You are on page 1of 14

Concise Definitive Review

Molecular biology of inflammation and sepsis: A primer*


Ismail Cinel, MD, PhD; Steven M. Opal, MD

Background: Remarkable progress has been made during the dysfunction, sepsis-related immunosuppression, late mediators of
last decade in defining the molecular mechanisms that underlie systemic inflammation, control mechanisms for coagulation, and
septic shock. This rapidly expanding field is leading to new reprogramming of immune response genes all have critical roles in
therapeutic opportunities in the management of severe sepsis. the development of sepsis.
Aim: To provide the clinician with a timely summary of the Conclusions: Many of these basic discoveries have direct impli-
molecular biology of sepsis and to better understand recent cations for the clinical management of sepsis. The translation of
advances in sepsis research. these “bench-to-bedside” findings into new therapeutic strategies is
Data Selection: Medline search of relevant publications in already underway. This brief review provides the clinician with a
basic mechanisms of sepsis/severe sepsis/septic shock, and se- primer into the basic mechanisms responsible for the molecular
lected literature review of other manuscripts about the signalo- biology of sepsis, severe sepsis, and septic shock.(Crit Care Med
some, inflammasome, apoptosis, or mechanisms of shock. 2009; 37:291–304)
Data Synthesis and Findings: The identification of the toll-like KEY WORDS: sepsis; inflammation; toll like receptors; inflamma-
receptors and the associated concept of innate immunity based upon some; signalosome; apoptosis; neutrophils; mitochondrial dys-
pathogen- or damage-associated molecular pattern molecules al- function; reactive oxygen species; nitric oxide; peroxynitrite; ac-
lowed significant advances in our understanding of the pathophys- tivated protein C; a disintegrin-like and metalloproteinase with
iology of sepsis. The essential elements of the inflammasome and thrombospondin type-1 motifs-13; pathogen associated molecular
signal transduction networks responsible for activation of the host patterns; danger associated molecular patterns
response have now been characterized. Apoptosis, mitochondrial

I n sepsis, the expected and appropri- damage. In addition, a reduction in their (TLRs); 2) nucleotide-oligomerization
ate inflammatory response to an in- antimicrobial capacity may set the stage for domain leucine-rich repeat (NOD-LRR)
fectious process becomes amplified opportunistic and/or super infections (2). proteins; and 3) cytoplasmic caspase ac-
leading to organ dysfunction or risk Severe sepsis may also be associated with tivation and recruiting domain heli-
for secondary infection. A continuum exists an exaggerated procoagulant state. This cases such as retinoic-acid-inducible
from a low grade systemic response associ- may lead to ischemic cell injury, an effect gene I (RIG-I)-like helicases (RLHs) (4,
ated with a self-limited infection to a that further amplifies the damage caused 5). These receptors initiate the innate
marked systemic response with solitary or by inappropriate inflammation. A micro- immune response and regulate the
multiorgan dysfunction, i.e., severe sepsis. vasculature injured by inflammation and adaptive immune response to infection
As a clinical syndrome, sepsis occurs when ischemia, in turn, further deranges the or tissue injury.
an infection is associated with the systemic host response by altering leukocyte traffick- Gram-positive and Gram-negative bac-
inflammatory response. ing, generating apoptotic microparticles, teria, viruses, parasites, and fungi all pos-
The complex toll-like receptor signaling and increasing cellular hypoxia (3). Mito- sess a limited number of unique cellular
and associated downstream regulators of chondrial dysfunction, an acquired intrin- constituents not found in vertebrate ani-
immune cell functions play a crucial role in sic defect in cellular respiration termed “cy- mals. These elements are now referred to as
the innate system as a first line of defense topathic hypoxia,” also has an important PAMPs, or more appropriately microbial-
against pathogens (1). However, signaling role by decreasing cellular oxygen con- associated molecular patterns, as these
is sometimes conflicting and a sustained sumption in this chaotic process. In this molecules are also common in nonpatho-
inflammatory response can result in tissue review, we highlight the current under- genic and commensal bacteria (6). PAMPs
standing of the basic molecular mecha- bind to PRRs, such as TLRs, expressed on
nisms that modulate these events so as to the surface of host cells. Cytoplasmic PRRs
From the Division of Critical Care Medicine (IC), The produce sepsis. exist to detect invasive intracellular patho-
Robert Wood Johnson School of Medicine, The University gens (7). The NOD proteins recognize com-
of Medicine and Dentistry of New Jersey, Camden NJ; mon fragments of bacterial peptidoglycan.
and The Infectious Disease Division (SMO), Memorial
Pattern Recognition Receptors,
Hospital of RI, The Warren Alpert School of Medicine of Pathogen-Associated Molecular Diamino-pimelate from Gram-negative
Brown University, Providence, RI. Patterns (PAMPs) and Danger- bacteria is the ligand for NOD1 and mu-
Dr. Opal has received grants from Wyeth and Eisai
Associated Molecular Patterns ramyl dipeptide from peptidoglycan is the
Inc. Dr. Cinel has not disclosed any potential conflicts ligand for NOD2 in the cytosol (see Figs. 1
of interest. (DAMPs)
For information regarding this article, E-mail:
and 2). The PRRs also recognized damage
cinel-ismail@cooperhealth.edu The initiation of the host response signals from the release of endogenous pep-
Copyright © 2008 by the Society of Critical Care during sepsis or tissue injury involves tides and glycosaminoglycans from apopto-
Medicine and Lippincott Williams & Wilkins three families of pattern recognition re- tic or necrotic host cells (8 –10) Caspase
DOI: 10.1097/CCM.0b013e31819267fb ceptors (PRRs): 1) toll-like receptors activation and recruiting domain helicases

Crit Care Med 2009 Vol. 37, No. 1 291


(LPS)-induced mortality (15). Multiple
positive feedback loops between danger-
associated molecular patterns and
PAMPs, and their overlapping receptors
temporally and spatially drive these pro-
cesses and may represent the molecular
basis for the observation that infections,
as well as nonspecific stress factors, can
trigger flares in systemic inflammatory
response. The degree to which TLR reg-
ulation mediates the ultimate outcome in
sepsis in individual patients remains elu-
sive and is an active area of clinical in-
vestigation (13, 16).

Inflammasome and Signalosome


Pathways

Figure 1. Specific host immune response to each pathogen is mediated by various sets of pathogen TLRs induce pro-interleukin(IL)-1beta
associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) as detailed in Figure production and prime NLR-containing
1. PRRs are essential for initiating the host’s immune defenses against invading pathogens, yet they multiprotein complexes, termed “inflam-
can also contribute to persistent and deleterious systemic inflammation. PRRs also serve as receptors masomes,” to respond to bacterial prod-
for endogenous danger signals, hemodynamic changes in sepsis (tissue hypoperfusion and ischemia/ ucts and products of damaged cells (17,
reperfusion phenomenon), thus same signaling systems that alerts the highly advantageous, host 19). This results in caspase-1 activation
defense mechanisms, also contributes to the disadvantageous, pathologic events of systemic inflam- and the subsequent processing of pro-
mation, coagulation, tissue damage in target organs in sepsis. Heat shock proteins, fibrinogen,
IL-1␤ to its active extracellular form
fibronectin, hyaluran, biglycans and high mobility group box-1 (HMGB-1) have been defined as danger
associated molecular patterns (DAMPs) which are likely relevant for sepsis. Toll-like receptors (TLRs),
IL-1␤ (19). Caspases are a set of cysteine
especially TLR4, are involved in the recognition of these endogenous or harmful self-antigens ligands proteases that alter the enzymatic activity
which are released during noninfectious injury, such as trauma or ischemia/reperfusion suggesting of target proteins at specific peptide se-
their function may not be restricted to the recognition of extrinsic pathogens. NOD-LRR, nucleotide- quences adjacent to aspartate moieties.
oligomerization domain leucine-rich repeat; ASC, apoptosis-associated speck-like protein containing Caspases are important in process of ap-
caspase activation and recruiting domain; NF-␬␤, nuclear factor ␬␤. optosis, cellular regulation, and inflam-
mation (Figs. 1 and 3). One of the many
targets of the caspase cascade is caspase
activated DNase (CAD). CAD activation
induces DNA fragmentation characteris-
tic of programmed cell death (apoptosis).
The posttranslational activation of
caspase-1 is tightly regulated by inflam-
masome, the known components of
which include caspase-1, ASC (apoptosis-
associated speck-like protein containing a
caspase activation and recruiting do-
main), NALP1 (NACHT, leucine rich re-
peat and pyrin domain containing 1), and
caspase-5 (20). Additionally, alternate in-
Figure 2. Binding of toll-like receptors (TLRs) activates intracellular signal-transduction pathways that flammasome constructions have been
lead to the activation of transcriptional activators such as interferon regulator factors, phosphoino- suggested to contain pyrin, NALP3, and
sitide 3-kinase (PI3K)/Akt, activator protein-1, and cytosolic nuclear factor-kappa ␤ (NF-␬␤). Activated
other members of the NOD-LRR family
NF-␬␤ moves from the cytoplasm to the nucleus, binds to transcription sites and induces activation
of an array of genes for acute phase proteins, inducible nitric oxide synthase, coagulation factors,
(21–23). ASC facilitates inflammasome
proinflammatory cytokines, as well as enzymatic activation of cellular proteases. TLR9 DNA, TLR 3 assembly thus triggering caspase-1 acti-
dsRNA, and TLR7/8 ss RNA are endosomal. TLR 10 ligand is not defined and TLR1 forms heterodimers vation and IL-1␤ processing. Interest-
with TLR2. LPS, lipopolysaccharide; IRF, interferon regulatory factor; JNK; c Jun N-terminal kinase. ingly, ASC can also regulate the nuclear
factor (NF)-␬B pathway, thus linking the
inflammasome to the signalosome
primarily recognize viral nucleic acids and ates danger-associated molecular pat- (Fig. 1) (24).
activate antiviral measures including the terns (i.e., high mobility group box-1, The inflammasome pathways contrib-
type I interferons. heat shock proteins, S100 proteins, hya- ute to the inflammatory response in sep-
TLR expression is significantly up- luran, etc.) that augment TLR expression sis (25). Caspase-1 knock out mice are
regulated in experimental models of sep- like PAMPs. It also primes the innate im- protected from sepsis (26) while a natu-
sis and in patients with sepsis (11–14). mune system for enhanced TLR reactiv- rally occurring polymorphism for human
Trauma including thermal injury gener- ity, resulting in excess lipopolysaccharide caspase-12, a putative regulator of

