secreted form of endothelial-cell-specific molecule 1 by specific monoclonal antibodies. J Vasc Res 37:417–425.
  • Béchard D, Scherpereel A, Hammad H, Gentina T, Tsicopoulos A, Aumercier M, Pestel J, Dessaint JP, Tonnel AB, Lassalle P. (2001). Human endothelial-cell specific molecule-1 binds directly to the integrin CD11a/CD18 (LFA-1) and blocks binding to intercellular adhesion molecule-1. J Immunol 167:3099–3106.
  • Betsholtz C. (2003). Biology of platelet-derived growth factors in development. Birth Defects Res C Embryo Today 69:272–285.
  • Binder BR, Christ G, Gruber F, Grubic N, Hufnagl P, Krebs M, Mihaly J, Prager GW. (2002). Plasminogen activator inhibitor 1: physiological and pathophysiological roles. News Physiol Sci 17:56–61.
  • Brauner JS, Rohde LE, Clausell N. (2000). Circulating endothelin-1 and tumor necrosis factor-alpha: early predictors of mortality in patients with septic shock. Intensive Care Med 26:305–313.
  • Brueckmann M, Hoffmann U, Engelhardt C, Lang S, Fukudome K, Haase KK, Liebe V, Kaden JJ, Putensen C, Borggrefe M, Huhle G. (2007). Prognostic value of platelet-derived growth factor in patients with severe sepsis. Growth Factors 25:15–24.
  • Carlos TM, Harlan JM. (1994). Leukocyte-endothelial adhesion molecules. Blood 84:2068–2101.
  • Claus RA, Bockmeyer CL, Sossdorf M, Lösche W. (2010). The balance between von-Willebrand factor and its cleaving protease ADAMTS13: biomarker in systemic inflammation and development of organ failure? Curr Mol Med 10:236–248.
  • Clauss M, Gerlach M, Gerlach H, Brett J, Wang F, Familletti PC, Pan YC, Olander JV, Connolly DT, Stern D. (1990). Vascular permeability factor: a tumor-derived polypeptide that induces endothelial cell and monocyte procoagulant activity, and promotes monocyte migration. J Exp Med 172:1535–1545.
  • Cocks RA, Chan TY. (1997). Alteration in leukocyte adhesion molecule expression following minor, moderate and major trauma. Eur J Emerg Med 4:193–195.
  • Cook-Mills JM, Marchese M, Abdala-Valencia H. (2010). VCAM-1 expression and signaling during disease: regulation by reactive oxygen species and antioxidants. Antioxid Redox Signal.
  • Cummings CJ, Sessler CN, Beall LD, Fisher BJ, Best AM, Fowler AA 3rd. (1997). Soluble E-selectin levels in sepsis and critical illness. Correlation with infection and hemodynamic dysfunction. Am J Respir Crit Care Med 156:431–437.
  • Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC, Yancopoulos GD. (1996). Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell 87:1161–1169.
  • de Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N, Williams LT. (1992). The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science 255:989–991.
  • De Waele JJ. (2010). Early source control in sepsis. Langenbecks Arch Surg 395:489–494.
  • Deitcher SR, Eisenberg PR. (1993). Elevated concentrations of cross-linked fibrin degradation products in plasma. An early marker of gram-negative bacteremia. Chest 103:1107–1112.
  • Depontieu F, Grigoriu BD, Scherpereel A, Adam E, Delehedde M, Gosset P, Lassalle P. (2008). Loss of Endocan tumorigenic properties after alternative splicing of exon 2. BMC Cancer 8:14.
  • Dombrovskiy VY, Martin AA, Sunderram J, Paz HL. (2007). Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: a trend analysis from 1993 to 2003. Crit Care Med 35:1244–1250.
  • Donnelly SC, Haslett C, Dransfield I, Robertson CE, Carter DC, Ross JA, Grant IS, Tedder TF. (1994). Role of selectins in development of adult respiratory distress syndrome. Lancet 344:215–219.
  • Duffy MJ, Mullan BA, Craig TR, Shyamsundar M, Macsweeney RE, Thompson G, Stevenson M, McAuley DF. (2011). Impaired endothelium-dependent vasodilatation is a novel predictor of mortality in intensive care. Crit Care Med.
  • Dvorak HF, Brown LF, Detmar M, Dvorak AM. (1995). Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol 146:1029–1039.
