Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Review

This paper contains Video doi: 10.1111/joim.12331

Endothelial barrier dysfunction in septic shock


S. M. Opal1 & T. van der Poll2
From the 1Infectious Disease Division, Alpert Medical School of Brown University, Pawtucket, RI, USA; and 2Academic Medical Center,
Division of Infectious Diseases & The Center of Experimental and Molecular Medicine, University of Amsterdam, Amsterdam, the Netherlands

Abstract. Opal SM, van der Poll T (Alpert Medical innate immune and coagulation systems to
School of Brown University, Pawtucket, RI, USA; orchestrate the host response in sepsis. The
University of Amsterdam, Amsterdam; the barrier function of the endothelial surface is
Netherlands). Endothelial barrier dysfunction in almost uniformly impaired in septic shock, and
septic shock (Review). J Intern Med 2015; 277: it is likely that this contributes to adverse out-
277–293. comes. In this review, we will highlight recent
advances in the understanding of the signalling
The endothelium provides an essential and selec- events that regulate endothelial function and
tive membrane barrier that regulates the move- molecular events that induce endothelial dysfunc-
ment of water, solutes, gases, macromolecules tion in sepsis. Endothelial barrier repair strategies
and the cellular elements of the blood from the as a treatment for sepsis include modulation of
tissue compartment in health and disease. Its C5a, high-mobility group box 1 and VEGF recep-
structure and continuous function is essential for tor 2; stimulation of angiopoietin-1, sphingosine 1
life for all vertebrate organisms. Recent evidence phosphate receptor 1 and Slit; and a number of
indicates that the endothelial surface does not other innovative approaches.
have a passive role in systemic inflammatory
states such as septic shock. In fact, endothelial Keywords: endothelial barrier, endothelial junctions,
cells are in dynamic equilibrium with a myriad protease-activated receptors, sepsis, sepsis-induced
of inflammatory mediators and elements of the immunosuppression, septic shock.

understanding the basic pathophysiology of sepsis


Introduction
from the molecular level to the level of organ
Sepsis, severe sepsis and septic shock comprise a communication and systems integration [14–24].
spectrum of increasingly common, potentially
lethal, yet poorly understood clinical syndromes. Currently, the nature of sepsis is viewed as a
The rising incidence is well documented and fundamental disintegration of systems controls
thought to be attributable to the ageing of the from intercellular signalling networks to neuroen-
population, the prevalence of immunocompro- docrine and immune regulatory mechanisms dur-
mised patients and implantable medical devices ing the time of systemic injury and invasive
and the progressive increase in antimicrobial resis- microbial stress. Failure to maintain or reconsti-
tance among common bacterial pathogens [1–5]. tute homeostasis after a major systemic injury
Developing new therapeutic agents to manage ultimately determines outcome in sepsis [7, 19,
patients with sepsis has proven difficult with a 22]. The immediate threat in sepsis is invasive
large number of failed clinical trials [6–8]. infection, and the need to activate immune
defences to clear the pathogen before irreparable
Despite the repeated failure to demonstrate sur- damage occurs due to the microorganism and its
vival benefits for a number of promising novel toxins. However, in the process of eliminating the
therapeutic agents for the treatment of sepsis in pathogen, the systemic host response to general-
Phase III clinical trials, the news regarding clinical ized infection can lead to collateral damage to
outcomes in patients with sepsis is not all bad [6, normal tissues [4]. It is remarkable that many of
7, 9]. The mortality rate is certainly improving in the same pathways, pattern recognition receptors
many clinical studies worldwide, which is primarily and innate host responses seen in sepsis are also
a reflection of improved supportive care and early observed in other forms of severe injury and
diagnosis and treatment protocols [10–13]. Addi- systemic inflammation without concomitant
tionally, substantial progress has been made in infection [2, 4, 22].

ª 2014 The Association for the Publication of the Journal of Internal Medicine 277
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

The long-term consequences of sepsis can be in septic patients [7]. The histopathological find-
profound and disabling. These consequences are ings of autopsy examinations of patients who have
increasingly recognized as a condition now referred died as a result of multiorgan dysfunction due to
to as persistent critical illness (PCI). PCI may sepsis (and in experimental animal studies of
persist for months or even years and is character- sepsis) are often remarkably normal appearing,
ized by prolonged lengths of stay in the intensive characterized primarily by mild tissue oedema in
care unit, persistent organ dysfunction, dysfunc- the intracellular and extravascular interstitium.
tional host response to repeat infections, slow or These anatomical findings seem incongruous with
even permanent cognitive decline and losses of the clinical findings in such patients with evidence
overall sense of well-being and function in society of acute kidney injury, cholestatic jaundice, acute
[25–27]. Sepsis is frequently followed by a reduced myocardial dysfunction and similar dysfunctions
duration of healthy life expectancy, even if the in other organs. Despite the relative preservation of
patient’s overall lifespan is preserved. tissue morphology, tissue function is often mark-
edly impaired. Cardiac myocytes stop contracting
In this review, we will focus on recent findings normally, alveoli cease to maintain the air–liquid
regarding the basic elements that drive the septic barrier interface in lung tissue, hepatocytes no
process at the level of cellular communication, the longer secrete bilirubin, endothelial cells retract,
coagulation system and endothelial barrier func- become permeable to macromolecules and lose
tion. Of note, other research priorities are clearly their anti-adhesive and anticoagulant surface
important in understanding the basic mecha- characteristics, and so on. The entire process
nisms that underlie the pathophysiology of sepsis, seems to be a manifestation of poor cellular and
including disordered immune function [28–30], tissue barrier function, loss of specialized tissue
neuroendocrine [31–33], nutritional [34], immune actions and an acquired form of cellular hiberna-
metabolism [18], mitochondrial sparing [23, 35, tion. Tissues stop generating variability in the
36] and systems integration studies [14, 19]. integrated circuitry and stop generating cycles of
communication within and between tissues. The
The importance of immune dysfunction and ele- loss of specialized cell function and barrier func-
ments of immune depression in sepsis is increas- tion has been noted by many investigators, sug-
ingly being recognized, and they might prove to be gesting that this constitutes a common host
suitable targets for therapeutic intervention response to sepsis and other forms of critical
[28–30]. Loss of adaptive immune function and illness [7, 15, 22].
downregulation of the host innate immune
response have been well described and cause Here, we will review the evidence that loss of barrier
adverse consequences. For example, opportunistic function in general, and endothelial barrier func-
viral infections are reactivated in the majority of tion in particular, is central to the pathogenesis of
patients with severe sepsis, and these infections sepsis and highly integrated into the host systemic
can cause clinically significant conditions such as inflammatory and coagulopathic response. A num-
systemic cytomegalovirus infection and herpes ber of authors have recently developed this concept
simplex activation [37]. What is not clear is of the aberrant and dysfunctional endothelial bar-
whether sepsis-induced immune suppression is a rier as the central pathophysiological process in
necessary compensatory mechanism to regulate septic shock [40–44]. We will also discuss how
the systemic septic host response or a potentially endothelial barriers are influenced by coagulation
treatable, pathological state of immune suppres- factors and elements of the innate immune
sion increasing the risk of superinfections in response, and review novel findings concerning
patients [38, 39]. This question can only be monocyte/macrophage–endothelial interactions
answered by carefully controlled clinical trials in and signalling in sepsis.
well-characterized patient populations.
The vascular endothelium and haemostasis
We wish to highlight the loss of cellular barriers
and its consequences at the level of the endothelial Endothelial cells are at the interface between
membrane to make our point about loss of function inflammation and coagulation in sepsis [43]
of vascular integrity in sepsis. In a recent review, (Fig. 1). Dysfunction of the endothelium has an
Deutschman and Tracey emphasized that loss of important role in the disturbance of the haemo-
cell membrane barrier function is a general finding static balance in sepsis. Moreover, the endothelium

278 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

Coagulation ↑ Anticoagulation ↓
MyD88-ARNO-ARF6

TFPI ↓ Endothelial cell


Tissue factor ↑ TM ↓ EPCR ↓ Tie2 Robo4
Antithrombin↓ Protein C (↓)
Microcirculation

Angiopoietin-2 ↑
Monocyte
APC ↓
Blood pressure ↓
Fibrinolysis ↓
NETs with trapped
platelets PAI-1 ↑ RBC deformability↓
APC ↓ - Thrombin ↑

Thrombosis VE-cadherin↓ Cell death


Neutrophil PAR1 Tight junctions ↓ Cell shrinkage
S1P3/S1P1 ↑

Tissue hypoperfusion Loss of barrier function


Capillary leak
Interstitial oedema
Tissue

Tissue oxygenation ↓

Organ failure

Fig. 1 Dysfunction of the vascular endothelium in severe sepsis. Sepsis is associated with microvascular thrombosis due to
concurrent activation of coagulation (mediated by tissue factor) and impairment of anticoagulant mechanisms as a
consequence of reduced activity of endogenous anticoagulant pathways mediated by activated protein C (APC),
antithrombin and tissue factor pathway inhibitor (TFPI) plus impaired fibrinolysis due to enhanced release of plasminogen
activator inhibitor type I (PAI-1). The capacity to generate APC is impaired at least in part due to reduced expression of the
endothelial receptors thrombomodulin (TM) and endothelial protein C receptor (EPCR). Thrombus formation is further
facilitated by neutrophil extracellular traps (NETs) released from dying neutrophils. Thrombus formation results in tissue
hypoperfusion, which is aggravated by hypotension and reduced red blood cell (RBC) deformability. Tissue oxygenation is
further impaired by loss of barrier function of the endothelium due to loss of vascular endothelial (VE)-cadherin function,
alterations in the endothelial cytoskeleton, high angiopoietin-2 levels (which interact with the endothelial cell receptor Tie2),
and activation of Myd88–ARNO–ARF6 signalling. ARF6 is adenosine diphosphate-ribosylation factor 6; ARNO is ARF
nucleotide-binding site opener. A disturbed balance between sphingosine 1 phosphate receptor (S1P)1 and S1P3 within the
vascular wall at least in part due to preferential induction of S1P3 via protease-activated receptor 1 (PAR1) secondary to a
reduced APC/thrombin ratio. Robo4 is an endothelial cell receptor that protects barrier function.