292 Crit Care Med 2009 Vol. 37, No. 1


on host defense against several infections
up-regulating microbial killing mecha-
nisms such as the production of reactive
oxygen and nitrogen species (ROS and
RNS) (31, 32). Negative regulation of
TLRs and TLR-induced programmed cell
death has been defined (7). Apoptosis in-
duction is used by Pseudomonas aerugi-
nosa to inhibit the secretion of immune
and proinflammatory mediators by target
cells (33).
MyD88 protein and IL-1 receptor-
associated kinase pathways activate
NF-␬B during the innate immune re-
sponse (34). NF-␬B is a transcription fac-
tor that is constituted by homo- or het-
erodimers of the Rel protein family with a
pivotal role in inflammation, cell sur-
vival, and proliferation. In unstimulated
cells, NF-␬B is maintained in a latent
form in the cytoplasm by means of se-
questration by inhibitory ␬B (I␬B) pro-
teins. NF-␬B activating stimuli, such as
cytokines, viruses, and lipopolysaccharide
(LPS), induce the degradation of inhibi-
tory ␬Bs by the proteasome, unmasking
the nuclear localization signal of NF-␬B,
resulting in its nuclear translocation,
binding to NF-␬B motifs, and gene tran-
scription. Dynamic redox control of NF-
␬B through glutaredoxin-regulated S-
glutathionylation of inhibitory ␬Bs has
been demonstrated (35). NF-␬B, subject
to regulation by redox changes, has been
shown to be involved in the transcrip-
tional regulation of more than 150 genes
with a significant portion demonstrating
proinflammatory properties (36). NF-␬B
is readily activated upon intraperitoneally
LPS challenge within 4 hrs in lung, liver,
and spleen (37). A variant IL-1 receptor-
Figure 3. Pathogenic mechanisms during sepsis or in response to tissue injury can lead to organ associated kinase ⫺1 haplotype has re-
dysfunction. PRRs, pattern recognition receptors; TLRs, toll-like receptors; NOD-LRR, nucleotide- cently been demonstrated to affect the
oligomerization domain leucine-rich repeat protein receptors; RLHs; retinoic-acid-inducible gene I magnitude of NF-␬B activation and di-
(RIG-I)-like helicases; TNF-␣, tumor necrosis factor alpha; IL-1, interleukin 1; HMGB-1, high mobility
rectly correlates with an increased inci-
group box-1; LPS, lipopolysaccharide; LTA, lipoteichoic acid; PGN, peptidoglycan; VSMCs, vascular
dence of septic shock and significantly
smooth muscle cells; RBC, red blood cell; MPO, myeloperoxidase; XOR, xanthine oxidoreductase;
iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase; RAGE, receptor for advanced glycation reduced survival rates (38). The impact of
end products; ROCK; RhoA/Rho kinase; PARP-1, poly(ADP ribose) polymerase-1; PAR, protease NF-␬B signaling is tissue-specific as defi-
activated receptor; ROS/RNS, reactive oxygen and nitrogen species; NF, nuclear factor; NADPH, cient NF-␬B activation in intestinal epi-
nicotimanide adenosine dinucleotide phosphate; PAMPs, pathogen-associated molecular patterns; thelium is associated with increased in-
DAMPs, danger-associated molecular patterns. flammation in vivo (39, 40). Both studies
demonstrate that defects of NF-␬B signal-
ing cause immunosuppression which
caspase-1, has been linked to sepsis (27, monocyte deactivation process may be triggers and maintains inflammation.
28). Thus, caspase-1 activation appears to maladaptive in the later phases of sepsis Thus, it can be suggested that inhibition
be a prerequisite for a competent im- and predispose to secondary infection. of massive NF-␬B activation in vivo leads
mune response (29). Recently, the in- Caspase-1 and its proinflammatory cy- to reduced inflammatory responses, at
flammasome components have been tokine products are likely to contribute to least during certain phases and certain
shown to be significantly lower in septic the pathogenesis of sepsis in overwhelm- tissues (i.e., parenchymal tissues) in sep-
shock patients during the early stages of ing inflammation. However, like many sis. On the other hand, this activation is
systemic inflammatory response with el- other essential elements of innate immu- followed by negative NF-␬B regulation
evated plasma cytokines levels (30). This nity, caspase-1 also has a positive impact favoring apoptosis in immune cells which

Crit Care Med 2009 Vol. 37, No. 1 293


may lead to immunosuppression and fatal Akt in lymphocytes decreases lymphocyte box protein (FoxP3), which is induced by
outcome in severe sepsis. Recently, using apoptosis, a Th1 cytokine propensity, and the anti-inflammatory cytokine trans-
specific gene-targeted deletions, it has improve outcome in cecal ligation and forming growth factor-␤ (59, 60). TLR
been shown that deletion of MyD88 puncture-induced sepsis (51). Peptide- triggering induces dendritic cell matura-
caused a worsened survival in a model of mediated activation of Akt and extracel- tion also, which is essential for the induc-
severe peritonitis, despite the marked de- lular regulated kinase signaling protects tion of adaptive immune responses (61).
crease in sepsis-induced T and B lympho- lymphocytes from numerous apoptotic Studies have recently highlightened the
cyte apoptosis suggesting MyD88, like stimuli both in vitro and in vivo (52). importance of TLRs on Tregs (62). In this
NF-␬B, is also critical host survival in This suggests again the survival advan- regard, the dominant role of TLR2 signal-
sepsis (41). tage of PI3K/Akt pathway activation for ing on the Treg-mediated immune sup-
the later stages of sepsis. pression has been demonstrated (63, 64).
Toll-Like Receptor Signaling The Role of Rho GTPases. The Rho Tregs control inflammatory reactions
family of small GTPases is one of the to commensal bacteria and opportunist
The Role of Phosphoinositide 3-Kinase. master regulators of cell motility, as they pathogens and play a major role in sup-
Phosphoinositide 3-kinase (PI3K), a sig- control actin cytoskeleton remodeling. pressing immune reactivity, ranging
nal transduction enzyme, and the down- RhoA, Rac1, and CDc42 are the well from autoimmunity to infectious disease
stream serine/threonine kinase Akt (also known family members which act molec- (65) and to injury (66). Monneret et al
known as protein kinase B) have been ular switches regulating responses of in- (67) observed that sepsis increases
reported in cellular activation, inflamma- nate immune cells related to pathogen CD4⫹CD25⫹ T cells in the peripheral
tory responses, chemotaxis, and apoptosis sensing, intracellular uptake, and de- blood of septic patients. This was subse-
(Figs. 2 and 3) (42). PI3K can function struction (53). These GTPases play both quently found to be a relative increase in
either as a positive or negative regulator unique and overlapping roles in phago- Tregs due to a decrease in the CD4⫹CD25–
of TLR signaling. PI3Ks (three types) act cyte functions including migration, che- T effector cell populations (68). Further-
at several steps downstream of TLRs, de- motaxis, and optimal bacterial killing more, Treg-mediated induction of “alter-
pending on the cell type and/or the en- (54). It has been shown that PAMPs may natively activated” macrophages has been
gagement of a specific TLR regulating utilize GTPases through TLRs (i.e., demonstrated suggesting as one of the
downstream signaling to NF-KB transac- TLR-2, -4, -3, and -9) in which Rac1 ac- causes of immune dysfunction in sys-
tivation or to mitogen-activated phos- tivation is required for PI3K activation temic inflammatory response syndrome
phokinase activation (43, 44). As a posi- upon TLR2 stimulation (55). RhoA regu- and sepsis (69, 70). Targeting apoptosis of
tive mediator of TLR signaling, PI3K lates not only cytoskeletal events, which Tregs may be a new therapeutic approach
together with p38 and extracellular reg- mediate neutrophil migration, but also in preventing the continuum of sepsis to
ulated kinase (ERK)1/2 mitogen-acti- contributes to NF-␬␤-dependent proin- severe sepsis. However, it has been re-
vated phosphokinases, lead to production flammatory gene transcription (Fig. 3) ported that depletion of CD25⫹ cells be-
of proinflammatory cytokines IL-1␤, (56). It has been suggested that ROCK fore inducing sepsis did not alter septic
IL-6, and IL-8 upon microbial challenge inhibition could attenuate cytoskeletal mortality pointing the need of more stud-
(44, 45). In detail, PI3K activation ap- rearrangement of endothelial cells, lead- ies to clarify the significance of this cell
peared to have a significantly promoting ing to decreased neutrophil emigration population’s expansion in sepsis morbid-
function for these mediators in mono- into the lung parenchyma in LPS- ity (71, 72).
cytes, whereas activation appeared to induced lung injury (57). An important Adoptive transfer of Tregs before or
limit the LPS response for generation of role for rho kinase in leukocyte recruit- following the initiation of polymicrobial
these cytokines in neutrophils (46). How- ment is also supported in an endotoxemic sepsis improved survival by enhancing
ever, PI3K inhibition resulted in im- liver injury model (58). The recently tumor necrosis factor (TNF)-␣ produc-
paired oxidative burst and phagocytosis emerged connections between TLR sig- tion and bacterial clearance (73). Tregs
activity in both neutrophils and mono- naling and small Rho-GTPases seem to inhibit LPS-induced monocyte survival
cytes. By limiting C5a-mediated effects provide new therapeutic avenue of re- through the Fas/FasL dependent pro-
on neutrophil cytokine generation, and search in sepsis. apoptotic mechanism, which might play
promoting oxidative burst and phagocy- a role in the resolution of exaggerated
tosis, PI3K activation seems to be a ther- Toll-Like Receptor Signaling inflammation (74). The positive and neg-
apeutic approach for limiting inflamma- and Regulatory T Cells (Tregs) ative effects of TLR on Tregs are curious
tion in sepsis. in sepsis, and the dynamics of TLR ex-
On the other hand, PI3K/Akt signaling To balance self-tolerance and immu- pression on immune-suppressive Tregs
pathway acts as an endogenous negative nity against pathogens, the immune sys- upon inflammation or in relation to type
feedback mechanism that serves to limit tem depends on both up-regulatory and of pathogen are needed to illuminate.
proinflammatory and apoptotic events as down-regulatory mechanisms. Recent
seen in monocytes in response to endo- studies have suggested that several lym- Neutrophils and
toxin (47). The suppressive role in in- phocyte subpopulations (i.e., CD4⫹CD25⫹ Monocyte/Macrophages in
flammation and coagulation was con- Foxp3⫹ T regulatory-cell 关Tregs兴) may Inflammation
firmed in mouse model of endotoxemia have the capacity to actively suppress an
(48). It can also promote the generation adaptive immune response and may po- Myeloid cells including neutrophils
of anti-inflammatory cytokine IL-10 (49). tentially be involved in septic immune and elements of monocyte/macrophage
PI3K/Akt has an effect in balancing Th1 dysfunction. Naturally occurring Tregs lineage are heavily armed with large
vs. Th2 responses (50). Overexpression of express the transcription factor forkhead stores of proteolytic enzymes and with