  • Ebihara I, Hirayama K, Kaneko S, Nagai M, Ogawa Y, Fujita S, Usui J, Mase K, Yamagata K, Kobayashi M. (2008). Vascular endothelial growth factor and soluble fms-like tyrosine kinase-1 in septic shock patients treated with direct hemoperfusion with a polymyxin B-immobilized fiber column. Ther Apher Dial 12:285–291.
  • Engel C, Brunkhorst FM, Bone HG, Brunkhorst R, Gerlach H, Grond S, Gruendling M, Huhle G, Jaschinski U, John S, Mayer K, Oppert M, Olthoff D, Quintel M, Ragaller M, Rossaint R, Stuber F, Weiler N, Welte T, Bogatsch H, Hartog C, Loeffler M, Reinhart K. (2007). Epidemiology of sepsis in Germany: results from a national prospective multicenter study. Intensive Care Med 33:606–618.
  • Fareed J, Hoppensteadt DA, Leya F, Iqbal O, Wolf H, Bick R. (1998). Useful laboratory tests for studying thrombogenesis in acute cardiac syndromes. Clin Chem 44:1845–1853.
  • Ferrara N, Davis-Smyth T. (1997). The biology of vascular endothelial growth factor. Endocr Rev 18:4–25.
  • Ferreira FL, Bota DP, Bross A, Mélot C, Vincent JL. (2001). Serial evaluation of the SOFA score to predict outcome in critically ill patients. Jama 286:1754–1758.
  • Fiedler U, Augustin HG. (2006a). Angiopoietins: a link between angiogenesis and inflammation. Trends Immunol 12: 552–558.
  • Fiedler U, Krissl T, Koidl S, Weiss C, Koblizek T, Deutsch U, Martiny-Baron G, Marmé D, Augustin HG. (2003). Angiopoietin-1 and angiopoietin-2 share the same binding domains in the Tie-2 receptor involving the first Ig-like loop and the epidermal growth factor-like repeats. J Biol Chem 278:1721–1727.
  • Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S, Thurston G, Gale NW, Witzenrath M, Rosseau S, Suttorp N, Sobke A, Herrmann M, Preissner KT, Vajkoczy P, Augustin HG. (2006). Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med 12:235–239.
  • Figueras-Aloy J, Gómez-Lopez L, Rodríguéz-Miguélez JM, Jordán-García Y, Salvia-Roiges MD, Jiménez W, Carbonell-Estrany X. (2004). Plasma endothelin-1 and clinical manifestations of neonatal sepsis. J Perinat Med 32:522–526.
  • Figueras-Aloy J, Gómez-López L, Rodríguez-Miguélez JM, Salvia-Roiges MD, Jordán-García I, Ferrer-Codina I, Carbonell-Estrany X, Jiménez-González R. (2007). Serum soluble ICAM-1, VCAM-1, L-selectin, and P-selectin levels as markers of infection and their relation to clinical severity in neonatal sepsis. Am J Perinatol 24:331–338.
  • Filep JG. (2006). Endocan or endothelial cell-specific molecule-1: a novel prognostic marker of sepsis? Crit Care Med 34:574–575.
  • Furlan M, Lämmle B. (2001). Aetiology and pathogenesis of thrombotic thrombocytopenic purpura and haemolytic uraemic syndrome: the role of von Willebrand factor-cleaving protease. Best Pract Res Clin Haematol 14:437–454.
  • Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. (1996). Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 2:1096–1103.
  • Gaengel K, Genové G, Armulik A, Betsholtz C. (2009). Endothelial-mural cell signaling in vascular development and angiogenesis. Arterioscler Thromb Vasc Biol 29:630–638.
  • Gerber HP, Hillan KJ, Ryan AM, Kowalski J, Keller GA, Rangell L, Wright BD, Radtke F, Aguet M, Ferrara N. (1999). VEGF is required for growth and survival in neonatal mice. Development 126:1149–1159.
  • Gimbrone MA Jr, Buchanan MR. (1982). Interactions of platelets and leukocytes with vascular endothelium: in vitro studies. Ann N Y Acad Sci 401:171–183.
  • Grigoriu BD, Depontieu F, Scherpereel A, Gourcerol D, Devos P, Ouatas T, Lafitte JJ, Copin MC, Tonnel AB, Lassalle P. (2006). Endocan expression and relationship with survival in human non-small cell lung cancer. Clin Cancer Res 12:4575–4582.