is involved in all three major pathogenetic path- results in the generation of thrombin and fibrin.
ways associated with coagulopathy in sepsis: tis- The pivotal role of TF in sepsis-induced coagulation
sue factor (TF)-mediated thrombin generation, has been established in early clinical studies in
dysfunctional anticoagulation and impaired fibri- humans and primate models in which strategies
nolysis. that prevent the activation of the TF–factor VIIa
pathway abrogated the activation of the common
TF is the main initiator of coagulation activation in pathway of coagulation elicited by administration
sepsis. It is clear that monocytes and macrophages of lipopolysaccharide (LPS) or bacteria [45]. In
are major sources of TF in severe sepsis; however, addition, in lethal sepsis models in baboons, TF
endothelial cells also contribute to a considerable inhibition prevented multiple organ failure and
extent [44]. Endothelial cells activated by bacterial mortality [46, 47]. Similarly, in genetically deficient
products or proinflammatory cytokines such as mice with an almost complete lack of TF, coagula-
tumour necrosis factor (TNF)-a or interleukin (IL)- tion, inflammation and mortality induced by injec-
1b express TF at their surface. Expression of TF by tion of high-dose LPS were reduced relative to
endothelial cells in vivo is restricted to certain control mice [48]. In addition, in its cell-associated
organs and vascular beds [42]. TF binds to and form, TF can reside in microparticles (MPs) shed
activates clotting factor VII, which via factor X from haematopoietic and endothelial cells. MPs

ª 2014 The Association for the Publication of the Journal of Internal Medicine 279
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

have been implicated in the activation of both concentrations are able to modulate TF-mediated
coagulation and inflammation in sepsis [49]. Fur- coagulation [46, 47].
thermore, upon stimulation with proinflammatory
cytokines, endothelial cells are able to release The vascular endothelium serves an important role
alternatively spliced TF (lacking exon 5) which in the protein C system, which represents a crucial
circulates in a soluble form and may exert proco- endogenous anticoagulant mechanism by virtue of
agulant activity [50]. the ability of APC to proteolytically inactivate
coagulation cofactors Va and VIIIa. APC is formed
Coagulation is regulated by three main anticoagu- from protein C when thrombin binds to the throm-
lant mechanisms: antithrombin, the protein C bomodulin receptor present on endothelial cells.
system and TF pathway inhibitor (TFPI). Sepsis is The activation of protein C to APC by thrombo-
associated with impaired function of all three modulin-bound thrombin is augmented by the
pathways, primarily as a consequence of endothe- presence of the endothelial protein C receptor
lial dysfunction [44]. Resting endothelial cells gen- (EPCR). During sepsis, the protein C system is
erate activated protein C (APC) on the cell surface, impaired as a result of several factors, most nota-
produce tissue-type plasminogen activator to stim- bly decreased synthesis and increased consump-
ulate fibrinolysis and impede thrombin formation tion of protein C and decreased activation of
and platelet adhesion. Antithrombin is the main protein C due to reduced expression of thrombo-
inhibitor of thrombin and activated (a) factor X modulin and EPCR on endothelial cells [45]. The
(Xa). Under physiological conditions, glycosamino- anticoagulant strength of the protein C system has
glycans present on the vessel wall support anti- been demonstrated in many preclinical settings
thrombin-mediated inhibition of thrombin and [56].
other clotting enzymes. During sepsis, antithrom-
bin concentrations are strongly reduced due to In a landmark study conducted in the 1980s,
impaired production, enhanced degradation (at Taylor and colleagues demonstrated that infusion
least in part by elastase from activated neutrophils) of APC into septic baboons prevented DIC and
and consumption caused by sustained thrombin death and that inhibition of protein C activation
generation [45]. Antithrombin function is further aggravated the response to Escherichia coli and
compromised by the decreased production of transformed a sublethal effect into a lethal DIC-
glycosaminoglycans on the endothelial surface, associated state [57]. In accordance with an impor-
which is mediated by proinflammatory cytokines tant regulatory function of the endogenous protein
[43]. C system in sepsis, treatment of bacteraemic
baboons with an anti-EPCR monoclonal antibody
TFPI is the main inhibitor of the TF–factor VIIa exacerbated a sublethal E. coli infection leading to
complex, which is predominantly expressed by lethal sepsis with massive activation of coagulation
endothelial cells [51]. TFPI binds with high affinity [58]. The anticoagulant effects of APC in haemo-
both to TF–factor VIIa and to factor Xa present in stasis may be enhanced by its capacity to inhibit
plasma and on endothelial cells. Under normal fibrinolysis by suppressing the function of two
conditions, TFPI is attached to the endothelium via fibrinolysis inhibitors: thrombin activatable fibri-
proteoglycans, which facilitates its TF–factor VIIa– nolytic inhibitor (TAFI) and plasminogen activator
factor X-inhibiting properties on the endothelial inhibitor type I (PAI-1) [56].
surface [52]. TFPI function is inhibited in sepsis by
the reduced synthesis of glycosaminoglycans on Adhesion of cells to injured endothelium relies on
the endothelial surfaces. The importance of TFPI as adhesive proteins, such as von Willebrand factor.
an endogenous anticoagulant in sepsis is illus- Monomers of von Willebrand factor (280 kDa) can
trated by the fact that rabbits became more be linked by disulfide bonds to form ultra-large
susceptible to LPS-induced disseminated intravas- multimers, with a molecular mass of up to 106 Da
cular coagulation (DIC) and the generalized [59]. The assembly of von Willebrand factor multi-
Shwartzman reaction following inhibition of TFPI mers occurs in endothelial cells, where they are
[53, 54]. Administration of recombinant TFPI dose- stored in Weibel–Palade bodies. Large multimers of
dependently inhibited LPS-induced thrombin gen- von Willebrand factor, which are released upon
eration in humans [55]. Pharmacological doses of endothelial cell perturbation, can bind more effi-
TFPI can prevent mortality during systemic infec- ciently to platelet glycoprotein Iba. In normal hae-
tion and inflammation, indicating that high TFPI mostasis, large von Willebrand factor multimers

280 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

are cleaved by a protease termed a disintegrin and and histones [69]. Finally, NETs generated as a
metalloproteinase with a thrombospondin type 1 consequence of platelet–neutrophil interactions
motif, member 13 (ADAMTS13); this process is have been implicated in tissue damage in sepsis
stimulated under circumstances of high shear [67].
stress. Physiologically, von Willebrand factor acts
to stabilize the adhesion of platelets at sites of
Protease-activated receptors and endothelial barrier function
vascular injury. Sepsis is associated with a relative
deficiency of ADAMTS13, resulting in increased Thrombin and an array of circulating serine prote-
levels of ultra-large von Willebrand factor multi- ases can cleave a set of ubiquitously expressed,
mers, which facilitate platelet adhesion to injured seven-transmembrane receptors known as prote-
endothelium [60]. It has been suggested that the ase-activated receptors (PARs) [43, 44]. Four such
resulting thrombotic microangiopathy has an receptors (PAR1–4) are found in humans and can
important role in multiple organ dysfunction in either disrupt or protect endothelial barrier func-
sepsis [61]. tion, depending on which G-protein-linked intra-
cellular signalling pathway is activated. PARs are
Activated platelets form a strong link between the unusual receptors as the ectodomain contains the
processes of primary and secondary haemostasis internal, sequestered ligand that, through feed-
by providing the phospholipid layer necessary for back, activates its own receptor, but only if the N-
the assembly of activated coagulation factor com- terminus of the ectodomain is removed by partial
plexes [62]. Disruption of the vascular integrity in proteolysis by serine proteases such as thrombin
sepsis results in the adhesion of platelets at sites of [43–45]. Thrombin binds to PAR1 expressed on
injury, where they can contribute to endothelial endothelial cells in the early phase of sepsis,
cell activation through several mechanisms. For contributes to endothelial dysfunction by G12/13
example, stimulation of glycoprotein IIb/IIIa on Rho-dependent cytoskeletal derangements in
platelets enhances CD40 ligand expression on the endothelial cells and induces endothelial cell con-
platelet membrane, which activates endothelial traction and rounding [70, 71]. Endothelial cell
cells to express adhesion molecules and TF [63, contraction destabilizes cell-to-cell contacts, caus-
64]. Expression of P-selectin on the platelet mem- ing an increase in vascular permeability, which
brane mediates the adherence of platelets to leu- facilitates the passage of large molecules (albumin
cocytes and endothelial cells and enhances the and other plasma proteins) and leucocytes from the
expression of TF on monocytes [65]. blood into the subendothelial compartment (see
Fig. 2). Gaps in the endothelial barrier also expose
Vascular inflammation and coagulation are ampli- the fluid compartment of blood to the basement
fied by the release of so-called neutrophil extracel- membrane and vessel adventitia with abundant TF
lular traps (NETs) by neutrophils. NETs are highly for clot initiation and collagen fibres for von Wille-
charged mixtures of DNA and nuclear proteins brand factor to polymerize and for binding plate-
together with serine proteases such as elastase, lets.
cathepsin G and calprotectin [66]. NETs function to
entrap pathogens (an action that is facilitated by It is interesting that thrombin-induced stimulation
platelets) and are designed to mediate swift elim- of endothelial cells by PAR1 activation can be
ination of invasive microorganisms. However, as deleterious to barrier function or beneficial to
for many components of innate immunity, NETs endothelial function depending on the stage of
can also contribute to collateral tissue damage [67, progression of sepsis and severity of the disease
68]. Indeed, NETs are procoagulant by promoting state [72]. Over time, thrombin linked to PAR1 can
adhesion, activation and aggregation of platelets transactivate PAR2 into a PAR1–PAR2 heterodimer,
and by activating the contact system, leading to the which has a protective role in endothelial barrier
release of bradykinin and activation of the intrinsic function and survival in sepsis models. This ‘role
pathway of coagulation [68]. Moreover, by promot- reversal’ for PAR1 signalling from endothelial bar-
ing platelet and red blood cell adhesion, and by rier disruption to barrier protection is mediated by
engagement of clotting factors, NETs can provide a PAR1 to PAR2 combined signalling that switches
scaffold for thrombus formation. Additionally, coupling of the GTPase RhoA intracellular barrier
NETs can induce endothelial cell death, an effect disruptive pathway to an alternative PAR2-Gi-
that is likely to be mediated by NET-associated Rac1-mediated intracellular signalling pathway
proteases or cationic proteins such as defensins resulting in actin polymerization and improved