294 Crit Care Med 2009 Vol. 37, No. 1


the capacity to rapidly generate ROS and and cellular exudates that further impede liver (86). Apoptosis is a counter-regula-
RNS to degrade internalized pathogens. adequate tissue oxygenation. Neutrophils tor of the initial inflammatory response
The highly proapoptotic nature of neu- participate in these rheologic changes in sepsis (90). Neutrophils are constitu-
trophils is designed to maintain a balance through their augmented binding to blood tively proapoptotic and apoptosis is fun-
between antimicrobial effectiveness and vessel walls and through the formation of damental for the resolution of inflamma-
the potential for neutrophil-associated platelet-leukocyte aggregates (83). Neutro- tion and cell turnover. Neutrophils can
damage to the host in septic challenge or phil elastase, other proteases, glycases and undergo apoptosis via intrinsic and ex-
in other injurious processes, such as inflammatory cytokines degrade endoge- trinsic pathways; the latter also requires
trauma or ischemia/reperfusion (75). nous anticoagulant activity, and impair fi- mitochondrial amplification (Fig. 4) (91).
Host tissue damage in severe sepsis may brinolysis on endothelial surfaces favoring The role played by mitochondria in the
arise via a variety of mechanisms includ- a procoagulant state (1). regulation of neutrophil life span is more
ing premature neutrophil activation dur- The Role of Apoptosis. Sepsis-induced crucial than in other cell types in the
ing migration, extracellular release of cy- neutrophil-mediated tissue injury has body (92). As neutrophils kill pathogens
totoxic molecules and toxins during been demonstrated in a variety of organs using ROS and RNS and a mixture of lytic
microbial killing, removal of infected or including the lungs (84 – 86), diaphragm enzymes, delayed clearance of neutro-
damaged host cells or debris during host (87), kidneys (86), intestine (88, 89), and phils in sepsis can potentially contribute
tissue remodeling, and failure to termi-
nate acute inflammatory responses (76).
Therefore, to maximize host defense ca-
pabilities while minimizing damage to
host tissues, neutrophil microbial re-
sponses are tightly regulated. On the
other hand, quorum sensing (the ability
of bacteria to assess their population den-
sity) has been found to have a crucial role
in regulating tissue invasion by bacterial
pathogens, and inhibitors of quorum
sensing system provide new avenues for
intervention against invasive pathogens
(1). Evidence now exists that quorum
sensing system can even open up bidirec-
tional lines of communication between
bacteria and the human host.
Leukocyte-endothelial interactions,
which may also contribute to inflamma-
tion-mediated injury, involve two sets of
adhesion molecules, selectins, and inte- Figure 4. Summary of apoptotic signaling pathways as seen through activation of death receptor
grins (77, 78). P-selectin is expressed on (extrinsic) or mitochondrial (intrinsic) pathway. Extrinsic signals bind to their receptors and trigger
platelets; E- and P-selectins are expressed intracellular signaling, leading to caspase-8 activation. Activation of caspase-8 by extrinsic stimuli
by endothelial cells, whereas L-selectin is (such as tumor necrosis factor-␣ [TNF-␣], Fas ligand) involves mitochondria-dependent signaling in
expressed on leukocytes. Selectins mediate type II cells. In type I cells, on the other hand, execution of apoptosis occurs without significant
participation of mitochondria. MAPK, mitogen-activated phosphokinase; PI3K/Akt, phosphoinositide
neutrophil rolling along activated endothe-
3-kinase/Akt; APAF-1, apoptosis protease activating factor 1; ER, endoplasmic reticulum; IL, interleukin.
lial surfaces as circulating neutrophils de-
celerate to engage endothelial receptors.
The beta-2 intergrins (CD11/CD18 com-
plexes) mediate tight adhesion to endothe-
lial membranes, allowing subsequent
egress of neutrophils to extravascular sites
of inflammation (78). Activated neutrophils
stimulate transendothelial albumin trans-
port through intracellular adhesion mole-
cule-1 mediated, Scr-dependent caveolin
phosphorylation. The role of caveolin in
inflammatory response in sepsis has been
recently defined (Fig. 3) (79 – 82).
Neutrophils contribute to blood coagu-
lation in localized inflammation and in
generalized sepsis (1, 3, 75). During sys-
temic inflammation, homeostatic mecha- Figure 5. The recognition of apoptotic cells by macrophages is largely dependent on the cell surface
nisms are compromised in the microcircu- appearance of phosphatidylserine (PS). S-nitrosylation of critical cysteine residues inhibits aminophospho-
lation including endothelial hyperactivity, lipid translocase (APLT), leading to PS externalization. It generates an “eat me” signal during apoptosis.
fibrin deposition, microvascular occlusion, iNOS, inducible nitric oxide synthase; .NO, nitric oxide; O2, superoxide; ONOOⴚ, peroxynitrite.

Crit Care Med 2009 Vol. 37, No. 1 295


to cell/organ injury. Importantly, the
phagocytosis of bacteria and fungi accel-
erates neutrophil apoptosis. Apoptotic
cell clearance induces anti-inflammatory
effects in tissues. It has been shown that
intratracheal administration of killed E.
coli attenuated lung injury and improved
survival in an intestinal ischemia/reper-
fusion injury associated with marked pul-
monary neutrophil infiltration (93).
Cytokine-induced prolonged neutro-
phil survival is accompanied by evidence
of increased neutrophil activation, in-
cluding augmented respiratory burst ac-
tivity (90). Neutrophils from patients
with sepsis manifest markedly prolonged
survival in vitro in association with evi-
dence of cellular activation (94). Phago- Figure 6. Proinflammatory/anti-inflammatory cytokines and reactive oxygen and nitrogen species have
cytosis of apoptotic neutrophils by important effects within the microcirculatory unit: the arteriole, endothelial cell, capillary bed, and the
macrophages inhibits the release of venule. The arteriole is where the characteristic intractable vasodilation of sepsis occurs. The capillary bed
is where the effects of endothelial cell activation/dysfunction are most pronounced and microvascular
proinflammatory cytokines and promotes
thromboses are formed. The postcapillary venule is where leukocyte trafficking is most disordered. All of
the secretion of anti-inflammatory cyto- these causes impair flow through the microcirculation leading to microcirculatory dysfunction. TLR,
kines (95). In contrast, inefficient apopto- toll-like receptor; NOD-LRR, nucleotide-oligomerization domain leucine-rich repeat; RLH, (RIG-I)-like
tic cell clearance is proinflammatory and helicase; VSM, vascular smooth muscle; IL, interleukin; TGF, transforming growth factor; MIF, migration
immunogenic (96). The recognition of inhibitory factor; TNF, tumor necrosis factor; HMGB, high mobility group box; PAF, protease activating
apoptotic cells by macrophages is largely factor; sTNFR, soluble tumor necrosis factor receptor.
dependent on the cell surface appearance
of an anionic phospholipid, phosphatidyl-
serine (PS), which is normally confined second messengers for hormones and re- contributors of ROS production, has been
to the inner leaflet of the plasma mem- dox regulation. Despite their importance reported in adult and pediatric patients
brane (97). Asymmetric distribution of as a defense mechanism against invading with sepsis (104).
PS across the plasma membrane is pathogens, an overwhelming production O2ⴚ in the presence of nitric oxide,
mainly because of the activity of a spe- of ROS and RNS or a deficit in oxidant generates peroxynitrite (ONOO⫺), a key
cialized enzymatic mechanism, amin- scavenger and antioxidant defenses result player in the pathogenesis of sepsis-
ophospholipid translocase. S-nitrosyla- in oxidative/nitrosative stress, a key ele- induced organ dysfunction. ONOO⫺ can
tion of critical cysteine residues inhibits ment in the deleterious processes in sep- cause DNA strand breakage, which trig-
aminophospholipid translocase, leading sis (Fig. 6) (101, 102). gers the activation of DNA repair en-
to PS externalization. PS expression dur- Stimulated neutrophils produce ROS zymes such as poly (adenine dinucleotide
ing apoptosis generates an “eat-me” sig- and RNS through the nicotinamide ade- phosphate-ribose) polymerase (Fig. 3).
nal (Fig. 5), which in turn triggers clear- nine dinucleotide phosphate oxidase Poly (adenine dinucleotide phosphate-
ance of apoptotic cells and suppresses the complex, myeloperoxidase and xanthine ribose) polymerase inhibitors protect
inflammatory response (98). It has been oxidoreductase and represent a defense against oxidative and nitrosative stress-
demonstrated that S-nitrosylation of crit- mechanism against invading microor- induced organ dysfunction in endotox-
ical cysteine residues in aminophospho- ganisms (103). Lipopolysaccharide and emia (87– 89). Recently, the potential role
lipid translocase using a cell-permeable other proinflammatory mediators acti- of poly (adenine dinucleotide phosphate-
transnitrosylating agent, S-nitroso-acetyl- vate nicotinamide adenine dinucleotide ribose) polymerase activation has been
cysteine, resulted in egression of PS to phosphate oxidase to produce superoxide implicated in the pathogenesis of myo-
the outer surface of the plasma mem- radical (O2⫺). In aqueous environments, cardial contractile dysfunction associated
brane, rendering these cells recognizable superoxide radical is rapidly catalyzed by with human septic shock (105).
by macrophages (99). The therapeutic po- superoxide dismutase hydrogen peroxide
tential of regulating neutrophil life-span (H2O2) and hydroxyl radicals. Myeloper- Novel Cytokines in Inflammation
in sepsis remains to be determined. Care oxidase from neutrophil azurophilic
must be exercised in regulating of this granules produces hypochlorous acid High Mobility Group Box-1 and Re-
pathway, as sepsis enhances the capacity from hydrogen peroxide (H2O2) and chlo- ceptor for Advanced Glycation End-
of macrophages to clear expanded apo- ride anion (Clⴚ) during respiratory burst. Products. High mobility group box-1
ptotic populations, a mechanism contrib- These radicals are highly cytotoxic, and (HMGB-1) is a nonhistone, nuclear DNA-
uting to septic immune suppression neutrophils used them to kill bacteria binding protein involved in nucleosome
(100). and other pathogens. Expression of hu- stabilization and gene transcription. How-
The Role of ROS and RNS. ROS and man xanthine oxidoreductase is markedly ever, when HMGB-1 is released in large
RNS exert several beneficial physiologic up-regulated by hypoxia, ischemia/reper- quantities into the extracellular environ-
functions, such as intracellular signaling fusion, LPS, and TNF-␣. Increased activ- ment, it becomes a lethal mediator of sys-
for several cytokines and growth factors, ity of xanthine oxide, one the important temic inflammation (Fig. 3) (106). Indeed,