  • Guo M, Daines D, Tang J, Shen Q, Perrin RM, Takada Y, Yuan SY, Wu MH. (2009). Fibrinogen-gamma C-terminal fragments induce endothelial barrier dysfunction and microvascular leak via integrin-mediated and RhoA-dependent mechanism. Arterioscler Thromb Vasc Biol 29:394–400.
  • Hemsén A, Modin A, Weitzberg E. (1996). Increased concentrations of endothelin-1 messenger RNA in tissues and endothelin-1 peptide in plasma in septic pigs: modulation by betamethasone. Crit Care Med 24:1530–1536.
  • Huang GW, Tao YM, Ding X. (2009). Endocan expression correlated with poor survival in human hepatocellular carcinoma. Dig Dis Sci 54:389–394.
  • Iba T, Kidokoro A, Fukunaga M, Sugiyama K, Sawada T, Kato H. (2005). Association between the severity of sepsis and the changes in hemostatic molecular markers and vascular endothelial damage markers. Shock 23:25–29.
  • Ince C. (2002). The microcirculation unveiled. Am J Respir Crit Care Med 166:1–2.
  • Janke J, Engeli S, Gorzelniak K, Feldpausch M, Heintze U, Böhnke J, Wellner M, Herse F, Lassalle P, Luft FC, Sharma AM. (2006). Adipose tissue and circulating endothelial cell specific molecule-1 in human obesity. Horm Metab Res 38:28–33.
  • Kansas GS. (1996). Selectins and their ligands: current concepts and controversies. Blood 88:3259–3287.
  • Karlsson S, Pettilä V, Tenhunen J, Lund V, Hovilehto S, Ruokonen E; Finnsepsis Study Group. (2008). Vascular endothelial growth factor in severe sepsis and septic shock. Anesth Analg 106:1820–1826.
  • Kayal S, Jaïs JP, Aguini N, Chaudière J, Labrousse J. (1998). Elevated circulating E-selectin, intercellular adhesion molecule 1, and von Willebrand factor in patients with severe infection. Am J Respir Crit Care Med 157:776–784.
  • Kerner T, Ahlers O, Spielmann S, Keh D, Bührer C, Gerlach M, Höfler S, Gerlach H. (1999). L-selectin in trauma patients: a marker for organ dysfunction and outcome? Eur J Clin Invest 29:1077–1086.
  • Kido T, Sawamura T, Masaki T. (1998). The processing pathway of endothelin-1 production. J Cardiovasc Pharmacol 31 Suppl 1:S13–S15.
  • Knoebl P. (2010). Blood coagulation disorders in septic patients. Wien Med Wochenschr 160:129–138.
  • Kranidioti H, Orfanos SE, Vaki I, Kotanidou A, Raftogiannis M, Dimopoulou I, Kotsaki A, Savva A, Papapetropoulos A, Armaganidis A, Giamarellos-Bourboulis EJ. (2009). Angiopoietin-2 is increased in septic shock: evidence for the existence of a circulating factor stimulating its release from human monocytes. Immunol Lett 125:65–71.
  • Kumar A, Haery C, Paladugu B, Kumar A, Symeoneides S, Taiberg L, Osman J, Trenholme G, Opal SM, Goldfarb R, Parrillo JE. (2006). The duration of hypotension before the initiation of antibiotic treatment is a critical determinant of survival in a murine model of Escherichia coli septic shock: association with serum lactate and inflammatory cytokine levels. J Infect Dis 193:251–258.
  • Lassalle P, Molet S, Janin A, Heyden JV, Tavernier J, Fiers W, Devos R, Tonnel AB. (1996). ESM-1 is a novel human endothelial cell-specific molecule expressed in lung and regulated by cytokines. J Biol Chem 271:20458–20464.
  • Läubli H, Borsig L. (2010). Selectins promote tumor metastasis. Semin Cancer Biol 20:169–177.
  • Lee WL, Slutsky AS. (2010). Sepsis and endothelial permeability. N Engl J Med 363:689–691.
  • Lehr HA, Bittinger F, Kirkpatrick CJ. (2000). Microcirculatory dysfunction in sepsis: a pathogenetic basis for therapy? J Pathol 190:373–386.
  • Lever A, Mackenzie I. (2007). Sepsis: definition, epidemiology, and diagnosis. BMJ 335:879–883.
  • Levi M. (2010). The coagulant response in sepsis and inflammation. Hamostaseologie 1: 14–16.
