SlideShare a Scribd company logo
1 of 14
Download to read offline
Concise Definitive Review
Molecular biology of inflammation and sepsis: A primer*
Ismail Cinel, MD, PhD; Steven M. Opal, MD
I
n sepsis, the expected and appropri-
ate inflammatory response to an in-
fectious process becomes amplified
leading to organ dysfunction or risk
for secondary infection. A continuum exists
from a low grade systemic response associ-
ated with a self-limited infection to a
marked systemic response with solitary or
multiorgan dysfunction, i.e., severe sepsis.
As a clinical syndrome, sepsis occurs when
an infection is associated with the systemic
inflammatory response.
The complex toll-like receptor signaling
and associated downstream regulators of
immune cell functions play a crucial role in
the innate system as a first line of defense
against pathogens (1). However, signaling
is sometimes conflicting and a sustained
inflammatory response can result in tissue
damage. In addition, a reduction in their
antimicrobial capacity may set the stage for
opportunistic and/or super infections (2).
Severe sepsis may also be associated with
an exaggerated procoagulant state. This
may lead to ischemic cell injury, an effect
that further amplifies the damage caused
by inappropriate inflammation. A micro-
vasculature injured by inflammation and
ischemia, in turn, further deranges the
host response by altering leukocyte traffick-
ing, generating apoptotic microparticles,
and increasing cellular hypoxia (3). Mito-
chondrial dysfunction, an acquired intrin-
sic defect in cellular respiration termed “cy-
topathic hypoxia,” also has an important
role by decreasing cellular oxygen con-
sumption in this chaotic process. In this
review, we highlight the current under-
standing of the basic molecular mecha-
nisms that modulate these events so as to
produce sepsis.
Pattern Recognition Receptors,
Pathogen-Associated Molecular
Patterns (PAMPs) and Danger-
Associated Molecular Patterns
(DAMPs)
The initiation of the host response
during sepsis or tissue injury involves
three families of pattern recognition re-
ceptors (PRRs): 1) toll-like receptors
(TLRs); 2) nucleotide-oligomerization
domain leucine-rich repeat (NOD-LRR)
proteins; and 3) cytoplasmic caspase ac-
tivation and recruiting domain heli-
cases such as retinoic-acid-inducible
gene I (RIG-I)-like helicases (RLHs) (4,
5). These receptors initiate the innate
immune response and regulate the
adaptive immune response to infection
or tissue injury.
Gram-positive and Gram-negative bac-
teria, viruses, parasites, and fungi all pos-
sess a limited number of unique cellular
constituents not found in vertebrate ani-
mals. These elements are now referred to as
PAMPs, or more appropriately microbial-
associated molecular patterns, as these
molecules are also common in nonpatho-
genic and commensal bacteria (6). PAMPs
bind to PRRs, such as TLRs, expressed on
the surface of host cells. Cytoplasmic PRRs
exist to detect invasive intracellular patho-
gens (7). The NOD proteins recognize com-
mon fragments of bacterial peptidoglycan.
Diamino-pimelate from Gram-negative
bacteria is the ligand for NOD1 and mu-
ramyl dipeptide from peptidoglycan is the
ligand for NOD2 in the cytosol (see Figs. 1
and 2). The PRRs also recognized damage
signals from the release of endogenous pep-
tides and glycosaminoglycans from apopto-
tic or necrotic host cells (8–10) Caspase
activation and recruiting domain helicases
From the Division of Critical Care Medicine (IC), The
Robert Wood Johnson School of Medicine, The University
of Medicine and Dentistry of New Jersey, Camden NJ;
and The Infectious Disease Division (SMO), Memorial
Hospital of RI, The Warren Alpert School of Medicine of
Brown University, Providence, RI.
Dr. Opal has received grants from Wyeth and Eisai
Inc. Dr. Cinel has not disclosed any potential conflicts
of interest.
For information regarding this article, E-mail:
cinel-ismail@cooperhealth.edu
Copyright © 2008 by the Society of Critical Care
Medicine and Lippincott Williams & Wilkins
DOI: 10.1097/CCM.0b013e31819267fb
Background: Remarkable progress has been made during the
last decade in defining the molecular mechanisms that underlie
septic shock. This rapidly expanding field is leading to new
therapeutic opportunities in the management of severe sepsis.
Aim: To provide the clinician with a timely summary of the
molecular biology of sepsis and to better understand recent
advances in sepsis research.
Data Selection: Medline search of relevant publications in
basic mechanisms of sepsis/severe sepsis/septic shock, and se-
lected literature review of other manuscripts about the signalo-
some, inflammasome, apoptosis, or mechanisms of shock.
Data Synthesis and Findings: The identification of the toll-like
receptors and the associated concept of innate immunity based upon
pathogen- or damage-associated molecular pattern molecules al-
lowed significant advances in our understanding of the pathophys-
iology of sepsis. The essential elements of the inflammasome and
signal transduction networks responsible for activation of the host
response have now been characterized. Apoptosis, mitochondrial
dysfunction, sepsis-related immunosuppression, late mediators of
systemic inflammation, control mechanisms for coagulation, and
reprogramming of immune response genes all have critical roles in
the development of sepsis.
Conclusions: Many of these basic discoveries have direct impli-
cations for the clinical management of sepsis. The translation of
these “bench-to-bedside” findings into new therapeutic strategies is
already underway. This brief review provides the clinician with a
primer into the basic mechanisms responsible for the molecular
biology of sepsis, severe sepsis, and septic shock.(Crit Care Med
2009; 37:291–304)
KEY WORDS: sepsis; inflammation; toll like receptors; inflamma-
some; signalosome; apoptosis; neutrophils; mitochondrial dys-
function; reactive oxygen species; nitric oxide; peroxynitrite; ac-
tivated protein C; a disintegrin-like and metalloproteinase with
thrombospondin type-1 motifs-13; pathogen associated molecular
patterns; danger associated molecular patterns
291Crit Care Med 2009 Vol. 37, No. 1
primarily recognize viral nucleic acids and
activate antiviral measures including the
type I interferons.
TLR expression is significantly up-
regulated in experimental models of sep-
sis and in patients with sepsis (11–14).
Trauma including thermal injury gener-
ates danger-associated molecular pat-
terns (i.e., high mobility group box-1,
heat shock proteins, S100 proteins, hya-
luran, etc.) that augment TLR expression
like PAMPs. It also primes the innate im-
mune system for enhanced TLR reactiv-
ity, resulting in excess lipopolysaccharide
(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
TLRs induce pro-interleukin(IL)-1beta
production and prime NLR-containing
multiprotein complexes, termed “inflam-
masomes,” to respond to bacterial prod-
ucts and products of damaged cells (17,
19). This results in caspase-1 activation
and the subsequent processing of pro-
IL-1␤ to its active extracellular form
IL-1␤ (19). Caspases are a set of cysteine
proteases that alter the enzymatic activity
of target proteins at specific peptide se-
quences adjacent to aspartate moieties.
Caspases are important in process of ap-
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-
flammasome constructions have been
suggested to contain pyrin, NALP3, and
other members of the NOD-LRR family
(21–23). ASC facilitates inflammasome
assembly thus triggering caspase-1 acti-
vation and IL-1␤ processing. Interest-
ingly, ASC can also regulate the nuclear
factor (NF)-␬B pathway, thus linking the
inflammasome to the signalosome
(Fig. 1) (24).
The inflammasome pathways contrib-
ute to the inflammatory response in sep-
sis (25). Caspase-1 knock out mice are
protected from sepsis (26) while a natu-
rally occurring polymorphism for human
caspase-12, a putative regulator of
Figure 1. Specific host immune response to each pathogen is mediated by various sets of pathogen
associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) as detailed in Figure
1. PRRs are essential for initiating the host’s immune defenses against invading pathogens, yet they
can also contribute to persistent and deleterious systemic inflammation. PRRs also serve as receptors
for endogenous danger signals, hemodynamic changes in sepsis (tissue hypoperfusion and ischemia/
reperfusion phenomenon), thus same signaling systems that alerts the highly advantageous, host
defense mechanisms, also contributes to the disadvantageous, pathologic events of systemic inflam-
mation, coagulation, tissue damage in target organs in sepsis. Heat shock proteins, fibrinogen,
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),
especially TLR4, are involved in the recognition of these endogenous or harmful self-antigens ligands
which are released during noninfectious injury, such as trauma or ischemia/reperfusion suggesting
their function may not be restricted to the recognition of extrinsic pathogens. NOD-LRR, nucleotide-
oligomerization domain leucine-rich repeat; ASC, apoptosis-associated speck-like protein containing
caspase activation and recruiting domain; NF-␬␤, nuclear factor ␬␤.
Figure 2. Binding of toll-like receptors (TLRs) activates intracellular signal-transduction pathways that
lead to the activation of transcriptional activators such as interferon regulator factors, phosphoino-
sitide 3-kinase (PI3K)/Akt, activator protein-1, and cytosolic nuclear factor-kappa ␤ (NF-␬␤). Activated
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,
proinflammatory cytokines, as well as enzymatic activation of cellular proteases. TLR9 DNA, TLR 3
dsRNA, and TLR7/8 ss RNA are endosomal. TLR 10 ligand is not defined and TLR1 forms heterodimers
with TLR2. LPS, lipopolysaccharide; IRF, interferon regulatory factor; JNK; c Jun N-terminal kinase.
292 Crit Care Med 2009 Vol. 37, No. 1
caspase-1, has been linked to sepsis (27,
28). Thus, caspase-1 activation appears to
be a prerequisite for a competent im-
mune response (29). Recently, the in-
flammasome components have been
shown to be significantly lower in septic
shock patients during the early stages of
systemic inflammatory response with el-
evated plasma cytokines levels (30). This
monocyte deactivation process may be
maladaptive in the later phases of sepsis
and predispose to secondary infection.
Caspase-1 and its proinflammatory cy-
tokine products are likely to contribute to
the pathogenesis of sepsis in overwhelm-
ing inflammation. However, like many
other essential elements of innate immu-
nity, caspase-1 also has a positive impact
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-
associated kinase Ϫ1 haplotype has re-
cently been demonstrated to affect the
magnitude of NF-␬B activation and di-
rectly correlates with an increased inci-
dence of septic shock and significantly
reduced survival rates (38). The impact of
NF-␬B signaling is tissue-specific as defi-
cient NF-␬B activation in intestinal epi-
thelium is associated with increased in-
flammation in vivo (39, 40). Both studies
demonstrate that defects of NF-␬B signal-
ing cause immunosuppression which
triggers and maintains inflammation.
Thus, it can be suggested that inhibition
of massive NF-␬B activation in vivo leads
to reduced inflammatory responses, at
least during certain phases and certain
tissues (i.e., parenchymal tissues) in sep-
sis. On the other hand, this activation is
followed by negative NF-␬B regulation
favoring apoptosis in immune cells which
Figure 3. Pathogenic mechanisms during sepsis or in response to tissue injury can lead to organ
dysfunction. PRRs, pattern recognition receptors; TLRs, toll-like receptors; NOD-LRR, nucleotide-
oligomerization domain leucine-rich repeat protein receptors; RLHs; retinoic-acid-inducible gene I
(RIG-I)-like helicases; TNF-␣, tumor necrosis factor alpha; IL-1, interleukin 1; HMGB-1, high mobility
group box-1; LPS, lipopolysaccharide; LTA, lipoteichoic acid; PGN, peptidoglycan; VSMCs, vascular
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
end products; ROCK; RhoA/Rho kinase; PARP-1, poly(ADP ribose) polymerase-1; PAR, protease
activated receptor; ROS/RNS, reactive oxygen and nitrogen species; NF, nuclear factor; NADPH,
nicotimanide adenosine dinucleotide phosphate; PAMPs, pathogen-associated molecular patterns;
DAMPs, danger-associated molecular patterns.
293Crit Care Med 2009 Vol. 37, No. 1
may lead to immunosuppression and fatal
outcome in severe sepsis. Recently, using
specific gene-targeted deletions, it has
been shown that deletion of MyD88
caused a worsened survival in a model of
severe peritonitis, despite the marked de-
crease in sepsis-induced T and B lympho-
cyte apoptosis suggesting MyD88, like
NF-␬B, is also critical host survival in
sepsis (41).
Toll-Like Receptor Signaling
The Role of Phosphoinositide 3-Kinase.
Phosphoinositide 3-kinase (PI3K), a sig-
nal transduction enzyme, and the down-
stream serine/threonine kinase Akt (also
known as protein kinase B) have been
reported in cellular activation, inflamma-
tory responses, chemotaxis, and apoptosis
(Figs. 2 and 3) (42). PI3K can function
either as a positive or negative regulator
of TLR signaling. PI3Ks (three types) act
at several steps downstream of TLRs, de-
pending on the cell type and/or the en-
gagement of a specific TLR regulating
downstream signaling to NF-KB transac-
tivation or to mitogen-activated phos-
phokinase activation (43, 44). As a posi-
tive mediator of TLR signaling, PI3K
together with p38 and extracellular reg-
ulated kinase (ERK)1/2 mitogen-acti-
vated phosphokinases, lead to production
of proinflammatory cytokines IL-1␤,
IL-6, and IL-8 upon microbial challenge
(44, 45). In detail, PI3K activation ap-
peared to have a significantly promoting
function for these mediators in mono-
cytes, whereas activation appeared to
limit the LPS response for generation of
these cytokines in neutrophils (46). How-
ever, PI3K inhibition resulted in im-
paired oxidative burst and phagocytosis
activity in both neutrophils and mono-
cytes. By limiting C5a-mediated effects
on neutrophil cytokine generation, and
promoting oxidative burst and phagocy-
tosis, PI3K activation seems to be a ther-
apeutic approach for limiting inflamma-
tion in sepsis.
On the other hand, PI3K/Akt signaling
pathway acts as an endogenous negative
feedback mechanism that serves to limit
proinflammatory and apoptotic events as
seen in monocytes in response to endo-
toxin (47). The suppressive role in in-
flammation and coagulation was con-
firmed in mouse model of endotoxemia
(48). It can also promote the generation
of anti-inflammatory cytokine IL-10 (49).
PI3K/Akt has an effect in balancing Th1
vs. Th2 responses (50). Overexpression of
Akt in lymphocytes decreases lymphocyte
apoptosis, a Th1 cytokine propensity, and
improve outcome in cecal ligation and
puncture-induced sepsis (51). Peptide-
mediated activation of Akt and extracel-
lular regulated kinase signaling protects
lymphocytes from numerous apoptotic
stimuli both in vitro and in vivo (52).
This suggests again the survival advan-
tage of PI3K/Akt pathway activation for
the later stages of sepsis.
The Role of Rho GTPases. The Rho
family of small GTPases is one of the
master regulators of cell motility, as they
control actin cytoskeleton remodeling.
RhoA, Rac1, and CDc42 are the well
known family members which act molec-
ular switches regulating responses of in-
nate immune cells related to pathogen
sensing, intracellular uptake, and de-
struction (53). These GTPases play both
unique and overlapping roles in phago-
cyte functions including migration, che-
motaxis, and optimal bacterial killing
(54). It has been shown that PAMPs may
utilize GTPases through TLRs (i.e.,
TLR-2, -4, -3, and -9) in which Rac1 ac-
tivation is required for PI3K activation
upon TLR2 stimulation (55). RhoA regu-
lates not only cytoskeletal events, which
mediate neutrophil migration, but also
contributes to NF-␬␤-dependent proin-
flammatory gene transcription (Fig. 3)
(56). It has been suggested that ROCK
inhibition could attenuate cytoskeletal
rearrangement of endothelial cells, lead-
ing to decreased neutrophil emigration
into the lung parenchyma in LPS-
induced lung injury (57). An important
role for rho kinase in leukocyte recruit-
ment is also supported in an endotoxemic
liver injury model (58). The recently
emerged connections between TLR sig-
naling and small Rho-GTPases seem to
provide new therapeutic avenue of re-
search in sepsis.
Toll-Like Receptor Signaling
and Regulatory T Cells (Tregs)
To balance self-tolerance and immu-
nity against pathogens, the immune sys-
tem depends on both up-regulatory and
down-regulatory mechanisms. Recent
studies have suggested that several lym-
phocyte subpopulations (i.e., CD4ϩCD25ϩ
Foxp3ϩ T regulatory-cell ͓Tregs͔) may
have the capacity to actively suppress an
adaptive immune response and may po-
tentially be involved in septic immune
dysfunction. Naturally occurring Tregs
express the transcription factor forkhead
box protein (FoxP3), which is induced by
the anti-inflammatory cytokine trans-
forming growth factor-␤ (59, 60). TLR
triggering induces dendritic cell matura-
tion also, which is essential for the induc-
tion of adaptive immune responses (61).
Studies have recently highlightened the
importance of TLRs on Tregs (62). In this
regard, the dominant role of TLR2 signal-
ing on the Treg-mediated immune sup-
pression has been demonstrated (63, 64).
Tregs control inflammatory reactions
to commensal bacteria and opportunist
pathogens and play a major role in sup-
pressing immune reactivity, ranging
from autoimmunity to infectious disease
(65) and to injury (66). Monneret et al
(67) observed that sepsis increases
CD4ϩCD25ϩ T cells in the peripheral
blood of septic patients. This was subse-
quently found to be a relative increase in
Tregs due to a decrease in the CD4ϩCD25–
T effector cell populations (68). Further-
more, Treg-mediated induction of “alter-
natively activated” macrophages has been
demonstrated suggesting as one of the
causes of immune dysfunction in sys-
temic inflammatory response syndrome
and sepsis (69, 70). Targeting apoptosis of
Tregs may be a new therapeutic approach
in preventing the continuum of sepsis to
severe sepsis. However, it has been re-
ported that depletion of CD25ϩ cells be-
fore inducing sepsis did not alter septic
mortality pointing the need of more stud-
ies to clarify the significance of this cell
population’s expansion in sepsis morbid-
ity (71, 72).
Adoptive transfer of Tregs before or
following the initiation of polymicrobial
sepsis improved survival by enhancing
tumor necrosis factor (TNF)-␣ produc-
tion and bacterial clearance (73). Tregs
inhibit LPS-induced monocyte survival
through the Fas/FasL dependent pro-
apoptotic mechanism, which might play
a role in the resolution of exaggerated
inflammation (74). The positive and neg-
ative effects of TLR on Tregs are curious
in sepsis, and the dynamics of TLR ex-
pression on immune-suppressive Tregs
upon inflammation or in relation to type
of pathogen are needed to illuminate.
Neutrophils and
Monocyte/Macrophages in
Inflammation
Myeloid cells including neutrophils
and elements of monocyte/macrophage
lineage are heavily armed with large
stores of proteolytic enzymes and with
294 Crit Care Med 2009 Vol. 37, No. 1
the capacity to rapidly generate ROS and
RNS to degrade internalized pathogens.
The highly proapoptotic nature of neu-
trophils is designed to maintain a balance
between antimicrobial effectiveness and
the potential for neutrophil-associated
damage to the host in septic challenge or
in other injurious processes, such as
trauma or ischemia/reperfusion (75).
Host tissue damage in severe sepsis may
arise via a variety of mechanisms includ-
ing premature neutrophil activation dur-
ing migration, extracellular release of cy-
totoxic molecules and toxins during
microbial killing, removal of infected or
damaged host cells or debris during host
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-
grins (77, 78). P-selectin is expressed on
platelets; E- and P-selectins are expressed
by endothelial cells, whereas L-selectin is
expressed on leukocytes. Selectins mediate
neutrophil rolling along activated endothe-
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-
nisms are compromised in the microcircu-
lation including endothelial hyperactivity,
fibrin deposition, microvascular occlusion,
and cellular exudates that further impede
adequate tissue oxygenation. Neutrophils
participate in these rheologic changes
through their augmented binding to blood
vessel walls and through the formation of
platelet-leukocyte aggregates (83). Neutro-
phil elastase, other proteases, glycases and
inflammatory cytokines degrade endoge-
nous anticoagulant activity, and impair fi-
brinolysis on endothelial surfaces favoring
a procoagulant state (1).
The Role of Apoptosis. Sepsis-induced
neutrophil-mediated tissue injury has
been demonstrated in a variety of organs
including the lungs (84–86), diaphragm
(87), kidneys (86), intestine (88, 89), and
liver (86). Apoptosis is a counter-regula-
tor of the initial inflammatory response
in sepsis (90). Neutrophils are constitu-
tively proapoptotic and apoptosis is fun-
damental for the resolution of inflamma-
tion and cell turnover. Neutrophils can
undergo apoptosis via intrinsic and ex-
trinsic pathways; the latter also requires
mitochondrial amplification (Fig. 4) (91).
The role played by mitochondria in the
regulation of neutrophil life span is more
crucial than in other cell types in the
body (92). As neutrophils kill pathogens
using ROS and RNS and a mixture of lytic
enzymes, delayed clearance of neutro-
phils in sepsis can potentially contribute
Figure 4. Summary of apoptotic signaling pathways as seen through activation of death receptor
(extrinsic) or mitochondrial (intrinsic) pathway. Extrinsic signals bind to their receptors and trigger
intracellular signaling, leading to caspase-8 activation. Activation of caspase-8 by extrinsic stimuli
(such as tumor necrosis factor-␣ [TNF-␣], Fas ligand) involves mitochondria-dependent signaling in
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
3-kinase/Akt; APAF-1, apoptosis protease activating factor 1; ER, endoplasmic reticulum; IL, interleukin.
Figure 5. The recognition of apoptotic cells by macrophages is largely dependent on the cell surface
appearance of phosphatidylserine (PS). S-nitrosylation of critical cysteine residues inhibits aminophospho-
lipid translocase (APLT), leading to PS externalization. It generates an “eat me” signal during apoptosis.
iNOS, inducible nitric oxide synthase; .
NO, nitric oxide; O2, superoxide; ONOO؊
, peroxynitrite.
295Crit Care Med 2009 Vol. 37, No. 1
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-
cytosis of apoptotic neutrophils by
macrophages inhibits the release of
proinflammatory cytokines and promotes
the secretion of anti-inflammatory cyto-
kines (95). In contrast, inefficient apopto-
tic cell clearance is proinflammatory and
immunogenic (96). The recognition of
apoptotic cells by macrophages is largely
dependent on the cell surface appearance
of an anionic phospholipid, phosphatidyl-
serine (PS), which is normally confined
to the inner leaflet of the plasma mem-
brane (97). Asymmetric distribution of
PS across the plasma membrane is
mainly because of the activity of a spe-
cialized enzymatic mechanism, amin-
ophospholipid translocase. S-nitrosyla-
tion of critical cysteine residues inhibits
aminophospholipid translocase, leading
to PS externalization. PS expression dur-
ing apoptosis generates an “eat-me” sig-
nal (Fig. 5), which in turn triggers clear-
ance of apoptotic cells and suppresses the
inflammatory response (98). It has been
demonstrated that S-nitrosylation of crit-
ical cysteine residues in aminophospho-
lipid translocase using a cell-permeable
transnitrosylating agent, S-nitroso-acetyl-
cysteine, resulted in egression of PS to
the outer surface of the plasma mem-
brane, rendering these cells recognizable
by macrophages (99). The therapeutic po-
tential of regulating neutrophil life-span
in sepsis remains to be determined. Care
must be exercised in regulating of this
pathway, as sepsis enhances the capacity
of macrophages to clear expanded apo-
ptotic populations, a mechanism contrib-
uting to septic immune suppression
(100).
The Role of ROS and RNS. ROS and
RNS exert several beneficial physiologic
functions, such as intracellular signaling
for several cytokines and growth factors,
second messengers for hormones and re-
dox regulation. Despite their importance
as a defense mechanism against invading
pathogens, an overwhelming production
of ROS and RNS or a deficit in oxidant
scavenger and antioxidant defenses result
in oxidative/nitrosative stress, a key ele-
ment in the deleterious processes in sep-
sis (Fig. 6) (101, 102).
Stimulated neutrophils produce ROS
and RNS through the nicotinamide ade-
nine dinucleotide phosphate oxidase
complex, myeloperoxidase and xanthine
oxidoreductase and represent a defense
mechanism against invading microor-
ganisms (103). Lipopolysaccharide and
other proinflammatory mediators acti-