296 Crit Care Med 2009 Vol. 37, No. 1


it has been recently shown that it has a for this protein in the extracellular fluid. tion-induced apoptosis (135). High MIF
weak proinflammatory activity by itself and HMGB-1 might serve as a shuttle platform levels delay the removal of activated mono-
binding to bacterial substances including for LPS and other microbial mediators for cytes/macrophages by apoptosis. This pro-
lipid molecules such as phophatidylserine docking to CD14 and recognition by the longs monocyte/macrophage survival, in-
strengthens its effects (107). Interestingly, TLRs (108, 123) or through binding to creases cytokine production, and sustains
phosphatidylserine has been implicated in host-derived proinflammatory mediators, an ongoing proinflammatory response.
the regulation of inflammation and such as IL-1beta (124).
HMGB-1 might thus regulate its anti- Elevated levels of HMGB-1 are measur- Mitochondrial Dysfunction in
inflammatory activities (108). able in the majority of patients up to 1 wk Inflammation
HMGB-1 is released into the extracel- after the diagnosis of sepsis or septic shock
lular space through acetylation or phos- and are correlated with the degree of organ Although microvascular flow abnormal-
phorylation (109, 110). HMGB-1 is either dysfunction (125, 126). However, serum ities occur, findings of decreased oxygen
“passively released” from necrotic cells, HMGB-1 levels do not consistently identify consumption (136) and elevated tissue ox-
and a mechanism that represents a pro- nonsurvivors from survivors as a predictor ygen tension (137), yet minimal cell death
cess adopted by the innate immune sys- of hospital mortality (127). As HMGB-1 is despite functional and biochemical de-
tem to recognize damaged and necrotic late inflammatory cytokine of sepsis, it pro- rangements (138), suggest that the prob-
cells, or “actively secreted” by immune vides a wide therapeutic time window for lem lies more in cellular oxygen utilization
cells including macrophages and neutro- clinical intervention and remains an attrac- rather than a problem with oxygen delivery
phils to trigger inflammation (106). Re- tive target for sepsis treatment. (139). It is postulated that prolonged and
cently, HMGB-1 release from macro- RAGE, a member of the immunoglobu- systemic inflammatory insult is accompa-
phages has been shown during the course lin superfamily, is a pattern-recognition re- nied by a basic tissue survival response me-
of apoptosis as well as necrosis and de- ceptor that binds diverse classes of endog- diated by switching off its energy-consum-
fined as a downstream event of cell apo- enous molecules including HMGB-1. It has ing biophysiological processes. Recent
ptosis during severe sepsis (111, 112). been defined as part of a newly appreciated evidence suggests that sepsis and septic
After treatment with LPS or various cy- component of the innate immune system shock severely impair the mitochondria
tokines such as TNF-␣, IL-1␤, or IFN-␥, referred to as the danger associated molec- (140, 141), and the severity and outcome of
HMGB-1 is released from activated mac- ular pattern system (Fig. 3) (128). Mem- organ dysfunction could be related to mi-
rophages within 4 hrs and reaches a pla- brane bound and soluble forms of RAGE tochondrial dysfunction (142). Depleted
teau around 18 –24 hrs. It binds to several (sRAGE) have been detected in plasma. Sol- levels of reduced glutathione, an important
transmembrane receptors such as recep- uble RAGE, anti-RAGE antibody (Fab 2 intramitochondrial antioxidant, in combi-
tor for advanced glycation end products fragment) or data from RAGE⫺/⫺ animals nation with excess generation of ROS and
(RAGE), TLR-2, and -4, activating NF-␬B have been shown to decrease inflammation, RNS severely inhibit oxidative phosphory-
and extracellular regulated kinase 1/2 reduce neutrophil extravasation, and re- lation and ATP generation (142). This ac-
(113, 114). duce migration (129). Recently, the dem- quired intrinsic derangement in cellular
Although HMGB-1 was originally de- onstration of survival benefit after delayed energy metabolism which has also been
scribed as a late mediator of endotoxin- administration of anti-RAGE antibody in a termed “cytopathic hypoxia,” contributes to
induced lethality (115), recent studies in- murine model of polymicrobial sepsis, even reduced activities of mitochondrial electron
dicate a role for HMGB-1 in angiogenesis, when delayed up to 24 hrs, provides a ther- transport chain enzyme complexes and im-
tissue repair, and regeneration (116). apeutic rationale for the use of anti-RAGE paired ATP biosynthesis, potentially organ
HMGB-1 appears to be a novel myocardial mAb as a salvage therapy for established dysfunction in sepsis (141, 143, 144).
depressant factor upon release by resi- severe sepsis (130). Sepsis-related derangements in mito-
dent myocardial cells following tissue in- Macrophage Migration Inhibitory chondrial function can activate the ubiq-
jury. HMGB-1 might decrease energy uti- Factor. Migration inhibitory factor (MIF) uitin proteolytic pathway in skeletal mus-
lization in ischemic tissue, thereby acts as a stress response mediator and cle of septic patients (145). Mitochondrial
preventing injured myocytes from wors- proinflammatory cytokine upon induc- permeability transition pore seems to be
ening ATP depletion that eventuate in tion by glucocorticoids (131). This pro- involved in sepsis-induced mitochondrial
necrosis (117). On other hand, excessive tein is readily measurable in patients with damage, since its inhibition significantly
release of HMGB-1 in sepsis might con- sepsis, and MIF probably contributes to improved organ function and reduced
tribute to sustained inflammation and to the pathogenesis of sepsis. Inhibition of mortality in rodents (146). Whether sep-
profound myocardial depression. MIF or its targeted deletion attenuates sis-related reduction in energy supply
The cholinergic anti-inflammatory TNF-␣ and IL-1␤ expression and protects could result in a state of cellular shut-
pathway is a neural mechanism that in- mice from lethality is experimental sepsis down analogous to myocardial “stun-
hibits the expression of HMGB-1 and (132, 133). Systemic challenge of animals ning” (or hibernation) following coronary
other cytokines (118 –121). Signals trans- with MIF increases LPS-related lethality occlusion, allowing for eventual restora-
mitted via the vagus nerve, the principal (133). MIF promotes the expression of tion of organ function and survival, has
nerve of the parasympathetic nervous TLR4 on macrophages, and thereby sen- not yet been determined.
system, significantly attenuate the re- sitizes these immune effector cells to LPS
lease of HMGB-1 and other cytokines in (134). Impact of Inflammation on
inflammation in animal and human stud- The immunoregulatory effects of MIF Coagulation
ies (122). might be crucial to the control and resolu-
The remarkable binding characteristics tion of the inflammatory response as a con- The clotting system is almost invari-
of HMGB-1 suggest another important role sequence of its ability to regulate activa- ably activated by systemic microbial in-

Crit Care Med 2009 Vol. 37, No. 1 297


vasion (147). Clotting is one most prom-
inent features of sepsis. Coagulation
contributes significantly to the outcome
in sepsis with concurrent down-regula-
tion of anticoagulant systems and fibri-
nolysis (Fig. 7). Inflammation-induced
coagulation in turn contributes to fur-
ther inflammation (148). Indeed, collab-
oration between clotting and inflamma-
tion accounts for the basic survival
strategy of walling off the damaged and
infected tissues from the rest of the host
(149). The key determinant of survival in
sepsis is to limit excess systemic inflam-
matory and coagulopathic damage while
retaining the benefits of controlled anti-
microbial clearance and localized clot
formation (147, 150).
The inflammatory reaction to tissue
injury activates the clotting system, and
coagulation promotes inflammation (3,
151, 152). The role of procoagulant apo- Figure 7. The extrinsic pathway (tissue factor pathway) is the primary mechanism by which thrombin is
ptotic microparticles have also been dem- generated in sepsis. The intrinsic cascade (contact factor pathway) primarily serves an accessory role in
onstrated in sepsis (153–155). Linkage of amplifying the prothrombotic events that are initiated in sepsis. Thrombin, factor Xa and the TF-factor VIIa
coagulation enzymes with their serine complex interact with the PAR (protease activated receptors) system and directly activate endothelial cells,
protease activity with protease activated platelets and white blood cells, and induce a proinflammatory response. Platelet-derived microparticles
receptors (PAR 1-4) on endothelial sur- (MPs) express functional adhesion receptors including P-selectin on their surface, attach to the site of
injury on the vessel wall, and support the rolling of leukocytes in the presence of shear stress or severe
faces increases P-selectin, cytokine produc-
insult. When bearing appropriate counter-ligands, MPs can transfer their procoagulant potential to target
tion, and adhesion molecule expression, cells. Platelet-derived MPs can bind to soluble and immobilized fibrinogen, thus delivering procoagulant
leading to microcirculatory dysfunction in entities to the thrombus via the formation of aggregates. In vitro, interaction between endothelial MPs and
severe sepsis (156, 157). monocytes promotes TF mRNA expression and TF-dependent procoagulant activity. Activated platelets and
Activated Protein C. Activated protein platelet-derived MPs thus amplify leukocyte-mediated tissue injury in thrombotic and inflammatory
C (APC) is derived from its zymogen pro- disorders. EPCR, endothelial protein C receptor; PMN, polymorphonuclear leukocyte; APC, activated
tein C in contact with thrombin: thrombo- protein C; ADAMST13, a disintegrin and metalloproteinase with a thrombospondin type 1 motifs 13;
modulin complexes on endothelial sur- ULVWF, unusually large von Willebrand factor; ICAM, intracellular adhesion molecule; VCAM, vascular cell
faces. Protein C was originally thought to adhesion molecule-1; IL, interleukin; TNF, tumor necrosis factor; IFN, interferon.
be synthesized exclusively by the liver
(158). It has recently shown that it is
strongly expressed by the endothelium and
keratinocytes (159). The conversion to APC
is augmented by endothelial PC receptor
(EPCR) which is present on endothelial
cells, neutrophils, monocytes, and keratin-
ocytes (160, 161) whereas soluble EPCR
inhibits APC anticoagulant activity (162).
Soluble EPCR is released constitutively and
levels increase in patients with Gram-
negative sepsis (163). Levels of APC, protein
C, and its cofactor protein S are depleted in
sepsis (164, 165). Furthermore, peripheral
conversion of Protein C by the thrombin:
thrombomodulin complex is impaired in
sepsis, further contributing to microvascu-
lar thrombosis and vascular leakage (166).
APC is a central endogenous anticoag-
ulant protein with antithrombotic, anti-
inflammatory, antiapoptotic, and profi- Figure 8. The homeostatic balance between thrombin and activated protein C (APC) in coagulation and
brinolytic activities (Fig. 8) (167, 168). inflammation is given. APC is a central endogenous anticoagulant protein with antithrombotic, anti-
Although an anti-inflammatory role for inflammatory, anti-apoptotic and pro-fibrinolytic activities. The ligand occupancy of endothelial protein C
APC may be an indirect consequence of receptor (EPCR) switches the protease-activated receptor 1 (PAR-1)-dependent signaling specificity of thrombin
its ability to reduce thrombin generation, from a permeability-enhancing to a barrier-protective response. Other pleiotropic effects are also explained. TAFI,
APC also has direct anti-inflammatory thrombin activatable fibrinolysis inhibitor; NF-␬␤, nuclear factor ␬␤.