ª 2014 The Association for the Publication of the Journal of Internal Medicine 281
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

to biased-barrier breakdown signalling in endothe-


Tight endothelial barrier Dysfunctional barrier
lial cell membranes [76, 77]. This cleavage site is
Activated protein C C’5a, PAF, NO located two amino acids proximal to the N-termi-
Bradykinin
Atrial natriuretic peptide
VEGF receptor 2
nus site for thrombin cleavage at position R41–S42.
Robo4-Silt2N
Ang1-Tie2 Thrombin, FXa, MMP The MMP1 cleavage site generates a slightly longer,
Protease inhibitors HMGB1 tethered peptide ligand which mediates barrier
PAR1-2 VE-cadherin PAR1 disruption upon PAR1 activation [72, 77].
S1P3
S1P1
Rac1 RhoA
Tie2 Tie2 Macrophage–endothelial cell interactions
TF Ang-1 TF TF
TF TF
The occurrence of crosstalk between myeloid and
endothelial cells has been well known for many
Collagen fibres Actin polymer
cytoskeleton
Actin breakdown years as endothelial signals are critical for target-
ing neutrophils and circulating monocytes from the
Fig. 2 Endothelial barrier function and dysfunction in vascular lumen to endothelial surfaces expressing
health and disease. The left panel shows the factors that adhesion molecules in areas of acute inflammation
contribute to tight junctions, with well-formed and func- [65]. Activated endothelial cells secrete IL-8, mono-
tional endothelial cells covering the microvasculature. The cyte chemoattractant protein-1 and macrophage
right panel highlights those molecular signals that con- inhibitory protein-1 to attract monocytes to inflam-
tribute to loss of barrier function and endothelial cell matory foci. Intravascular, adherent, activated
rounding and retraction. Breakdown of tight junctions is monocytes express large amounts of TF to propa-
accompanied by loss of intravascular proteins to the
gate procoagulant activity at the site of infection or
interstitial spaces, procoagulant and proinflammatory
injury. However, the extent to which tissue macro-
processes, and loss of barrier integrity. Ang, angiopoietin;
Tie2, tyrosine kinase with immunoglobulin-like and epi- phages are involved in angiogenesis, tissue repair
thelial growth factor-like domains 2; VE, vascular endo- and maintenance of endothelial barrier function is
thelium; PAR1–2, protease-activated receptor type 1 and 2 just beginning to be understood.
heterodimer; Rac1–Cell division control protein; TF, tissue
factor; S1P, sphingosine 1 phosphate receptor; C’, comple- Macrophages are initially activated after tissue
ment; PAF, platelet-activating factor; NO, nitric oxide; FXa, injury by ischaemic necrosis of mesenchymal cells
activated factor X; MMP, matrix metalloprotease; VEGF in an IL-1a-dependent process. IL-1a primarily
receptor 2, vascular endothelial growth factor receptor-2; exists as an uncleaved but biologically active
HMGB1, high-mobility group box 1; RhoA, GTPase of the
precursor which is located within the intracellular
Ras homolog gene family.
space and nucleus where it regulates transcription
[78]. During necrotic, but not apoptotic, cell death,
barrier function. This dual signalling system might the IL-1a precursor is released from intracellular
be amenable to therapeutic intervention with stores within dying mesenchymal cells and binds
agents that target specific PARs at specific times to its type 1 IL-1 receptor expressed on adjacent
during the progression of sepsis (see Table 1 and tissue macrophages. By contrast, during apopto-
discussion below) [72–74]. sis, IL-1a precursor remains tightly bound to the
chromatin where it is then digested during the
Recently, matrix metalloprotease (MMP)1 has been apoptotic nuclear fragmentation process without
detected at high levels in the circulation in patients IL-1a release [79]. Necrotic cell release of IL-1a
with severe sepsis/septic shock (18-fold higher precursor is rapidly followed by inflammasome
than normal blood levels) [75]. This might have assembly within the macrophage cytosol, resulting
significant prognostic and therapeutic implica- in caspase-1 synthesis, IL-1b processing and
tions. MMP1 efficiently cleaved PAR1 on endothe- extracellular secretion with marked upregulation
lial cells but, in contrast to thrombin with dual of IL-1b -induced chemokine, cytokine and adhe-
signalling ability, MMP1–PAR1 signalling mediates sion molecule expression on nearby endothelial
only barrier disruption [75]. Endothelial-derived cells [79, 80]. Recruitment of circulating neutroph-
MMP1 activates noncanonical PAR1 signalling and ils and monocytes activates inducible nitric oxide
is associated with poor outcomes in experimental synthase with rapid efflux of nitric oxide followed
models of septic shock [76, 77]. MMP1 cleaves and by vasodilatation, opening of endothelial gaps and
activates PAR1 at a novel D39–P40 site, which leads loss of endothelial barrier function (Fig. 2) [78].

282 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

Table 1 Endothelial barrier disrupters and promoters

Barrier disrupters Barrier promoters


Molecule Mechanism Molecule Mechanism
C’ components Vasodilators, increase C’ regulators and Block anaphylatoxin
C3a, C5a vascular permeability carboxy-peptidases actions, degrade C3a and C5a
Bradykinin Vasodilator, increases Carboxy-peptidases Degrade bradykinin
permeability
PAF Vasodilator, increases PAF-acetyl hydrolase Degrades PAF in the
permeability circulation
Proinflammatory Increase surface expression Anti-inflammatory Limit expression of
cytokines of adhesins, cytokines inflammatory cytokines
PMN–EC interaction
Ang-2 Inhibitor of Ang-1, pro-apoptotic Ang-1 Inhibits NF-jB signalling,
inhibits apoptosis
Tie1 Specific inhibitor of Tie 2 Tie2 Tyrosine kinase receptor
interaction with Ang-1 for Ang-1, anti-apoptotic
RhoA GTPase Breaks down actin cytoskeleton, Rac1 Stabilizes actin cytoskeleton
internalizes VE-cadherin
PAR1 signals Pro-apoptotic, activate RhoA PAR1–PAR2 transactivation Anti-apoptotic, promotes Rac1
MMPs Activate PAR-1 barrier disruption Protease inhibitors Limit activity of MMPs
and RhoA
S1P3 Activates RhoA, de-polymerizes S1P1 Membrane stabilizer,
actin cytoskeleton promotes VE-cadherins,
actin polymerization
Vascular endothelial Activates Src kinase to Atrial natriuretic peptide Activates Rac1, anti-
growth factor 2 phosphorylate and internalize inflammatory, limits NF-jB
VE-cadherin
b-arrestin-mediated Internalizes phosphorylated Slit–Robo4 Inhibits VE-cadherin
endocytosis VE-cadherin, impairs phosphorylation by
endothelial barrier p120-catenin, blocks
apoptosis
HMGB1 Breaks down VE-cadherin, HMGB1 mAb Inhibits HMGB1-dependent
promotes cytokine and alteration of endothelial
chemokine generation barrier function

Ang, angiopoietin; S1P, sphingosine 1 phosphate receptor; NF-jB, nuclear factor kappa light-chain enhancer of B cells;
VE, vascular endothelial; C’, complement; PMN, polymorphonuclear cell; PAF, platelet-activating factor; EC, endothelial
cell; Tie 1 and Tie 2, tyrosine kinase with immunoglobulin-like and epithelial growth factor-like domains; GTPase,
guanosine triphosphatase; RhoA, GTPase of the Ras homolog gene family; Rac1, a subfamily of GTPases; PAR, protease-
activated receptor; MMP, matrix metalloprotease; Src kinase, sarcoma-related nonreceptor protein kinase; HMGB1, high-
mobility group box 1; mAb, monoclonal antibody.

Extra-luminal macrophages support endothelial synthesis and release of soluble growth factors
cell growth, limit apoptosis and remodel the into the microenvironment, (ii) local release of
extracellular matrix via expression of MMPs to macrophage exosomes and (iii) direct cell-to-cell
support endothelial membrane structure and contact with endothelial cells (Fig. 3). Vascular
function [81–83]. Tissue macrophages signal to endothelial growth factor A, colony-stimulating
endothelial cells by one of three mechanisms: (i) factor 1, IL-1b and TNF-a are secreted by

ª 2014 The Association for the Publication of the Journal of Internal Medicine 283
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

Endothelial barrier

Activated TNF
Capillary lumen
monocytes HMGB1 MCP1
MIP-1 IL-1α
TF TF IL-1β
MCP1 TF RAGE
IL- 8 P-selectin TLR4
PSGL1

Soluble Ang2
Exosomes cytokines Tie2

Direct cell-to-
cell contact

Tissue macrophages
activating ECs

Fig. 3 Interactions between tissue macrophages in the perivascular space, circulating monocytes and endothelial cell
surfaces. High-mobility group box 1 (HMGB1) is a late cytokine-like mediator in sepsis that increases endothelial permeability
and induces inflammatory cytokine synthesis by endothelial cells (ECs). PSGL1, P (platelet)-selectin glycopeptide ligand 1;
MIP1, macrophage inhibitory protein 1; MCP1, monocyte chemoattractant protein 1; IL, interleukin; TNF, tumour necrosis
factor; TF, tissue factor; TLR4, Toll-like receptor 4; RAGE, receptor for advanced glycation end products.