vate nicotinamide adenine dinucleotide
phosphate oxidase to produce superoxide
radical (O2
Ϫ
). In aqueous environments,
superoxide radical is rapidly catalyzed by
superoxide dismutase hydrogen peroxide
(H2O2) and hydroxyl radicals. Myeloper-
oxidase from neutrophil azurophilic
granules produces hypochlorous acid
from hydrogen peroxide (H2O2) and chlo-
ride anion (Cl؊
) during respiratory burst.
These radicals are highly cytotoxic, and
neutrophils used them to kill bacteria
and other pathogens. Expression of hu-
man xanthine oxidoreductase is markedly
up-regulated by hypoxia, ischemia/reper-
fusion, LPS, and TNF-␣. Increased activ-
ity of xanthine oxide, one the important
contributors of ROS production, has been
reported in adult and pediatric patients
with sepsis (104).
O2
؊
in the presence of nitric oxide,
generates peroxynitrite (ONOOϪ
), a key
player in the pathogenesis of sepsis-
induced organ dysfunction. ONOOϪ
can
cause DNA strand breakage, which trig-
gers the activation of DNA repair en-
zymes such as poly (adenine dinucleotide
phosphate-ribose) polymerase (Fig. 3).
Poly (adenine dinucleotide phosphate-
ribose) polymerase inhibitors protect
against oxidative and nitrosative stress-
induced organ dysfunction in endotox-
emia (87–89). Recently, the potential role
of poly (adenine dinucleotide phosphate-
ribose) polymerase activation has been
implicated in the pathogenesis of myo-
cardial contractile dysfunction associated
with human septic shock (105).
Novel Cytokines in Inflammation
High Mobility Group Box-1 and Re-
ceptor for Advanced Glycation End-
Products. High mobility group box-1
(HMGB-1) is a nonhistone, nuclear DNA-
binding protein involved in nucleosome
stabilization and gene transcription. How-
ever, when HMGB-1 is released in large
quantities into the extracellular environ-
ment, it becomes a lethal mediator of sys-
temic inflammation (Fig. 3) (106). Indeed,
Figure 6. Proinflammatory/anti-inflammatory cytokines and reactive oxygen and nitrogen species have
important effects within the microcirculatory unit: the arteriole, endothelial cell, capillary bed, and the
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
thromboses are formed. The postcapillary venule is where leukocyte trafficking is most disordered. All of
these causes impair flow through the microcirculation leading to microcirculatory dysfunction. TLR,
toll-like receptor; NOD-LRR, nucleotide-oligomerization domain leucine-rich repeat; RLH, (RIG-I)-like
helicase; VSM, vascular smooth muscle; IL, interleukin; TGF, transforming growth factor; MIF, migration
inhibitory factor; TNF, tumor necrosis factor; HMGB, high mobility group box; PAF, protease activating
factor; sTNFR, soluble tumor necrosis factor receptor.
296 Crit Care Med 2009 Vol. 37, No. 1
it has been recently shown that it has a
weak proinflammatory activity by itself and
binding to bacterial substances including
lipid molecules such as phophatidylserine
strengthens its effects (107). Interestingly,
phosphatidylserine has been implicated in
the regulation of inflammation and
HMGB-1 might thus regulate its anti-
inflammatory activities (108).
HMGB-1 is released into the extracel-
lular space through acetylation or phos-
phorylation (109, 110). HMGB-1 is either
“passively released” from necrotic cells,
and a mechanism that represents a pro-
cess adopted by the innate immune sys-
tem to recognize damaged and necrotic
cells, or “actively secreted” by immune
cells including macrophages and neutro-
phils to trigger inflammation (106). Re-
cently, HMGB-1 release from macro-
phages has been shown during the course
of apoptosis as well as necrosis and de-
fined as a downstream event of cell apo-
ptosis during severe sepsis (111, 112).
After treatment with LPS or various cy-
tokines such as TNF-␣, IL-1␤, or IFN-␥,
HMGB-1 is released from activated mac-
rophages within 4 hrs and reaches a pla-
teau around 18–24 hrs. It binds to several
transmembrane receptors such as recep-
tor for advanced glycation end products
(RAGE), TLR-2, and -4, activating NF-␬B
and extracellular regulated kinase 1/2
(113, 114).
Although HMGB-1 was originally de-
scribed as a late mediator of endotoxin-
induced lethality (115), recent studies in-
dicate a role for HMGB-1 in angiogenesis,
tissue repair, and regeneration (116).
HMGB-1 appears to be a novel myocardial
depressant factor upon release by resi-
dent myocardial cells following tissue in-
jury. HMGB-1 might decrease energy uti-
lization in ischemic tissue, thereby
preventing injured myocytes from wors-
ening ATP depletion that eventuate in
necrosis (117). On other hand, excessive
release of HMGB-1 in sepsis might con-
tribute to sustained inflammation and to
profound myocardial depression.
The cholinergic anti-inflammatory
pathway is a neural mechanism that in-
hibits the expression of HMGB-1 and
other cytokines (118–121). Signals trans-
mitted via the vagus nerve, the principal
nerve of the parasympathetic nervous
system, significantly attenuate the re-
lease of HMGB-1 and other cytokines in
inflammation in animal and human stud-
ies (122).
The remarkable binding characteristics
of HMGB-1 suggest another important role
for this protein in the extracellular fluid.
HMGB-1 might serve as a shuttle platform
for LPS and other microbial mediators for
docking to CD14 and recognition by the
TLRs (108, 123) or through binding to
host-derived proinflammatory mediators,
such as IL-1beta (124).
Elevated levels of HMGB-1 are measur-
able in the majority of patients up to 1 wk
after the diagnosis of sepsis or septic shock
and are correlated with the degree of organ
dysfunction (125, 126). However, serum
HMGB-1 levels do not consistently identify
nonsurvivors from survivors as a predictor
of hospital mortality (127). As HMGB-1 is
late inflammatory cytokine of sepsis, it pro-
vides a wide therapeutic time window for
clinical intervention and remains an attrac-
tive target for sepsis treatment.
RAGE, a member of the immunoglobu-
lin superfamily, is a pattern-recognition re-
ceptor that binds diverse classes of endog-
enous molecules including HMGB-1. It has
been defined as part of a newly appreciated
component of the innate immune system
referred to as the danger associated molec-
ular pattern system (Fig. 3) (128). Mem-
brane bound and soluble forms of RAGE
(sRAGE) have been detected in plasma. Sol-
uble RAGE, anti-RAGE antibody (Fab 2
fragment) or data from RAGEϪ/Ϫ animals
have been shown to decrease inflammation,
reduce neutrophil extravasation, and re-
duce migration (129). Recently, the dem-
onstration of survival benefit after delayed
administration of anti-RAGE antibody in a
murine model of polymicrobial sepsis, even
when delayed up to 24 hrs, provides a ther-
apeutic rationale for the use of anti-RAGE
mAb as a salvage therapy for established
severe sepsis (130).
Macrophage Migration Inhibitory
Factor. Migration inhibitory factor (MIF)
acts as a stress response mediator and
proinflammatory cytokine upon induc-
tion by glucocorticoids (131). This pro-
tein is readily measurable in patients with
sepsis, and MIF probably contributes to
the pathogenesis of sepsis. Inhibition of
MIF or its targeted deletion attenuates
TNF-␣ and IL-1␤ expression and protects
mice from lethality is experimental sepsis
(132, 133). Systemic challenge of animals
with MIF increases LPS-related lethality
(133). MIF promotes the expression of
TLR4 on macrophages, and thereby sen-
sitizes these immune effector cells to LPS
(134).
The immunoregulatory effects of MIF
might be crucial to the control and resolu-
tion of the inflammatory response as a con-
sequence of its ability to regulate activa-
tion-induced apoptosis (135). High MIF
levels delay the removal of activated mono-
cytes/macrophages by apoptosis. This pro-
longs monocyte/macrophage survival, in-
creases cytokine production, and sustains
an ongoing proinflammatory response.
Mitochondrial Dysfunction in
Inflammation
Although microvascular flow abnormal-
ities occur, findings of decreased oxygen
consumption (136) and elevated tissue ox-
ygen tension (137), yet minimal cell death
despite functional and biochemical de-
rangements (138), suggest that the prob-
lem lies more in cellular oxygen utilization
rather than a problem with oxygen delivery
(139). It is postulated that prolonged and
systemic inflammatory insult is accompa-
nied by a basic tissue survival response me-
diated by switching off its energy-consum-
ing biophysiological processes. Recent
evidence suggests that sepsis and septic
shock severely impair the mitochondria
(140, 141), and the severity and outcome of
organ dysfunction could be related to mi-
tochondrial dysfunction (142). Depleted
levels of reduced glutathione, an important
intramitochondrial antioxidant, in combi-
nation with excess generation of ROS and
RNS severely inhibit oxidative phosphory-
lation and ATP generation (142). This ac-
quired intrinsic derangement in cellular
energy metabolism which has also been
termed “cytopathic hypoxia,” contributes to
reduced activities of mitochondrial electron
transport chain enzyme complexes and im-
paired ATP biosynthesis, potentially organ
dysfunction in sepsis (141, 143, 144).
Sepsis-related derangements in mito-
chondrial function can activate the ubiq-
uitin proteolytic pathway in skeletal mus-
cle of septic patients (145). Mitochondrial
permeability transition pore seems to be
involved in sepsis-induced mitochondrial
damage, since its inhibition significantly
improved organ function and reduced
mortality in rodents (146). Whether sep-
sis-related reduction in energy supply
could result in a state of cellular shut-
down analogous to myocardial “stun-
ning” (or hibernation) following coronary
occlusion, allowing for eventual restora-
tion of organ function and survival, has
not yet been determined.
Impact of Inflammation on
Coagulation
The clotting system is almost invari-
ably activated by systemic microbial in-
297Crit Care Med 2009 Vol. 37, No. 1
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-
ptotic microparticles have also been dem-
onstrated in sepsis (153–155). Linkage of
coagulation enzymes with their serine
protease activity with protease activated
receptors (PAR 1-4) on endothelial sur-
faces increases P-selectin, cytokine produc-
tion, and adhesion molecule expression,
leading to microcirculatory dysfunction in
severe sepsis (156, 157).
Activated Protein C. Activated protein
C (APC) is derived from its zymogen pro-
tein C in contact with thrombin: thrombo-
modulin complexes on endothelial sur-
faces. Protein C was originally thought to
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-
brinolytic activities (Fig. 8) (167, 168).
Although an anti-inflammatory role for
APC may be an indirect consequence of
its ability to reduce thrombin generation,
APC also has direct anti-inflammatory
Figure 7. The extrinsic pathway (tissue factor pathway) is the primary mechanism by which thrombin is
generated in sepsis. The intrinsic cascade (contact factor pathway) primarily serves an accessory role in
amplifying the prothrombotic events that are initiated in sepsis. Thrombin, factor Xa and the TF-factor VIIa
complex interact with the PAR (protease activated receptors) system and directly activate endothelial cells,
platelets and white blood cells, and induce a proinflammatory response. Platelet-derived microparticles
(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
insult. When bearing appropriate counter-ligands, MPs can transfer their procoagulant potential to target
cells. Platelet-derived MPs can bind to soluble and immobilized fibrinogen, thus delivering procoagulant
entities to the thrombus via the formation of aggregates. In vitro, interaction between endothelial MPs and
monocytes promotes TF mRNA expression and TF-dependent procoagulant activity. Activated platelets and
platelet-derived MPs thus amplify leukocyte-mediated tissue injury in thrombotic and inflammatory
disorders. EPCR, endothelial protein C receptor; PMN, polymorphonuclear leukocyte; APC, activated
protein C; ADAMST13, a disintegrin and metalloproteinase with a thrombospondin type 1 motifs 13;
ULVWF, unusually large von Willebrand factor; ICAM, intracellular adhesion molecule; VCAM, vascular cell
adhesion molecule-1; IL, interleukin; TNF, tumor necrosis factor; IFN, interferon.
Figure 8. The homeostatic balance between thrombin and activated protein C (APC) in coagulation and
inflammation is given. APC is a central endogenous anticoagulant protein with antithrombotic, anti-
inflammatory, anti-apoptotic and pro-fibrinolytic activities. The ligand occupancy of endothelial protein C
receptor (EPCR) switches the protease-activated receptor 1 (PAR-1)-dependent signaling specificity of thrombin
from a permeability-enhancing to a barrier-protective response. Other pleiotropic effects are also explained. TAFI,
thrombin activatable fibrinolysis inhibitor; NF-␬␤, nuclear factor ␬␤.
298 Crit Care Med 2009 Vol. 37, No. 1
properties (169). APC can cleave and ac-
tivate PAR1-dependent cellular pathways
(170). APC competes for PAR-1 binding
with thrombin, but in vitro studies sug-
gest that APC is 103
- to 104
-fold less po-
tent that thrombin in cleaving PAR-1.
The critical receptors required for both
PC activation and APC cellular signaling
(i.e., thrombomodulin, EPCR and PAR-1)
are co-localized in lipid rafts on endothe-
lial cells (171). EPCR is associated with
caveolin-1 on lipid rafts and EPCR bind-
ing to the gamma-carboxyglutamic acid
domain of protein C/APC leads to its dis-
sociation from caveolin-1 (Fig. 7). APC
then engages PAR-1 generating a protec-
tive signaling pathway through coupling
of PAR-1 to the pertussis toxin-sensitive
G(i)-protein. Thus, when EPCR is bound
by protein C, the PAR-1-dependent pro-
tective signaling responses in endothelial
cells can be mediated by either thrombin
or APC. These results explain how PAR-1
and EPCR participate in protective sig-
naling events in endothelial cells (172).
Therapeutic administration of recom-
binant human APC (rhAPC) is currently
in use as a treatment strategy for severe
sepsis patients with a high risk of death
(173, 174). Genetically engineered vari-
ants of APC have been designed with
greater antiapoptotic activity and reduced
anticoagulant activity relative to wild-
type APC to increase the risk/benefit ratio
of rhAPC regarding the bleeding compli-
cation (175). A nonanticoagulant from of
APC reduces mortality in experimental
models of endotoxemia and sepsis (176).
von Willebrand Factor, A Disintegrin-Like
and Metalloproteinase with Thrombospondin
Type-1 Motifs 13 and Ashwell Receptor.
The discovery of a disintegrin-like and met-
alloproteinase with thrombospondin type-1
motifs 13 (ADAMTS-13) has provided new
insights in pathogenesis of thrombosis in
sepsis. ADAMTS-13, the principal physio-
logic modulator of von Willebrand factor
(VWF) is produced mainly stellate cells in
the liver (177). VWF is synthesized in vas-
cular endothelial cells and released into the
plasma as unusually large VWF multimers
which are rapidly degraded into smaller
VWF multimers by ADAMTS-13. Deficiency
of the ADAMTS-13, as observed in most
forms of thrombotic thrombocytopenic
purpura, increases the level of unusually
large VWF multimers in plasma and leads
to platelet aggregation and/or thrombus
formation, especially in small arterioles, re-
sulting in microvascular failure (178–180).
Recently, inflammation associated-
ADAMTS-13 deficiency has been de-
scribed in patients with systemic inflam-
matory response syndrome and severe
sepsis (Fig. 7) (181–185). Decreased lev-
els of ADAMTS-13 have been reported in
healthy volunteers following endotoxin
infusion (186). Furthermore, reduced
ADAMTS-13 levels are associated with dif-
ferences in morbidity, mortality, and
variables of inflammation and endothelial
dysregulation in severe sepsis patients
(182, 187).
The Ashwell receptor, which is the
major lectin of hepatocytes, modulates
VWF homeostasis. It has been recently
demonstrated that the marked thrombo-
cytopenia associated with S. pneumoniae
sepsis is the result of Ashwell receptor-
dependent clearance of platelets (188).
The ensuing reduction in platelet counts
at the onset of sepsis protects the host
against the development of disseminated
intravascular coagulation. Homeostatic
adaptation by this receptor moderates the
onset and severity of disseminated intra-
vascular coagulation during sepsis sug-
gesting the improvement in host survival
probability (189). At present, it remains
unclear whether blocking the Ashwell re-
ceptor may have a beneficial effect in
severe sepsis.
CONCLUSION
A unifying concept of innate immu-
nity is based upon pathogen-, or damage-
associated molecular pattern molecules
and downstream signaling pathways. This
has facilitated significant advances in our
understanding of the pathophysiology of
sepsis and led to a multitude of clinical
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 Cϩ Erythropoietin Coagulation and inflammation LHRI III Recruiting
Recombinant activated Protein C Coagulation Lilly III Recruiting
Recombinant plasma gelsolin Replacement of circulating
actin-binding protein
CBC II Recruiting
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
Chicago, USA
IV Recruiting
Rosuvastatin Pleitrophic effects UASLP, Mexico and Beth
Israel
II Ongoing
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.
299Crit Care Med 2009 Vol. 37, No. 1
trials (Table 1). However, further identi-
fication of the critical elements in severe
sepsis that drives the transition from lo-
calized inflammation to deleterious host
response remains incompletely illumi-
nated. The limitations in our current
knowledge of the molecular mechanisms
in sepsis have made the design of inter-
vention trials in clinical sepsis challeng-
ing (190). New findings as to biomarkers
and measures to detect genetic signa-
tures of sepsis are now making their way
into clinical trial designs. It is anticipated
that such innovations will improve the
outlook for successful development of
new sepsis treatments tailored to individ-
ual patient needs.
REFERENCES
1. van der Poll T, Opal SM: Host-pathogen
interactions in sepsis. Lancet Infect Dis
2008; 8:32–43
2. Cinel I, Dellinger RP: Making a difference in
outcome in severe sepsis: The organism, the
host response and the treating clinician.
Emerg Med Crit Care Rev 2007; 1:1–3
3. Cinel I, Dellinger RP: Advances in patho-
genesis and management of sepsis. Curr
Opin Infect Dis 2007; 20:345–352
4. Uematsu S, Akira S: Toll-like receptors and
innate immunity. J Mol Med 2007; 84:
712–725
5. Creagh EM, O’Neill LA: TLRs, NLRs and
RLRs: A trinity of pathogen sensors that
co-operate in innate immunity. Trends
Immunol 2006; 27:352–357
6. Granucci F, Foti M, Ricciardi-Castagnoli P:
Dendritic cell biology. Adv Immunol 2005;
88:193–233
7. Liew FY, Xu D, Brint EK, et al: Negative
regulation of toll-like receptor-mediated
immune responses. Nat Rev Immunol 2005;
5:446–458
8. Akira S, Uematsu S, Takeuchi O: Pathogen
recognition and innate immunity. Cell
2006; 124:783–801
9. Bianchi ME: DAMPs PAM. Ps and alarmins:
All we need to know about danger. J Leukoc
Biol 2007; 81:1–5
10. Mollen KP, Anand RJ, Tsung A, et al:
Emerging paradigm: Toll-like receptor
4-sentinel for the detection of tissue dam-
age. Shock 2006; 26:430–437
11. Tsujimoto H, Ono S, Hiraki S, et al: He-
moperfusion with polymyxin B-immobi-
lized fibers reduced the number of CD16ϩ
CD14ϩ monocytes in patients with septic
shock. J Endotoxin Res 2004; 10:229–237
12. Armstrong L, Medford AR, Hunter KJ, et al:
Differential expression of toll-like receptor
(TLR)-2 and TLR-4 on monocytes in human
sepsis. Clin Exp Immunol 2004; 136:
312–319
13. Tsujimoto H, Ono S, Majima T, et al: Neu-
trophil elastase, MIP-2, and TLR-4 expres-
sion during human and experimental sep-
sis. Shock 2005; 23:39–44
14. Williams DL, Ha T, Li C, et al: Modulation of
tissue toll-like receptor 2 and 4 during the
early phases of polymicrobial sepsis corre-
lates with mortality. Crit Care Med 2003;
31:1808–1818
15. Paterson HM, Murphy TJ, Purcell EJ, et al:
Injury primes the innate immune system
for enhanced toll-like receptor reactivity.
J Immunol 2003; 171:1473–1483
16. Tsujimoto H, Ono S, Efron PA, et al: Role of
toll like receptors in the development of
sepsis. Shock 2008; 29:315–321
17. Ogura Y, Sutterwala FS, Flavell RA: The
inflammasome: First line of the immune
response to cell stress. Cell 2006; 126:
659–662
18. Drenth JP, van der Meer JW: The inflamma-
some—A linebacker of innate defense.
N Engl J Med 2006; 355:730–732
19. Dinarello CA: Interleukin-1␤, interleukin-
18, and the interleukin-1␤ converting en-
zyme. Ann NY Acad Sci 1998; 856:1–11
20. Martinon F, Burns K, Tschopp J: The in-
flammasome. A molecular platform trigger-
ing activation of inflammatory caspases and
processing of proIL-1␤. MolCell 2002; 10:
417–426
21. Yu JW, Wu J, Zhang Z, et al: Cryopyrin and
pyrin activate caspase-1, but not NF-
kappaB, via ASC oligomerization. Cell
Death Differ 2005; 13:236–249
22. Seshadri S, Duncan MD, Hart JM, et al:
Pyrin levels in human monocytes and
monocyte-derived macrophages regulate
IL-1␤ processing and release. J Immunol
2007; 179:1274–1281
23. Tschopp J, Martinon F, Burns K: NALPs: A
novel protein family involved in inflamma-
tion. NatRevMol Cell Biol 2003; 4:95–104
24. Sarkar A, Duncan M, Hart J, et al: ASC
directs NF-␬B activation by regulating re-
ceptor interacting protein-2 (RIP2)
caspase-1 interactions. J Immunol 2006;
176:4979–4986
25. Scott AM, Saleh M: The inflammatory
caspases: Guardians against infections and
sepsis. Cell Death Differ 2007; 14:23–31
26. Sarkar A, Hall MW, Exline M, et al:
Caspase-1 regulates E. coli sepsis and
splenic B cell apoptosis independently of
IL-1␤ and IL-18. Am J Respir Crit Care Med
2006; 174:1003–1010
27. Saleh M, Vaillancourt JP, Graham RK, et al:
Differential modulation of endotoxin re-
sponsiveness by human caspase-12 poly-
morphisms. Nature 2004; 429:75–79
28. Saleh M, Mathison JC, Wolinski MK, et al:
Enhanced bacterial clearance and sepsis re-
sistance in caspase-12-deficient mice. Na-
ture 2006; 440:1064–1068
29. Joshi VD, Kalvakolanu DV, Hasday JD, et al:
IL-18 levels and the outcome of innate im-
mune response to lipopolysaccharide: Im-
portance of a positive feedback loop with
caspase-1 in IL-18 expression. J Immunol
2002; 169:2536–2544
30. Fahy RJ, Exline MC, Gavrilin MA, et al:
Inflammasome mRNA expression in human
monocytes during early septic shock. Am J
Resp Crit Care Med 2008; 177:983–988.
31. Lara-Tejero M, Sutterwala FS, Ogura Y, et
al: Role of the caspase-1 inflammasome in
Salmonella typhimurium pathogenesis.
J Exp Med 2006; 203:1407–1412
32. Brightbill HD, Libraty DH, Krutzik SR, et
al: Host defense mechanisms triggered by
microbial lipoproteins through toll-like re-
ceptors. Science 1999; 285:732–736
33. Bruno TF, Woods DE, Mody CH: Exoen-
zyme S from Pseudomonas aeruginosa in-
duces apoptosis in T lymphocytes. J Leukoc
Biol 2000; 67:808–816
34. Wesche H, Henzel WJ, Shillinglaw W, et al:
MyD88: An adapter that recruits IRAK to
the IL-1 receptor complex. Immunity 1997;
7:837–847
35. Reynaert NL, van der Vliet A, Guala AS, et
al: Dynamic redox control of NF-kappaB
through glutaredoxin-regulated S-gluta-
thionylation of inhibitory kappaB kinase
beta. Proc Natl Acad Sci USA 2006; 103:
13086–13091
36. Pahl HL: Activators and target genes of rel/
NF-kappaB transcription factors. Oncogene
1999; 18:6853–6866
37. Blackwell TS, Yull FE, Chen CL, et al: Mul-
tiorgan nuclear factor kappa B activation in
a transgenic mouse model of systemic in-
flammation. Am J Respir Crit Care Med
2000; 162:1095–1101
38. Arcaroli J, Silva E, Maloney JP, et al: Variant
IRAK-1 haplotype is associated with in-
creased nuclear factor-kappaB activation
and worse outcomes in sepsis. Am J Respir
Crit Care Med 2006; 173:1335–1341
39. Zaph C, Troy AE, Taylor BC, et al: Epithelial
cell-intrinsic IKK-beta expression regulates
intestinal immune homeostasis. Nature
2007; 446:552–556
40. Liu YJ, Soumelis V, Watanabe N, et al:
TSLP: An epithelial cell cytokine that regu-
lates T cell differentiation by conditioning
dendritic cell maturation. Annu Rev Immu-
nol 2007; 25:193–219
41. Peck-Palmer OM, Unsinger J, Chang KC, et
al: Deletion of MyD88 markedly attenuates
sepsis-induced T and B lymphocyte apopto-
sis but worsens survival. J Leukoc Biol
2008; 83:1009–1018
42. Cantley LC. The phosphoinositide 3-kinase
pathway. Science 2002; 296:1655–1657
43. Okugawa S, Ota Y, Kitazawa T, et al: Janus
kinase2 is involved in lipopolysaccharide-
induced activation of macrophages. Am J
Physiol Cell Physiol 2003; 285:C399–C408
44. Ojaniemi M, Glumoff V, Harju K, et al:
Phosphatidylinositol kinase is involved in
Toll-like receptor 4-mediated cytokine ex-
pression in mouse macrophages. Eur J Im-
munol 2003; 335:97–605
45. Guillot L, Le GofficR, BlochS, et al: Involve-
ment of toll-like receptor 3 in the immune
response of lung epithelial cells to double-
300 Crit Care Med 2009 Vol. 37, No. 1
stranded RNA and influenza A virus. J Biol
Chem 2005; 280:5571–5580
46. Wrann CD, Tabriz NA, Barkhausen T, et al:
The phosphatidylinositol 3-kinase signaling
pathway exerts protective effects during