298 Crit Care Med 2009 Vol. 37, No. 1


Table 1. Ongoing and upcoming clinical studies with molecular targets

Target Sponsor or Institution Phase Comments

CytoFab (AZD9773) Fabs of IgG that bind to TNF-␣ AstraZeneca II Recruiting


Recombinant human lactoferrin LPS-neutralization Agennix II Recruiting
Lipid emulsion (GR-270773) TLR4 Glaxo Smith Kline II Suspended
E5564 TLR4 Eiasi III Ongoing
TAK-242 TLR4 Takeda III Recruiting
Recombinant human soluble thrombomodulin Coagulation Artisan Pharma II Recruiting
Recombinant antithrombin Coagulation Leo Pharma II Recruiting
Recombinant tissue factor pathway inhibitor Coagulation Novartis III Completed
Recombinant activated Protein C⫹ Erythropoietin Coagulation and inflammation LHRI III Recruiting
Recombinant activated Protein C Coagulation Lilly III Recruiting
Recombinant plasma gelsolin Replacement of circulating CBC II Recruiting
actin-binding protein
L-Citrulline supplementation NO pathway MU II Not yet open for
participant
recruitment
Inhaled nitric oxide NO pathway NIMS III Not yet open for
participant
recruitment
Simvastatin Pleitrophic effects MUV, Austria and IV Recruiting
Chicago, USA
Rosuvastatin Pleitrophic effects UASLP, Mexico and Beth II Ongoing
Israel
Atorvastatin Pleitrophic effects HCPA, Brazil II Recruiting

TNF, tumor necrosis factor; LPS, lipopolysaccharide; NO, nitric oxide; TLR, toll-like receptor, LHRI, Lawson Human Resources Institute; CBC, Critical
Biologics Corporation; MU, Maastricht University; NIMS; National Institute of Medical Sciences; MUV, Medical University of Vienna; UASLP, Universidad
Autonoma de San Luis Potosi; HCPA, Hospital de Clinical de Porto Alegre.

properties (169). APC can cleave and ac- of rhAPC regarding the bleeding compli- ADAMTS-13 levels are associated with dif-
tivate PAR1-dependent cellular pathways cation (175). A nonanticoagulant from of ferences in morbidity, mortality, and
(170). APC competes for PAR-1 binding APC reduces mortality in experimental variables of inflammation and endothelial
with thrombin, but in vitro studies sug- models of endotoxemia and sepsis (176). dysregulation in severe sepsis patients
gest that APC is 103- to 104-fold less po- von Willebrand Factor, A Disintegrin-Like (182, 187).
tent that thrombin in cleaving PAR-1. and Metalloproteinase with Thrombospondin The Ashwell receptor, which is the
The critical receptors required for both Type-1 Motifs 13 and Ashwell Receptor. major lectin of hepatocytes, modulates
PC activation and APC cellular signaling The discovery of a disintegrin-like and met- VWF homeostasis. It has been recently
(i.e., thrombomodulin, EPCR and PAR-1) alloproteinase with thrombospondin type-1 demonstrated that the marked thrombo-
are co-localized in lipid rafts on endothe- motifs 13 (ADAMTS-13) has provided new cytopenia associated with S. pneumoniae
lial cells (171). EPCR is associated with insights in pathogenesis of thrombosis in sepsis is the result of Ashwell receptor-
caveolin-1 on lipid rafts and EPCR bind- sepsis. ADAMTS-13, the principal physio- dependent clearance of platelets (188).
ing to the gamma-carboxyglutamic acid logic modulator of von Willebrand factor The ensuing reduction in platelet counts
domain of protein C/APC leads to its dis- (VWF) is produced mainly stellate cells in at the onset of sepsis protects the host
sociation from caveolin-1 (Fig. 7). APC the liver (177). VWF is synthesized in vas- against the development of disseminated
then engages PAR-1 generating a protec- cular endothelial cells and released into the intravascular coagulation. Homeostatic
tive signaling pathway through coupling plasma as unusually large VWF multimers adaptation by this receptor moderates the
of PAR-1 to the pertussis toxin-sensitive which are rapidly degraded into smaller onset and severity of disseminated intra-
G(i)-protein. Thus, when EPCR is bound VWF multimers by ADAMTS-13. Deficiency vascular coagulation during sepsis sug-
by protein C, the PAR-1-dependent pro- of the ADAMTS-13, as observed in most gesting the improvement in host survival
tective signaling responses in endothelial forms of thrombotic thrombocytopenic probability (189). At present, it remains
cells can be mediated by either thrombin purpura, increases the level of unusually unclear whether blocking the Ashwell re-
or APC. These results explain how PAR-1 large VWF multimers in plasma and leads ceptor may have a beneficial effect in
and EPCR participate in protective sig- to platelet aggregation and/or thrombus severe sepsis.
naling events in endothelial cells (172). formation, especially in small arterioles, re-
Therapeutic administration of recom- sulting in microvascular failure (178 –180). CONCLUSION
binant human APC (rhAPC) is currently Recently, inflammation associated-
in use as a treatment strategy for severe ADAMTS-13 deficiency has been de- A unifying concept of innate immu-
sepsis patients with a high risk of death scribed in patients with systemic inflam- nity is based upon pathogen-, or damage-
(173, 174). Genetically engineered vari- matory response syndrome and severe associated molecular pattern molecules
ants of APC have been designed with sepsis (Fig. 7) (181–185). Decreased lev- and downstream signaling pathways. This
greater antiapoptotic activity and reduced els of ADAMTS-13 have been reported in has facilitated significant advances in our
anticoagulant activity relative to wild- healthy volunteers following endotoxin understanding of the pathophysiology of
type APC to increase the risk/benefit ratio infusion (186). Furthermore, reduced sepsis and led to a multitude of clinical

Crit Care Med 2009 Vol. 37, No. 1 299


trials (Table 1). However, further identi- sion during human and experimental sep- 30. Fahy RJ, Exline MC, Gavrilin MA, et al:
fication of the critical elements in severe sis. Shock 2005; 23:39 – 44 Inflammasome mRNA expression in human
sepsis that drives the transition from lo- 14. Williams DL, Ha T, Li C, et al: Modulation of monocytes during early septic shock. Am J
calized inflammation to deleterious host tissue toll-like receptor 2 and 4 during the Resp Crit Care Med 2008; 177:983–988.
early phases of polymicrobial sepsis corre- 31. Lara-Tejero M, Sutterwala FS, Ogura Y, et
response remains incompletely illumi-
lates with mortality. Crit Care Med 2003; al: Role of the caspase-1 inflammasome in
nated. The limitations in our current 31:1808 –1818 Salmonella typhimurium pathogenesis.
knowledge of the molecular mechanisms 15. Paterson HM, Murphy TJ, Purcell EJ, et al: J Exp Med 2006; 203:1407–1412
in sepsis have made the design of inter- Injury primes the innate immune system 32. Brightbill HD, Libraty DH, Krutzik SR, et
vention trials in clinical sepsis challeng- for enhanced toll-like receptor reactivity. al: Host defense mechanisms triggered by
ing (190). New findings as to biomarkers J Immunol 2003; 171:1473–1483 microbial lipoproteins through toll-like re-
and measures to detect genetic signa- 16. Tsujimoto H, Ono S, Efron PA, et al: Role of ceptors. Science 1999; 285:732–736
tures of sepsis are now making their way toll like receptors in the development of 33. Bruno TF, Woods DE, Mody CH: Exoen-
into clinical trial designs. It is anticipated sepsis. Shock 2008; 29:315–321 zyme S from Pseudomonas aeruginosa in-
that such innovations will improve the 17. Ogura Y, Sutterwala FS, Flavell RA: The duces apoptosis in T lymphocytes. J Leukoc
inflammasome: First line of the immune Biol 2000; 67:808 – 816
outlook for successful development of
response to cell stress. Cell 2006; 126: 34. Wesche H, Henzel WJ, Shillinglaw W, et al:
new sepsis treatments tailored to individ- 659 – 662
ual patient needs. MyD88: An adapter that recruits IRAK to
18. Drenth JP, van der Meer JW: The inflamma- the IL-1 receptor complex. Immunity 1997;
some—A linebacker of innate defense. 7:837– 847
REFERENCES N Engl J Med 2006; 355:730 –732 35. Reynaert NL, van der Vliet A, Guala AS, et
19. Dinarello CA: Interleukin-1␤, interleukin- al: Dynamic redox control of NF-kappaB
1. van der Poll T, Opal SM: Host-pathogen 18, and the interleukin-1␤ converting en- through glutaredoxin-regulated S-gluta-
interactions in sepsis. Lancet Infect Dis zyme. Ann NY Acad Sci 1998; 856:1–11 thionylation of inhibitory kappaB kinase
2008; 8:32– 43 20. Martinon F, Burns K, Tschopp J: The in- beta. Proc Natl Acad Sci USA 2006; 103:
2. Cinel I, Dellinger RP: Making a difference in flammasome. A molecular platform trigger- 13086 –13091
outcome in severe sepsis: The organism, the ing activation of inflammatory caspases and 36. Pahl HL: Activators and target genes of rel/
host response and the treating clinician. processing of proIL-1␤. MolCell 2002; 10: NF-kappaB transcription factors. Oncogene
Emerg Med Crit Care Rev 2007; 1:1–3 417– 426
1999; 18:6853– 6866
3. Cinel I, Dellinger RP: Advances in patho- 21. Yu JW, Wu J, Zhang Z, et al: Cryopyrin and
37. Blackwell TS, Yull FE, Chen CL, et al: Mul-
genesis and management of sepsis. Curr pyrin activate caspase-1, but not NF-
tiorgan nuclear factor kappa B activation in
Opin Infect Dis 2007; 20:345–352 kappaB, via ASC oligomerization. Cell
a transgenic mouse model of systemic in-
4. Uematsu S, Akira S: Toll-like receptors and Death Differ 2005; 13:236 –249
flammation. Am J Respir Crit Care Med
innate immunity. J Mol Med 2007; 84: 22. Seshadri S, Duncan MD, Hart JM, et al:
2000; 162:1095–1101
712–725 Pyrin levels in human monocytes and
38. Arcaroli J, Silva E, Maloney JP, et al: Variant
5. Creagh EM, O’Neill LA: TLRs, NLRs and monocyte-derived macrophages regulate
IRAK-1 haplotype is associated with in-
RLRs: A trinity of pathogen sensors that IL-1␤ processing and release. J Immunol
creased nuclear factor-kappaB activation
co-operate in innate immunity. Trends 2007; 179:1274 –1281
and worse outcomes in sepsis. Am J Respir
Immunol 2006; 27:352–357 23. Tschopp J, Martinon F, Burns K: NALPs: A
Crit Care Med 2006; 173:1335–1341
6. Granucci F, Foti M, Ricciardi-Castagnoli P: novel protein family involved in inflamma-
39. Zaph C, Troy AE, Taylor BC, et al: Epithelial
Dendritic cell biology. Adv Immunol 2005; tion. NatRevMol Cell Biol 2003; 4:95–104
cell-intrinsic IKK-beta expression regulates
88:193–233 24. Sarkar A, Duncan M, Hart J, et al: ASC
directs NF-␬B activation by regulating re- intestinal immune homeostasis. Nature
7. Liew FY, Xu D, Brint EK, et al: Negative
ceptor interacting protein-2 (RIP2) 2007; 446:552–556
regulation of toll-like receptor-mediated
caspase-1 interactions. J Immunol 2006; 40. Liu YJ, Soumelis V, Watanabe N, et al:
immune responses. Nat Rev Immunol 2005;
176:4979 – 4986 TSLP: An epithelial cell cytokine that regu-
5:446 – 458
8. Akira S, Uematsu S, Takeuchi O: Pathogen 25. Scott AM, Saleh M: The inflammatory lates T cell differentiation by conditioning
recognition and innate immunity. Cell caspases: Guardians against infections and dendritic cell maturation. Annu Rev Immu-
2006; 124:783– 801 sepsis. Cell Death Differ 2007; 14:23–31 nol 2007; 25:193–219
9. Bianchi ME: DAMPs PAM. Ps and alarmins: 26. Sarkar A, Hall MW, Exline M, et al: 41. Peck-Palmer OM, Unsinger J, Chang KC, et
All we need to know about danger. J Leukoc Caspase-1 regulates E. coli sepsis and al: Deletion of MyD88 markedly attenuates
Biol 2007; 81:1–5 splenic B cell apoptosis independently of sepsis-induced T and B lymphocyte apopto-
10. Mollen KP, Anand RJ, Tsung A, et al: IL-1␤ and IL-18. Am J Respir Crit Care Med sis but worsens survival. J Leukoc Biol
Emerging paradigm: Toll-like receptor 2006; 174:1003–1010 2008; 83:1009 –1018
4-sentinel for the detection of tissue dam- 27. Saleh M, Vaillancourt JP, Graham RK, et al: 42. Cantley LC. The phosphoinositide 3-kinase
age. Shock 2006; 26:430 – 437 Differential modulation of endotoxin re- pathway. Science 2002; 296:1655–1657
11. Tsujimoto H, Ono S, Hiraki S, et al: He- sponsiveness by human caspase-12 poly- 43. Okugawa S, Ota Y, Kitazawa T, et al: Janus
moperfusion with polymyxin B-immobi- morphisms. Nature 2004; 429:75–79 kinase2 is involved in lipopolysaccharide-
lized fibers reduced the number of CD16⫹ 28. Saleh M, Mathison JC, Wolinski MK, et al: induced activation of macrophages. Am J
CD14⫹ monocytes in patients with septic Enhanced bacterial clearance and sepsis re- Physiol Cell Physiol 2003; 285:C399 –C408
shock. J Endotoxin Res 2004; 10:229 –237 sistance in caspase-12-deficient mice. Na- 44. Ojaniemi M, Glumoff V, Harju K, et al:
12. Armstrong L, Medford AR, Hunter KJ, et al: ture 2006; 440:1064 –1068 Phosphatidylinositol kinase is involved in
Differential expression of toll-like receptor 29. Joshi VD, Kalvakolanu DV, Hasday JD, et al: Toll-like receptor 4-mediated cytokine ex-
(TLR)-2 and TLR-4 on monocytes in human IL-18 levels and the outcome of innate im- pression in mouse macrophages. Eur J Im-
sepsis. Clin Exp Immunol 2004; 136: mune response to lipopolysaccharide: Im- munol 2003; 335:97– 605
312–319 portance of a positive feedback loop with 45. Guillot L, Le GofficR, BlochS, et al: Involve-
13. Tsujimoto H, Ono S, Majima T, et al: Neu- caspase-1 in IL-18 expression. J Immunol ment of toll-like receptor 3 in the immune
trophil elastase, MIP-2, and TLR-4 expres- 2002; 169:2536 –2544 response of lung epithelial cells to double-