activated tissue macrophages. These cytokines increasing cell migration [81, 86]. It is tempting to
and growth factors bind to their cognate receptors speculate that macrophage-derived exosomes are
expressed on endothelial cells to support growth generated in systemic inflammatory states and
and to promote expression on vascular endothe- disrupt endothelial barrier function during sepsis,
lial surfaces of adhesion molecules such as but this has not yet been confirmed within the
P-selectin, E-selectin, intercellular adhesion microcirculation and perivascular space in
molecule 1 (ICAM1) and vascular cell adhesion patients with sepsis [81].
molecule (VCAM) [81].
Macrophages appear to have the capacity to
Recent evidence suggests that exosomes (i.e. small directly influence endothelial cell function and
microvesicles with cytosolic contents enclosed structure by direct cell-to-cell contact with endo-
within a lipid bilayer) are generated by perivascular thelial cells. Macrophages express the angiopoietin
macrophages and can influence endothelial behav- receptor, which is a tyrosine kinase known as Tie2
iour [84, 85]. Exosomes encase RNA species from (discussed in detail below). Interaction between
donor macrophages including microRNAs [83] and angiopoietin (Ang)-2 and Tie2 promotes endothelial
can fuse with the cellular membrane of adjacent growth, cell survival and angiogenesis. This direct
endothelial cells and deliver their contents directly cell contact between macrophages and endothelial
into target cells. MicroRNAs are short sequences of cells might be important in tissue repair and for re-
RNA (20–25 nucleotides in length) that bind to the establishing endothelial barrier function during
untranslated tails of specific complementary mes- the recovery phase of sepsis [81, 82]. Reciprocal
senger RNA (mRNA) sequences and limit further interactions between endothelial cells and macro-
translation of or degrade peptide sequences from phages maintain tissue macrophage viability and
the mRNA template. These microRNAs have the promote a microenvironment for macrophage dif-
capacity to simultaneously inhibit the peptide ferentiation and maturation [81, 87] Endothelial
synthesis of multiple mRNA targets. One microRNA cells promote differentiation to M2-like macro-
known as miR-150 has been successfully delivered phages, which have an anti-inflammatory pheno-
from exosomes to endothelial monolayers in vitro type and rely on oxidative metabolism to support
and found to alter endothelial cell behaviour by resolution of tissue injury and repair.

284 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

organ [98]. Microvascular dysfunction has been


Endothelial cell MPs
linked to organ failure and mortality in patients
MPs are vesicles that range in size from 0.1 to 2 lm with sepsis [96]; disturbed function of the vascular
and are shed from the plasma membrane of endothelium plays a pivotal role.
multiple cell types upon activation or apoptosis
[88, 89]. MPs contain many biologically active The glycocalyx is a thin layer of glucosaminogly-
molecules, including proteins, lipids, mRNAs and cans that covers the surface of the vascular endo-
microRNAs. The cellular source of circulating MPs thelium [43]. The glycocalyx contains several
is revealed by the antigens they express, which components that are essential for normal homeo-
allows investigation of the function of cell-specific stasis, including antithrombin and superoxide
MPs. It was shown that MPs are present at low dismutase. One function of the glycocalyx is to
levels in the circulation of healthy subjects and reduce adhesion of leucocytes and platelets to
originate predominantly from platelets [90]. Proin- endothelial cells. Sepsis results in the degradation
flammatory cytokines and bacterial products can of the glycocalyx, leading to enhanced adhesion of
induce shedding of MPs from endothelial cells that inflammatory cells and injury [44]. Other patho-
contain ultra-large von Willebrand factor multi- logical responses implicated in microvascular dys-
mers, which potently promote the formation of function include activation of coagulation
platelet aggregates and increase their stability [91]. (resulting in thrombosis in the microvasculature),
Activated monocytes release MPs expressing TF alterations in red cell deformability and impaired
[92]. Patients with septic shock had elevated total release of nitric oxide [99, 100]. Although these
MP levels regardless of the presence of DIC, responses that contribute to microvascular dys-
whereas levels of endothelium- and leucocyte- function are clearly detrimental in the context of
derived MPs were positively correlated with DIC severe sepsis, they are important for host defence
status [93]. Of note, MPs can also exert anticoag- against invasive infection by limiting dissemination
ulant effects. Anionic phospholipids exposed by of the infection and recruiting cells to the infectious
MPs can promote the assembly of anticoagulant site. Of note, it has been reported that microcircu-
proteins such as TFPI, thrombomodulin, EPCR and latory function is improved by several anticoagu-
protein S. APC can stimulate the release of MPs lants, including APC, antithrombin and low
from endothelial cells, which facilitates the efficient molecular weight heparin [101–104]. It is interest-
inactivation of factors Va and VIIIa by MP-derived ing that a modified antithrombin that is unable to
EPCR. Several cytoprotective properties associated bind to the vascular endothelium but with intact
with APC could be reproduced by APC-positive MPs anticoagulant properties failed to improve the
in vitro [94]. Additionally, recombinant human microcirculation in animals with sepsis, suggesting
APC, which until recently was registered for the that the anticoagulant effect per se is not sufficient
treatment of severe sepsis, has been found to to improve microcirculatory function [103].
induce an increase in circulating APC-positive
MPs. This suggests a functional role of anticoagu-
Endothelial barrier function
lant and cytoprotective MPs in the in vivo effects of
APC [95]. Endothelial barrier dysfunction and microvascular
leak critically contribute to the pathogenesis of
organ failure in sepsis and of sepsis-related com-
The endothelium and the microcirculation
plications such as acute lung injury [105]. The
The endothelium plays a pivotal role in the normal vascular barrier consists of endothelial cells,
function of the microcirculation. Sepsis is associ- together with cell–cell junctions and extracellular
ated with a decreased flow velocity in the microcir- components such as the glycocalyx. Cell–cell junc-
culation and a reduced density of perfused tions include adherens junctions, mainly com-
capillaries [96]. Alterations in the microcirculation posed of vascular endothelial (VE)-cadherin, and
in sepsis are characterized in particular by heter- tight junctions (the zona occludens), predomi-
ogeneous perfusion of tissues due to a lack, or nantly consisting of occludins and claudins [106].
intermittent perfusion, of capillaries adjacent to Under normal conditions, the endothelial barrier is
those with normal perfusion [97]. Heterogeneous semi-permeable, allowing transport of fluids and
perfusion of the microvascular circulation disturbs solutes from blood to tissues. In sepsis, however,
tissue oxygenation and leads to hypoxic areas even barrier function is disturbed, resulting in
in the presence of preserved total blood flow to an enhanced passage of proteins and solutes outside

ª 2014 The Association for the Publication of the Journal of Internal Medicine 285
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

also transactivate S1P1 signalling to promote


endothelial barrier function [109]. For example,
PAR1 can influence vascular stability and function
via S1P1 signalling. APC potently inhibits throm-
bin-induced vascular hyperpermeability by a
mechanism dependent on transactivation of S1P1
[110], whereas thrombin-induced vascular hyper-
permeability is dependent on another S1P receptor,
S1P3 [111]. Notably, activation of PAR1 by low
doses of thrombin can (like APC) lead to a barrier
protective effect [112] and thrombin can transacti-
vate PAR1–PAR2 heterodimers in late sepsis, which
is also barrier protective [72]. Recently, a key role
Animation 1 This animation describes in a time lag
for S1P2 in the permeability and inflammatory
fashion the pathophysiology behind endothelial barrier
dysfunction in septic shock. Watch the animation here. responses of the vascular endothelium during
Please note that the version of the video with narration will endotoxaemia was revealed [113]. Downstream sig-
be up shortly. nalling of S1P2 in vascular inflammation included
the activation of the stress-activated protein kinase
the circulation, and oedema (Animation 1). Several and nuclear factor kappa light-chain enhancer of
mediators are involved in maintaining vascular B-cell (NF-jB) pathways. Several S1P analogues
barrier function (see Table 1). have been developed, with the aim of preserving
vascular integrity, for possible clinical use [109].
Vascular endothelial growth factor
Fibrinopeptide Bb15–42
Vascular endothelial growth factor (VEGF) is a
glycoprotein produced by endothelial and lung Fibrinopeptide Bb15–42 is a cleavage product of
epithelial cells, as well as by leucocytes and plate- fibrin that binds to VE-cadherin and stabilizes
lets [106]. VEGF regulates vascular permeability interendothelial junctions [114]. Administration of
through binding to VEGF receptors 1–3, which are fibrinopeptide Bb15–42 may be of therapeutic value
endothelial-cell-specific membrane tyrosine kinase in sepsis. Treatment with this peptide preserved
receptors [107]. Activation of VEGF receptor 2 endothelial barrier function in two different shock
dissociates VE-cadherin from the adherens junc- models, induced by Dengue virus and LPS, by
tion resulting in increased vascular permeability inhibiting stress-induced opening of endothelial
[108]. Although the results of preclinical studies to cell adherens junctions; fibrinopeptide Bb15–42-
investigate the role of VEGF in sepsis have been treated animals showed improved survival rates
inconsistent [106], a pilot study to test an anti- and reduced haemoconcentration and fibrinogen
VEGF antibody (bevacizumab) in patients with consumption [115]. In a pig model of haemorrhagic
septic shock was announced in 2010 (clinicaltri- shock, fibrinopeptide Bb15–42 improved pulmonary
als.gov identifier NCT01063010); the status of this and circulatory function and reduced plasma IL-6
trial is currently unknown. levels and neutrophil influx into the myocardium,
liver and small intestine [116]. Moreover, in a
murine polymicrobial sepsis model, fibrinopeptide
Sphingosine-1-phosphate
Bb15–42 treatment reduced proinflammatory cyto-
Sphingosine-1-phosphate (S1P) is an endogenous kine levels in the lung, liver and blood, decreased
bioactive sphingolipid produced in many types of neutrophil infiltration into the lung and attenuated
cells that are highly abundant in plasma and liver damage, possibly through maintaining vascu-
regulate endothelial barrier function by the activa- lar integrity and suppressing vascular leakage
tion of its G-protein-coupled receptor S1P1 [109]. [117]. In several models of acute lung injury,
The interaction between S1P and S1P1 on endo- produced by airway exposure to LPS or acid,
thelial cells enhances (i) vascular barrier function fibrinopeptide Bb15–42 reduced proinflammatory
by downstream activation of small GTPases, (ii) cytokine levels, neutrophil influx and vascular
cytoskeletal reorganization, (iii) adherens junction leak; moreover, fibrinopeptide Bb15–42 enhanced
and tight junction assembly and (iv) focal adhesion bacterial clearance and survival in mice with acid
formation. Other barrier-enhancing agents may aspiration-induced lung injury subsequently