sepsis by controlling C5a-mediated activa-
tion of innate immune functions. J Immu-
nol 2007; 178:5940–5948
47. Guha M, Mackman N: The PI3K-Akt path-
way limits LPS activation of signaling path-
ways and expression of inflammatory medi-
ators in human monocytic cells. J Biol
Chem 2002; 277:32124–32132
48. Schabbaue G, Tencati M, Pedersen B, et al:
PI3K-Akt pathway suppresses coagulation
and inflammation in endotoxemic mice. Ar-
terioscler Thromb Vasc Biol 2004; 24:
1963–1969
49. Pengal RA, Ganesan LP, Wei G, et al: Li-
popolysaccharide-induced production of in-
terleukin-10 is promoted by the serin thre-
onine kinase Akt. Mol Immunol 2006; 43:
1557–1156
50. Fukao T, Koyasu S: PI3K and negative reg-
ulation of TLR signaling. Trends Immunol
2003; 24:358–363
51. Bommhardt U, Chang KC, Swanson PE, et
al: Akt decreases lymphocyte apoptosis and
improves survival in sepsis. J Immunol
2004; 172:7583–7591
52. Mc Dunn JE, Muenzer JT, Rachdi L, et al:
Peptide-mediated activation of Akt and ex-
tracellular regulated kinase signaling pre-
vents lymphocyte apoptosis. FASEB J 2008;
22:561–568
53. Hall A: Rho GT. Pases and the actin cy-
toskeleton. Science 1998; 279:509–514
54. Ruse M, Knaus UG: New players in TLR-
mediated innate immunity. Immunologic
Res 2006; 34:33–48
55. Arbibe L, Mira JP, Teusch N, et al: Toll-like
receptor 2-mediated NF-kappa B activation
requires a Racl dependenpt pathway. Nat
Immunol 2000; 1:533–540
56. Honing H, van den Berg TK, van der Pol
SM, et al: Rho activation promotes transen-
dothelial migration of monocytes via
ROCK. J Leukoc Biol 2004; 75:523–528
57. Tasaka S, Koh H, Yamada W, et al: Attenu-
ation of endotoxin-induced acute lung in-
jury by the Rho-associated kinase inhibitor,
Y-27632. Am J Respir Cell Mol Biol 2005;
32:504–510
58. Thorlacius K, Slotta JE, Laschke MW, et al:
Protective effect of fasudil, a Rho-kinase
inhibitor, on chemokine expression, leuko-
cyte recruitment, and hepatocellular apo-
ptosis in septic liver injury. J Leukoc Biol
2006; 79:923–931
59. Hori S, Nomura T, Sakaguchi S: Control of
regulatory T cell development by the tran-
scription factor FoxP3. Science 2003; 299:
1057–1061
60. Zheng SG, Wang JH, Koss MN, et al: CD4ϩ
and CD8ϩ regulatory T cells generated ex
vivo with IL-2 and TGF-beta suppress a
stimulatory Graft-versus- Host disease with
a Lupus-like syndrome. J Immunol 2004;
172:1531–1539
61. Ausubel FM: Are innate immune signaling
pathways in plants and animals conserved?
Nat Immunol 2005; 6:973–979
62. Caramalho I, Lopes-Carvalho T, Ostler D, et
al: Regulatory T cells selectively express
toll-like receptors and are activated by lipo-
polysaccharide. J Exp Med 2003; 197:
403–411
63. Sutmuller RPM, den Brok MH, Kramer M,
et al: Toll-like receptor 2 controls expansion
and function of regulatory T cells. J Clin
Invest 2006; 116:485–494
64. Liu H, Komai-Koma M, Xu D, et al: Toll-like
receptor 2 signaling modulates the func-
tions of CD4ϩCD25ϩ regulatory T cells.
Proc Natl Acad Sci USA 2006; 103:
7048–7053
65. McHugh RS, Shevach EM: The role of sup-
pressor T cells in regulation of immune
responses. J Allergy Clin Immunol 2002;
110:693–702
66. Murphy TJ, Choileain NN, Zang Y, et al:
CD4ϩ
CD25ϩ
regulatory T cells control in-
nate immune reactivity after injury. J Im-
munol 2005; 174:2957–2963
67. Monneret G, Debard AL, Venet F, et al:
Marked elevation of human circulating
CD4ϩCD25ϩ regulatory T cells in sepsis-
induced immunoparalysis. Crit Care Med
2003; 31:2068–2071
68. Venet F, Pachot A, Debard AL, et al: In-
creased percentage of CD4ϩCD25ϩ regula-
tory T cells during septic shock is due to the
decrease of CD4ϩCD25-lymphocytes. Crit
Care Med 2004; 32:2329–2331
69. Takahashi H, Tsuda Y, Takeuchi D, et al:
Influence of systemic inflammatory re-
sponse syndrome on host resistance against
bacterial infections. Crit Care Med 2004;
32:1879–1885
70. Tiemessen MM, Jagger AL, Evans HG, et al:
CD4ϩCD25ϩFoxp3ϩ regulatory T cells in-
duce alternative activation of human mono-
cytes/macrophages. Proc Natl Acad Sci USA
2007; 104:19446–19451
71. Wisnoski N, Chung CS, Chen Y, et al: The
contribution of CD4ϩ CD25ϩ T-regulato-
ry-cells to immune suppression in sepsis.
Shock 2007; 27:251–257
72. van Maren WW, Jacobs JF, de Vries IJ, et al:
Toll-like receptor signalling on Tregs: To
suppress or not to suppress? Immunology
2008; 124:445–452.
73. Heuer JG, Zhang T, Zhao J, et al: Adoptive
transfer of in vitro-stimulated CD4ϩ
CD25ϩ
regulatory T cells increases bacterial clear-
ance and improves survival in polymicrobial
sepsis. J Immunol 2005; 174:7141–7146
74. Venet F, Pachot A, Debard AL, et al: Human
CD4ϩCD25ϩ regulatory T lymphocytes in-
hibit lipopolysaccharide-induced monocyte
survival through a Fas/Fas ligand-depen-
dent mechanism. J Immunol 2006; 177:
6540–6547
75. Brown KA, Brain SD, Pearson JD, et al:
Neutrophils in development of multiple or-
gan failure in sepsis. Lancet 2006; 368:
157–169
76. Marshal JC: The pathogenesis and molecu-
lar biology of sepsis. Crit Care Resusc 2006;
8:227–229
77. Chandra A, Enkhbaatar P, Nakano Y, et al:
Sepsis: Emerging role of nitric oxide and
selectins. Clinics 2006; 61:71–76
78. Hu G, Predescu D, Vogel SM: ICAM-1 de-
pendent neutrophil adhesion to endothelial
cells increases caveolae-mediated pulmo-
nary vascular albumin permeability. Crit
Care Med 2007; 34 (Suppl 12): A31
79. Garrean S, Gao X-P, Brovkovych V, et al:
Caveolin-1 regulates NF-kB activation and
lung inflammatory response to sepsis in-
duced by lipopolysaccharide. J Immunol
2006 177: 4853–4860
80. Wang XM, Kim HP, Song R, et al: Caveo-
lin-1 confers antiinflammatory effects in
murine macrophages via the MKK3/p38
MAPK pathway. Am J Respir Cell Mol Biol
2006; 34:434–442
81. Medina FA, de Almeida CJ, Dew E, et al:
Caveolin-1 deficient mice show defects in
innate immunity and inflammatory im-
mune response during Salmonella enterica
serovar typhimurium infection. Infect Im-
mun 2006; 74:6665–6674
82. Medina FA, Cohen AW, de Almeida CJ, et al:
Immune dysfunction in caveolin-1 null
mice following infection with Trypanosoma
cruzi (Tulahuen strain). Microbes Infect
2007; 9:325–333
83. Astiz ME, DeGent GE, Lin RY, et al: Micro-
vascular function and rheologic changes in
hyperdynamic sepsis. Crit Care Med 1995;
23:265–271
84. Ozdulger A, Cinel I, Koksel O, et al: The
protective effect of N-acetylcysteine on ap-
optotic lung injury in cecal ligation and
puncture-induced sepsis model. Shock
2003; 19:366–372
85. Stehr SN, Knels L, Weissflog C, et al: Effects
of IgM enriched solution on polymorpho-
nuclear neutrophil function, bacterial
clearance, and lung histology in endotox-
emia. Shock 2007; 29:167–172
86. Kabay B, Kocaefe C, Baykal A, et al: Inter-
leukin-10 gene transfer: Prevention of mul-
tiple organ injury in a murine cecal ligation
and puncture model of sepsis. World J Surg
2007; 31:105–115
87. Ozdulger A, Cinel I, Unlu A, et al: Poly
(ADP-ribose) synthetase inhibition prevents
lipopolysaccharide-induced peroxynitrite
mediated damage in diaphragm. Pharmacol
Res 2002; 46:67–73
88. Taner S, Cinel I, Ozer L, et al: Poly (ADP-
ribose) synthetase inhibition reduces bacte-
rial translocation in rats after endotoxin
challenge. Shock 2001; 16:159–162
89. Cinel I, Buyukafsar K, Cinel L, et al: The
role of poly (ADP-ribose) synthetase inhibi-
tion in preventing endotoxemia-induced in-
testinal epithelial apoptosis. Pharmacol Res
2002; 46:119–127
301Crit Care Med 2009 Vol. 37, No. 1
90. Savill J: Apoptosis in resolution of inflam-
mation. J Leukoc Biol 1997; 61:375–380
91. Maianski NA, Geissler J, Srinivasula SM, et
al: Functional characterization of mito-
chondria in neutrophils: A role restricted to
apoptosis. Cell Death Differ 2004; 11:
143–153
92. van Raam BJ, Verhoeven AJ, Kuijpers TW:
Mitochondria in neutrophil apoptosis. Int
J Hematol 2006; 84:199–204
93. Sookhai S, Wang JJ, McCourt M, et al: A
novel therapeutic strategy for attenuating
neutrophil-mediated lung injury in vivo.
Ann Surg 2002; 235:285–291
94. Taneja R, Parodo J, Kapus A, et al: Delayed
neutrophil apoptosis in sepsis is associated
with maintenance of mitochondrial trans-
membrane potential and reduced caspase-9
activity. Crit Care Med 2004; 32:1460–1469
95. Fadok VA, Bratton DL, Konowal A, et al:
Macrophages that have ingested apoptotic
cells in vitro inhibit proinflammatory cyto-
kine production through autocrine/para-
crine mechanisms involving TGF, PGE2,
and PAF. J Clin Invest 1998; 101:890–898
96. Sanford AN, Suriano AR, Herche D, et al:
Abnormal apoptosis in chronic granuloma-
tous disease and autoantibody production
characteristic of lupus. Rheumatology
2006; 45:178–181
97. Daleke DL: Regulation of transbilayer
plasma membrane phospholipid asymme-
try. J Lipid Res 2003; 44:233–242
98. Bayir H, Kagan VE: Bench-to-bedside re-
view: Mitochondrial injury, oxidative stress
and apoptosis—there is nothing more prac-
tical than a good theory. Crit Care 2008;
12:206
99. Tyurina YY, Basova LV, Konduru NV, et al:
Nitrosative stress inhibits the aminophos-
pholipid translocase resulting in phosphati-
dylserine externalization and macrophage
engulfment: Implications for the resolution
of inflammation. J Biol Chem 2007; 282:
8498–8509
100. Swan R, Chung CS, Albina J, et al: Polymi-
crobial sepsis enhances clearance of apopto-
tic immune cells by splenic macrophages.
Surgery 2007; 142:253–261
101. Macdonald J, Galley HF, Webster NR: Oxi-
dative stress and gene expression in sepsis.
Br J Anaest 2003; 90:221–232
102. Matejovic M, Krouzecky A, Rokyta R Jr, et
al: Effects of combining inducible nitric ox-
ide synthase inhibitor and radical scavenger
during porcine bacteremia. Shock 2007; 27:
61–68
103. Fialkow L, Wang Y, Downey GP: Reactive
oxygen and nitrogen species as signaling
molecules regulating neutrophil function.
Free Radic Biol Med 2007; 42:153–164
104. Galley HF, Davies MJ, Webster NR: Xan-
thine oxidase activity and free radical gen-
eration in patients with sepsis syndrome.
Crit Care Med 1996; 24:1649–1653
105. Soriano FG, Nogueira AC, Caldini EG, et al:
Potential role of poly (adenosine 5Ј-
diphosphate-ribose) polymerase activation
in the pathogenesis of myocardial contrac-
tile dysfunction associated with human sep-
tic shock. Crit Care Med 2006; 34:
1073–1079
106. Ulloa L, Tracey KJ: The “cytokine profile”: A
code for sepsis. Trends Mol Med 2005; 11:
56–63
107. Zimmermann K, Volkel D, Pable S, et al:
Native versus recombinant high-mobility
group B1 proteins: functional activity in
vitro. Inflammation 2004; 28:221–229
108. Rouhiainen A, Tumova S, Valmu L, et al:
Pivotal advance: Analysis of proinflamma-
tory activity of highly purified eukaryotic
recombinant HMGB1 (amphoterin). J Leu-
kocyte Biol 2007; 81:49–58
109. Bonaldi T, Talamo F, Scaffidi P, et al: Mono-
cytic cells hyperacetylate chromatin protein
HMGB1 to redirect it towards secretion.
EMBO J 2003; 22:5551–5560
110. Youn JH, Shin JS: Nucleocytoplasmic shut-
tling of HMGB1 is regulated by phosphory-
lation that redirects it toward secretion.
J Immunol 2006; 177:7889–7897
111. Bell CW, Jiang W, Reich CF III, et al: The
extracellular release of HMGB1 during ap-
optotic cell death. Am J Physiol Cell Physiol
2006; 291:C1318–C1325
112. Qin S, Wang H, Yuan R, et al: Role of
HMGB1 in apoptosis-mediated sepsis lethal-
ity. J Exp Med 2006; 203:1637–1642
113. Hori O, Brett J, Slattery T, et al: The recep-
tor for advanced glycation end products
(RAGE) is a cellular binding site for ampho-
terin: Mediation of neurite outgrowth and
co-expression of rage and amphoterin in the
developing nervous system. J Biol Chem
1995; 270:25752–25761
114. Park JS, Svetkauskaite D, He Q, et al: In-
volvement of toll-like receptors 2 and 4 in
cellular activation by high mobility group
box 1 protein. J Biol Chem 2004; 279:
7370–7377
115. Wang H, Bloom O, Zhang M, et al: HMG-1
as a late mediator of endotoxin lethality in
mice. Science 1999; 285:248–251
116. Mitola S, Belleri M, Urbinati C, et al: Cut-
ting edge: Extracellular high mobility
group box-1 protein is a proangiogenic cy-
tokine. J Immunol 2006; 176:12–15
117. Tzeng HP, Fan J, Vallejo JG, et al: Negative
inotropic effects of high-mobility group box
1 protein in isolated contracting cardiac
myocytes. Am J Physiol Heart Circ Physiol
2008; 294:H1490–H1496
118. Wang H, Yu M, Ochani M, et al: Nicotinic
acetylcholine receptor ␣7 receptor is an es-
sential regulator of inflammation. Nature
2003; 421:384–388
119. Pavlov VA, Wang H, Czura CJ, et al: The
cholinergic anti-inflammatory pathway: A
missing link in neuromodulation. Mol Med
2003; 9:125–134
120. Wang H, Liao H, Ochani M, et al: Cholin-
ergic agonists inhibit HMGB1 release and
improve survival in experimental sepsis.
Nat Med 2004; 10:1216–1221
121. Huston JM, Gallowitsch-Puerta M, Ochani
M, et al: Transcutaneous vagus nerve stim-
ulation reduces serum high mobility group
box 1 levels and improves survival in mu-
rine sepsis. Crit Care Med 2007; 35:
2762–2768
122. Goldstein RS, Bruchfeld A, Yang L, et al:
Cholinergic anti-inflammatory pathway ac-
tivity and High Mobility Group Box-1
(HMGB1) serum levels in patients with
rheumatoid arthritis. Mol Med 2007;
13(3–4):210–215
123. Youn JH, Oh YJ, Kim ES, et al: High Mo-
bility Group Box 1 protein binding to lipo-
polysaccharide facilitates transfer of lipo-
polysaccharide to CD14 and enhances
lipopolysaccharide-mediated TNF-{alpha}
production in human monocytes. J Immu-
nol 2008; 180:5067–5074
124. Sha Y, Zmijewski J, Xu Z, et al: HMGB1
develops enhanced proinflammatory activ-
ity by binding to cytokines. J Immunol
2008; 180:2531–2537
125. Sunde´n-Cullberg J, Norrby-Teglund A, Rou-
hiainen A, et al: Persistent elevation of high
mobility group box-1 protein (HMGB1) in
patients with severe sepsis and septic shock.
Crit Care Med 2005; 33: 564–573
126. Gibot S, Massin F, Cravoisy A, et al: High-
mobility group box 1 protein plasma con-
centrations during septic shock. Intensive
Care Med 2007; 33:1347–1353
127. Karlsson S, Pettila¨ V, Tenhunen J, et al:
HMGB1 as a predictor of organ dysfunction
and outcome in patients with severe sepsis.
Intensive Care Med 2008; 34:1046–1053.
128. Harris HE, Raucci A: Alarmin(g) news about
danger. EMBO Rep 2006; 7:774–778
129. Chavakis T, Bierhaus A, Al-Fakhri N, et al:
The pattern recognition receptor (RAGE) is
a counterreceptor for leukocyte integrins: A
novel pathway for inflammatory cell re-
cruitment. J Exp Med 2003; 198:1507–1515
130. Lutterloh EC, Opal SM, Pittman DD, et al:
Inhibition of the RAGE products increases
survival in experimental models of severe
sepsis and systemic infection. Crit Care
2007; 11:R122
131. Calandra T, Bernhagen J, Metz CN, et al:
MIF as a glucocorticoid-induced modulator
of cytokine production. Nature 1995; 377:
68–71
132. Calandra T, Echtenacher B, Roy DL, et al:
Protection from septic shock by neutraliza-
tion of macrophage migration inhibitory
factor. Nat Med 2000; 6:164–170
133. Bozza M, Satoskar AR, Lin G, et al: Targeted
disruption of migration inhibitory factor
gene reveals its critical role in sepsis. J Exp
Med 1999; 189:341–346
134. Roger T, David J, Glauser MP, et al: MIF
regulates innate immune responses
through modulation of Toll-like receptor
4. Nature 2001; 414:920–924
135. Mitchell RA, Liao H, Chesney J, et al: Mac-
rophage migration inhibitory factor (MIF)
sustains macrophage proinflammatory
function by inhibiting p53: Regulatory role
302 Crit Care Med 2009 Vol. 37, No. 1
in the innate immune response. Proc Natl
Acad Sci USA 2002; 99:345–350
136. Kreymann G, Grosser S, Buggisch P, et al:
Oxygen consumption and resting metabolic
rate in sepsis, sepsis syndrome, and septic
shock. Crit Care Med 1993; 21:1012–1019
137. Rosser DM, Stidwill RP, Jacobson D, et al:
Oxygen tension in the bladder epithelium
rises in both high and low cardiac output
endotoxemic sepsis. J Appl Physiol 1995;
79:1878–1882
138. Hotchkiss RS, Swanson PE, Freeman BD, et
al: Apoptotic cell death in patients with sep-
sis, shock, and multiple organ dysfunction.
Crit Care Med 1999; 27:1230–1251
139. Brealey D, et al: Mitochondrial dysfunction
in a long-term rodent model of sepsis and
organ failure. Am J Physiol Regul Integr
Comp Physiol 2004; 286:R491–R497
140. Crouser ED: Mitochondrial dysfunction in
septic shock and multiple organ dysfunc-
tion syndrome. Mitochondrion 2004;
4:729–741
141. Fink MP: Cytopathic hypoxia. Mitochon-
drial dysfunction as mechanism contribut-
ing to organ dysfunction in sepsis. Crit Care
Clinics 2001; 17:219–237
142. Brealey D, Brand M, Hargreaves I, et al:
Association between mitochondrial dys-
function and severity and outcome of septic
shock. Lancet 2002; 360:219–223
143. Crouser ED, Julian MW, Blaho DV, et al:
Endotoxin-induced mitochondrial damage
correlates with impaired respiratory activ-
ity. Crit Care Med 2002; 30:276–284
144. Levy RJ, Vijayasarathy C, Raj NR, et al:
Competitive and noncompetitive inhibition
of myocardial cytochrome C oxidase in sep-
sis. Shock 2004; 21:110–114
145. Rabuel C, Renaud E, Brealey D, et al: Hu-
man septic myopathy: Induction of cycloox-
ygenase, heme oxygenase and activation of
the ubiquitin proteolytic pathway. Anesthe-
siology 2004; 101:583–590
146. Larche J, Lancel S, Hassoun SM, et al: In-
hibition of mitochondrial permeability
transition prevents sepsis-induced myocar-
dial dysfunction and mortality. J Am Coll
Cardiol 2006; 48:377–385
147. Levi M, Ten Cate H: Disseminated intravas-
cular coagulation. N Engl J Med 1999; 341:
586–592
148. Schouten M, Wiersinga WJ, Levi M, et al:
Inflammation, endothelium, and coagula-
tion in sepsis. J Leukoc Biol 2008; 83:
536–545
149. Opal SM: The nexus between systemic in-
flammation and disordered coagulation in
sepsis. J Endotoxin Res 2004; 10:125–129
150. Esmon CT: Are natural anticoagulants can-
didates for modulating the inflammatory
response to endotoxin? Blood 2000; 95:
1113–1116
151. Johnson K, Choi Y, DeGroot E, et al: Po-
tential mechanisms for a proinflamma-
tory vascular cytokine response to coagu-
lation activation. J Immunol 1998; 160:
5130–5135
152. Stouthard JM, Levi M, Hack CE, et al: In-
terleukin-6 stimulates coagulation, not fi-
brinolysis, in humans. Thromb Haemost
1996; 76:738–742
153. Aupeix K, Hugel B, Martin T, et al: The
significance of shed membrane particles
during programmed cell death in vitro, and
in vivo, in HIV-1 infection. J Clin Invest
1997; 99:1546–1554
154. Soriano AO, Jy W, Chirinos JA, et al: Levels
of endothelial and platelet microparticles
and their interactions with leukocytes neg-
atively correlate with organ dysfunction and
predict mortality in severe sepsis. Crit Care
Med 2005; 33:2540–2546
155. Fujimi S, Ogura H, Tanaka H, et al: Acti-
vated polymorphonuclear leukocytes en-
hance production of leukocyte micropar-
ticles with increased adhesion molecules in
patients with sepsis. J Trauma 2002; 52:
443–448
156. Amaral A, Opal SM, Vincent JL: Coagulation
in sepsis. Intensive Care Med 2004; 30:
1032–1040
157. Vervloet MG, Thijs LG, Hack CE: Derange-
ments of coagulation and fibrinolysis in
critically ill patients with sepsis and septic
shock. Semin Thromb Hemost 1998; 24:
33–44
158. Yan SB, Dhainut JF: Activated protein C
versus protein C in severe sepsis. Crit Care
Med 2001; 29(Suppl):S69–S74
159. Xue M, Campbell D, Jackson CJ: Protein C
is an autocrine growth factor for human
skin keratinocytes, J Biol Chem 2007; 282:
13610–13616
160. Xue M, March L, Sambrook PN, et al:
Endothelial protein C receptor is overex-
pressed in rheumatoid arthritic (RA) sy-
novium and mediates the anti-inflamma-
tory effects of activated protein C in RA
monocytes. Ann Rheum Dis 2007; 66:
1574–1580
161. Regan LM, Stearns-Kurosawa DJ, Kurosawa
S, et al: The endothelial cell protein C re-
ceptor: Inhibition of activated protein C an-
ticoagulant function without modulation of
reaction with proteinase inhibitors. J Biol
Chem 1996; 271:17499–17503
162. Kurosawa S, Stearns-Kurosawa DJ, Carson
CW, et al: Plasma levels of endothelial cell
protein C receptor are elevated in patients
with sepsis and systemic lupus erythemato-
sus: Lack of correlation with thrombo-
modulin suggests involvement of different
pathological processes. Blood 1998; 91:
725–727
163. Yan SB, Helterbrand JD, Hartman DL, et al:
Low levels of protein C are associated with
poor outcome in severe sepsis. Chest 2001;
120:915–922
164. LaRosa SP, Opal SM, Utterback B, et al:
Decreased protein C, protein S, and anti-
thrombin levels are predictive of poor out-
come in Gram-negative sepsis caused by
Burkholderia pseudomallei. Int J Infect Dis
2006; 10:25–31
165. Yan SB, Dhainaut JF: Activated protein C
versus protein C in severe sepsis. Crit Care
Med 2001; 29(Suppl):S69–S74
166. Joyce DE, Gelbert L, Ciaccia A, et al: Gene
expression profile of antithrombotic protein
C defines new mechanisms modulating in-
flammation and apoptosis. J Biol Chem
2001; 14:11199–111203
167. Jackson CJ, Xue M: Activated protein C-An
anticoagulant that does more than stop
clots. Int J Biochem Cell Biol 2008; 40:
2692–2697
168. Dutt T, Toh CH: The yin-yang of thrombin
and activated protein C. Br J Haematology
2008; 140:505–515
169. Riewald M, Ruf W: Protease-activated recep-
tor-1 signaling by activated protein C in
cytokine-perturbed endothelial cells is dis-
tinct from thrombin signaling. J Biol Chem
2005; 280:19808–19814
170. Bae JS, Yang L, Rezaie AR: Receptors of the
protein C activation and activated protein C
signaling pathways are colocalized in lipid
rafts of endothelial cells. Proc Natl Acad Sci
USA 2007; 104:2867–2872
171. Bae JS, Yang L, Manithody C, et al: The
ligand occupancy of endothelial protein
C receptor switches the protease-activated
receptor 1-dependent signaling specificity
of thrombin from a permeability-enhanc-
ing to a barrier-protective response
in endothelial cells. Blood 2007; 110:
3909–3916
172. Shorr AF, Bernard GR, Dhainaut JF, et al:
Protein C concentrations in severe sepsis:
An early directional change in plasma levels
predicts outcome. Crit Care 2006; 10:R92
173. Cinel I, Dellinger RP: Current treatment of
severe sepsis. Curr Infect Dis Rep 2006;
8:358–365
174. Dellinger RP, Levy MM, Carlet JM, et al:
Surviving Sepsis Campaign: International
guidelines for management of severe sepsis
and septic shock: 2008. Crit Care Med 2008;
36:296–327
175. Bae JS, Yang L, Manithody C, et al: Engi-
neering a disulfide bond to stabilize the
calcium binding loop of activated protein C
eliminates its anticoagulant but not protec-
tive signaling properties. J Biol Chem 2007;
282:9251–9259
176. Kerschen EJ, Fernandez JA, Cooley BC, et
al: Endotoxemia and sepsis mortality reduc-
tion by non-anticoagulant activated protein
C. J Exp Med 2007; 204:2439–2448
177. Moake JL: Thrombotic microangiopathies.
N Engl J Med 2002; 347:589–600
178. Ruggeri ZM: Von Willebrand factor, plate-
lets and endothelial cell interactions.
J Thromb Haemost 2003; 1:1335–1342
179. Furlan M, Robles R, Galbusera M, et al: von
Willebrand factor-cleaving protease in
thrombotic thrombocytopenic purpura and
the hemolytic-uremic syndrome. New Engl
J Med 1998; 339:1578–1584
180. Arya M, Anvari B, Romo GM, et al: Ultra-
large multimers of von Willebrand factor
form spontaneous high-strength bonds
303Crit Care Med 2009 Vol. 37, No. 1
with the platelet glycoprotein Ib-IX com-
plex: Studies using optical tweezers. Blood
2002; 99:3971–3977
181. Bianchi V, Robles R, Alberio L, et al: Von
Willebrand factor-cleaving protease (AD-
AMTS13) in thrombocytopenic disorders: A
severely deficient activity is specific for
thrombotic thrombocytopenic purpura.
Blood 2002; 100:710–713
182. Ono T, Mimuro J, Madoiwa S, et al: Severe
secondary deficiency of von Willebrand fac-
tor-cleaving protease (ADAMTS13) in pa-
tients with sepsis-induced disseminated in-
travascular coagulation: Its correlation with
development of renal failure. Blood 2006;
107:528–534
183. Nguyen TC, Liu A, Liu L, et al: Acquired
ADAMTS-13 deficiency in pediatric pa-
tients with severe sepsis. Haematologica
2007; 92:121–124
184. Kremer Hovinga JA, Zeerleder S, Kessler
P, et al: ADAMTS-13, von Willebrand fac-
tor and related parameters in severe sep-
sis and septic shock. J Thromb Haemost
2007; 5:2284–2290
185. Bockmeyer CL, Claus RA, Budde U, et al:
Inflammation-associated ADAMTS13 defi-
ciency promotes formation of ultra-large
von Willebrand factor. Haematologica
2008; 93:137–140
186. Reiter RA, Varadi K, Turecek PL, et al:
Changes in ADAMTS13 (von-Willebrand
factor-cleaving protease) activity after in-
duced release of von Willebrand factor
during acute systemic inflammation.
Thromb Haemost 2005; 93:554–558
187. Martin K, Borgel D, Lerolle N, et al: De-
creased ADAMTS-13 (A disintegrin-like and
metalloprotease with thrombospondin type
1 repeats) is associated with a poor progno-
sis in sepsis-induced organ failure. Crit
Care Med 2007; 35:2375–2382
188. Grewal PK, Uchiyama S, Ditto D, et al: The
Ashwell receptor mitigates the lethal coagulopa-
thy of sepsis. Nat Med 2008; 14:648–655
189. van’t Veer C, van der Poll T. Keeping blood
clots at bay in sepsis. Nat Med 2008; 14:
606–608
190. Treziack S, Cinel I, Dellinger RP, et al:
Resuscitating the microcirculation in se-
vere sepsis: The central role of nitric oxide,
emerging concepts for novel therapies, and
challenges for clinical trials. Acad Emerg
Med 2008; 15:1–15
304 Crit Care Med 2009 Vol. 37, No. 1