300 Crit Care Med 2009 Vol. 37, No. 1


stranded RNA and influenza A virus. J Biol a Lupus-like syndrome. J Immunol 2004; gan failure in sepsis. Lancet 2006; 368:
Chem 2005; 280:5571–5580 172:1531–1539 157–169
46. Wrann CD, Tabriz NA, Barkhausen T, et al: 61. Ausubel FM: Are innate immune signaling 76. Marshal JC: The pathogenesis and molecu-
The phosphatidylinositol 3-kinase signaling pathways in plants and animals conserved? lar biology of sepsis. Crit Care Resusc 2006;
pathway exerts protective effects during Nat Immunol 2005; 6:973–979 8:227–229
sepsis by controlling C5a-mediated activa- 62. Caramalho I, Lopes-Carvalho T, Ostler D, et 77. Chandra A, Enkhbaatar P, Nakano Y, et al:
tion of innate immune functions. J Immu- al: Regulatory T cells selectively express Sepsis: Emerging role of nitric oxide and
nol 2007; 178:5940 –5948 toll-like receptors and are activated by lipo- selectins. Clinics 2006; 61:71–76
47. Guha M, Mackman N: The PI3K-Akt path- polysaccharide. J Exp Med 2003; 197: 78. Hu G, Predescu D, Vogel SM: ICAM-1 de-
way limits LPS activation of signaling path- 403– 411 pendent neutrophil adhesion to endothelial
ways and expression of inflammatory medi- 63. Sutmuller RPM, den Brok MH, Kramer M, cells increases caveolae-mediated pulmo-
ators in human monocytic cells. J Biol et al: Toll-like receptor 2 controls expansion nary vascular albumin permeability. Crit
Chem 2002; 277:32124 –32132 and function of regulatory T cells. J Clin Care Med 2007; 34 (Suppl 12): A31
48. Schabbaue G, Tencati M, Pedersen B, et al: Invest 2006; 116:485– 494 79. Garrean S, Gao X-P, Brovkovych V, et al:
PI3K-Akt pathway suppresses coagulation 64. Liu H, Komai-Koma M, Xu D, et al: Toll-like Caveolin-1 regulates NF-kB activation and
and inflammation in endotoxemic mice. Ar- receptor 2 signaling modulates the func- lung inflammatory response to sepsis in-
terioscler Thromb Vasc Biol 2004; 24: tions of CD4⫹CD25⫹ regulatory T cells. duced by lipopolysaccharide. J Immunol
1963–1969 Proc Natl Acad Sci USA 2006; 103: 2006 177: 4853– 4860
49. Pengal RA, Ganesan LP, Wei G, et al: Li- 7048 –7053 80. Wang XM, Kim HP, Song R, et al: Caveo-
popolysaccharide-induced production of in- 65. McHugh RS, Shevach EM: The role of sup- lin-1 confers antiinflammatory effects in
terleukin-10 is promoted by the serin thre- pressor T cells in regulation of immune murine macrophages via the MKK3/p38
onine kinase Akt. Mol Immunol 2006; 43: responses. J Allergy Clin Immunol 2002; MAPK pathway. Am J Respir Cell Mol Biol
1557–1156 110:693–702 2006; 34:434 – 442
50. Fukao T, Koyasu S: PI3K and negative reg- 66. Murphy TJ, Choileain NN, Zang Y, et al: 81. Medina FA, de Almeida CJ, Dew E, et al:
ulation of TLR signaling. Trends Immunol CD4⫹CD25⫹ regulatory T cells control in- Caveolin-1 deficient mice show defects in
2003; 24:358 –363 nate immune reactivity after injury. J Im- innate immunity and inflammatory im-
51. Bommhardt U, Chang KC, Swanson PE, et munol 2005; 174:2957–2963 mune response during Salmonella enterica
al: Akt decreases lymphocyte apoptosis and 67. Monneret G, Debard AL, Venet F, et al: serovar typhimurium infection. Infect Im-
Marked elevation of human circulating
improves survival in sepsis. J Immunol mun 2006; 74:6665– 6674
CD4⫹CD25⫹ regulatory T cells in sepsis-
2004; 172:7583–7591 82. Medina FA, Cohen AW, de Almeida CJ, et al:
induced immunoparalysis. Crit Care Med
52. Mc Dunn JE, Muenzer JT, Rachdi L, et al: Immune dysfunction in caveolin-1 null
2003; 31:2068 –2071
Peptide-mediated activation of Akt and ex- mice following infection with Trypanosoma
68. Venet F, Pachot A, Debard AL, et al: In-
tracellular regulated kinase signaling pre- cruzi (Tulahuen strain). Microbes Infect
creased percentage of CD4⫹CD25⫹ regula-
vents lymphocyte apoptosis. FASEB J 2008; 2007; 9:325–333
tory T cells during septic shock is due to the
22:561–568 83. Astiz ME, DeGent GE, Lin RY, et al: Micro-
decrease of CD4⫹CD25-lymphocytes. Crit
53. Hall A: Rho GT. Pases and the actin cy- vascular function and rheologic changes in
Care Med 2004; 32:2329 –2331
toskeleton. Science 1998; 279:509 –514 hyperdynamic sepsis. Crit Care Med 1995;
69. Takahashi H, Tsuda Y, Takeuchi D, et al:
54. Ruse M, Knaus UG: New players in TLR- 23:265–271
Influence of systemic inflammatory re-
mediated innate immunity. Immunologic 84. Ozdulger A, Cinel I, Koksel O, et al: The
sponse syndrome on host resistance against
Res 2006; 34:33– 48 protective effect of N-acetylcysteine on ap-
bacterial infections. Crit Care Med 2004;
55. Arbibe L, Mira JP, Teusch N, et al: Toll-like optotic lung injury in cecal ligation and
32:1879 –1885
receptor 2-mediated NF-kappa B activation 70. Tiemessen MM, Jagger AL, Evans HG, et al: puncture-induced sepsis model. Shock
requires a Racl dependenpt pathway. Nat CD4⫹CD25⫹Foxp3⫹ regulatory T cells in- 2003; 19:366 –372
Immunol 2000; 1:533–540 duce alternative activation of human mono- 85. Stehr SN, Knels L, Weissflog C, et al: Effects
56. Honing H, van den Berg TK, van der Pol cytes/macrophages. Proc Natl Acad Sci USA of IgM enriched solution on polymorpho-
SM, et al: Rho activation promotes transen- 2007; 104:19446 –19451 nuclear neutrophil function, bacterial
dothelial migration of monocytes via 71. Wisnoski N, Chung CS, Chen Y, et al: The clearance, and lung histology in endotox-
ROCK. J Leukoc Biol 2004; 75:523–528 contribution of CD4⫹ CD25⫹ T-regulato- emia. Shock 2007; 29:167–172
57. Tasaka S, Koh H, Yamada W, et al: Attenu- ry-cells to immune suppression in sepsis. 86. Kabay B, Kocaefe C, Baykal A, et al: Inter-
ation of endotoxin-induced acute lung in- Shock 2007; 27:251–257 leukin-10 gene transfer: Prevention of mul-
jury by the Rho-associated kinase inhibitor, 72. van Maren WW, Jacobs JF, de Vries IJ, et al: tiple organ injury in a murine cecal ligation
Y-27632. Am J Respir Cell Mol Biol 2005; Toll-like receptor signalling on Tregs: To and puncture model of sepsis. World J Surg
32:504 –510 suppress or not to suppress? Immunology 2007; 31:105–115
58. Thorlacius K, Slotta JE, Laschke MW, et al: 2008; 124:445– 452. 87. Ozdulger A, Cinel I, Unlu A, et al: Poly
Protective effect of fasudil, a Rho-kinase 73. Heuer JG, Zhang T, Zhao J, et al: Adoptive (ADP-ribose) synthetase inhibition prevents
inhibitor, on chemokine expression, leuko- transfer of in vitro-stimulated CD4⫹CD25⫹ lipopolysaccharide-induced peroxynitrite
cyte recruitment, and hepatocellular apo- regulatory T cells increases bacterial clear- mediated damage in diaphragm. Pharmacol
ptosis in septic liver injury. J Leukoc Biol ance and improves survival in polymicrobial Res 2002; 46:67–73
2006; 79:923–931 sepsis. J Immunol 2005; 174:7141–7146 88. Taner S, Cinel I, Ozer L, et al: Poly (ADP-
59. Hori S, Nomura T, Sakaguchi S: Control of 74. Venet F, Pachot A, Debard AL, et al: Human ribose) synthetase inhibition reduces bacte-
regulatory T cell development by the tran- CD4⫹CD25⫹ regulatory T lymphocytes in- rial translocation in rats after endotoxin
scription factor FoxP3. Science 2003; 299: hibit lipopolysaccharide-induced monocyte challenge. Shock 2001; 16:159 –162
1057–1061 survival through a Fas/Fas ligand-depen- 89. Cinel I, Buyukafsar K, Cinel L, et al: The
60. Zheng SG, Wang JH, Koss MN, et al: CD4⫹ dent mechanism. J Immunol 2006; 177: role of poly (ADP-ribose) synthetase inhibi-
and CD8⫹ regulatory T cells generated ex 6540 – 6547 tion in preventing endotoxemia-induced in-
vivo with IL-2 and TGF-beta suppress a 75. Brown KA, Brain SD, Pearson JD, et al: testinal epithelial apoptosis. Pharmacol Res
stimulatory Graft-versus- Host disease with Neutrophils in development of multiple or- 2002; 46:119 –127