286 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

challenged with Pseudomonas aeruginosa [118]. was shown to induce endothelial permeability via
Because of its myocardial protective effects in activation of the adaptor MyD88 by a route that did
experimental ischaemia–reperfusion injury [114], not rely on NF-jB activation. Rather, MyD88,
fibrinopeptide Bb15–42 (FX06) has been evaluated in which also functions as the common adaptor of
a multicentre Phase IIa clinical trial in patients with Toll-like receptor (TLR) signalling, activated a dis-
myocardial infarction; in this study, fibrinopeptide tinct NF-jB-independent pathway, through the
Bb15–42 significantly reduced the size of the necrotic small GTPase ADP-ribosylation factor 6 (ARF)6
core zone of infarcts [119]. As such, fibrinopeptide and its activator ARF nucleotide-binding site
Bb15–42 is a promising therapeutic agent, although opener (ARNO; also known as CYTH2). ARNO binds
its mechanism of action remains unclear. directly to MyD88, suggesting that MyD88–ARNO–
ARF6 functions as a proximal IL-1b and TLR
signalling pathway distinct from that mediated by
Robo4 and Slit
NF-jB. SecinH3, an inhibitor of ARF guanine
Endothelial cells express the receptor Robo4 nucleotide-exchange factors such as ARNO,
which, after binding to its ligand Slit, inhibits enhanced vascular stability and improved out-
inflammation-induced endothelial permeability by comes in animal models of sterile inflammation
consolidation of adherens junctions and modifica- [122]. The relevance of MyD88–ARNO–ARF6 sig-
tion of cytoskeletal dynamics [41, 120, 121]. It is nalling and the effect of SecinH3 have not yet been
likely that Slit stabilizes the vasculature by investigated in human sepsis.
enhancing VE-cadherin localization to the cell
surface. Indeed, it was shown that Slit2N, an active
Ang-1 and Ang-2
fragment of Slit2, increased VE-cadherin and
p120-catenin localization to the endothelial cell Ang-1 and Ang-2 are widely studied biomarkers of
surface [41]. Accordingly, Slit2N reduced endothe- endothelial activation and dysfunction in sepsis
lial cell monolayer permeability in vitro caused by [123]. Ang-1 is produced constitutively, in partic-
inflammatory mediators, including VEGF, LPS, ular by pericytes and smooth muscle cells, whereas
TNF-a and IL-1b [41, 121]. In vivo, Slit2N attenu- Ang-2 is produced by endothelial cells where it is
ated the accumulation of neutrophils and protein stored in Weibel–Palade bodies for quick release
exudates in the alveolar space of LPS-treated mice. upon exposure to inflammatory stimuli. Ang-1 and
This effect of Slit2N was absent in Robo4-deficient Ang-2 bind to and inhibit the endothelial cell Tie2
mice, demonstrating the importance of this recep- receptor. Under physiological conditions, circulat-
tor and also suggesting that the effect of Slit2N is ing Ang-1 levels exceed those of Ang-2, enabling
endothelial cell specific as Robo4 expression was preferential interaction between Ang-1 and the
restricted to this cell type [121]. In addition, the Tie2 receptor. This triggers pro-survival pathways
Slit2N-dependent reduction in endothelial cell and inhibits pro-inflammatory responses, resulting
monolayer permeability was inhibited in the pres- in endothelial cell quiescence.
ence of a VE-cadherin blocking antibody, confirm-
ing the importance of the action of Slit2N on The inflammatory response that accompanies sep-
VE-cadherin in vivo. Similarly, in mice with sis causes exocytosis of Weibel–Palade bodies and
abdominal sepsis caused by caecal ligation and release of Ang-2, which results in a shift towards
puncture, Slit2N inhibited the leakage of Evans Ang-2–Tie2 interaction. This interaction promotes
Blue dye in the kidney and spleen, indicating pro-inflammatory and pro-thrombotic pathways,
preserved vascular barrier function and improved microvascular leak and angiogenic stimuli. In
survival. Slit2N also inhibited lung permeability in clinical sepsis, a rise in the Ang-2:Ang-1 ratio,
a mouse model of avian H5N1 influenza [41]. normally low under nonstressed conditions, is an
Hence, the Slit–Robo4 interaction is important for indicator of acute vascular dysfunction [106]. In
maintaining vascular integrity in multiple settings, accordance with this, elevated Ang-2 levels were
and Slit2N may be an attractive therapeutic agent found to be correlated with severity of illness and
in conditions associated with disruption of the adverse outcomes in patients with sepsis [124,
endothelial barrier, including sepsis. 125]. The functional role of Ang-2 was illustrated
by experiments with heterozygous mice missing
Recently, a novel pathway important for endothe- one Ang-2 allele; these animals were partially
lial cell integrity that functions independently from protected against vascular leak, acute lung injury
NF-jB has been identified [122]. Specifically, IL-1b and death in models of caecal ligation and

ª 2014 The Association for the Publication of the Journal of Internal Medicine 287
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

puncture and LPS-induced toxicity [126]. Ang-1– vasculotide protected against vascular leak and
Tie2-targeted strategies have been tested in pre- mortality in polymicrobial sepsis in mice [127].
clinical sepsis models. The synthetic Tie2 agonist Similarly, vasculotide and the Ang-1-mimetic

Table 2 Potential new treatment options to prevent or treat endothelial barrier dysfunction in sepsis

Therapeutic agent Proposed mechanism of action Current developmental status


Selepressin and other Potent vasopressors that specifically Phase II clinical trials [138]
V1A receptor agonists bind to V1A receptors reducing
permeability and limiting
oedema formation
C’5a monoclonal antibody Blocks C’5a anaphylotoxin Early clinical trials planned
from binding to its receptor for sepsis [139]
and prevents excess
endothelial permeability
and C5a-induced apoptosis
Recombinant human Promote APC formation which Available in Japan, in Phase III
thrombomodulin and prevents endothelial apoptosis; clinical trials in Europe and North
APC variants induce carboxypeptidase activity and South America [140]
to degrade C’5a
Ang-1/Tie2 agents Inhibit NF-jB signalling and Preclinical studies [127, 128]
possess endothelial cell
anti-apoptotic activity
S1P1 agonists Stimulate VE-cadherins, Preclinical studies [109]
actin polymerization at
adherens junctions of the
endothelium
Fibrinopeptide Bb15–42 (FXO6) Fibrin cleavage product that Phase II clinical trials in myocardial
binds VE-cadherin and infarction, sepsis preclinical and
stabilizes interendothelial junctions early clinical studies [118, 119]
Slit2N agonists Stabilize tight junctions between Preclinical studies [41]
endothelial cells
Pepducins Lipidated peptides that are super-agonists Preclinical studies [136]
of PAR2, inducing Rac-mediated
barrier stabilization
Anti-HMGB1 mAb Blocks HMGB1-mediated loss of Preclinical studies [131–135]
endothelial barrier function and
upregulation of cytokine generation
and of adhesion molecules
Anti-VEGF receptor 2 mAb Blocks VEGF2-mediated breakdown Early clinical trials [108]
of VE-cadherin from the adherens
junction in endothelial cells

Ang, angiopoietin; S1P, sphingosine 1 phosphate receptor; V1A, vasopressin 1A receptor; C’, complement; APC, activated
protein C; NF-jB, nuclear factor kappa light-chain enhancer of B cells; VE, vascular endothelium; Tie2, tyrosine kinase
with immunoglobulin-like and epithelial growth factor-like domains; PAR, protease-activated receptor; Rac, a subfamily of
GTPases; HMGB1, high-mobility group box 1; VEGF, vascular endothelial growth factor; mAb, monoclonal antibody.