More Related Content

What's hot

Tlr and inflammation 2013
Tlr and inflammation 2013Tlr and inflammation 2013
Tlr and inflammation 2013Elsa von Licy
 
Nucleic acid recognition by the innate immune system
Nucleic acid recognition by the innate immune systemNucleic acid recognition by the innate immune system
Nucleic acid recognition by the innate immune systemANKUR SHARMA
 
“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...
“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...
“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...Nicholas Young
 
Phsi2005 notes (immune) 1
Phsi2005 notes (immune) 1Phsi2005 notes (immune) 1
Phsi2005 notes (immune) 1julialoiko
 
Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2
Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2
Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2QIAGEN
 
Innate immunity and adjuvants
Innate immunity and adjuvantsInnate immunity and adjuvants
Innate immunity and adjuvantsSukhbir Brar
 
BIO2098 immunity essay
BIO2098 immunity essayBIO2098 immunity essay
BIO2098 immunity essayVanessa Otti
 
art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)Lior Lalush
 
Seminario biomol
Seminario biomolSeminario biomol
Seminario biomolSydgold15
 
Toll like receptors in periodontology
Toll like receptors in periodontology Toll like receptors in periodontology
Toll like receptors in periodontology Krupali Gandhi
 
Engineering of phage derived lytic enzymes
Engineering of phage derived lytic enzymesEngineering of phage derived lytic enzymes
Engineering of phage derived lytic enzymesPig Farm Solution
 
Eicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantationEicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantationFederal University of Bahia
 
Systemic acquired resistance (SAR): A novel strategy for plant protection.
Systemic acquired resistance (SAR): A novel strategy for plant protection.Systemic acquired resistance (SAR): A novel strategy for plant protection.
Systemic acquired resistance (SAR): A novel strategy for plant protection.mohd younus wani
 

What's hot (20)

Tlr and inflammation 2013
Tlr and inflammation 2013Tlr and inflammation 2013
Tlr and inflammation 2013
 
Nucleic acid recognition by the innate immune system
Nucleic acid recognition by the innate immune systemNucleic acid recognition by the innate immune system
Nucleic acid recognition by the innate immune system
 
“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...
“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...
“Estrogen-mediated TLR8 expression via STAT1 facilitates endogenous miRokine ...
 
Toll like receptors
Toll like receptors   Toll like receptors
Toll like receptors
 
Phsi2005 notes (immune) 1
Phsi2005 notes (immune) 1Phsi2005 notes (immune) 1
Phsi2005 notes (immune) 1
 
Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2
Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2
Toll-like Receptors in Inflammation: Host Defense Webinar Series Part 2
 
Innate immunity and adjuvants
Innate immunity and adjuvantsInnate immunity and adjuvants
Innate immunity and adjuvants
 
BIO2098 immunity essay
BIO2098 immunity essayBIO2098 immunity essay
BIO2098 immunity essay
 
Toll-like receptors
Toll-like receptors Toll-like receptors
Toll-like receptors
 
art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)
 
Seminario biomol
Seminario biomolSeminario biomol
Seminario biomol
 
TLR
TLRTLR
TLR
 
Sepsis
SepsisSepsis
Sepsis
 
1.1 philippe sansonetti
1.1 philippe sansonetti1.1 philippe sansonetti
1.1 philippe sansonetti
 
Toll like receptors in periodontology
Toll like receptors in periodontology Toll like receptors in periodontology
Toll like receptors in periodontology
 
Inmunidad Basica
Inmunidad BasicaInmunidad Basica
Inmunidad Basica
 
Engineering of phage derived lytic enzymes
Engineering of phage derived lytic enzymesEngineering of phage derived lytic enzymes
Engineering of phage derived lytic enzymes
 
Eicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantationEicosanoids: lipid mediators of inflammation in transplantation
Eicosanoids: lipid mediators of inflammation in transplantation
 
Systemic acquired resistance (SAR): A novel strategy for plant protection.
Systemic acquired resistance (SAR): A novel strategy for plant protection.Systemic acquired resistance (SAR): A novel strategy for plant protection.
Systemic acquired resistance (SAR): A novel strategy for plant protection.
 