Crit Care Med 2009 Vol. 37, No. 1 301


90. Savill J: Apoptosis in resolution of inflam- in the pathogenesis of myocardial contrac- M, et al: Transcutaneous vagus nerve stim-
mation. J Leukoc Biol 1997; 61:375–380 tile dysfunction associated with human sep- ulation reduces serum high mobility group
91. Maianski NA, Geissler J, Srinivasula SM, et tic shock. Crit Care Med 2006; 34: box 1 levels and improves survival in mu-
al: Functional characterization of mito- 1073–1079 rine sepsis. Crit Care Med 2007; 35:
chondria in neutrophils: A role restricted to 106. Ulloa L, Tracey KJ: The “cytokine profile”: A 2762–2768
apoptosis. Cell Death Differ 2004; 11: code for sepsis. Trends Mol Med 2005; 11: 122. Goldstein RS, Bruchfeld A, Yang L, et al:
143–153 56 – 63 Cholinergic anti-inflammatory pathway ac-
92. van Raam BJ, Verhoeven AJ, Kuijpers TW: 107. Zimmermann K, Volkel D, Pable S, et al: tivity and High Mobility Group Box-1
Mitochondria in neutrophil apoptosis. Int Native versus recombinant high-mobility (HMGB1) serum levels in patients with
J Hematol 2006; 84:199 –204 group B1 proteins: functional activity in rheumatoid arthritis. Mol Med 2007;
93. Sookhai S, Wang JJ, McCourt M, et al: A vitro. Inflammation 2004; 28:221–229 13(3– 4):210 –215
novel therapeutic strategy for attenuating 108. Rouhiainen A, Tumova S, Valmu L, et al: 123. Youn JH, Oh YJ, Kim ES, et al: High Mo-
neutrophil-mediated lung injury in vivo. Pivotal advance: Analysis of proinflamma- bility Group Box 1 protein binding to lipo-
Ann Surg 2002; 235:285–291 tory activity of highly purified eukaryotic polysaccharide facilitates transfer of lipo-
94. Taneja R, Parodo J, Kapus A, et al: Delayed recombinant HMGB1 (amphoterin). J Leu- polysaccharide to CD14 and enhances
neutrophil apoptosis in sepsis is associated kocyte Biol 2007; 81:49 –58 lipopolysaccharide-mediated TNF-{alpha}
with maintenance of mitochondrial trans- 109. Bonaldi T, Talamo F, Scaffidi P, et al: Mono- production in human monocytes. J Immu-
membrane potential and reduced caspase-9 cytic cells hyperacetylate chromatin protein nol 2008; 180:5067–5074
activity. Crit Care Med 2004; 32:1460 –1469 HMGB1 to redirect it towards secretion. 124. Sha Y, Zmijewski J, Xu Z, et al: HMGB1
95. Fadok VA, Bratton DL, Konowal A, et al: EMBO J 2003; 22:5551–5560 develops enhanced proinflammatory activ-
Macrophages that have ingested apoptotic 110. Youn JH, Shin JS: Nucleocytoplasmic shut- ity by binding to cytokines. J Immunol
cells in vitro inhibit proinflammatory cyto- tling of HMGB1 is regulated by phosphory- 2008; 180:2531–2537
kine production through autocrine/para- lation that redirects it toward secretion. 125. Sundén-Cullberg J, Norrby-Teglund A, Rou-
crine mechanisms involving TGF, PGE2, J Immunol 2006; 177:7889 –7897 hiainen A, et al: Persistent elevation of high
and PAF. J Clin Invest 1998; 101:890 – 898 111. Bell CW, Jiang W, Reich CF III, et al: The mobility group box-1 protein (HMGB1) in
96. Sanford AN, Suriano AR, Herche D, et al: extracellular release of HMGB1 during ap- patients with severe sepsis and septic shock.
Abnormal apoptosis in chronic granuloma- optotic cell death. Am J Physiol Cell Physiol Crit Care Med 2005; 33: 564 –573
tous disease and autoantibody production 2006; 291:C1318 –C1325 126. Gibot S, Massin F, Cravoisy A, et al: High-
characteristic of lupus. Rheumatology 112. Qin S, Wang H, Yuan R, et al: Role of
mobility group box 1 protein plasma con-
2006; 45:178 –181 HMGB1 in apoptosis-mediated sepsis lethal-
centrations during septic shock. Intensive
97. Daleke DL: Regulation of transbilayer ity. J Exp Med 2006; 203:1637–1642
Care Med 2007; 33:1347–1353
plasma membrane phospholipid asymme- 113. Hori O, Brett J, Slattery T, et al: The recep-
127. Karlsson S, Pettilä V, Tenhunen J, et al:
try. J Lipid Res 2003; 44:233–242 tor for advanced glycation end products
HMGB1 as a predictor of organ dysfunction
98. Bayir H, Kagan VE: Bench-to-bedside re- (RAGE) is a cellular binding site for ampho-
and outcome in patients with severe sepsis.
view: Mitochondrial injury, oxidative stress terin: Mediation of neurite outgrowth and
Intensive Care Med 2008; 34:1046 –1053.
and apoptosis—there is nothing more prac- co-expression of rage and amphoterin in the
128. Harris HE, Raucci A: Alarmin(g) news about
tical than a good theory. Crit Care 2008; developing nervous system. J Biol Chem
danger. EMBO Rep 2006; 7:774 –778
12:206 1995; 270:25752–25761
129. Chavakis T, Bierhaus A, Al-Fakhri N, et al:
99. Tyurina YY, Basova LV, Konduru NV, et al: 114. Park JS, Svetkauskaite D, He Q, et al: In-
The pattern recognition receptor (RAGE) is
Nitrosative stress inhibits the aminophos- volvement of toll-like receptors 2 and 4 in
a counterreceptor for leukocyte integrins: A
pholipid translocase resulting in phosphati- cellular activation by high mobility group
novel pathway for inflammatory cell re-
dylserine externalization and macrophage box 1 protein. J Biol Chem 2004; 279:
engulfment: Implications for the resolution 7370 –7377 cruitment. J Exp Med 2003; 198:1507–1515
of inflammation. J Biol Chem 2007; 282: 115. Wang H, Bloom O, Zhang M, et al: HMG-1 130. Lutterloh EC, Opal SM, Pittman DD, et al:
8498 – 8509 as a late mediator of endotoxin lethality in Inhibition of the RAGE products increases
100. Swan R, Chung CS, Albina J, et al: Polymi- mice. Science 1999; 285:248 –251 survival in experimental models of severe
crobial sepsis enhances clearance of apopto- 116. Mitola S, Belleri M, Urbinati C, et al: Cut- sepsis and systemic infection. Crit Care
tic immune cells by splenic macrophages. ting edge: Extracellular high mobility 2007; 11:R122
Surgery 2007; 142:253–261 group box-1 protein is a proangiogenic cy- 131. Calandra T, Bernhagen J, Metz CN, et al:
101. Macdonald J, Galley HF, Webster NR: Oxi- tokine. J Immunol 2006; 176:12–15 MIF as a glucocorticoid-induced modulator
dative stress and gene expression in sepsis. 117. Tzeng HP, Fan J, Vallejo JG, et al: Negative of cytokine production. Nature 1995; 377:
Br J Anaest 2003; 90:221–232 inotropic effects of high-mobility group box 68 –71
102. Matejovic M, Krouzecky A, Rokyta R Jr, et 1 protein in isolated contracting cardiac 132. Calandra T, Echtenacher B, Roy DL, et al:
al: Effects of combining inducible nitric ox- myocytes. Am J Physiol Heart Circ Physiol Protection from septic shock by neutraliza-
ide synthase inhibitor and radical scavenger 2008; 294:H1490 –H1496 tion of macrophage migration inhibitory
during porcine bacteremia. Shock 2007; 27: 118. Wang H, Yu M, Ochani M, et al: Nicotinic factor. Nat Med 2000; 6:164 –170
61– 68 acetylcholine receptor ␣7 receptor is an es- 133. Bozza M, Satoskar AR, Lin G, et al: Targeted
103. Fialkow L, Wang Y, Downey GP: Reactive sential regulator of inflammation. Nature disruption of migration inhibitory factor
oxygen and nitrogen species as signaling 2003; 421:384 –388 gene reveals its critical role in sepsis. J Exp
molecules regulating neutrophil function. 119. Pavlov VA, Wang H, Czura CJ, et al: The Med 1999; 189:341–346
Free Radic Biol Med 2007; 42:153–164 cholinergic anti-inflammatory pathway: A 134. Roger T, David J, Glauser MP, et al: MIF
104. Galley HF, Davies MJ, Webster NR: Xan- missing link in neuromodulation. Mol Med regulates innate immune responses
thine oxidase activity and free radical gen- 2003; 9:125–134 through modulation of Toll-like receptor
eration in patients with sepsis syndrome. 120. Wang H, Liao H, Ochani M, et al: Cholin- 4. Nature 2001; 414:920 –924
Crit Care Med 1996; 24:1649 –1653 ergic agonists inhibit HMGB1 release and 135. Mitchell RA, Liao H, Chesney J, et al: Mac-
105. Soriano FG, Nogueira AC, Caldini EG, et al: improve survival in experimental sepsis. rophage migration inhibitory factor (MIF)
Potential role of poly (adenosine 5⬘- Nat Med 2004; 10:1216 –1221 sustains macrophage proinflammatory
diphosphate-ribose) polymerase activation 121. Huston JM, Gallowitsch-Puerta M, Ochani function by inhibiting p53: Regulatory role