  • Levi M. (2007). Disseminated intravascular coagulation. Crit Care Med 35:2191–2195.
  • Levi M, Ten Cate H. (1999). Disseminated intravascular coagulation. N Engl J Med 341:586–592.
  • Levi M, ten Cate H, van der Poll T. (2002). Endothelium: interface between coagulation and inflammation. Crit Care Med 30:S220–S224.
  • Levi M, van der Poll T. (2010). Inflammation and coagulation. Crit Care Med 38:S26–S34.
  • Ley K. (2003). The role of selectins in inflammation and disease. Trends Mol Med 9:263–268.
  • Lord ST. (2007). Fibrinogen and fibrin: scaffold proteins in hemostasis. Curr Opin Hematol 14:236–241.
  • Madoiwa S, Nunomiya S, Ono T, Shintani Y, Ohmori T, Mimuro J, Sakata Y. (2006). Plasminogen activator inhibitor 1 promotes a poor prognosis in sepsis-induced disseminated intravascular coagulation. Int J Hematol 84:398–405.
  • Maekawa K, Futami S, Nishida M, Terada T, Inagawa H, Suzuki S, Ono K. (1998). Effects of trauma and sepsis on soluble L-selectin and cell surface expression of L-selectin and CD11b. J Trauma 44:460–468.
  • Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN, Yancopoulos GD. (1997). Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 277:55–60.
  • 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.
  • Mankhambo LA, Banda DL, Jeffers G, White SA, Balmer P, Nkhoma S, Phiri H, Molyneux EM, Hart CA, Molyneux ME, Heyderman RS, Carrol ED; IPD Study Group. (2010). The role of angiogenic factors in predicting clinical outcome in severe bacterial infection in Malawian children. Crit Care 14:R91.
  • McCarter SD, Mei SH, Lai PF, Zhang QW, Parker CH, Suen RS, Hood RD, Zhao YD, Deng Y, Han RN, Dumont DJ, Stewart DJ. (2007). Cell-based angiopoietin-1 gene therapy for acute lung injury. Am J Respir Crit Care Med 175:1014–1026.
  • McDonald JR, Friedman ND, Stout JE, Sexton DJ, Kaye KS. (2005). Risk factors for ineffective therapy in patients with bloodstream infection. Arch Intern Med 165:308–313.
  • McEver RP. (1997). Selectin-carbohydrate interactions during inflammation and metastasis. Glycoconj J 14:585–591.
  • McGill SN, Ahmed NA, Christou NV. (1998). Increased plasma von Willebrand factor in the systemic inflammatory response syndrome is derived from generalized endothelial cell activation. Crit Care Med 26:296–300.
  • McKeating EG, Andrews PJ, Mascia L. (1998). Leukocyte adhesion molecule profiles and outcome after traumatic brain injury. Acta Neurochir Suppl 71:200–202.
  • Mimuro J, Niimura M, Kashiwakura Y, Ishiwata A, Ono T, Ohmori T, Madoiwa S, Okada K, Matsuo O, Sakata Y. (2008). Unbalanced expression of ADAMTS13 and von Willebrand factor in mouse endotoxinemia. Thromb Res 122:91–97.
  • Morigi M, Zoja C, Figliuzzi M, Foppolo M, Micheletti G, Bontempelli M, Saronni M, Remuzzi G, Remuzzi A. (1995). Fluid shear stress modulates surface expression of adhesion molecules by endothelial cells. Blood 85:1696–1703.
  • Mosesson MW. (2005). Fibrinogen and fibrin structure and functions. J Thromb Haemost 3:1894–1904.
  • Moss M, Gillespie MK, Ackerson L, Moore FA, Moore EE, Parsons PE. (1996). Endothelial cell activity varies in patients at risk for the adult respiratory distress syndrome. Crit Care Med 24:1782–1786.
  • Nawroth PP, Stern DM. (1985). A pathway of coagulation on endothelial cells. J Cell Biochem 28:253–264.
  • Newman W, Beall LD, Carson CW, Hunder GG, Graben N, Randhawa ZI, Gopal TV, Wiener-Kronish J, Matthay MA. (1993). Soluble E-selectin is found in supernatants of activated endothelial cells and is elevated in the serum of patients with septic shock. J Immunol 150:644–654.
  • Olexa SA, Budzynski AZ, Doolittle RF, Cottrell BA, Greene TC. (1981). Structure of fragment E species from human cross-linked fibrin. Biochemistry 20:6139–6145.