288 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

MAT.Ang-1 attenuated vascular leak induced by remove their anticoagulant properties and promote
LPS in mice [128, 129]. Strategies that either enhance their cytoprotective capacity, are possible novel
the Ang-1–Tie2 interaction or inhibit the effects of treatments for sepsis [110]. Agents are also in
Ang-2 warrant further investigation in sepsis. development that directly protect the endothelial
barrier, including Ang-1 and Tie2 agonists
[127–129], S1P1 agonists [109], fibrinopeptide
High-mobility group box 1
Bb15–42 therapy [118, 119] and Slit2N agonists,
High-mobility group box 1 (HMGB1), first by stabilizing tight junctions between endothelial
described as a DNA-binding nuclear protein in cells [121]. Another strategy is the use of pepduc-
eukaryotic cells, regulates chromosomal replica- ins, small lipidated peptides that can readily cross
tion, transcription and DNA repair [130]. It has cell membranes and either block PAR1/RhoA bar-
subsequently been found that HMGB1 has a crit- rier disruption or function as super-agonists of
ical role in the innate immune response to infection PAR2/Rac-mediated barrier stabilization, as
and injury as a late-acting, proinflammatory cyto- potential therapies for septic shock [136, 137].
kine-like protein. HMGB1 release from either acti- Table 2 provides a summary of the novel treatment
vated or necrotic cells (particularly myeloid and options now in clinical development that specifi-
endothelial cells), but not from apoptotic cells, cally target endothelial barrier function in sepsis
results in this inflammatory action [131, 132]. The [138–140].
endothelial lining is both a source of HMGB1
release in sepsis and a target of HMGB1-mediated Novel interventions directed at re-establishing
inflammatory actions during septic shock or other endothelial barrier function in sepsis could be a
severe forms of noninfectious tissue injury. major addition to the therapeutic armamentarium
HMGB1 functions as an ‘alarmin’, or damage- at a time when antibiotics are failing and results
associated molecular pattern, inducing and main- with other adjuvant therapies have been disap-
taining the acute inflammatory response. Elevated pointing. This is an exciting area of research at
circulating levels of HMGB1 are detected by a present, and clinical trials should soon provide
series of receptors expressed within the microcir- information to determine whether targeting the
culation including the receptor for advanced gly- endothelium will benefit patients in septic shock.
cated end products, TLR2 and TLR4 [130, 131].
HMGB1 disrupts endothelial barriers, alters the
Conflict of interest statement
actin filament cytoskeleton, impairs tight junc-
tions, promotes the release of large quantities of IL- Our institution receives a grant from Asahi Kasei to
1a as well as an array of other cytokines and run the clinical coordinating centre for soluble
chemokines, and stimulates enhanced expression thrombomodulin.
of cell surface adhesion components such as
ICAM1 and VCAM1 on endothelial membranes
[133–135]. Antibodies directed against HMGB1
References
can reverse many of these changes seen in exper-
imental animal models of sepsis, indicating that 1 McEvoy C, Kollef MH. Determinants of hospital mortality
clinical development of HMGB1 inhibitors should among patients with sepsis or septic shock receiving appro-
be considered (see Fig. 3 and Table 2). priate antibiotic treatment. Curr Infect Dis Rep 2013; 15:
400–6.
2 Angus DC, van der Poll T. Severe sepsis and septic shock. N
Conclusion Engl J Med 2013; 369: 840–51.
3 Bosmann M, Ward PA. The inflammatory response in sepsis.
Recovery and maintenance of the endothelial bar- Trends Immunol 2013; 34: 129–36.
rier is critical to survival in sepsis. New therapeutic 4 Vincent J-L, Opal SM, Marshall JC, Tracey KJ. Sepsis
targets for the management of sepsis at the level of definitions: time for change. Lancet 2013; 381: 774–5.
5 Dellinger RP, Levy MM, Rhodes A et al. The surviving sepsis
the endothelium might become clinically applica-
guidelines: 2012. Crit Care Med 2013; 41: 580–637.
ble as the cellular and molecular mechanisms that 6 Opal SM, Dellinger RP, Vincent J-L, Masur H, Angus DC.
cause barrier dysfunction become better under- The next generation of sepsis clinical trial designs: what is
stood. Agents are in development that promote cell next after the demise of recombinant human activated
survival by targeting mitochondria and cellular protein C? Crit Care Med 2014; 42: 1714–21.
energetics [7, 35, 36]. Recombinant variants of 7 Deutchman CS, Tracey KJ. Sepsis: current dogma and new
perspectives. Immunity 2014; 40: 463–75.
APC, which have been specifically engineered to

ª 2014 The Association for the Publication of the Journal of Internal Medicine 289
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

8 Hotchkiss RS, Opal SM. Immunotherapy for sepsis: a new 30 Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome:
approach against an ancient foe. N Engl J Med 2010; 363: a sensor for metabolic danger? Science 2010; 327: 296–300.
87–9. 31 Borovikova LV, Ivanova S, Zhang M et al. Vagus nerve
9 Angus DC. The search for effective therapy for sepsis: back stimulation attenuates the systemic inflammatory response
to the drawing board? JAMA 2011; 306: 2614–5. to endotoxin. Nature 2000; 405: 458–62.
10 Ranieri VM, Thompson BT, Barie PS et al. Drotrecogin alfa 32 Chiu IMI, Heesters BA, Ghasemlou N et al. Bacteria activate
(activated) for adults with septic shock. N Engl J Med 2012; sensory neurons that modulate pain and inflammation.
366: 2055–64. Nature 2013; 501: 52–7.
11 Kaukonen K-M, Bailey M, Suzuki S, Pilcher D, Bellomo R. 33 Rosas-Ballina M, Ochani M, Parrish WR et al. Splenic nerve
Mortality related to severe sepsis and septic shock among is required for cholinergic anti-inflammatory pathway con-
critically ill patients in Australia and New Zealand. JAMA trol of TNF in endotoxemia. Proc Natl Acad Sci USA 2008;
2014; 331: 1308–16. 105: 11008–13.
12 Galbois A, Aegerter P, Martel-Samb P et al. Improved prog- 34 Casaer MP, van den Berghe G. Nutrition in the acute phase
nosis of septic shock in patients with cirrhosis: a multicenter of critical illness. N Engl J Med 2014; 78: 1227–35.
study. Crit Care Med 2014; 42: 1666–75. 35 Levy RJ, Deutschman CS. Cytochrome c oxidase dysfunc-
13 Yealy DM, Kellum JA, Huang DT et al. A randomized trial of tion in sepsis. Crit Care Med Suppl 2007; 35: S468–75.
protocol-based care for early septic shock. N Engl J Med 36 Horng T. Calcium signaling and mitochondrial destabiliza-
2014; 370: 1683–93. tion in the triggering of the NLRP3 inflammasome. Trends
14 Andersson U, Tracey KJ. Neural reflexes in inflammation Immunol 2014; 35: 253–61.
and immunity. J Exp Med 2012; 209: 1057–68. 37 Walton AH, Muenzer JT, Rasche D et al. Reactivation of
15 Brealey D, Brand M, Hargreaves I et al. Association between multiple viruses in patients with sepsis. PLoS ONE 2014; 9:
mitochondrial dysfunction and severity and outcome of e98819.
septic shock. Lancet 2002; 360: 219–23. 38 Cavaillon J-M, Eisen D, Annane D. Is boosting the immune
16 Haneklaus M, O’Neill LA, Coli RC. Modulatory mechanisms system in sepsis appropriate? Crit Care 2014; 18: 216.
controlling the NLRP3 inflammasome in inflammation: 39 Jani IV, Peter TF. How point-of-care testing could drive
recent developments. Curr Opin Immunol 2013; 25: 40–5. innovation in global health. N Engl J Med 2013; 368: 2319–
17 Iskander KN, Osuchowski MF, Stearns-Kurosawa DJ et al. 24.
Sepsis: multiple abnormalities, heterogeneous responses, 40 Lee WL, Slutsky AS. Sepsis and endothelial permeability. N
and evolving understanding. Physiol Rev 2013; 93: 1247– Engl J Med 2010; 363: 689–91.
88. 41 London NR, Zhu W, Bozza FA et al. Targeting Robo4-
18 McGettrick AF, O’Neill LA. How metabolism generates dependent Slit signaling to survive the cytokine storm in
signals during innate immunity and inflammation. J Biol sepsis and influenza. Sci Transl Med 2010; 2: 23ra19.
Chem 2013; 288: 22893–8. 42 Aird WC. The role of the endothelium in severe sepsis and
19 Medzhitov R, Schneider DS, Soares MP. Disease tolerance as multiple organ dysfunction syndrome. Blood 2003; 101:
a defense strategy. Science 2012; 335: 936–41. 3765–77.
20 Ward PA, Bosmann M. A historical perspective on sepsis. Am 43 Schouten M, Wiersinga WJ, Levi M, van der Poll T. Inflam-
J Pathol 2012; 181: 2–7. mation, endothelium, and coagulation in sepsis. J Leukoc
21 Nakahira K, Haspel JA, Rathinam VAK et al. Autophagy Biol 2008; 83: 536–45.
proteins regulate innate immune responses by inhibiting the 44 Levi M, van der Poll T. Endothelial injury in sepsis. Intensive
release of mitochondrial DNA mediated by the NALP3 Care Med 2013; 39: 1839–42.
inflammasome. Nat Immunol 2011; 12: 222–30. 45 Levi M, van der Poll T. Inflammation and coagulation. Crit
22 Nathan C, Ding A. Nonresolving inflammation. Cell 2010; Care Med 2010; 38(2 Suppl): S26–34.
140: 871–82. 46 Creasey AA, Chang AC, Feigen L, Wun TC, Taylor FB Jr,
23 Singer M. The role of mitochondrial dysfunction in sepsis- Hinshaw LB. Tissue factor pathway inhibitor reduces mor-
induced multi-organ failure. Virulence 2014; 5: 66–72. tality from Escherichia coli septic shock. J Clin Invest 1993;
24 Xiao W, Mindrinros MN, Seok J et al. A genomic storm in 91: 2850–6.
critical injured humans. J Exp Med 2011; 208: 2581–90. 47 Carr C, Bild GS, Chang AC et al. Recombinant E. coli-derived
25 Chavan SS, Huerta PT, Robbiati S et al. HMGB1 mediates tissue factor pathway inhibitor reduces coagulopathic and
cognitive impairment in sepsis survivors. Mol Med 2012; 18: lethal effects in the baboon gram-negative model of septic
930–7. shock. Circ Shock 1994; 44: 126–37.
26 Andersson U, Tracey KJ. HMGB1 is a therapeutic target for 48 Pawlinski R, Pedersen B, Schabbauer G et al. Role of tissue
sterile inflammation and infection. Annu Rev Immunol 2011; factor and protease-activated receptors in a mouse model of
29: 139–62. endotoxemia. Blood 2004; 103: 1342–7.
27 Angus DC. The lingering consequences of sepsis: the Hidden 49 Meziani F, Delabranche X, Asfar P, Toti F. Bench-to-bedside
public health disaster? JAMA 2010; 304: 1833–4. review: circulating microparticles–a new player in sepsis?
28 Boomer JS, To K, Chang KC et al. Immunosuppression in Crit Care 2010; 14: 236.
patients who die of sepsis and multiple organ failure. JAMA 50 Szotowski B, Antoniak S, Rauch U. Alternatively spliced
2011; 306: 2594–605. tissue factor: a previously unknown piece in the puzzle of
29 Felmet KA, Hall MW, Clark RSB, Jaffe R, Carcillo JA. hemostasis. Trends Cardiovasc Med 2006; 16: 177–82.
Prolonged lymphopenia, lymphoid depletion, and hypoprol- 51 Monroe DM, Key NS. The tissue factor-factor VIIa complex:
actinemia in children with nosocomial sepsis and multiple procoagulant activity, regulation, and multitasking.
organ failure. J Immunol 2005; 174: 3765–72. J Thromb Haemost 2007; 5: 1097–105.