302 Crit Care Med 2009 Vol. 37, No. 1


in the innate immune response. Proc Natl 152. Stouthard JM, Levi M, Hack CE, et al: In- versus protein C in severe sepsis. Crit Care
Acad Sci USA 2002; 99:345–350 terleukin-6 stimulates coagulation, not fi- Med 2001; 29(Suppl):S69 –S74
136. Kreymann G, Grosser S, Buggisch P, et al: brinolysis, in humans. Thromb Haemost 166. Joyce DE, Gelbert L, Ciaccia A, et al: Gene
Oxygen consumption and resting metabolic 1996; 76:738 –742 expression profile of antithrombotic protein
rate in sepsis, sepsis syndrome, and septic 153. Aupeix K, Hugel B, Martin T, et al: The C defines new mechanisms modulating in-
shock. Crit Care Med 1993; 21:1012–1019 significance of shed membrane particles flammation and apoptosis. J Biol Chem
137. Rosser DM, Stidwill RP, Jacobson D, et al: during programmed cell death in vitro, and 2001; 14:11199 –111203
Oxygen tension in the bladder epithelium in vivo, in HIV-1 infection. J Clin Invest 167. Jackson CJ, Xue M: Activated protein C-An
rises in both high and low cardiac output 1997; 99:1546 –1554 anticoagulant that does more than stop
endotoxemic sepsis. J Appl Physiol 1995; 154. Soriano AO, Jy W, Chirinos JA, et al: Levels clots. Int J Biochem Cell Biol 2008; 40:
79:1878 –1882 of endothelial and platelet microparticles 2692–2697
138. Hotchkiss RS, Swanson PE, Freeman BD, et and their interactions with leukocytes neg- 168. Dutt T, Toh CH: The yin-yang of thrombin
al: Apoptotic cell death in patients with sep- atively correlate with organ dysfunction and and activated protein C. Br J Haematology
sis, shock, and multiple organ dysfunction. predict mortality in severe sepsis. Crit Care 2008; 140:505–515
Crit Care Med 1999; 27:1230 –1251 Med 2005; 33:2540 –2546 169. Riewald M, Ruf W: Protease-activated recep-
139. Brealey D, et al: Mitochondrial dysfunction 155. Fujimi S, Ogura H, Tanaka H, et al: Acti- tor-1 signaling by activated protein C in
in a long-term rodent model of sepsis and vated polymorphonuclear leukocytes en- cytokine-perturbed endothelial cells is dis-
organ failure. Am J Physiol Regul Integr hance production of leukocyte micropar- tinct from thrombin signaling. J Biol Chem
Comp Physiol 2004; 286:R491–R497 ticles with increased adhesion molecules in 2005; 280:19808 –19814
140. Crouser ED: Mitochondrial dysfunction in patients with sepsis. J Trauma 2002; 52: 170. Bae JS, Yang L, Rezaie AR: Receptors of the
septic shock and multiple organ dysfunc- 443– 448 protein C activation and activated protein C
tion syndrome. Mitochondrion 2004; 156. Amaral A, Opal SM, Vincent JL: Coagulation signaling pathways are colocalized in lipid
4:729 –741 in sepsis. Intensive Care Med 2004; 30: rafts of endothelial cells. Proc Natl Acad Sci
141. Fink MP: Cytopathic hypoxia. Mitochon- 1032–1040 USA 2007; 104:2867–2872
drial dysfunction as mechanism contribut- 157. Vervloet MG, Thijs LG, Hack CE: Derange- 171. Bae JS, Yang L, Manithody C, et al: The
ing to organ dysfunction in sepsis. Crit Care ments of coagulation and fibrinolysis in ligand occupancy of endothelial protein
Clinics 2001; 17:219 –237 critically ill patients with sepsis and septic C receptor switches the protease-activated
142. Brealey D, Brand M, Hargreaves I, et al: shock. Semin Thromb Hemost 1998; 24: receptor 1-dependent signaling specificity
Association between mitochondrial dys- 33– 44
of thrombin from a permeability-enhanc-
function and severity and outcome of septic 158. Yan SB, Dhainut JF: Activated protein C
ing to a barrier-protective response
shock. Lancet 2002; 360:219 –223 versus protein C in severe sepsis. Crit Care
in endothelial cells. Blood 2007; 110:
143. Crouser ED, Julian MW, Blaho DV, et al: Med 2001; 29(Suppl):S69 –S74
3909 –3916
Endotoxin-induced mitochondrial damage 159. Xue M, Campbell D, Jackson CJ: Protein C
172. Shorr AF, Bernard GR, Dhainaut JF, et al:
correlates with impaired respiratory activ- is an autocrine growth factor for human
Protein C concentrations in severe sepsis:
ity. Crit Care Med 2002; 30:276 –284 skin keratinocytes, J Biol Chem 2007; 282:
An early directional change in plasma levels
144. Levy RJ, Vijayasarathy C, Raj NR, et al: 13610 –13616
predicts outcome. Crit Care 2006; 10:R92
Competitive and noncompetitive inhibition 160. Xue M, March L, Sambrook PN, et al:
173. Cinel I, Dellinger RP: Current treatment of
of myocardial cytochrome C oxidase in sep- Endothelial protein C receptor is overex-
severe sepsis. Curr Infect Dis Rep 2006;
sis. Shock 2004; 21:110 –114 pressed in rheumatoid arthritic (RA) sy-
8:358 –365
145. Rabuel C, Renaud E, Brealey D, et al: Hu- novium and mediates the anti-inflamma-
174. Dellinger RP, Levy MM, Carlet JM, et al:
man septic myopathy: Induction of cycloox- tory effects of activated protein C in RA
Surviving Sepsis Campaign: International
ygenase, heme oxygenase and activation of monocytes. Ann Rheum Dis 2007; 66:
the ubiquitin proteolytic pathway. Anesthe- 1574 –1580 guidelines for management of severe sepsis
siology 2004; 101:583–590 161. Regan LM, Stearns-Kurosawa DJ, Kurosawa and septic shock: 2008. Crit Care Med 2008;
146. Larche J, Lancel S, Hassoun SM, et al: In- S, et al: The endothelial cell protein C re- 36:296 –327
hibition of mitochondrial permeability ceptor: Inhibition of activated protein C an- 175. Bae JS, Yang L, Manithody C, et al: Engi-
transition prevents sepsis-induced myocar- ticoagulant function without modulation of neering a disulfide bond to stabilize the
dial dysfunction and mortality. J Am Coll reaction with proteinase inhibitors. J Biol calcium binding loop of activated protein C
Cardiol 2006; 48:377–385 Chem 1996; 271:17499 –17503 eliminates its anticoagulant but not protec-
147. Levi M, Ten Cate H: Disseminated intravas- 162. Kurosawa S, Stearns-Kurosawa DJ, Carson tive signaling properties. J Biol Chem 2007;
cular coagulation. N Engl J Med 1999; 341: CW, et al: Plasma levels of endothelial cell 282:9251–9259
586 –592 protein C receptor are elevated in patients 176. Kerschen EJ, Fernandez JA, Cooley BC, et
148. Schouten M, Wiersinga WJ, Levi M, et al: with sepsis and systemic lupus erythemato- al: Endotoxemia and sepsis mortality reduc-
Inflammation, endothelium, and coagula- sus: Lack of correlation with thrombo- tion by non-anticoagulant activated protein
tion in sepsis. J Leukoc Biol 2008; 83: modulin suggests involvement of different C. J Exp Med 2007; 204:2439 –2448
536 –545 pathological processes. Blood 1998; 91: 177. Moake JL: Thrombotic microangiopathies.
149. Opal SM: The nexus between systemic in- 725–727 N Engl J Med 2002; 347:589 – 600
flammation and disordered coagulation in 163. Yan SB, Helterbrand JD, Hartman DL, et al: 178. Ruggeri ZM: Von Willebrand factor, plate-
sepsis. J Endotoxin Res 2004; 10:125–129 Low levels of protein C are associated with lets and endothelial cell interactions.
150. Esmon CT: Are natural anticoagulants can- poor outcome in severe sepsis. Chest 2001; J Thromb Haemost 2003; 1:1335–1342
didates for modulating the inflammatory 120:915–922 179. Furlan M, Robles R, Galbusera M, et al: von
response to endotoxin? Blood 2000; 95: 164. LaRosa SP, Opal SM, Utterback B, et al: Willebrand factor-cleaving protease in
1113–1116 Decreased protein C, protein S, and anti- thrombotic thrombocytopenic purpura and
151. Johnson K, Choi Y, DeGroot E, et al: Po- thrombin levels are predictive of poor out- the hemolytic-uremic syndrome. New Engl
tential mechanisms for a proinflamma- come in Gram-negative sepsis caused by J Med 1998; 339:1578 –1584
tory vascular cytokine response to coagu- Burkholderia pseudomallei. Int J Infect Dis 180. Arya M, Anvari B, Romo GM, et al: Ultra-
lation activation. J Immunol 1998; 160: 2006; 10:25–31 large multimers of von Willebrand factor
5130 –5135 165. Yan SB, Dhainaut JF: Activated protein C form spontaneous high-strength bonds

Crit Care Med 2009 Vol. 37, No. 1 303


with the platelet glycoprotein Ib-IX com- tients with severe sepsis. Haematologica 187. Martin K, Borgel D, Lerolle N, et al: De-
plex: Studies using optical tweezers. Blood 2007; 92:121–124 creased ADAMTS-13 (A disintegrin-like and
2002; 99:3971–3977 184. Kremer Hovinga JA, Zeerleder S, Kessler metalloprotease with thrombospondin type
181. Bianchi V, Robles R, Alberio L, et al: Von P, et al: ADAMTS-13, von Willebrand fac- 1 repeats) is associated with a poor progno-
Willebrand factor-cleaving protease (AD- tor and related parameters in severe sep- sis in sepsis-induced organ failure. Crit
AMTS13) in thrombocytopenic disorders: A sis and septic shock. J Thromb Haemost Care Med 2007; 35:2375–2382
severely deficient activity is specific for 2007; 5:2284 –2290 188. Grewal PK, Uchiyama S, Ditto D, et al: The
thrombotic thrombocytopenic purpura. 185. Bockmeyer CL, Claus RA, Budde U, et al: Ashwell receptor mitigates the lethal coagulopa-
Blood 2002; 100:710 –713 Inflammation-associated ADAMTS13 defi- thy of sepsis. Nat Med 2008; 14:648–655
182. Ono T, Mimuro J, Madoiwa S, et al: Severe ciency promotes formation of ultra-large 189. van’t Veer C, van der Poll T. Keeping blood
secondary deficiency of von Willebrand fac- von Willebrand factor. Haematologica clots at bay in sepsis. Nat Med 2008; 14:
tor-cleaving protease (ADAMTS13) in pa- 2008; 93:137–140 606 – 608
tients with sepsis-induced disseminated in- 186. Reiter RA, Varadi K, Turecek PL, et al: 190. Treziack S, Cinel I, Dellinger RP, et al:
travascular coagulation: Its correlation with Changes in ADAMTS13 (von-Willebrand Resuscitating the microcirculation in se-
development of renal failure. Blood 2006; factor-cleaving protease) activity after in- vere sepsis: The central role of nitric oxide,
107:528 –534 duced release of von Willebrand factor emerging concepts for novel therapies, and
183. Nguyen TC, Liu A, Liu L, et al: Acquired during acute systemic inflammation. challenges for clinical trials. Acad Emerg
ADAMTS-13 deficiency in pediatric pa- Thromb Haemost 2005; 93:554 –558 Med 2008; 15:1–15

304 Crit Care Med 2009 Vol. 37, No. 1

You might also like