  • Ono T, Mimuro J, Madoiwa S, Soejima K, Kashiwakura Y, Ishiwata A, Takano K, Ohmori T, Sakata Y. (2006). Severe secondary deficiency of von Willebrand factor-cleaving protease (ADAMTS13) in patients with sepsis-induced disseminated intravascular coagulation: its correlation with development of renal failure. Blood 107:528–534.
  • Orfanos SE, Kotanidou A, Glynos C, Athanasiou C, Tsigkos S, Dimopoulou I, Sotiropoulou C, Zakynthinos S, Armaganidis A, Papapetropoulos A, Roussos C. (2007). Angiopoietin-2 is increased in severe sepsis: correlation with inflammatory mediators. Crit Care Med 35:199–206.
  • Papapetropoulos A, Fulton D, Mahboubi K, Kalb RG, O’Connor DS, Li F, Altieri DC, Sessa WC. (2000). Angiopoietin-1 inhibits endothelial cell apoptosis via the Akt/survivin pathway. J Biol Chem 275:9102–9105.
  • Petzelbauer P, Zacharowski PA, Miyazaki Y, Friedl P, Wickenhauser G, Castellino FJ, Gröger M, Wolff K, Zacharowski K. (2005). The fibrin-derived peptide Bbeta15-42 protects the myocardium against ischemia-reperfusion injury. Nat Med 11:298–304.
  • Pickkers P, Sprong T, Eijk L, Hoeven H, Smits P, Deuren M. (2005). Vascular endothelial growth factor is increased during the first 48 hours of human septic shock and correlates with vascular permeability. Shock 24:508–512.
  • Piechota M, Banach M, Irzmanski R, Barylski M, Piechota-Urbanska M, Kowalski J, Pawlicki L. (2007). Plasma endothelin-1 levels in septic patients. J Intensive Care Med 22:232–239.
  • Pierrakos C, Vincent JL. (2010). Sepsis biomarkers: a review. Crit Care 14:R15.
  • Rao RM, Yang L, Garcia-Cardena G, Luscinskas FW. (2007). Endothelial-dependent mechanisms of leukocyte recruitment to the vascular wall. Circ Res 101:234–247.
  • Reigstad LJ, Varhaug JE, Lillehaug JR. (2005). Structural and functional specificities of PDGF-C and PDGF-D, the novel members of the platelet-derived growth factors family. Febs J 272:5723–5741.
  • Reinhart K, Bayer O, Brunkhorst F, Meisner M. (2002). Markers of endothelial damage in organ dysfunction and sepsis. Crit Care Med 30:S302–S312.
  • Ricciuto DR, Dos Santos CC, Hawkes M, Toltl LJ, Conroy AL, Rajwans N, Lafferty EI, Cook DJ, Fox-Robichaud A, Kahnamoui K, Kain KC, Liaw PC, Liles WC. (2011). Angiopoietin-1 and angiopoietin-2 as clinically informative prognostic biomarkers of morbidity and mortality in severe sepsis. Crit Care Med.
  • Rivera-Chavez F, Toledo-Pereyra LH, Nora DT, Bachulis B, Ilgenfritz F, Dean RE. (1998). P-selectin blockade is beneficial after uncontrolled hemorrhagic shock. J Trauma 45:440–445.
  • Rubin DB, Wiener-Kronish JP, Murray JF, Green DR, Turner J, Luce JM, Montgomery AB, Marks JD, Matthay MA. (1990). Elevated von Willebrand factor antigen is an early plasma predictor of acute lung injury in nonpulmonary sepsis syndrome. J Clin Invest 86:474–480.
  • Russel JA. (2008). The current management of septic shock. Minerva Med 99:431–458.
  • Sadler JE. (1998). Biochemistry and genetics of von Willebrand factor. Annu Rev Biochem 67:395–424.
  • Sakamaki F, Ishizaka A, Handa M, Fujishima S, Urano T, Sayama K, Nakamura H, Kanazawa M, Kawashiro T, Katayama M. (1995). Soluble form of P-selectin in plasma is elevated in acute lung injury. Am J Respir Crit Care Med 151:1821–1826.
  • Sarrazin S, Adam E, Lyon M, Depontieu F, Motte V, Landolfi C, Lortat-Jacob H, Bechard D, Lassalle P, Delehedde M. (2006). Endocan or endothelial cell specific molecule-1 (ESM-1): a potential novel endothelial cell marker and a new target for cancer therapy. Biochim Biophys Acta 1765:25–37.