290 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

52 Ott I, Miyagi Y, Miyazaki K et al. Reversible regulation of 69 Saffarzadeh M, Juenemann C, Queisser MA et al. Neutrophil
tissue factor-induced coagulation by glycosyl phosphatidyl- extracellular traps directly induce epithelial and endothelial
inositol-anchored tissue factor pathway inhibitor. Arterios- cell death: a predominant role of histones. PLoS ONE 2012;
cler Thromb Vasc Biol 2000; 20: 874–82. 7: e32366.
53 Sandset PM, Warn-Cramer BJ, Maki SL, Rapaport SI. 70 Riewald M, Ruf W. Mechanistic coupling of protease signal-
Immunodepletion of extrinsic pathway inhibitor sensitizes ing and initiation of coagulation by tissue factor. Proc Natl
rabbits to endotoxin-induced intravascular coagulation and Acad Sci USA 2001; 98: 7742–7.
the generalized Shwartzman reaction. Blood 1991; 78: 71 Riewald M, Petrovan RJ, Donner A, Mueller BM, Ruf W.
1496–502. Activation of endothelial cell protease activated receptor 1 by
54 Sandset PM, Warn-Cramer BJ, Rao LV, Maki SL, Rapaport the protein C pathway. Science 2002; 296: 1880–2.
SI. Depletion of extrinsic pathway inhibitor (EPI) sensitizes 72 Kaneider NC, Leger AJ, Agarwal A et al. ‘Role reversal’ for the
rabbits to disseminated intravascular coagulation induced receptor PAR1 in sepsis-induced vascular damage. Nat
with tissue factor: evidence supporting a physiologic role for Immunol 2007; 8: 1303–12.
EPI as a natural anticoagulant. Proc Natl Acad Sci USA 1991; 73 Covic L, Gresser AL, Talavera J, Swift S, Kuliopulos A.
88: 708–12. Activation and inhibition of G protein-coupled receptors by
55 de Jonge E, Dekkers PE, Creasey AA et al. Tissue factor cell-penetrating membrane-tethered peptides. Proc Natl
pathway inhibitor dose-dependently inhibits coagulation Acad Sci U S A 2002; 99: 643–8.
activation without influencing the fibrinolytic and cytokine 74 Wielders SJ, Bennaghmouch A, Reutelingsperger CP, Bevers
response during human endotoxemia. Blood 2000; 95: EM, Lindhout T. Anticoagulant and antithrombotic proper-
1124–9. ties of intracellular protease-activated receptor antagonists.
56 Danese S, Vetrano S, Zhang L, Poplis VA, Castellino FJ. The J Thromb Haemos 2007; 5: 571–6.
protein C pathway in tissue inflammation and injury: 75 Tressel SL, Kaneider NC, Kasuda S et al. A matrix metallo-
pathogenic role and therapeutic implications. Blood 2010; protease-PAR1 system regulates vascular integrity, systemic
115: 1121–30. inflammation and death in sepsis. EMBO Mol Med 2011; 3:
57 Taylor FB Jr, Chang A, Esmon CT, D’Angelo A, Vigano- 370–84.
D’Angelo S, Blick KE. Protein C prevents the coagulopathic 76 Austin KM, Covic L, Kuliopulos A. Matrix metalloproteases
and lethal effects of Escherichia coli infusion in the baboon. and PAR1 activation. Blood 2013; 121: 431–9.
J Clin Invest 1987; 79: 918–25. 77 Austin KM, Nguyen N, Javid G, Covic L, Kuliopulos A.
58 Taylor FB Jr, Stearns-Kurosawa DJ, Kurosawa S et al. The Noncanonical matrix metalloprotease-1-protease-activated
endothelial cell protein C receptor aids in host defense receptor-1 signaling triggers vascular smooth muscle cell
against Escherichia coli sepsis. Blood 2000; 95: 1680–6. dedifferentiation and arterial stenosis. J Biol Chem 2013;
59 Moake JL. Thrombotic microangiopathies. N Engl J Med 288: 23105–15.
2002; 347: 589–600. 78 Dinarello CA. Interleukin-1 in the pathogenesis and treat-
60 Bockmeyer CL, Claus RA, Budde U et al. Inflammation- ment of inflammatory diseases. Blood 2011; 117: 3720–32.
associated ADAMTS13 deficiency promotes formation of 79 Cohen I, Rider P, Carmi Y et al. Differential release of
ultra-large von Willebrand factor. Haematologica 2008; 93: chromatin-bound IL-1 alpha discriminates between necrotic
137–40. and apoptotic cell death by the ability to induce sterile
61 Booth KK, Terrell DR, Vesely SK, George JN. Systemic inflammation. Proc Natl Acad Sci USA 2010; 107: 2574–9.
infections mimicking thrombotic thrombocytopenic pur- 80 Eigenbrod T, Park JH, Harder J, Iwakura Y, Nunez G.
pura. Am J Hematol 2011; 86: 743–51. Cutting edge: critical role of mesothelial cells in necrosis-
62 de Stoppelaar SF, van ‘t Veer C, van der Poll T. The role induced inflammation through the recognition of IL-1
of platelets in sepsis. Thromb Haemost 2014; 26: 112 alpha released from dying cells. J Immunol 2008; 181:
(2). 8194–8.
63 Henn V, Slupsky JR, Grafe M et al. CD40 ligand on activated 81 Baer C, Squadrito ML, Iruela-Arispe ML, DePalma M.
platelets triggers an inflammatory reaction of endothelial Reciprocal interactions between endothelial cells and mac-
cells. Nature 1998; 391: 591–4. rophages in angiogenic vascular niches. Expert Cell Res
64 May AE, Kalsch T, Massberg S, Herouy Y, Schmidt R, Gawaz 2013; 319: 1626–34.
M. Engagement of glycoprotein IIb/IIIa (alpha(IIb)beta3) on 82 Parikh SM, Mammoto T, Schultz H et al. Excess circulating
platelets upregulates CD40L and triggers CD40L-dependent angiopoietin-2 may contribute to pulmonary vascular leak in
matrix degradation by endothelial cells. Circulation 2002; sepsis in humans. PLoS Med 2006; 3: e46.
106: 2111–7. 83 Huges DP, Marron MB, Brindle NP. The anti-inflammatory
65 Furie B, Furie BC. Role of platelet P-selectin and micropar- endothelial tyrosine kinase Tie2 interacts with a novel
ticle PSGL-1 in thrombus formation. Trends Mol Med 2004; nuclear factor-Kappa B inhibitor ABIN-2. Cir Res 2003; 92:
10: 171–8. 630–6.
66 Gardiner EE, Andrews RK. Neutrophil extracellular traps 84 Thery C, Zitvogel L, Amigorena S. Exosomes: composition,
(NETs) and infection-related vascular dysfunction. Blood Rev biogenesis and function. Nat Rev Immunol 2002; 2: 569–79.
2012; 26: 255–9. 85 Simons M, Raposo G. Exosomes-vesicular carriers of inter-
67 Kaplan MJ, Radic M. Neutrophil extracellular traps: double- cellular communication. Curr Opin Cell Biol 2009; 21: 575–
edged swords of innate immunity. J Immunol 2012; 189: 81.
2689–95. 86 Zhang Y, Liu D, Chen X et al. Secreted monocytic miR-150
68 Martinod K, Wagner DD. Thrombosis: tangled up in NETs. enhances targeted endothelial cell migration. Mol Cell 2010;
Blood 2014; 123: 2768–76. 39: 133–44.

ª 2014 The Association for the Publication of the Journal of Internal Medicine 291
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