Innate immunity
Innate immunityInnate immunity
Innate immunity
 

Viewers also liked

Viewers also liked (12)

Sepsis seminar final
Sepsis seminar   finalSepsis seminar   final
Sepsis seminar final
 
Sepsis Bp Ad
Sepsis Bp AdSepsis Bp Ad
Sepsis Bp Ad
 
The Microcirculation in Sepsis
The Microcirculation in SepsisThe Microcirculation in Sepsis
The Microcirculation in Sepsis
 
ACTEP2014: Sepsis management has anything change
ACTEP2014: Sepsis management has anything change ACTEP2014: Sepsis management has anything change
ACTEP2014: Sepsis management has anything change
 
Best Practice in Sepsis
Best Practice in SepsisBest Practice in Sepsis
Best Practice in Sepsis
 
ACTEP2014: Sepsis marker in clinical use
ACTEP2014: Sepsis marker in clinical useACTEP2014: Sepsis marker in clinical use
ACTEP2014: Sepsis marker in clinical use
 
Sepsis
SepsisSepsis
Sepsis
 
Evidence-based management of CHF
Evidence-based management of CHFEvidence-based management of CHF
Evidence-based management of CHF
 
Biomarkers in sepsis
Biomarkers in sepsisBiomarkers in sepsis
Biomarkers in sepsis
 
Sepsis 2016
Sepsis 2016 Sepsis 2016
Sepsis 2016
 
Sepsis
SepsisSepsis
Sepsis
 
Sepsis and HRRP
Sepsis and HRRPSepsis and HRRP
Sepsis and HRRP
 

Similar to Sepsis primer

Innate immunity & inflammation1
Innate immunity & inflammation1Innate immunity & inflammation1
Innate immunity & inflammation1AjayKawale2601
 
Membrane-Bound-Pattern-Recognition-Receptors.pptx
Membrane-Bound-Pattern-Recognition-Receptors.pptxMembrane-Bound-Pattern-Recognition-Receptors.pptx
Membrane-Bound-Pattern-Recognition-Receptors.pptxAnumol Lorance
 
Perio 2
Perio 2Perio 2
Perio 2soamia
 
Why do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdf
Why do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdfWhy do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdf
Why do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdfaroraopticals15
 
Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...
Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...
Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...dr.Ihsan alsaimary
 
Vih y paciente septico
Vih y paciente septicoVih y paciente septico
Vih y paciente septicoAivan Lima
 
238833220 1srier-sle
238833220 1srier-sle238833220 1srier-sle
238833220 1srier-slehomeworkping4
 
Immuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal diseaseImmuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal diseaseGanesh Nair
 
RECEPTOR DIVERSITY OF INNATE IMMUNITY.pptx
RECEPTOR DIVERSITY OF INNATE IMMUNITY.pptxRECEPTOR DIVERSITY OF INNATE IMMUNITY.pptx
RECEPTOR DIVERSITY OF INNATE IMMUNITY.pptxJustinMutua
 
The liver as a lymphoid organ annu rev
The liver as a lymphoid organ annu revThe liver as a lymphoid organ annu rev
The liver as a lymphoid organ annu revmaxcueva13
 
Host microbial interactions in periodontal diseases
Host microbial interactions in periodontal diseasesHost microbial interactions in periodontal diseases
Host microbial interactions in periodontal diseasesDr Heena Sharma
 

Similar to Sepsis primer (20)

Mecanismo les
Mecanismo lesMecanismo les
Mecanismo les
 
Innate immunity & inflammation1
Innate immunity & inflammation1Innate immunity & inflammation1
Innate immunity & inflammation1
 
TLR.pdf
TLR.pdfTLR.pdf
TLR.pdf
 
journal.pbio.3001136.pdf
journal.pbio.3001136.pdfjournal.pbio.3001136.pdf
journal.pbio.3001136.pdf
 
Membrane-Bound-Pattern-Recognition-Receptors.pptx
Membrane-Bound-Pattern-Recognition-Receptors.pptxMembrane-Bound-Pattern-Recognition-Receptors.pptx
Membrane-Bound-Pattern-Recognition-Receptors.pptx
 
3 nod
3 nod3 nod
3 nod
 
CNS IRIS.pptx
CNS IRIS.pptxCNS IRIS.pptx
CNS IRIS.pptx
 
CNS IRIS.pptx
CNS IRIS.pptxCNS IRIS.pptx
CNS IRIS.pptx
 
Perio 2
Perio 2Perio 2
Perio 2
 
Why do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdf
Why do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdfWhy do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdf
Why do viral particles, zymosan on fungi, endotoxin (LPS) from gram .pdf
 
Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...
Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...
Evaluation and importance of innate & adaptive immunity Dr. ihsan edan ab...
 
Vih y paciente septico
Vih y paciente septicoVih y paciente septico
Vih y paciente septico
 
238833220 1srier-sle
238833220 1srier-sle238833220 1srier-sle
238833220 1srier-sle
 
Immuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal diseaseImmuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal disease
 
Phagocytosis and tl rs
 Phagocytosis and tl rs Phagocytosis and tl rs
Phagocytosis and tl rs
 
RECEPTOR DIVERSITY OF INNATE IMMUNITY.pptx
RECEPTOR DIVERSITY OF INNATE IMMUNITY.pptxRECEPTOR DIVERSITY OF INNATE IMMUNITY.pptx
RECEPTOR DIVERSITY OF INNATE IMMUNITY.pptx
 
The liver as a lymphoid organ annu rev
The liver as a lymphoid organ annu revThe liver as a lymphoid organ annu rev
The liver as a lymphoid organ annu rev
 
Host microbial interactions in periodontal diseases
Host microbial interactions in periodontal diseasesHost microbial interactions in periodontal diseases
Host microbial interactions in periodontal diseases
 
plospathogen
plospathogenplospathogen
plospathogen
 
Innate immunity
Innate immunityInnate immunity
Innate immunity
 

More from Aivan Lima

Ventilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétricaVentilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétricaAivan Lima
 
Manejo inicial gran quemado ii
Manejo inicial gran quemado iiManejo inicial gran quemado ii
Manejo inicial gran quemado iiAivan Lima
 
Manejo inicial gran quemado i
Manejo inicial gran quemado iManejo inicial gran quemado i
Manejo inicial gran quemado iAivan Lima
 
Manejo ambulatorio de quemaduras
Manejo ambulatorio de quemadurasManejo ambulatorio de quemaduras
Manejo ambulatorio de quemadurasAivan Lima
 
Lra que hay de nuevo
Lra que hay de nuevoLra que hay de nuevo
Lra que hay de nuevoAivan Lima
 
Nice sugar nejm 2009
Nice sugar nejm 2009Nice sugar nejm 2009
Nice sugar nejm 2009Aivan Lima
 
Hiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensivaHiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensivaAivan Lima
 
Tratamiento del choque septico
Tratamiento del choque septicoTratamiento del choque septico
Tratamiento del choque septicoAivan Lima
 
Liquidos en el paciente septico
Liquidos en el paciente septicoLiquidos en el paciente septico
Liquidos en el paciente septicoAivan Lima
 
The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014Aivan Lima
 
Surviving sepsis guidelines
Surviving sepsis guidelinesSurviving sepsis guidelines
Surviving sepsis guidelinesAivan Lima
 
Severe sepsis
Severe sepsis Severe sepsis
Severe sepsis Aivan Lima
 

More from Aivan Lima (20)

lra.pptx
lra.pptxlra.pptx
lra.pptx
 
Anestesia
AnestesiaAnestesia
Anestesia
 
Neuro
NeuroNeuro
Neuro
 
Renal
RenalRenal
Renal
 
Ventilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétricaVentilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétrica
 
Hipertensivas
HipertensivasHipertensivas
Hipertensivas
 
Manejo inicial gran quemado ii
Manejo inicial gran quemado iiManejo inicial gran quemado ii
Manejo inicial gran quemado ii
 
Manejo inicial gran quemado i
Manejo inicial gran quemado iManejo inicial gran quemado i
Manejo inicial gran quemado i
 
Manejo ambulatorio de quemaduras
Manejo ambulatorio de quemadurasManejo ambulatorio de quemaduras
Manejo ambulatorio de quemaduras
 
Ngal lra
Ngal lraNgal lra
Ngal lra
 
Lra que hay de nuevo
Lra que hay de nuevoLra que hay de nuevo
Lra que hay de nuevo
 
Advances aki
Advances akiAdvances aki
Advances aki
 
Lra lancet
Lra lancetLra lancet
Lra lancet
 
Nice sugar nejm 2009
Nice sugar nejm 2009Nice sugar nejm 2009
Nice sugar nejm 2009
 
Hiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensivaHiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensiva
 
Tratamiento del choque septico
Tratamiento del choque septicoTratamiento del choque septico
Tratamiento del choque septico
 
Liquidos en el paciente septico
Liquidos en el paciente septicoLiquidos en el paciente septico
Liquidos en el paciente septico
 
The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014
 
Surviving sepsis guidelines
Surviving sepsis guidelinesSurviving sepsis guidelines
Surviving sepsis guidelines
 
Severe sepsis
Severe sepsis Severe sepsis
Severe sepsis
 

Recently uploaded

(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...indiancallgirl4rent
 
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...vidya singh
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeTop Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeCall Girls Delhi
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...Taniya Sharma
 
Bangalore Call Girls Nelamangala Number 9332606886 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 9332606886  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 9332606886  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 9332606886 Meetin With Bangalore Esc...narwatsonia7
 
Call Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...narwatsonia7
 
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Dipal Arora
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...chandars293
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...Taniya Sharma
 
Lucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel roomLucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel roomdiscovermytutordmt
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...hotbabesbook
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Dipal Arora
 

Recently uploaded (20)

(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
 
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
 
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeTop Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
 
Bangalore Call Girls Nelamangala Number 9332606886 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 9332606886  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 9332606886  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 9332606886 Meetin With Bangalore Esc...
 
Call Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Aurangabad Just Call 9907093804 Top Class Call Girl Service Available
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
 
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
 
Lucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel roomLucknow Call girls - 8800925952 - 24x7 service with hotel room
Lucknow Call girls - 8800925952 - 24x7 service with hotel room
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
 