  • Scherpereel A, Depontieu F, Grigoriu B, Cavestri B, Tsicopoulos A, Gentina T, Jourdain M, Pugin J, Tonnel AB, Lassalle P. (2006). Endocan, a new endothelial marker in human sepsis. Crit Care Med 34:532–537.
  • Scherpereel A, Gentina T, Grigoriu B, Sénéchal S, Janin A, Tsicopoulos A, Plénat F, Béchard D, Tonnel AB, Lassalle P. (2003). Overexpression of endocan induces tumor formation. Cancer Res 63:6084–6089.
  • Schuetz P, Christ-Crain M, Morgenthaler NG, Struck J, Bergmann A, Müller B. (2007). Circulating precursor levels of endothelin-1 and adrenomedullin, two endothelium-derived, counteracting substances, in sepsis. Endothelium 14:345–351.
  • Schuetz P, Christ-Crain M, Müller B. (2009). Procalcitonin and other biomarkers to improve assessment and antibiotic stewardship in infections–hope for hype? Swiss Med Wkly 139:318–326.
  • Schuetz P, Christ-Crain M, Zimmerli W, Mueller B. (2011). Repeated measurements of endothelin-1 precursor peptides predict the outcome in community-acquired pneumonia. Intensive Care Med 37:970–980.
  • Schuetz P, Stolz D, Mueller B, Morgenthaler NG, Struck J, Mueller C, Bingisser R, Tamm M, Christ-Crain M. (2008). Endothelin-1 precursor peptides correlate with severity of disease and outcome in patients with community acquired pneumonia. BMC Infect Dis 8:22.
  • Schweizer A, Valdenaire O, Nelböck P, Deuschle U, Dumas Milne Edwards JB, Stumpf JG, Löffler BM. (1997). Human endothelin-converting enzyme (ECE-1): three isoforms with distinct subcellular localizations. Biochem J 328 (Pt 3):871–877.
  • Seidelin JB, Nielsen OH, Strøm J. (2002). Soluble L-selectin levels predict survival in sepsis. Intensive Care Med 28:1613–1618.
  • Semeraro N, Colucci M. (1992). Changes in the coagulation-fibrinolysis balance of endothelial cells and mononuclear phagocytes: role in disseminated intravascular coagulation associated with infectious diseases. Int J Clin Lab Res 21:214–220.
  • Shapiro NI, Schuetz P, Yano K, Sorasaki M, Parikh SM, Jones AE, Trzeciak S, Ngo L, Aird WC. (2010). The association of endothelial cell signaling, severity of illness, and organ dysfunction in sepsis. Crit Care 14:R182.
  • Siemiatkowski A, Rogowski F, Wereszczynska-Siemiatkowska U, Malinowska L, Borkowski J. (2001). Soluble selectin profiles associated with severe trauma. Arch Immunol Ther Exp (Warsz) 49:317–324.
  • Slade E, Tamber PS, Vincent JL. (2003). The Surviving Sepsis Campaign: raising awareness to reduce mortality. Crit Care 7:1–2.
  • Smith EB, Keen GA, Grant A, Stirk C. (1990). Fate of fibrinogen in human arterial intima. Arteriosclerosis 10:263–275.
  • Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN, Yancopoulos GD. (1996). Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 87:1171–1180.
  • Tallquist M, Kazlauskas A. (2004). PDGF signaling in cells and mice. Cytokine Growth Factor Rev 15:205–213.
  • Taylor FB Jr, Toh CH, Hoots WK, Wada H, Levi M; Scientific Subcommittee on Disseminated Intravascular Coagulation (DIC) of the International Society on Thrombosis and Haemostasis (ISTH). (2001). Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb Haemost 86:1327–1330.
  • Terman BI, Dougher-Vermazen M, Carrion ME, Dimitrov D, Armellino DC, Gospodarowicz D, Böhlen P. (1992). Identification of the KDR tyrosine kinase as a receptor for vascular endothelial cell growth factor. Biochem Biophys Res Commun 187:1579–1586.
  • Thommen R, Humar R, Misevic G, Pepper MS, Hahn AW, John M, Battegay EJ. (1997). PDGF-BB increases endothelial migration on cord movements during angiogenesis in vitro. J Cell Biochem 64:403–413.
  • 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.