87 He H, Xu J, Carmen M et al. Endothelial cells provide an action with microvascular endothelium. Crit Care Med 2002;
instructive niche for the differentiation and functional 30: 218–25.
polarization of M2-like macrophages. Blood 2012; 120: 104 Iba T, Okamoto K, Ohike T et al. Enoxaparin and fondapar-
3152–62. inux attenuates endothelial damage in endotoxemic rats. J
88 Burnier L, Fontana P, Kwak BR, Angelillo-Scherrer A. Cell- Trauma Acute Care Surg 2012; 72: 177–82.
derived microparticles in haemostasis and vascular medi- 105 Goldenberg NM, Steinberg BE, Slutsky AS, Lee WL. Broken
cine. Thromb Haemost 2009; 101: 439–51. barriers: a new take on sepsis pathogenesis. Sci Transl Med
89 Norling LV, Dalli J. Microparticles are novel effectors of 2011; 22: 3 (88):88 ps25.
immunity. Curr Opin Pharmacol 2013; 13: 570–5. 106 Darwish I, Liles WC. Emerging therapeutic strategies to
90 Berckmans RJ, Nieuwland R, Boing AN, Romijn FP, Hack prevent infection-related microvascular endothelial activa-
CE, Sturk A. Cell-derived microparticles circulate in healthy tion and dysfunction. Virulence 2013; 4: 572–82.
humans and support low grade thrombin generation. 107 Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF
Thromb Haemost 2001; 85: 639–46. receptor signalling - in control of vascular function. Nat Rev
91 Jy W, Jimenez JJ, Mauro LM et al. Endothelial microparti- Mol Cell Biol 2006; 7: 359–71.
cles induce formation of platelet aggregates via a von 108 Gavard J, Gutkind JS. VEGF controls endothelial-cell per-
Willebrand factor/ristocetin dependent pathway, rendering meability by promoting the beta-arrestin-dependent endo-
them resistant to dissociation. J Thromb Haemost 2005; 3: cytosis of VE-cadherin. Nat Cell Biol 2006; 8: 1223–34.
1301–8. 109 Wang L, Dudek SM. Regulation of vascular permeability by
92 Satta N, Toti F, Feugeas O et al. Monocyte vesiculation is a sphingosine 1-phosphate. Microvasc Res 2009; 77: 39–45.
possible mechanism for dissemination of membrane-associ- 110 Feistritzer C, Riewald M. Endothelial barrier protection by
ated procoagulant activities and adhesion molecules after activated protein C through PAR1-dependent sphingosine 1-
stimulation by lipopolysaccharide. J Immunol 1994; 153: phosphate receptor-1 crossactivation. Blood 2005; 105:
3245–55. 3178–84.
93 Delabranche X, Boisrame-Helms J, Asfar P et al. Micropar- 111 Singleton PA, Moreno-Vinasco L, Sammani S, Wanderling
ticles are new biomarkers of septic shock-induced dissem- SL, Moss J, Garcia JG. Attenuation of vascular permeability
inated intravascular coagulopathy. Intensive Care Med by methylnaltrexone: role of mOP-R and S1P3 transactiva-
2013; 39: 1695–703. tion. Am J Respir Cell Mol Biol 2007; 37: 222–31.
94 Perez-Casal M, Downey C, Cutillas-Moreno B, Zuzel M, 112 Bae JS, Kim YU, Park MK, Rezaie AR. Concentration
Fukudome K, Toh CH. Microparticle-associated endothelial dependent dual effect of thrombin in endothelial cells via
protein C receptor and the induction of cytoprotective and Par-1 and Pi3 Kinase. J Cell Physiol 2009; 219: 744–51.
anti-inflammatory effects. Haematologica 2009; 94: 387–94. 113 Zhang G, Yang L, Kim GS et al. Critical role of sphingosine-
95 Perez-Casal M, Thompson V, Downey C et al. The clinical 1-phosphate receptor 2 (S1PR2) in acute vascular inflam-
and functional relevance of microparticles induced by acti- mation. Blood 2013; 122: 443–55.
vated protein C treatment in sepsis. Crit Care 2011; 15: 114 Petzelbauer P, Zacharowski PA, Miyazaki Y et al. The fibrin-
R195. derived peptide Bbeta15-42 protects the myocardium against
96 Lundy DJ, Trzeciak S. Microcirculatory dysfunction in ischemia-reperfusion injury. Nat Med 2005; 11: 298–304.
sepsis. Crit Care Nurs Clin North Am 2011; 23: 67–77. 115 Groger M, Pasteiner W, Ignatyev G et al. Peptide Bbeta(15-
97 De Backer D, Orbegozo Cortes D, Donadello K, Vincent JL. 42) preserves endothelial barrier function in shock. PLoS
Pathophysiology of microcirculatory dysfunction and the ONE 2009; 4: e5391.
pathogenesis of septic shock. Virulence 2014; 5: 73–9. 116 Roesner JP, Petzelbauer P, Koch A et al. Bbeta15-42 (FX06)
98 Edul VS, Enrico C, Laviolle B, Vazquez AR, Ince C, Dubin A. reduces pulmonary, myocardial, liver, and small intestine
Quantitative assessment of the microcirculation in healthy damage in a pig model of hemorrhagic shock and reperfu-
volunteers and in patients with septic shock. Crit Care Med sion. Crit Care Med 2009; 37: 598–605.
2012; 40: 1443–8. 117 Jennewein C, Mehring M, Tran N et al. The fibrinopeptide
99 De Backer D, Donadello K, Sakr Y et al. Microcirculatory bbeta15-42 reduces inflammation in mice subjected to
alterations in patients with severe sepsis: impact of time of polymicrobial sepsis. Shock 2012; 38: 275–80.
assessment and relationship with outcome. Crit Care Med 118 Matt U, Warszawska JM, Bauer M et al. Bbeta(15-42)
2013; 41: 791–9. protects against acid-induced acute lung injury and sec-
100 Piagnerelli M, Boudjeltia KZ, Vanhaeverbeek M, Vincent JL. ondary pseudomonas pneumonia in vivo. Am J Respir Crit
Red blood cell rheology in sepsis. Intensive Care Med 2003; Care Med 2009; 180: 1208–17.
29: 1052–61. 119 Atar D, Petzelbauer P, Schwitter J et al. Effect of intravenous
101 Gierer P, Hoffmann JN, Mahr F et al. Activated protein C FX06 as an adjunct to primary percutaneous coronary
reduces tissue hypoxia, inflammation, and apoptosis in intervention for acute ST-segment elevation myocardial
traumatized skeletal muscle during endotoxemia. Crit Care infarction results of the F.I.R.E. (Efficacy of FX06 in the
Med 2007; 35: 1966–71. Prevention of Myocardial Reperfusion Injury) trial. J Am Coll
102 De Backer D, Verdant C, Chierego M, Koch M, Gullo A, Cardiol 2009; 53: 720–9.
Vincent JL. Effects of drotrecogin alfa activated on micro- 120 Jones CA, London NR, Chen H et al. Robo4 stabilizes the
circulatory alterations in patients with severe sepsis. Crit vascular network by inhibiting pathologic angiogenesis and
Care Med 2006; 34: 1918–24. endothelial hyperpermeability. Nat Med 2008; 14: 448–53.
103 Hoffmann JN, Vollmar B, Romisch J, Inthorn D, Schildberg 121 Jones CA, Nishiya N, London NR et al. Slit2-Robo4 signal-
FW, Menger MD. Antithrombin effects on endotoxin-induced ling promotes vascular stability by blocking Arf6 activity.
microcirculatory disorders are mediated mainly by its inter- Nat Cell Biol 2009; 11: 1325–31.

292 ª 2014 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2015, 277; 277–293
S. M. Opal & T. van der Poll Review: Endothelial dysfunction in sepsis

122 Zhu W, London NR, Gibson CC et al. Interleukin receptor cell-penetrating pepducins. Proc Natl Acad Sci U S A 2011;
activates a MYD88-ARNO-ARF6 cascade to disrupt vascular 108: 8491–6.
stability. Nature 2012; 492: 252–5. 133 Boucheix OB, Milano SP, Henriksson M, Reinheimer TM.
123 Page AV, Liles WC. Biomarkers of endothelial activation/ Selepressin, a new V1A receptor agonist: hemodynamic
dysfunction in infectious diseases. Virulence 2013; 4: 507–16. comparison to vasopressin in dogs. Shock 2013; 39: 533–8.
124 Parikh SM, Mammoto T, Schultz A et al. Excess circulating 134 Ward P. The harmful role of C5a on innate immunity in
angiopoietin-2 may contribute to pulmonary vascular leak sepsis. J Innate Immun 2010; 2: 439–45.
in sepsis in humans. PLoS Med 2006; 3: e46. 135 Vincent J-L, Ramesh MK, Ernest D et al. A randomized,
125 Kumpers P, Lukasz A, David S et al. Excess circulating double-blind, placebo-controlled, phase 2b study to evalu-
angiopoietin-2 is a strong predictor of mortality in critically ate the safety and efficacy of recombinant human soluble
ill medical patients. Crit Care 2008; 12: R147. thrombomodulin, ART-123, in patients with sepsis and
126 David S, Mukherjee A, Ghosh CC et al. Angiopoietin-2 may suspected disseminated intravascular coagulation. Crit Care
contribute to multiple organ dysfunction and death in Med 2013; 41: 2069–79.
sepsis*. Crit Care Med 2012; 40: 3034–41. 136 Denk S, Perl M, Huber-Lang M. Damage and pathogen
127 Kumpers P, Gueler F, David S et al. The synthetic tie2 associated molecular patterns and alarmins: keys to sepsis?
agonist peptide vasculotide protects against vascular leak- Eur Surg Res 2014; 48: 171–9.
age and reduces mortality in murine abdominal sepsis. Crit 137 Wolfson RK, Chiang ET, Garcia JGN. HMGB1 induces
Care 2011; 15: R261. human lung endothelial cell cytoskeletal rearrangement
128 David S, Ghosh CC, Kumpers P et al. Effects of a synthetic and barrier disruption. Microvasc Res 2011; 81: 189–97.
PEG-ylated Tie-2 agonist peptide on endotoxemic lung 138 Huang W, Liu Y, Li L et al. HMGB1 increases permeability of
injury and mortality. Am J Physiol Lung Cell Mol Physiol the endothelial cell monolayer via RAGE and Src family
2011; 300: L851–62. tyrosine kinases. Inflammation 2012; 35: 350–62.
129 Alfieri A, Watson JJ, Kammerer RA et al. Angiopoietin-1 139 Fiuza C, Bustin M, Talwar S et al. Inflammation-promoting
variant reduces LPS-induced microvascular dysfunction in a activity of HMGB1 on human microvascular endothelial
murine model of sepsis. Crit Care 2012; 16: R182. cells. Blood 2003; 101: 2652–60.
130 Lamkanfi M, Sarkar A, Walle LV et al. Inflammasome- 140 Rao DA, Tracey KJ, Pober JS. IL-1alpha and IL-1beta are
dependent release of the alarmin HMGB1 in endotoxemia. endogenous mediators linking cell injury to adaptive allore-
J Immunol 2010; 185: 4385–92. active responses. J Immunol 2007; 179: 6536–46.
131 Cisowski J, O’Callaghan K, Kuliopulos A et al. Targeting
protease-activated receptor-1 with cell-penetrating Correspondence: Steven M. Opal MD, Chief, Infectious Disease
pepducins in lung cancer. Am J Pathol 2011; 179: 513–23. Division, Memorial Hospital of Rhode Island, 111 Brewster Street,
132 Sevigny LM, Zhang P, Bohm A et al. Interdicting Pawtucket, RI 02860, USA.
protease-activated receptor-2-driven inflammation with (fax: 401-729-2795; e-mail: Steven_Opal@brown.edu).

ª 2014 The Association for the Publication of the Journal of Internal Medicine 293
Journal of Internal Medicine, 2015, 277; 277–293

You might also like