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
 
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
 

Sepsis primer

  • 1. Concise Definitive Review Molecular biology of inflammation and sepsis: A primer* Ismail Cinel, MD, PhD; Steven M. Opal, MD I n sepsis, the expected and appropri- ate inflammatory response to an in- fectious process becomes amplified leading to organ dysfunction or risk for secondary infection. A continuum exists from a low grade systemic response associ- ated with a self-limited infection to a marked systemic response with solitary or multiorgan dysfunction, i.e., severe sepsis. As a clinical syndrome, sepsis occurs when an infection is associated with the systemic inflammatory response. The complex toll-like receptor signaling and associated downstream regulators of immune cell functions play a crucial role in the innate system as a first line of defense against pathogens (1). However, signaling is sometimes conflicting and a sustained inflammatory response can result in tissue damage. In addition, a reduction in their antimicrobial capacity may set the stage for opportunistic and/or super infections (2). Severe sepsis may also be associated with an exaggerated procoagulant state. This may lead to ischemic cell injury, an effect that further amplifies the damage caused by inappropriate inflammation. A micro- vasculature injured by inflammation and ischemia, in turn, further deranges the host response by altering leukocyte traffick- ing, generating apoptotic microparticles, and increasing cellular hypoxia (3). Mito- chondrial dysfunction, an acquired intrin- sic defect in cellular respiration termed “cy- topathic hypoxia,” also has an important role by decreasing cellular oxygen con- sumption in this chaotic process. In this review, we highlight the current under- standing of the basic molecular mecha- nisms that modulate these events so as to produce sepsis. Pattern Recognition Receptors, Pathogen-Associated Molecular Patterns (PAMPs) and Danger- Associated Molecular Patterns (DAMPs) The initiation of the host response during sepsis or tissue injury involves three families of pattern recognition re- ceptors (PRRs): 1) toll-like receptors (TLRs); 2) nucleotide-oligomerization domain leucine-rich repeat (NOD-LRR) proteins; and 3) cytoplasmic caspase ac- tivation and recruiting domain heli- cases such as retinoic-acid-inducible gene I (RIG-I)-like helicases (RLHs) (4, 5). These receptors initiate the innate immune response and regulate the adaptive immune response to infection or tissue injury. Gram-positive and Gram-negative bac- teria, viruses, parasites, and fungi all pos- sess a limited number of unique cellular constituents not found in vertebrate ani- mals. These elements are now referred to as PAMPs, or more appropriately microbial- associated molecular patterns, as these molecules are also common in nonpatho- genic and commensal bacteria (6). PAMPs bind to PRRs, such as TLRs, expressed on the surface of host cells. Cytoplasmic PRRs exist to detect invasive intracellular patho- gens (7). The NOD proteins recognize com- mon fragments of bacterial peptidoglycan. Diamino-pimelate from Gram-negative bacteria is the ligand for NOD1 and mu- ramyl dipeptide from peptidoglycan is the ligand for NOD2 in the cytosol (see Figs. 1 and 2). The PRRs also recognized damage signals from the release of endogenous pep- tides and glycosaminoglycans from apopto- tic or necrotic host cells (8–10) Caspase activation and recruiting domain helicases From the Division of Critical Care Medicine (IC), The Robert Wood Johnson School of Medicine, The University of Medicine and Dentistry of New Jersey, Camden NJ; and The Infectious Disease Division (SMO), Memorial Hospital of RI, The Warren Alpert School of Medicine of Brown University, Providence, RI. Dr. Opal has received grants from Wyeth and Eisai Inc. Dr. Cinel has not disclosed any potential conflicts of interest. For information regarding this article, E-mail: cinel-ismail@cooperhealth.edu Copyright © 2008 by the Society of Critical Care Medicine and Lippincott Williams & Wilkins DOI: 10.1097/CCM.0b013e31819267fb Background: Remarkable progress has been made during the last decade in defining the molecular mechanisms that underlie septic shock. This rapidly expanding field is leading to new therapeutic opportunities in the management of severe sepsis. Aim: To provide the clinician with a timely summary of the molecular biology of sepsis and to better understand recent advances in sepsis research. Data Selection: Medline search of relevant publications in basic mechanisms of sepsis/severe sepsis/septic shock, and se- lected literature review of other manuscripts about the signalo- some, inflammasome, apoptosis, or mechanisms of shock. Data Synthesis and Findings: The identification of the toll-like receptors and the associated concept of innate immunity based upon pathogen- or damage-associated molecular pattern molecules al- lowed significant advances in our understanding of the pathophys- iology of sepsis. The essential elements of the inflammasome and signal transduction networks responsible for activation of the host response have now been characterized. Apoptosis, mitochondrial dysfunction, sepsis-related immunosuppression, late mediators of systemic inflammation, control mechanisms for coagulation, and reprogramming of immune response genes all have critical roles in the development of sepsis. Conclusions: Many of these basic discoveries have direct impli- cations for the clinical management of sepsis. The translation of these “bench-to-bedside” findings into new therapeutic strategies is already underway. This brief review provides the clinician with a primer into the basic mechanisms responsible for the molecular biology of sepsis, severe sepsis, and septic shock.(Crit Care Med 2009; 37:291–304) KEY WORDS: sepsis; inflammation; toll like receptors; inflamma- some; signalosome; apoptosis; neutrophils; mitochondrial dys- function; reactive oxygen species; nitric oxide; peroxynitrite; ac- tivated protein C; a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs-13; pathogen associated molecular patterns; danger associated molecular patterns 291Crit Care Med 2009 Vol. 37, No. 1
  • 2. primarily recognize viral nucleic acids and activate antiviral measures including the type I interferons. TLR expression is significantly up- regulated in experimental models of sep- sis and in patients with sepsis (11–14). Trauma including thermal injury gener- ates danger-associated molecular pat- terns (i.e., high mobility group box-1, heat shock proteins, S100 proteins, hya- luran, etc.) that augment TLR expression like PAMPs. It also primes the innate im- mune system for enhanced TLR reactiv- ity, resulting in excess lipopolysaccharide (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 TLRs induce pro-interleukin(IL)-1beta production and prime NLR-containing multiprotein complexes, termed “inflam- masomes,” to respond to bacterial prod- ucts and products of damaged cells (17, 19). This results in caspase-1 activation and the subsequent processing of pro- IL-1␤ to its active extracellular form IL-1␤ (19). Caspases are a set of cysteine proteases that alter the enzymatic activity of target proteins at specific peptide se- quences adjacent to aspartate moieties. Caspases are important in process of ap- 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- flammasome constructions have been suggested to contain pyrin, NALP3, and other members of the NOD-LRR family (21–23). ASC facilitates inflammasome assembly thus triggering caspase-1 acti- vation and IL-1␤ processing. Interest- ingly, ASC can also regulate the nuclear factor (NF)-␬B pathway, thus linking the inflammasome to the signalosome (Fig. 1) (24). The inflammasome pathways contrib- ute to the inflammatory response in sep- sis (25). Caspase-1 knock out mice are protected from sepsis (26) while a natu- rally occurring polymorphism for human caspase-12, a putative regulator of Figure 1. Specific host immune response to each pathogen is mediated by various sets of pathogen associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) as detailed in Figure 1. PRRs are essential for initiating the host’s immune defenses against invading pathogens, yet they can also contribute to persistent and deleterious systemic inflammation. PRRs also serve as receptors for endogenous danger signals, hemodynamic changes in sepsis (tissue hypoperfusion and ischemia/ reperfusion phenomenon), thus same signaling systems that alerts the highly advantageous, host defense mechanisms, also contributes to the disadvantageous, pathologic events of systemic inflam- mation, coagulation, tissue damage in target organs in sepsis. Heat shock proteins, fibrinogen, 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), especially TLR4, are involved in the recognition of these endogenous or harmful self-antigens ligands which are released during noninfectious injury, such as trauma or ischemia/reperfusion suggesting their function may not be restricted to the recognition of extrinsic pathogens. NOD-LRR, nucleotide- oligomerization domain leucine-rich repeat; ASC, apoptosis-associated speck-like protein containing caspase activation and recruiting domain; NF-␬␤, nuclear factor ␬␤. Figure 2. Binding of toll-like receptors (TLRs) activates intracellular signal-transduction pathways that lead to the activation of transcriptional activators such as interferon regulator factors, phosphoino- sitide 3-kinase (PI3K)/Akt, activator protein-1, and cytosolic nuclear factor-kappa ␤ (NF-␬␤). Activated 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, proinflammatory cytokines, as well as enzymatic activation of cellular proteases. TLR9 DNA, TLR 3 dsRNA, and TLR7/8 ss RNA are endosomal. TLR 10 ligand is not defined and TLR1 forms heterodimers with TLR2. LPS, lipopolysaccharide; IRF, interferon regulatory factor; JNK; c Jun N-terminal kinase. 292 Crit Care Med 2009 Vol. 37, No. 1
  • 3. caspase-1, has been linked to sepsis (27, 28). Thus, caspase-1 activation appears to be a prerequisite for a competent im- mune response (29). Recently, the in- flammasome components have been shown to be significantly lower in septic shock patients during the early stages of systemic inflammatory response with el- evated plasma cytokines levels (30). This monocyte deactivation process may be maladaptive in the later phases of sepsis and predispose to secondary infection. Caspase-1 and its proinflammatory cy- tokine products are likely to contribute to the pathogenesis of sepsis in overwhelm- ing inflammation. However, like many other essential elements of innate immu- nity, caspase-1 also has a positive impact 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- associated kinase Ϫ1 haplotype has re- cently been demonstrated to affect the magnitude of NF-␬B activation and di- rectly correlates with an increased inci- dence of septic shock and significantly reduced survival rates (38). The impact of NF-␬B signaling is tissue-specific as defi- cient NF-␬B activation in intestinal epi- thelium is associated with increased in- flammation in vivo (39, 40). Both studies demonstrate that defects of NF-␬B signal- ing cause immunosuppression which triggers and maintains inflammation. Thus, it can be suggested that inhibition of massive NF-␬B activation in vivo leads to reduced inflammatory responses, at least during certain phases and certain tissues (i.e., parenchymal tissues) in sep- sis. On the other hand, this activation is followed by negative NF-␬B regulation favoring apoptosis in immune cells which Figure 3. Pathogenic mechanisms during sepsis or in response to tissue injury can lead to organ dysfunction. PRRs, pattern recognition receptors; TLRs, toll-like receptors; NOD-LRR, nucleotide- oligomerization domain leucine-rich repeat protein receptors; RLHs; retinoic-acid-inducible gene I (RIG-I)-like helicases; TNF-␣, tumor necrosis factor alpha; IL-1, interleukin 1; HMGB-1, high mobility group box-1; LPS, lipopolysaccharide; LTA, lipoteichoic acid; PGN, peptidoglycan; VSMCs, vascular 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 end products; ROCK; RhoA/Rho kinase; PARP-1, poly(ADP ribose) polymerase-1; PAR, protease activated receptor; ROS/RNS, reactive oxygen and nitrogen species; NF, nuclear factor; NADPH, nicotimanide adenosine dinucleotide phosphate; PAMPs, pathogen-associated molecular patterns; DAMPs, danger-associated molecular patterns. 293Crit Care Med 2009 Vol. 37, No. 1
  • 4. may lead to immunosuppression and fatal outcome in severe sepsis. Recently, using specific gene-targeted deletions, it has been shown that deletion of MyD88 caused a worsened survival in a model of severe peritonitis, despite the marked de- crease in sepsis-induced T and B lympho- cyte apoptosis suggesting MyD88, like NF-␬B, is also critical host survival in sepsis (41). Toll-Like Receptor Signaling The Role of Phosphoinositide 3-Kinase. Phosphoinositide 3-kinase (PI3K), a sig- nal transduction enzyme, and the down- stream serine/threonine kinase Akt (also known as protein kinase B) have been reported in cellular activation, inflamma- tory responses, chemotaxis, and apoptosis (Figs. 2 and 3) (42). PI3K can function either as a positive or negative regulator of TLR signaling. PI3Ks (three types) act at several steps downstream of TLRs, de- pending on the cell type and/or the en- gagement of a specific TLR regulating downstream signaling to NF-KB transac- tivation or to mitogen-activated phos- phokinase activation (43, 44). As a posi- tive mediator of TLR signaling, PI3K together with p38 and extracellular reg- ulated kinase (ERK)1/2 mitogen-acti- vated phosphokinases, lead to production of proinflammatory cytokines IL-1␤, IL-6, and IL-8 upon microbial challenge (44, 45). In detail, PI3K activation ap- peared to have a significantly promoting function for these mediators in mono- cytes, whereas activation appeared to limit the LPS response for generation of these cytokines in neutrophils (46). How- ever, PI3K inhibition resulted in im- paired oxidative burst and phagocytosis activity in both neutrophils and mono- cytes. By limiting C5a-mediated effects on neutrophil cytokine generation, and promoting oxidative burst and phagocy- tosis, PI3K activation seems to be a ther- apeutic approach for limiting inflamma- tion in sepsis. On the other hand, PI3K/Akt signaling pathway acts as an endogenous negative feedback mechanism that serves to limit proinflammatory and apoptotic events as seen in monocytes in response to endo- toxin (47). The suppressive role in in- flammation and coagulation was con- firmed in mouse model of endotoxemia (48). It can also promote the generation of anti-inflammatory cytokine IL-10 (49). PI3K/Akt has an effect in balancing Th1 vs. Th2 responses (50). Overexpression of Akt in lymphocytes decreases lymphocyte apoptosis, a Th1 cytokine propensity, and improve outcome in cecal ligation and puncture-induced sepsis (51). Peptide- mediated activation of Akt and extracel- lular regulated kinase signaling protects lymphocytes from numerous apoptotic stimuli both in vitro and in vivo (52). This suggests again the survival advan- tage of PI3K/Akt pathway activation for the later stages of sepsis. The Role of Rho GTPases. The Rho family of small GTPases is one of the master regulators of cell motility, as they control actin cytoskeleton remodeling. RhoA, Rac1, and CDc42 are the well known family members which act molec- ular switches regulating responses of in- nate immune cells related to pathogen sensing, intracellular uptake, and de- struction (53). These GTPases play both unique and overlapping roles in phago- cyte functions including migration, che- motaxis, and optimal bacterial killing (54). It has been shown that PAMPs may utilize GTPases through TLRs (i.e., TLR-2, -4, -3, and -9) in which Rac1 ac- tivation is required for PI3K activation upon TLR2 stimulation (55). RhoA regu- lates not only cytoskeletal events, which mediate neutrophil migration, but also contributes to NF-␬␤-dependent proin- flammatory gene transcription (Fig. 3) (56). It has been suggested that ROCK inhibition could attenuate cytoskeletal rearrangement of endothelial cells, lead- ing to decreased neutrophil emigration into the lung parenchyma in LPS- induced lung injury (57). An important role for rho kinase in leukocyte recruit- ment is also supported in an endotoxemic liver injury model (58). The recently emerged connections between TLR sig- naling and small Rho-GTPases seem to provide new therapeutic avenue of re- search in sepsis. Toll-Like Receptor Signaling and Regulatory T Cells (Tregs) To balance self-tolerance and immu- nity against pathogens, the immune sys- tem depends on both up-regulatory and down-regulatory mechanisms. Recent studies have suggested that several lym- phocyte subpopulations (i.e., CD4ϩCD25ϩ Foxp3ϩ T regulatory-cell ͓Tregs͔) may have the capacity to actively suppress an adaptive immune response and may po- tentially be involved in septic immune dysfunction. Naturally occurring Tregs express the transcription factor forkhead box protein (FoxP3), which is induced by the anti-inflammatory cytokine trans- forming growth factor-␤ (59, 60). TLR triggering induces dendritic cell matura- tion also, which is essential for the induc- tion of adaptive immune responses (61). Studies have recently highlightened the importance of TLRs on Tregs (62). In this regard, the dominant role of TLR2 signal- ing on the Treg-mediated immune sup- pression has been demonstrated (63, 64). Tregs control inflammatory reactions to commensal bacteria and opportunist pathogens and play a major role in sup- pressing immune reactivity, ranging from autoimmunity to infectious disease (65) and to injury (66). Monneret et al (67) observed that sepsis increases CD4ϩCD25ϩ T cells in the peripheral blood of septic patients. This was subse- quently found to be a relative increase in Tregs due to a decrease in the CD4ϩCD25– T effector cell populations (68). Further- more, Treg-mediated induction of “alter- natively activated” macrophages has been demonstrated suggesting as one of the causes of immune dysfunction in sys- temic inflammatory response syndrome and sepsis (69, 70). Targeting apoptosis of Tregs may be a new therapeutic approach in preventing the continuum of sepsis to severe sepsis. However, it has been re- ported that depletion of CD25ϩ cells be- fore inducing sepsis did not alter septic mortality pointing the need of more stud- ies to clarify the significance of this cell population’s expansion in sepsis morbid- ity (71, 72). Adoptive transfer of Tregs before or following the initiation of polymicrobial sepsis improved survival by enhancing tumor necrosis factor (TNF)-␣ produc- tion and bacterial clearance (73). Tregs inhibit LPS-induced monocyte survival through the Fas/FasL dependent pro- apoptotic mechanism, which might play a role in the resolution of exaggerated inflammation (74). The positive and neg- ative effects of TLR on Tregs are curious in sepsis, and the dynamics of TLR ex- pression on immune-suppressive Tregs upon inflammation or in relation to type of pathogen are needed to illuminate. Neutrophils and Monocyte/Macrophages in Inflammation Myeloid cells including neutrophils and elements of monocyte/macrophage lineage are heavily armed with large stores of proteolytic enzymes and with 294 Crit Care Med 2009 Vol. 37, No. 1
  • 5. the capacity to rapidly generate ROS and RNS to degrade internalized pathogens. The highly proapoptotic nature of neu- trophils is designed to maintain a balance between antimicrobial effectiveness and the potential for neutrophil-associated damage to the host in septic challenge or in other injurious processes, such as trauma or ischemia/reperfusion (75). Host tissue damage in severe sepsis may arise via a variety of mechanisms includ- ing premature neutrophil activation dur- ing migration, extracellular release of cy- totoxic molecules and toxins during microbial killing, removal of infected or damaged host cells or debris during host 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- grins (77, 78). P-selectin is expressed on platelets; E- and P-selectins are expressed by endothelial cells, whereas L-selectin is expressed on leukocytes. Selectins mediate neutrophil rolling along activated endothe- 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- nisms are compromised in the microcircu- lation including endothelial hyperactivity, fibrin deposition, microvascular occlusion, and cellular exudates that further impede adequate tissue oxygenation. Neutrophils participate in these rheologic changes through their augmented binding to blood vessel walls and through the formation of platelet-leukocyte aggregates (83). Neutro- phil elastase, other proteases, glycases and inflammatory cytokines degrade endoge- nous anticoagulant activity, and impair fi- brinolysis on endothelial surfaces favoring a procoagulant state (1). The Role of Apoptosis. Sepsis-induced neutrophil-mediated tissue injury has been demonstrated in a variety of organs including the lungs (84–86), diaphragm (87), kidneys (86), intestine (88, 89), and liver (86). Apoptosis is a counter-regula- tor of the initial inflammatory response in sepsis (90). Neutrophils are constitu- tively proapoptotic and apoptosis is fun- damental for the resolution of inflamma- tion and cell turnover. Neutrophils can undergo apoptosis via intrinsic and ex- trinsic pathways; the latter also requires mitochondrial amplification (Fig. 4) (91). The role played by mitochondria in the regulation of neutrophil life span is more crucial than in other cell types in the body (92). As neutrophils kill pathogens using ROS and RNS and a mixture of lytic enzymes, delayed clearance of neutro- phils in sepsis can potentially contribute Figure 4. Summary of apoptotic signaling pathways as seen through activation of death receptor (extrinsic) or mitochondrial (intrinsic) pathway. Extrinsic signals bind to their receptors and trigger intracellular signaling, leading to caspase-8 activation. Activation of caspase-8 by extrinsic stimuli (such as tumor necrosis factor-␣ [TNF-␣], Fas ligand) involves mitochondria-dependent signaling in 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 3-kinase/Akt; APAF-1, apoptosis protease activating factor 1; ER, endoplasmic reticulum; IL, interleukin. Figure 5. The recognition of apoptotic cells by macrophages is largely dependent on the cell surface appearance of phosphatidylserine (PS). S-nitrosylation of critical cysteine residues inhibits aminophospho- lipid translocase (APLT), leading to PS externalization. It generates an “eat me” signal during apoptosis. iNOS, inducible nitric oxide synthase; . NO, nitric oxide; O2, superoxide; ONOO؊ , peroxynitrite. 295Crit Care Med 2009 Vol. 37, No. 1