  • Thurston G, Suri C, Smith K, McClain J, Sato TN, Yancopoulos GD, McDonald DM. (1999). Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Science 286:2511–2514.
  • Trojanowska M. (2008). Role of PDGF in fibrotic diseases and systemic sclerosis. Rheumatology (Oxford) 47 Suppl 5:v2–v4.
  • Tschaikowsky K, Sägner S, Lehnert N, Kaul M, Ritter J. (2000). Endothelin in septic patients: effects on cardiovascular and renal function and its relationship to proinflammatory cytokines. Crit Care Med 28:1854–1860.
  • Tyagi A, Sethi AK, Girotra G, Mohta M. (2009). The microcirculation in sepsis. Indian J Anaesth 53:281–293.
  • van der Flier M, van Leeuwen HJ, van Kessel KP, Kimpen JL, Hoepelman AI, Geelen SP. (2005). Plasma vascular endothelial growth factor in severe sepsis. Shock 23:35–38.
  • Walker JB, Nesheim ME. (1999). The molecular weights, mass distribution, chain composition, and structure of soluble fibrin degradation products released from a fibrin clot perfused with plasmin. J Biol Chem 274:5201–5212.
  • Wei CM, Lerman A, Rodeheffer RJ, McGregor CG, Brandt RR, Wright S, Heublein DM, Kao PC, Edwards WD, Burnett JC Jr. (1994). Endothelin in human congestive heart failure. Circulation 89:1580–1586.
  • Wellner M, Herse F, Janke J, Gorzelniak K, Engeli S, Bechart D, Lasalle P, Luft FC, Sharma AM. (2003). Endothelial cell specific molecule-1–a newly identified protein in adipocytes. Horm Metab Res 35:217–221.
  • Whalen MJ, Doughty LA, Carlos TM, Wisniewski SR, Kochanek PM, Carcillo JA. (2000). Intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 are increased in the plasma of children with sepsis-induced multiple organ failure. Crit Care Med 28:2600–2607.
  • Witz IP. (2008). The selectin-selectin ligand axis in tumor progression. Cancer Metastasis Rev 27:19–30.
  • Witzenbichler B, Westermann D, Knueppel S, Schultheiss HP, Tschope C. (2005). Protective role of angiopoietin-1 in endotoxic shock. Circulation 111:97–105.
  • Wong AL, Haroon ZA, Werner S, Dewhirst MW, Greenberg CS, Peters KG. (1997). Tie2 expression and phosphorylation in angiogenic and quiescent adult tissues. Circ Res 81:567–574.
  • Yaguchi A, Lobo FL, Vincent JL, Pradier O. (2004). Platelet function in sepsis. J Thromb Haemost 2:2096–2102.
  • Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, Yazaki Y, Goto K, Masaki T. (1988). A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature 332:411–415.
  • Yang KY, Liu KT, Chen YC, Chen CS, Lee YC, Perng RP, Feng JY. (2011). Plasma soluble vascular endothelial growth factor receptor-1 levels predict outcomes of pneumonia-related septic shock patients: a prospective observational study. Crit Care 15:R11.
  • Yano K, Liaw PC, Mullington JM, Shih SC, Okada H, Bodyak N, Kang PM, Toltl L, Belikoff B, Buras J, Simms BT, Mizgerd JP, Carmeliet P, Karumanchi SA, Aird WC. (2006). Vascular endothelial growth factor is an important determinant of sepsis morbidity and mortality. J Exp Med 203:1447–1458.
  • Yuan HT, Suri C, Landon DN, Yancopoulos GD, Woolf AS. (2000). Angiopoietin-2 is a site-specific factor in differentiation of mouse renal vasculature. J Am Soc Nephrol 11:1055–1066.
  • Zambon M, Ceola M, Almeida-de-Castro R, Gullo A, Vincent JL. (2008). Implementation of the Surviving Sepsis Campaign guidelines for severe sepsis and septic shock: we could go faster. J Crit Care 23:455–460.
  • Zhang WJ, Wojta J, Binder BR. (1997). Notoginsenoside R1 counteracts endotoxin-induced activation of endothelial cells in vitro and endotoxin-induced lethality in mice in vivo. Arterioscler Thromb Vasc Biol 17:465–474.
  • Zuo L, Zhang SM, Hu RL, Zhu HQ, Zhou Q, Gui SY, Wu Q, Wang Y. (2008). Correlation between expression and differentiation of endocan in colorectal cancer. World J Gastroenterol 14:4562–4568.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.