  • 6. 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- cytosis of apoptotic neutrophils by macrophages inhibits the release of proinflammatory cytokines and promotes the secretion of anti-inflammatory cyto- kines (95). In contrast, inefficient apopto- tic cell clearance is proinflammatory and immunogenic (96). The recognition of apoptotic cells by macrophages is largely dependent on the cell surface appearance of an anionic phospholipid, phosphatidyl- serine (PS), which is normally confined to the inner leaflet of the plasma mem- brane (97). Asymmetric distribution of PS across the plasma membrane is mainly because of the activity of a spe- cialized enzymatic mechanism, amin- ophospholipid translocase. S-nitrosyla- tion of critical cysteine residues inhibits aminophospholipid translocase, leading to PS externalization. PS expression dur- ing apoptosis generates an “eat-me” sig- nal (Fig. 5), which in turn triggers clear- ance of apoptotic cells and suppresses the inflammatory response (98). It has been demonstrated that S-nitrosylation of crit- ical cysteine residues in aminophospho- lipid translocase using a cell-permeable transnitrosylating agent, S-nitroso-acetyl- cysteine, resulted in egression of PS to the outer surface of the plasma mem- brane, rendering these cells recognizable by macrophages (99). The therapeutic po- tential of regulating neutrophil life-span in sepsis remains to be determined. Care must be exercised in regulating of this pathway, as sepsis enhances the capacity of macrophages to clear expanded apo- ptotic populations, a mechanism contrib- uting to septic immune suppression (100). The Role of ROS and RNS. ROS and RNS exert several beneficial physiologic functions, such as intracellular signaling for several cytokines and growth factors, second messengers for hormones and re- dox regulation. Despite their importance as a defense mechanism against invading pathogens, an overwhelming production of ROS and RNS or a deficit in oxidant scavenger and antioxidant defenses result in oxidative/nitrosative stress, a key ele- ment in the deleterious processes in sep- sis (Fig. 6) (101, 102). Stimulated neutrophils produce ROS and RNS through the nicotinamide ade- nine dinucleotide phosphate oxidase complex, myeloperoxidase and xanthine oxidoreductase and represent a defense mechanism against invading microor- ganisms (103). Lipopolysaccharide and other proinflammatory mediators acti- vate nicotinamide adenine dinucleotide phosphate oxidase to produce superoxide radical (O2 Ϫ ). In aqueous environments, superoxide radical is rapidly catalyzed by superoxide dismutase hydrogen peroxide (H2O2) and hydroxyl radicals. Myeloper- oxidase from neutrophil azurophilic granules produces hypochlorous acid from hydrogen peroxide (H2O2) and chlo- ride anion (Cl؊ ) during respiratory burst. These radicals are highly cytotoxic, and neutrophils used them to kill bacteria and other pathogens. Expression of hu- man xanthine oxidoreductase is markedly up-regulated by hypoxia, ischemia/reper- fusion, LPS, and TNF-␣. Increased activ- ity of xanthine oxide, one the important contributors of ROS production, has been reported in adult and pediatric patients with sepsis (104). O2 ؊ in the presence of nitric oxide, generates peroxynitrite (ONOOϪ ), a key player in the pathogenesis of sepsis- induced organ dysfunction. ONOOϪ can cause DNA strand breakage, which trig- gers the activation of DNA repair en- zymes such as poly (adenine dinucleotide phosphate-ribose) polymerase (Fig. 3). Poly (adenine dinucleotide phosphate- ribose) polymerase inhibitors protect against oxidative and nitrosative stress- induced organ dysfunction in endotox- emia (87–89). Recently, the potential role of poly (adenine dinucleotide phosphate- ribose) polymerase activation has been implicated in the pathogenesis of myo- cardial contractile dysfunction associated with human septic shock (105). Novel Cytokines in Inflammation High Mobility Group Box-1 and Re- ceptor for Advanced Glycation End- Products. High mobility group box-1 (HMGB-1) is a nonhistone, nuclear DNA- binding protein involved in nucleosome stabilization and gene transcription. How- ever, when HMGB-1 is released in large quantities into the extracellular environ- ment, it becomes a lethal mediator of sys- temic inflammation (Fig. 3) (106). Indeed, Figure 6. Proinflammatory/anti-inflammatory cytokines and reactive oxygen and nitrogen species have important effects within the microcirculatory unit: the arteriole, endothelial cell, capillary bed, and the 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 thromboses are formed. The postcapillary venule is where leukocyte trafficking is most disordered. All of these causes impair flow through the microcirculation leading to microcirculatory dysfunction. TLR, toll-like receptor; NOD-LRR, nucleotide-oligomerization domain leucine-rich repeat; RLH, (RIG-I)-like helicase; VSM, vascular smooth muscle; IL, interleukin; TGF, transforming growth factor; MIF, migration inhibitory factor; TNF, tumor necrosis factor; HMGB, high mobility group box; PAF, protease activating factor; sTNFR, soluble tumor necrosis factor receptor. 296 Crit Care Med 2009 Vol. 37, No. 1
  • 7. it has been recently shown that it has a weak proinflammatory activity by itself and binding to bacterial substances including lipid molecules such as phophatidylserine strengthens its effects (107). Interestingly, phosphatidylserine has been implicated in the regulation of inflammation and HMGB-1 might thus regulate its anti- inflammatory activities (108). HMGB-1 is released into the extracel- lular space through acetylation or phos- phorylation (109, 110). HMGB-1 is either “passively released” from necrotic cells, and a mechanism that represents a pro- cess adopted by the innate immune sys- tem to recognize damaged and necrotic cells, or “actively secreted” by immune cells including macrophages and neutro- phils to trigger inflammation (106). Re- cently, HMGB-1 release from macro- phages has been shown during the course of apoptosis as well as necrosis and de- fined as a downstream event of cell apo- ptosis during severe sepsis (111, 112). After treatment with LPS or various cy- tokines such as TNF-␣, IL-1␤, or IFN-␥, HMGB-1 is released from activated mac- rophages within 4 hrs and reaches a pla- teau around 18–24 hrs. It binds to several transmembrane receptors such as recep- tor for advanced glycation end products (RAGE), TLR-2, and -4, activating NF-␬B and extracellular regulated kinase 1/2 (113, 114). Although HMGB-1 was originally de- scribed as a late mediator of endotoxin- induced lethality (115), recent studies in- dicate a role for HMGB-1 in angiogenesis, tissue repair, and regeneration (116). HMGB-1 appears to be a novel myocardial depressant factor upon release by resi- dent myocardial cells following tissue in- jury. HMGB-1 might decrease energy uti- lization in ischemic tissue, thereby preventing injured myocytes from wors- ening ATP depletion that eventuate in necrosis (117). On other hand, excessive release of HMGB-1 in sepsis might con- tribute to sustained inflammation and to profound myocardial depression. The cholinergic anti-inflammatory pathway is a neural mechanism that in- hibits the expression of HMGB-1 and other cytokines (118–121). Signals trans- mitted via the vagus nerve, the principal nerve of the parasympathetic nervous system, significantly attenuate the re- lease of HMGB-1 and other cytokines in inflammation in animal and human stud- ies (122). The remarkable binding characteristics of HMGB-1 suggest another important role for this protein in the extracellular fluid. HMGB-1 might serve as a shuttle platform for LPS and other microbial mediators for docking to CD14 and recognition by the TLRs (108, 123) or through binding to host-derived proinflammatory mediators, such as IL-1beta (124). Elevated levels of HMGB-1 are measur- able in the majority of patients up to 1 wk after the diagnosis of sepsis or septic shock and are correlated with the degree of organ dysfunction (125, 126). However, serum HMGB-1 levels do not consistently identify nonsurvivors from survivors as a predictor of hospital mortality (127). As HMGB-1 is late inflammatory cytokine of sepsis, it pro- vides a wide therapeutic time window for clinical intervention and remains an attrac- tive target for sepsis treatment. RAGE, a member of the immunoglobu- lin superfamily, is a pattern-recognition re- ceptor that binds diverse classes of endog- enous molecules including HMGB-1. It has been defined as part of a newly appreciated component of the innate immune system referred to as the danger associated molec- ular pattern system (Fig. 3) (128). Mem- brane bound and soluble forms of RAGE (sRAGE) have been detected in plasma. Sol- uble RAGE, anti-RAGE antibody (Fab 2 fragment) or data from RAGEϪ/Ϫ animals have been shown to decrease inflammation, reduce neutrophil extravasation, and re- duce migration (129). Recently, the dem- onstration of survival benefit after delayed administration of anti-RAGE antibody in a murine model of polymicrobial sepsis, even when delayed up to 24 hrs, provides a ther- apeutic rationale for the use of anti-RAGE mAb as a salvage therapy for established severe sepsis (130). Macrophage Migration Inhibitory Factor. Migration inhibitory factor (MIF) acts as a stress response mediator and proinflammatory cytokine upon induc- tion by glucocorticoids (131). This pro- tein is readily measurable in patients with sepsis, and MIF probably contributes to the pathogenesis of sepsis. Inhibition of MIF or its targeted deletion attenuates TNF-␣ and IL-1␤ expression and protects mice from lethality is experimental sepsis (132, 133). Systemic challenge of animals with MIF increases LPS-related lethality (133). MIF promotes the expression of TLR4 on macrophages, and thereby sen- sitizes these immune effector cells to LPS (134). The immunoregulatory effects of MIF might be crucial to the control and resolu- tion of the inflammatory response as a con- sequence of its ability to regulate activa- tion-induced apoptosis (135). High MIF levels delay the removal of activated mono- cytes/macrophages by apoptosis. This pro- longs monocyte/macrophage survival, in- creases cytokine production, and sustains an ongoing proinflammatory response. Mitochondrial Dysfunction in Inflammation Although microvascular flow abnormal- ities occur, findings of decreased oxygen consumption (136) and elevated tissue ox- ygen tension (137), yet minimal cell death despite functional and biochemical de- rangements (138), suggest that the prob- lem lies more in cellular oxygen utilization rather than a problem with oxygen delivery (139). It is postulated that prolonged and systemic inflammatory insult is accompa- nied by a basic tissue survival response me- diated by switching off its energy-consum- ing biophysiological processes. Recent evidence suggests that sepsis and septic shock severely impair the mitochondria (140, 141), and the severity and outcome of organ dysfunction could be related to mi- tochondrial dysfunction (142). Depleted levels of reduced glutathione, an important intramitochondrial antioxidant, in combi- nation with excess generation of ROS and RNS severely inhibit oxidative phosphory- lation and ATP generation (142). This ac- quired intrinsic derangement in cellular energy metabolism which has also been termed “cytopathic hypoxia,” contributes to reduced activities of mitochondrial electron transport chain enzyme complexes and im- paired ATP biosynthesis, potentially organ dysfunction in sepsis (141, 143, 144). Sepsis-related derangements in mito- chondrial function can activate the ubiq- uitin proteolytic pathway in skeletal mus- cle of septic patients (145). Mitochondrial permeability transition pore seems to be involved in sepsis-induced mitochondrial damage, since its inhibition significantly improved organ function and reduced mortality in rodents (146). Whether sep- sis-related reduction in energy supply could result in a state of cellular shut- down analogous to myocardial “stun- ning” (or hibernation) following coronary occlusion, allowing for eventual restora- tion of organ function and survival, has not yet been determined. Impact of Inflammation on Coagulation The clotting system is almost invari- ably activated by systemic microbial in- 297Crit Care Med 2009 Vol. 37, No. 1
  • 8. 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- ptotic microparticles have also been dem- onstrated in sepsis (153–155). Linkage of coagulation enzymes with their serine protease activity with protease activated receptors (PAR 1-4) on endothelial sur- faces increases P-selectin, cytokine produc- tion, and adhesion molecule expression, leading to microcirculatory dysfunction in severe sepsis (156, 157). Activated Protein C. Activated protein C (APC) is derived from its zymogen pro- tein C in contact with thrombin: thrombo- modulin complexes on endothelial sur- faces. Protein C was originally thought to 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- brinolytic activities (Fig. 8) (167, 168). Although an anti-inflammatory role for APC may be an indirect consequence of its ability to reduce thrombin generation, APC also has direct anti-inflammatory Figure 7. The extrinsic pathway (tissue factor pathway) is the primary mechanism by which thrombin is generated in sepsis. The intrinsic cascade (contact factor pathway) primarily serves an accessory role in amplifying the prothrombotic events that are initiated in sepsis. Thrombin, factor Xa and the TF-factor VIIa complex interact with the PAR (protease activated receptors) system and directly activate endothelial cells, platelets and white blood cells, and induce a proinflammatory response. Platelet-derived microparticles (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 insult. When bearing appropriate counter-ligands, MPs can transfer their procoagulant potential to target cells. Platelet-derived MPs can bind to soluble and immobilized fibrinogen, thus delivering procoagulant entities to the thrombus via the formation of aggregates. In vitro, interaction between endothelial MPs and monocytes promotes TF mRNA expression and TF-dependent procoagulant activity. Activated platelets and platelet-derived MPs thus amplify leukocyte-mediated tissue injury in thrombotic and inflammatory disorders. EPCR, endothelial protein C receptor; PMN, polymorphonuclear leukocyte; APC, activated protein C; ADAMST13, a disintegrin and metalloproteinase with a thrombospondin type 1 motifs 13; ULVWF, unusually large von Willebrand factor; ICAM, intracellular adhesion molecule; VCAM, vascular cell adhesion molecule-1; IL, interleukin; TNF, tumor necrosis factor; IFN, interferon. Figure 8. The homeostatic balance between thrombin and activated protein C (APC) in coagulation and inflammation is given. APC is a central endogenous anticoagulant protein with antithrombotic, anti- inflammatory, anti-apoptotic and pro-fibrinolytic activities. The ligand occupancy of endothelial protein C receptor (EPCR) switches the protease-activated receptor 1 (PAR-1)-dependent signaling specificity of thrombin from a permeability-enhancing to a barrier-protective response. Other pleiotropic effects are also explained. TAFI, thrombin activatable fibrinolysis inhibitor; NF-␬␤, nuclear factor ␬␤. 298 Crit Care Med 2009 Vol. 37, No. 1
  • 9. properties (169). APC can cleave and ac- tivate PAR1-dependent cellular pathways (170). APC competes for PAR-1 binding with thrombin, but in vitro studies sug- gest that APC is 103 - to 104 -fold less po- tent that thrombin in cleaving PAR-1. The critical receptors required for both PC activation and APC cellular signaling (i.e., thrombomodulin, EPCR and PAR-1) are co-localized in lipid rafts on endothe- lial cells (171). EPCR is associated with caveolin-1 on lipid rafts and EPCR bind- ing to the gamma-carboxyglutamic acid domain of protein C/APC leads to its dis- sociation from caveolin-1 (Fig. 7). APC then engages PAR-1 generating a protec- tive signaling pathway through coupling of PAR-1 to the pertussis toxin-sensitive G(i)-protein. Thus, when EPCR is bound by protein C, the PAR-1-dependent pro- tective signaling responses in endothelial cells can be mediated by either thrombin or APC. These results explain how PAR-1 and EPCR participate in protective sig- naling events in endothelial cells (172). Therapeutic administration of recom- binant human APC (rhAPC) is currently in use as a treatment strategy for severe sepsis patients with a high risk of death (173, 174). Genetically engineered vari- ants of APC have been designed with greater antiapoptotic activity and reduced anticoagulant activity relative to wild- type APC to increase the risk/benefit ratio of rhAPC regarding the bleeding compli- cation (175). A nonanticoagulant from of APC reduces mortality in experimental models of endotoxemia and sepsis (176). von Willebrand Factor, A Disintegrin-Like and Metalloproteinase with Thrombospondin Type-1 Motifs 13 and Ashwell Receptor. The discovery of a disintegrin-like and met- alloproteinase with thrombospondin type-1 motifs 13 (ADAMTS-13) has provided new insights in pathogenesis of thrombosis in sepsis. ADAMTS-13, the principal physio- logic modulator of von Willebrand factor (VWF) is produced mainly stellate cells in the liver (177). VWF is synthesized in vas- cular endothelial cells and released into the plasma as unusually large VWF multimers which are rapidly degraded into smaller VWF multimers by ADAMTS-13. Deficiency of the ADAMTS-13, as observed in most forms of thrombotic thrombocytopenic purpura, increases the level of unusually large VWF multimers in plasma and leads to platelet aggregation and/or thrombus formation, especially in small arterioles, re- sulting in microvascular failure (178–180). Recently, inflammation associated- ADAMTS-13 deficiency has been de- scribed in patients with systemic inflam- matory response syndrome and severe sepsis (Fig. 7) (181–185). Decreased lev- els of ADAMTS-13 have been reported in healthy volunteers following endotoxin infusion (186). Furthermore, reduced ADAMTS-13 levels are associated with dif- ferences in morbidity, mortality, and variables of inflammation and endothelial dysregulation in severe sepsis patients (182, 187). The Ashwell receptor, which is the major lectin of hepatocytes, modulates VWF homeostasis. It has been recently demonstrated that the marked thrombo- cytopenia associated with S. pneumoniae sepsis is the result of Ashwell receptor- dependent clearance of platelets (188). The ensuing reduction in platelet counts at the onset of sepsis protects the host against the development of disseminated intravascular coagulation. Homeostatic adaptation by this receptor moderates the onset and severity of disseminated intra- vascular coagulation during sepsis sug- gesting the improvement in host survival probability (189). At present, it remains unclear whether blocking the Ashwell re- ceptor may have a beneficial effect in severe sepsis. CONCLUSION A unifying concept of innate immu- nity is based upon pathogen-, or damage- associated molecular pattern molecules and downstream signaling pathways. This has facilitated significant advances in our understanding of the pathophysiology of sepsis and led to a multitude of clinical 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 Cϩ Erythropoietin Coagulation and inflammation LHRI III Recruiting Recombinant activated Protein C Coagulation Lilly III Recruiting Recombinant plasma gelsolin Replacement of circulating actin-binding protein CBC II Recruiting 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 Chicago, USA IV Recruiting Rosuvastatin Pleitrophic effects UASLP, Mexico and Beth Israel II Ongoing 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. 299Crit Care Med 2009 Vol. 37, No. 1
  • 10. trials (Table 1). However, further identi- fication of the critical elements in severe sepsis that drives the transition from lo- calized inflammation to deleterious host response remains incompletely illumi- nated. The limitations in our current knowledge of the molecular mechanisms in sepsis have made the design of inter- vention trials in clinical sepsis challeng- ing (190). New findings as to biomarkers and measures to detect genetic signa- tures of sepsis are now making their way into clinical trial designs. It is anticipated that such innovations will improve the outlook for successful development of new sepsis treatments tailored to individ- ual patient needs. REFERENCES 1. van der Poll T, Opal SM: Host-pathogen interactions in sepsis. Lancet Infect Dis 2008; 8:32–43 2. Cinel I, Dellinger RP: Making a difference in outcome in severe sepsis: The organism, the host response and the treating clinician. Emerg Med Crit Care Rev 2007; 1:1–3 3. Cinel I, Dellinger RP: Advances in patho- genesis and management of sepsis. Curr Opin Infect Dis 2007; 20:345–352 4. Uematsu S, Akira S: Toll-like receptors and innate immunity. J Mol Med 2007; 84: 712–725 5. Creagh EM, O’Neill LA: TLRs, NLRs and RLRs: A trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol 2006; 27:352–357 6. Granucci F, Foti M, Ricciardi-Castagnoli P: Dendritic cell biology. Adv Immunol 2005; 88:193–233 7. Liew FY, Xu D, Brint EK, et al: Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol 2005; 5:446–458 8. Akira S, Uematsu S, Takeuchi O: Pathogen recognition and innate immunity. Cell 2006; 124:783–801 9. Bianchi ME: DAMPs PAM. Ps and alarmins: All we need to know about danger. J Leukoc Biol 2007; 81:1–5 10. Mollen KP, Anand RJ, Tsung A, et al: Emerging paradigm: Toll-like receptor 4-sentinel for the detection of tissue dam- age. Shock 2006; 26:430–437 11. Tsujimoto H, Ono S, Hiraki S, et al: He- moperfusion with polymyxin B-immobi- lized fibers reduced the number of CD16ϩ CD14ϩ monocytes in patients with septic shock. J Endotoxin Res 2004; 10:229–237 12. Armstrong L, Medford AR, Hunter KJ, et al: Differential expression of toll-like receptor (TLR)-2 and TLR-4 on monocytes in human sepsis. Clin Exp Immunol 2004; 136: 312–319 13. Tsujimoto H, Ono S, Majima T, et al: Neu- trophil elastase, MIP-2, and TLR-4 expres- sion during human and experimental sep- sis. Shock 2005; 23:39–44 14. Williams DL, Ha T, Li C, et al: Modulation of tissue toll-like receptor 2 and 4 during the early phases of polymicrobial sepsis corre- lates with mortality. Crit Care Med 2003; 31:1808–1818 15. Paterson HM, Murphy TJ, Purcell EJ, et al: Injury primes the innate immune system for enhanced toll-like receptor reactivity. J Immunol 2003; 171:1473–1483 16. Tsujimoto H, Ono S, Efron PA, et al: Role of toll like receptors in the development of sepsis. Shock 2008; 29:315–321 17. Ogura Y, Sutterwala FS, Flavell RA: The inflammasome: First line of the immune response to cell stress. Cell 2006; 126: 659–662 18. Drenth JP, van der Meer JW: The inflamma- some—A linebacker of innate defense. N Engl J Med 2006; 355:730–732 19. Dinarello CA: Interleukin-1␤, interleukin- 18, and the interleukin-1␤ converting en- zyme. Ann NY Acad Sci 1998; 856:1–11 20. Martinon F, Burns K, Tschopp J: The in- flammasome. A molecular platform trigger- ing activation of inflammatory caspases and processing of proIL-1␤. MolCell 2002; 10: 417–426 21. Yu JW, Wu J, Zhang Z, et al: Cryopyrin and pyrin activate caspase-1, but not NF- kappaB, via ASC oligomerization. Cell Death Differ 2005; 13:236–249 22. Seshadri S, Duncan MD, Hart JM, et al: Pyrin levels in human monocytes and monocyte-derived macrophages regulate IL-1␤ processing and release. J Immunol 2007; 179:1274–1281 23. Tschopp J, Martinon F, Burns K: NALPs: A novel protein family involved in inflamma- tion. NatRevMol Cell Biol 2003; 4:95–104 24. Sarkar A, Duncan M, Hart J, et al: ASC directs NF-␬B activation by regulating re- ceptor interacting protein-2 (RIP2) caspase-1 interactions. J Immunol 2006; 176:4979–4986 25. Scott AM, Saleh M: The inflammatory caspases: Guardians against infections and sepsis. Cell Death Differ 2007; 14:23–31 26. Sarkar A, Hall MW, Exline M, et al: Caspase-1 regulates E. coli sepsis and splenic B cell apoptosis independently of IL-1␤ and IL-18. Am J Respir Crit Care Med 2006; 174:1003–1010 27. Saleh M, Vaillancourt JP, Graham RK, et al: Differential modulation of endotoxin re- sponsiveness by human caspase-12 poly- morphisms. Nature 2004; 429:75–79 28. Saleh M, Mathison JC, Wolinski MK, et al: Enhanced bacterial clearance and sepsis re- sistance in caspase-12-deficient mice. Na- ture 2006; 440:1064–1068 29. Joshi VD, Kalvakolanu DV, Hasday JD, et al: IL-18 levels and the outcome of innate im- mune response to lipopolysaccharide: Im- portance of a positive feedback loop with caspase-1 in IL-18 expression. J Immunol 2002; 169:2536–2544 30. Fahy RJ, Exline MC, Gavrilin MA, et al: Inflammasome mRNA expression in human monocytes during early septic shock. Am J Resp Crit Care Med 2008; 177:983–988. 31. Lara-Tejero M, Sutterwala FS, Ogura Y, et al: Role of the caspase-1 inflammasome in Salmonella typhimurium pathogenesis. J Exp Med 2006; 203:1407–1412 32. Brightbill HD, Libraty DH, Krutzik SR, et al: Host defense mechanisms triggered by microbial lipoproteins through toll-like re- ceptors. Science 1999; 285:732–736 33. Bruno TF, Woods DE, Mody CH: Exoen- zyme S from Pseudomonas aeruginosa in- duces apoptosis in T lymphocytes. J Leukoc Biol 2000; 67:808–816 34. Wesche H, Henzel WJ, Shillinglaw W, et al: MyD88: An adapter that recruits IRAK to the IL-1 receptor complex. Immunity 1997; 7:837–847 35. Reynaert NL, van der Vliet A, Guala AS, et al: Dynamic redox control of NF-kappaB through glutaredoxin-regulated S-gluta- thionylation of inhibitory kappaB kinase beta. Proc Natl Acad Sci USA 2006; 103: 13086–13091 36. Pahl HL: Activators and target genes of rel/ NF-kappaB transcription factors. Oncogene 1999; 18:6853–6866 37. Blackwell TS, Yull FE, Chen CL, et al: Mul- tiorgan nuclear factor kappa B activation in a transgenic mouse model of systemic in- flammation. Am J Respir Crit Care Med 2000; 162:1095–1101 38. Arcaroli J, Silva E, Maloney JP, et al: Variant IRAK-1 haplotype is associated with in- creased nuclear factor-kappaB activation and worse outcomes in sepsis. Am J Respir Crit Care Med 2006; 173:1335–1341 39. Zaph C, Troy AE, Taylor BC, et al: Epithelial cell-intrinsic IKK-beta expression regulates intestinal immune homeostasis. Nature 2007; 446:552–556 40. Liu YJ, Soumelis V, Watanabe N, et al: TSLP: An epithelial cell cytokine that regu- lates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immu- nol 2007; 25:193–219 41. Peck-Palmer OM, Unsinger J, Chang KC, et al: Deletion of MyD88 markedly attenuates sepsis-induced T and B lymphocyte apopto- sis but worsens survival. J Leukoc Biol 2008; 83:1009–1018 42. Cantley LC. The phosphoinositide 3-kinase pathway. Science 2002; 296:1655–1657 43. Okugawa S, Ota Y, Kitazawa T, et al: Janus kinase2 is involved in lipopolysaccharide- induced activation of macrophages. Am J Physiol Cell Physiol 2003; 285:C399–C408 44. Ojaniemi M, Glumoff V, Harju K, et al: Phosphatidylinositol kinase is involved in Toll-like receptor 4-mediated cytokine ex- pression in mouse macrophages. Eur J Im- munol 2003; 335:97–605 45. Guillot L, Le GofficR, BlochS, et al: Involve- ment of toll-like receptor 3 in the immune response of lung epithelial cells to double- 300 Crit Care Med 2009 Vol. 37, No. 1
  • 11. stranded RNA and influenza A virus. J Biol Chem 2005; 280:5571–5580 46. Wrann CD, Tabriz NA, Barkhausen T, et al: The phosphatidylinositol 3-kinase signaling pathway exerts protective effects during sepsis by controlling C5a-mediated activa- tion of innate immune functions. J Immu- nol 2007; 178:5940–5948 47. Guha M, Mackman N: The PI3K-Akt path- way limits LPS activation of signaling path- ways and expression of inflammatory medi- ators in human monocytic cells. J Biol Chem 2002; 277:32124–32132 48. Schabbaue G, Tencati M, Pedersen B, et al: PI3K-Akt pathway suppresses coagulation and inflammation in endotoxemic mice. Ar- terioscler Thromb Vasc Biol 2004; 24: 1963–1969 49. Pengal RA, Ganesan LP, Wei G, et al: Li- popolysaccharide-induced production of in- terleukin-10 is promoted by the serin thre- onine kinase Akt. Mol Immunol 2006; 43: 1557–1156 50. Fukao T, Koyasu S: PI3K and negative reg- ulation of TLR signaling. Trends Immunol 2003; 24:358–363 51. Bommhardt U, Chang KC, Swanson PE, et al: Akt decreases lymphocyte apoptosis and improves survival in sepsis. J Immunol 2004; 172:7583–7591 52. Mc Dunn JE, Muenzer JT, Rachdi L, et al: Peptide-mediated activation of Akt and ex- tracellular regulated kinase signaling pre- vents lymphocyte apoptosis. FASEB J 2008; 22:561–568 53. Hall A: Rho GT. Pases and the actin cy- toskeleton. Science 1998; 279:509–514 54. Ruse M, Knaus UG: New players in TLR- mediated innate immunity. Immunologic Res 2006; 34:33–48 55. Arbibe L, Mira JP, Teusch N, et al: Toll-like receptor 2-mediated NF-kappa B activation requires a Racl dependenpt pathway. Nat Immunol 2000; 1:533–540 56. Honing H, van den Berg TK, van der Pol SM, et al: Rho activation promotes transen- dothelial migration of monocytes via ROCK. J Leukoc Biol 2004; 75:523–528 57. Tasaka S, Koh H, Yamada W, et al: Attenu- ation of endotoxin-induced acute lung in- jury by the Rho-associated kinase inhibitor, Y-27632. Am J Respir Cell Mol Biol 2005; 32:504–510 58. Thorlacius K, Slotta JE, Laschke MW, et al: Protective effect of fasudil, a Rho-kinase inhibitor, on chemokine expression, leuko- cyte recruitment, and hepatocellular apo- ptosis in septic liver injury. J Leukoc Biol 2006; 79:923–931 59. Hori S, Nomura T, Sakaguchi S: Control of regulatory T cell development by the tran- scription factor FoxP3. Science 2003; 299: 1057–1061 60. Zheng SG, Wang JH, Koss MN, et al: CD4ϩ and CD8ϩ regulatory T cells generated ex vivo with IL-2 and TGF-beta suppress a stimulatory Graft-versus- Host disease with a Lupus-like syndrome. J Immunol 2004; 172:1531–1539 61. Ausubel FM: Are innate immune signaling pathways in plants and animals conserved? Nat Immunol 2005; 6:973–979 62. Caramalho I, Lopes-Carvalho T, Ostler D, et al: Regulatory T cells selectively express toll-like receptors and are activated by lipo- polysaccharide. J Exp Med 2003; 197: 403–411 63. Sutmuller RPM, den Brok MH, Kramer M, et al: Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest 2006; 116:485–494 64. Liu H, Komai-Koma M, Xu D, et al: Toll-like receptor 2 signaling modulates the func- tions of CD4ϩCD25ϩ regulatory T cells. Proc Natl Acad Sci USA 2006; 103: 7048–7053 65. McHugh RS, Shevach EM: The role of sup- pressor T cells in regulation of immune responses. J Allergy Clin Immunol 2002; 110:693–702 66. Murphy TJ, Choileain NN, Zang Y, et al: CD4ϩ CD25ϩ regulatory T cells control in- nate immune reactivity after injury. J Im- munol 2005; 174:2957–2963 67. Monneret G, Debard AL, Venet F, et al: Marked elevation of human circulating CD4ϩCD25ϩ regulatory T cells in sepsis- induced immunoparalysis. Crit Care Med 2003; 31:2068–2071 68. Venet F, Pachot A, Debard AL, et al: In- creased percentage of CD4ϩCD25ϩ regula- tory T cells during septic shock is due to the decrease of CD4ϩCD25-lymphocytes. Crit Care Med 2004; 32:2329–2331 69. Takahashi H, Tsuda Y, Takeuchi D, et al: Influence of systemic inflammatory re- sponse syndrome on host resistance against bacterial infections. Crit Care Med 2004; 32:1879–1885 70. Tiemessen MM, Jagger AL, Evans HG, et al: CD4ϩCD25ϩFoxp3ϩ regulatory T cells in- duce alternative activation of human mono- cytes/macrophages. Proc Natl Acad Sci USA 2007; 104:19446–19451 71. Wisnoski N, Chung CS, Chen Y, et al: The contribution of CD4ϩ CD25ϩ T-regulato- ry-cells to immune suppression in sepsis. Shock 2007; 27:251–257 72. van Maren WW, Jacobs JF, de Vries IJ, et al: Toll-like receptor signalling on Tregs: To suppress or not to suppress? Immunology 2008; 124:445–452. 73. Heuer JG, Zhang T, Zhao J, et al: Adoptive transfer of in vitro-stimulated CD4ϩ CD25ϩ regulatory T cells increases bacterial clear- ance and improves survival in polymicrobial sepsis. J Immunol 2005; 174:7141–7146 74. Venet F, Pachot A, Debard AL, et al: Human CD4ϩCD25ϩ regulatory T lymphocytes in- hibit lipopolysaccharide-induced monocyte survival through a Fas/Fas ligand-depen- dent mechanism. J Immunol 2006; 177: 6540–6547 75. Brown KA, Brain SD, Pearson JD, et al: Neutrophils in development of multiple or- gan failure in sepsis. Lancet 2006; 368: 157–169 76. Marshal JC: The pathogenesis and molecu- lar biology of sepsis. Crit Care Resusc 2006; 8:227–229 77. Chandra A, Enkhbaatar P, Nakano Y, et al: Sepsis: Emerging role of nitric oxide and selectins. Clinics 2006; 61:71–76 78. Hu G, Predescu D, Vogel SM: ICAM-1 de- pendent neutrophil adhesion to endothelial cells increases caveolae-mediated pulmo- nary vascular albumin permeability. Crit Care Med 2007; 34 (Suppl 12): A31 79. Garrean S, Gao X-P, Brovkovych V, et al: Caveolin-1 regulates NF-kB activation and lung inflammatory response to sepsis in- duced by lipopolysaccharide. J Immunol 2006 177: 4853–4860 80. Wang XM, Kim HP, Song R, et al: Caveo- lin-1 confers antiinflammatory effects in murine macrophages via the MKK3/p38 MAPK pathway. Am J Respir Cell Mol Biol 2006; 34:434–442 81. Medina FA, de Almeida CJ, Dew E, et al: Caveolin-1 deficient mice show defects in innate immunity and inflammatory im- mune response during Salmonella enterica serovar typhimurium infection. Infect Im- mun 2006; 74:6665–6674 82. Medina FA, Cohen AW, de Almeida CJ, et al: Immune dysfunction in caveolin-1 null mice following infection with Trypanosoma cruzi (Tulahuen strain). Microbes Infect 2007; 9:325–333 83. Astiz ME, DeGent GE, Lin RY, et al: Micro- vascular function and rheologic changes in hyperdynamic sepsis. Crit Care Med 1995; 23:265–271 84. Ozdulger A, Cinel I, Koksel O, et al: The protective effect of N-acetylcysteine on ap- optotic lung injury in cecal ligation and puncture-induced sepsis model. Shock 2003; 19:366–372 85. Stehr SN, Knels L, Weissflog C, et al: Effects of IgM enriched solution on polymorpho- nuclear neutrophil function, bacterial clearance, and lung histology in endotox- emia. Shock 2007; 29:167–172 86. Kabay B, Kocaefe C, Baykal A, et al: Inter- leukin-10 gene transfer: Prevention of mul- tiple organ injury in a murine cecal ligation and puncture model of sepsis. World J Surg 2007; 31:105–115 87. Ozdulger A, Cinel I, Unlu A, et al: Poly (ADP-ribose) synthetase inhibition prevents lipopolysaccharide-induced peroxynitrite mediated damage in diaphragm. Pharmacol Res 2002; 46:67–73 88. Taner S, Cinel I, Ozer L, et al: Poly (ADP- ribose) synthetase inhibition reduces bacte- rial translocation in rats after endotoxin challenge. Shock 2001; 16:159–162 89. Cinel I, Buyukafsar K, Cinel L, et al: The role of poly (ADP-ribose) synthetase inhibi- tion in preventing endotoxemia-induced in- testinal epithelial apoptosis. Pharmacol Res 2002; 46:119–127 301Crit Care Med 2009 Vol. 37, No. 1
  • 12. 90. Savill J: Apoptosis in resolution of inflam- mation. J Leukoc Biol 1997; 61:375–380 91. Maianski NA, Geissler J, Srinivasula SM, et al: Functional characterization of mito- chondria in neutrophils: A role restricted to apoptosis. Cell Death Differ 2004; 11: 143–153 92. van Raam BJ, Verhoeven AJ, Kuijpers TW: Mitochondria in neutrophil apoptosis. Int J Hematol 2006; 84:199–204 93. Sookhai S, Wang JJ, McCourt M, et al: A novel therapeutic strategy for attenuating neutrophil-mediated lung injury in vivo. Ann Surg 2002; 235:285–291 94. Taneja R, Parodo J, Kapus A, et al: Delayed neutrophil apoptosis in sepsis is associated with maintenance of mitochondrial trans- membrane potential and reduced caspase-9 activity. Crit Care Med 2004; 32:1460–1469 95. Fadok VA, Bratton DL, Konowal A, et al: Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cyto- kine production through autocrine/para- crine mechanisms involving TGF, PGE2, and PAF. J Clin Invest 1998; 101:890–898 96. Sanford AN, Suriano AR, Herche D, et al: Abnormal apoptosis in chronic granuloma- tous disease and autoantibody production characteristic of lupus. Rheumatology 2006; 45:178–181 97. Daleke DL: Regulation of transbilayer plasma membrane phospholipid asymme- try. J Lipid Res 2003; 44:233–242 98. Bayir H, Kagan VE: Bench-to-bedside re- view: Mitochondrial injury, oxidative stress and apoptosis—there is nothing more prac- tical than a good theory. Crit Care 2008; 12:206 99. Tyurina YY, Basova LV, Konduru NV, et al: Nitrosative stress inhibits the aminophos- pholipid translocase resulting in phosphati- dylserine externalization and macrophage engulfment: Implications for the resolution of inflammation. J Biol Chem 2007; 282: 8498–8509 100. Swan R, Chung CS, Albina J, et al: Polymi- crobial sepsis enhances clearance of apopto- tic immune cells by splenic macrophages. Surgery 2007; 142:253–261 101. Macdonald J, Galley HF, Webster NR: Oxi- dative stress and gene expression in sepsis. Br J Anaest 2003; 90:221–232 102. Matejovic M, Krouzecky A, Rokyta R Jr, et al: Effects of combining inducible nitric ox- ide synthase inhibitor and radical scavenger during porcine bacteremia. Shock 2007; 27: 61–68 103. Fialkow L, Wang Y, Downey GP: Reactive oxygen and nitrogen species as signaling molecules regulating neutrophil function. Free Radic Biol Med 2007; 42:153–164 104. Galley HF, Davies MJ, Webster NR: Xan- thine oxidase activity and free radical gen- eration in patients with sepsis syndrome. Crit Care Med 1996; 24:1649–1653 105. Soriano FG, Nogueira AC, Caldini EG, et al: Potential role of poly (adenosine 5Ј- diphosphate-ribose) polymerase activation in the pathogenesis of myocardial contrac- tile dysfunction associated with human sep- tic shock. Crit Care Med 2006; 34: 1073–1079 106. Ulloa L, Tracey KJ: The “cytokine profile”: A code for sepsis. Trends Mol Med 2005; 11: 56–63 107. Zimmermann K, Volkel D, Pable S, et al: Native versus recombinant high-mobility group B1 proteins: functional activity in vitro. Inflammation 2004; 28:221–229 108. Rouhiainen A, Tumova S, Valmu L, et al: Pivotal advance: Analysis of proinflamma- tory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). J Leu- kocyte Biol 2007; 81:49–58 109. Bonaldi T, Talamo F, Scaffidi P, et al: Mono- cytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J 2003; 22:5551–5560 110. Youn JH, Shin JS: Nucleocytoplasmic shut- tling of HMGB1 is regulated by phosphory- lation that redirects it toward secretion. J Immunol 2006; 177:7889–7897 111. Bell CW, Jiang W, Reich CF III, et al: The extracellular release of HMGB1 during ap- optotic cell death. Am J Physiol Cell Physiol 2006; 291:C1318–C1325 112. Qin S, Wang H, Yuan R, et al: Role of HMGB1 in apoptosis-mediated sepsis lethal- ity. J Exp Med 2006; 203:1637–1642 113. Hori O, Brett J, Slattery T, et al: The recep- tor for advanced glycation end products (RAGE) is a cellular binding site for ampho- terin: Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J Biol Chem 1995; 270:25752–25761 114. Park JS, Svetkauskaite D, He Q, et al: In- volvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem 2004; 279: 7370–7377 115. Wang H, Bloom O, Zhang M, et al: HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999; 285:248–251 116. Mitola S, Belleri M, Urbinati C, et al: Cut- ting edge: Extracellular high mobility group box-1 protein is a proangiogenic cy- tokine. J Immunol 2006; 176:12–15 117. Tzeng HP, Fan J, Vallejo JG, et al: Negative inotropic effects of high-mobility group box 1 protein in isolated contracting cardiac myocytes. Am J Physiol Heart Circ Physiol 2008; 294:H1490–H1496 118. Wang H, Yu M, Ochani M, et al: Nicotinic acetylcholine receptor ␣7 receptor is an es- sential regulator of inflammation. Nature 2003; 421:384–388 119. Pavlov VA, Wang H, Czura CJ, et al: The cholinergic anti-inflammatory pathway: A missing link in neuromodulation. Mol Med 2003; 9:125–134 120. Wang H, Liao H, Ochani M, et al: Cholin- ergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat Med 2004; 10:1216–1221 121. Huston JM, Gallowitsch-Puerta M, Ochani M, et al: Transcutaneous vagus nerve stim- ulation reduces serum high mobility group box 1 levels and improves survival in mu- rine sepsis. Crit Care Med 2007; 35: 2762–2768 122. Goldstein RS, Bruchfeld A, Yang L, et al: Cholinergic anti-inflammatory pathway ac- tivity and High Mobility Group Box-1 (HMGB1) serum levels in patients with rheumatoid arthritis. Mol Med 2007; 13(3–4):210–215 123. Youn JH, Oh YJ, Kim ES, et al: High Mo- bility Group Box 1 protein binding to lipo- polysaccharide facilitates transfer of lipo- polysaccharide to CD14 and enhances lipopolysaccharide-mediated TNF-{alpha} production in human monocytes. J Immu- nol 2008; 180:5067–5074 124. Sha Y, Zmijewski J, Xu Z, et al: HMGB1 develops enhanced proinflammatory activ- ity by binding to cytokines. J Immunol 2008; 180:2531–2537 125. Sunde´n-Cullberg J, Norrby-Teglund A, Rou- hiainen A, et al: Persistent elevation of high mobility group box-1 protein (HMGB1) in patients with severe sepsis and septic shock. Crit Care Med 2005; 33: 564–573 126. Gibot S, Massin F, Cravoisy A, et al: High- mobility group box 1 protein plasma con- centrations during septic shock. Intensive Care Med 2007; 33:1347–1353 127. Karlsson S, Pettila¨ V, Tenhunen J, et al: HMGB1 as a predictor of organ dysfunction and outcome in patients with severe sepsis. Intensive Care Med 2008; 34:1046–1053. 128. Harris HE, Raucci A: Alarmin(g) news about danger. EMBO Rep 2006; 7:774–778 129. Chavakis T, Bierhaus A, Al-Fakhri N, et al: The pattern recognition receptor (RAGE) is a counterreceptor for leukocyte integrins: A novel pathway for inflammatory cell re- cruitment. J Exp Med 2003; 198:1507–1515 130. Lutterloh EC, Opal SM, Pittman DD, et al: Inhibition of the RAGE products increases survival in experimental models of severe sepsis and systemic infection. Crit Care 2007; 11:R122 131. Calandra T, Bernhagen J, Metz CN, et al: MIF as a glucocorticoid-induced modulator of cytokine production. Nature 1995; 377: 68–71 132. Calandra T, Echtenacher B, Roy DL, et al: Protection from septic shock by neutraliza- tion of macrophage migration inhibitory factor. Nat Med 2000; 6:164–170 133. Bozza M, Satoskar AR, Lin G, et al: Targeted disruption of migration inhibitory factor gene reveals its critical role in sepsis. J Exp Med 1999; 189:341–346 134. Roger T, David J, Glauser MP, et al: MIF regulates innate immune responses through modulation of Toll-like receptor 4. Nature 2001; 414:920–924 135. Mitchell RA, Liao H, Chesney J, et al: Mac- rophage migration inhibitory factor (MIF) sustains macrophage proinflammatory function by inhibiting p53: Regulatory role 302 Crit Care Med 2009 Vol. 37, No. 1
  • 13. in the innate immune response. Proc Natl Acad Sci USA 2002; 99:345–350 136. Kreymann G, Grosser S, Buggisch P, et al: Oxygen consumption and resting metabolic rate in sepsis, sepsis syndrome, and septic shock. Crit Care Med 1993; 21:1012–1019 137. Rosser DM, Stidwill RP, Jacobson D, et al: Oxygen tension in the bladder epithelium rises in both high and low cardiac output endotoxemic sepsis. J Appl Physiol 1995; 79:1878–1882 138. Hotchkiss RS, Swanson PE, Freeman BD, et al: Apoptotic cell death in patients with sep- sis, shock, and multiple organ dysfunction. Crit Care Med 1999; 27:1230–1251 139. Brealey D, et al: Mitochondrial dysfunction in a long-term rodent model of sepsis and organ failure. Am J Physiol Regul Integr Comp Physiol 2004; 286:R491–R497 140. Crouser ED: Mitochondrial dysfunction in septic shock and multiple organ dysfunc- tion syndrome. Mitochondrion 2004; 4:729–741 141. Fink MP: Cytopathic hypoxia. Mitochon- drial dysfunction as mechanism contribut- ing to organ dysfunction in sepsis. Crit Care Clinics 2001; 17:219–237 142. Brealey D, Brand M, Hargreaves I, et al: Association between mitochondrial dys- function and severity and outcome of septic shock. Lancet 2002; 360:219–223 143. Crouser ED, Julian MW, Blaho DV, et al: Endotoxin-induced mitochondrial damage correlates with impaired respiratory activ- ity. Crit Care Med 2002; 30:276–284 144. Levy RJ, Vijayasarathy C, Raj NR, et al: Competitive and noncompetitive inhibition of myocardial cytochrome C oxidase in sep- sis. Shock 2004; 21:110–114 145. Rabuel C, Renaud E, Brealey D, et al: Hu- man septic myopathy: Induction of cycloox- ygenase, heme oxygenase and activation of the ubiquitin proteolytic pathway. Anesthe- siology 2004; 101:583–590 146. Larche J, Lancel S, Hassoun SM, et al: In- hibition of mitochondrial permeability transition prevents sepsis-induced myocar- dial dysfunction and mortality. J Am Coll Cardiol 2006; 48:377–385 147. Levi M, Ten Cate H: Disseminated intravas- cular coagulation. N Engl J Med 1999; 341: 586–592 148. Schouten M, Wiersinga WJ, Levi M, et al: Inflammation, endothelium, and coagula- tion in sepsis. J Leukoc Biol 2008; 83: 536–545 149. Opal SM: The nexus between systemic in- flammation and disordered coagulation in sepsis. J Endotoxin Res 2004; 10:125–129 150. Esmon CT: Are natural anticoagulants can- didates for modulating the inflammatory response to endotoxin? Blood 2000; 95: 1113–1116 151. Johnson K, Choi Y, DeGroot E, et al: Po- tential mechanisms for a proinflamma- tory vascular cytokine response to coagu- lation activation. J Immunol 1998; 160: 5130–5135 152. Stouthard JM, Levi M, Hack CE, et al: In- terleukin-6 stimulates coagulation, not fi- brinolysis, in humans. Thromb Haemost 1996; 76:738–742 153. Aupeix K, Hugel B, Martin T, et al: The significance of shed membrane particles during programmed cell death in vitro, and in vivo, in HIV-1 infection. J Clin Invest 1997; 99:1546–1554 154. Soriano AO, Jy W, Chirinos JA, et al: Levels of endothelial and platelet microparticles and their interactions with leukocytes neg- atively correlate with organ dysfunction and predict mortality in severe sepsis. Crit Care Med 2005; 33:2540–2546 155. Fujimi S, Ogura H, Tanaka H, et al: Acti- vated polymorphonuclear leukocytes en- hance production of leukocyte micropar- ticles with increased adhesion molecules in patients with sepsis. J Trauma 2002; 52: 443–448 156. Amaral A, Opal SM, Vincent JL: Coagulation in sepsis. Intensive Care Med 2004; 30: 1032–1040 157. Vervloet MG, Thijs LG, Hack CE: Derange- ments of coagulation and fibrinolysis in critically ill patients with sepsis and septic shock. Semin Thromb Hemost 1998; 24: 33–44 158. Yan SB, Dhainut JF: Activated protein C versus protein C in severe sepsis. Crit Care Med 2001; 29(Suppl):S69–S74 159. Xue M, Campbell D, Jackson CJ: Protein C is an autocrine growth factor for human skin keratinocytes, J Biol Chem 2007; 282: 13610–13616 160. Xue M, March L, Sambrook PN, et al: Endothelial protein C receptor is overex- pressed in rheumatoid arthritic (RA) sy- novium and mediates the anti-inflamma- tory effects of activated protein C in RA monocytes. Ann Rheum Dis 2007; 66: 1574–1580 161. Regan LM, Stearns-Kurosawa DJ, Kurosawa S, et al: The endothelial cell protein C re- ceptor: Inhibition of activated protein C an- ticoagulant function without modulation of reaction with proteinase inhibitors. J Biol Chem 1996; 271:17499–17503 162. Kurosawa S, Stearns-Kurosawa DJ, Carson CW, et al: Plasma levels of endothelial cell protein C receptor are elevated in patients with sepsis and systemic lupus erythemato- sus: Lack of correlation with thrombo- modulin suggests involvement of different pathological processes. Blood 1998; 91: 725–727 163. Yan SB, Helterbrand JD, Hartman DL, et al: Low levels of protein C are associated with poor outcome in severe sepsis. Chest 2001; 120:915–922 164. LaRosa SP, Opal SM, Utterback B, et al: Decreased protein C, protein S, and anti- thrombin levels are predictive of poor out- come in Gram-negative sepsis caused by Burkholderia pseudomallei. Int J Infect Dis 2006; 10:25–31 165. Yan SB, Dhainaut JF: Activated protein C versus protein C in severe sepsis. Crit Care Med 2001; 29(Suppl):S69–S74 166. Joyce DE, Gelbert L, Ciaccia A, et al: Gene expression profile of antithrombotic protein C defines new mechanisms modulating in- flammation and apoptosis. J Biol Chem 2001; 14:11199–111203 167. Jackson CJ, Xue M: Activated protein C-An anticoagulant that does more than stop clots. Int J Biochem Cell Biol 2008; 40: 2692–2697 168. Dutt T, Toh CH: The yin-yang of thrombin and activated protein C. Br J Haematology 2008; 140:505–515 169. Riewald M, Ruf W: Protease-activated recep- tor-1 signaling by activated protein C in cytokine-perturbed endothelial cells is dis- tinct from thrombin signaling. J Biol Chem 2005; 280:19808–19814 170. Bae JS, Yang L, Rezaie AR: Receptors of the protein C activation and activated protein C signaling pathways are colocalized in lipid rafts of endothelial cells. Proc Natl Acad Sci USA 2007; 104:2867–2872 171. Bae JS, Yang L, Manithody C, et al: The ligand occupancy of endothelial protein C receptor switches the protease-activated receptor 1-dependent signaling specificity of thrombin from a permeability-enhanc- ing to a barrier-protective response in endothelial cells. Blood 2007; 110: 3909–3916 172. Shorr AF, Bernard GR, Dhainaut JF, et al: Protein C concentrations in severe sepsis: An early directional change in plasma levels predicts outcome. Crit Care 2006; 10:R92 173. Cinel I, Dellinger RP: Current treatment of severe sepsis. Curr Infect Dis Rep 2006; 8:358–365 174. Dellinger RP, Levy MM, Carlet JM, et al: Surviving Sepsis Campaign: International guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med 2008; 36:296–327 175. Bae JS, Yang L, Manithody C, et al: Engi- neering a disulfide bond to stabilize the calcium binding loop of activated protein C eliminates its anticoagulant but not protec- tive signaling properties. J Biol Chem 2007; 282:9251–9259 176. Kerschen EJ, Fernandez JA, Cooley BC, et al: Endotoxemia and sepsis mortality reduc- tion by non-anticoagulant activated protein C. J Exp Med 2007; 204:2439–2448 177. Moake JL: Thrombotic microangiopathies. N Engl J Med 2002; 347:589–600 178. Ruggeri ZM: Von Willebrand factor, plate- lets and endothelial cell interactions. J Thromb Haemost 2003; 1:1335–1342 179. Furlan M, Robles R, Galbusera M, et al: von Willebrand factor-cleaving protease in thrombotic thrombocytopenic purpura and the hemolytic-uremic syndrome. New Engl J Med 1998; 339:1578–1584 180. Arya M, Anvari B, Romo GM, et al: Ultra- large multimers of von Willebrand factor form spontaneous high-strength bonds 303Crit Care Med 2009 Vol. 37, No. 1
  • 14. with the platelet glycoprotein Ib-IX com- plex: Studies using optical tweezers. Blood 2002; 99:3971–3977 181. Bianchi V, Robles R, Alberio L, et al: Von Willebrand factor-cleaving protease (AD- AMTS13) in thrombocytopenic disorders: A severely deficient activity is specific for thrombotic thrombocytopenic purpura. Blood 2002; 100:710–713 182. Ono T, Mimuro J, Madoiwa S, et al: Severe secondary deficiency of von Willebrand fac- tor-cleaving protease (ADAMTS13) in pa- tients with sepsis-induced disseminated in- travascular coagulation: Its correlation with development of renal failure. Blood 2006; 107:528–534 183. Nguyen TC, Liu A, Liu L, et al: Acquired ADAMTS-13 deficiency in pediatric pa- tients with severe sepsis. Haematologica 2007; 92:121–124 184. Kremer Hovinga JA, Zeerleder S, Kessler P, et al: ADAMTS-13, von Willebrand fac- tor and related parameters in severe sep- sis and septic shock. J Thromb Haemost 2007; 5:2284–2290 185. Bockmeyer CL, Claus RA, Budde U, et al: Inflammation-associated ADAMTS13 defi- ciency promotes formation of ultra-large von Willebrand factor. Haematologica 2008; 93:137–140 186. Reiter RA, Varadi K, Turecek PL, et al: Changes in ADAMTS13 (von-Willebrand factor-cleaving protease) activity after in- duced release of von Willebrand factor during acute systemic inflammation. Thromb Haemost 2005; 93:554–558 187. Martin K, Borgel D, Lerolle N, et al: De- creased ADAMTS-13 (A disintegrin-like and metalloprotease with thrombospondin type 1 repeats) is associated with a poor progno- sis in sepsis-induced organ failure. Crit Care Med 2007; 35:2375–2382 188. Grewal PK, Uchiyama S, Ditto D, et al: The Ashwell receptor mitigates the lethal coagulopa- thy of sepsis. Nat Med 2008; 14:648–655 189. van’t Veer C, van der Poll T. Keeping blood clots at bay in sepsis. Nat Med 2008; 14: 606–608 190. Treziack S, Cinel I, Dellinger RP, et al: Resuscitating the microcirculation in se- vere sepsis: The central role of nitric oxide, emerging concepts for novel therapies, and challenges for clinical trials. Acad Emerg Med 2008; 15:1–15 304 Crit Care Med 2009 Vol. 37, No. 1