You are on page 1of 12

Semin Immunopathol

DOI 10.1007/s00281-017-0639-8

REVIEW

Cytokine storm and sepsis disease pathogenesis


Benjamin G. Chousterman 1,2 & Filip K. Swirski 3 & Georg F. Weber 4

Received: 11 December 2016 / Accepted: 5 May 2017


# Springer-Verlag Berlin Heidelberg 2017

Abstract Infectious diseases are a leading cause of death Keywords Cytokine . Inflammation . Innate immunity .
worldwide. Sepsis is a severe clinical syndrome related to Sepsis . Endothelium
the host response to infection. The severity of infections is
due to an activation cascade that will lead to an
autoamplifying cytokine production: the cytokine storm. Introduction
Cytokines are a broad category of relatively small proteins
(<40 kDa) that are produced and released with the aim of cell The World Health Organization (WHO) identifies infectious
signaling. Our understanding of the processes that trigger this diseases as a major cause of death worldwide, especially in
tremendous amount of cytokine production has made dramat- low-income countries where they represent the main cause of
ic progress over the last decades, but unfortunately, these find- death in 2012. Since the Italian physician Girolamo Fracastoro
ings could not translate yet into effective treatments; so far, all hypothesized in 1546 that some diseases could be caused by
clinical trials targeting cytokine production or effects failed. Bgerms^ and the work of Louis Pasteur and Robert Koch on
This review aims to summarize the pathophysiology of the the transmission of infectious disease, the understanding of
cytokine storm; to describe the type, effects, and kinetics of infection pathophysiology has been a major domain of re-
cytokine production; and to discuss the therapeutic challenges search for scientists and physicians.
of targeting cytokines. New promising therapeutic strategies The technical and conceptual advances of the last decades
focusing on the endothelium, as a source and a target of cyto- accelerated the discovery of mechanisms involved in the host
kines, are described. response to infection; this has generated a tremendous amount
of data that is still accumulating at an exponential rhythm.
The Bbig picture^ is still blurry, but it appears that infec-
tions cause damage not only because of the virulence of the
This article is a contribution to the special issue on Cytokine Storm in germ but also because of the host response after the detection
Infectious Diseases - Guest Editor: John Teijaro of molecular patterns that can be found in the microorganisms
(the pathogen-associated molecular patterns (PAMPs)).
* Benjamin G. Chousterman Infection will cause a local reaction that will extend to the
benjamin.chousterman@aphp.fr entire body mainly through the circulation of immune cells
and soluble mediators.
1
Département d’Anesthésie-Réanimation, Hôpitaux Universitaires Host response to infection is therefore not only an imme-
Lariboisière–Saint-Louis, AP-HP, Paris, France diate and local process but a complex time- and space-
2
Inserm U1160, Hôpital Saint-Louis, Paris, France compartmentalized process that involves various cell types
3
Center for Systems Biology, Department of Imaging, Massachusetts
(especially leukocytes and endothelial cells). The release of
General Hospital, Harvard Medical School, Boston, MA 02114, Binflammatory^ cytokines will induce new cytokine produc-
USA tion and release that will in turn cause cell and organ damage.
4
Department of Surgery, University of Erlangen-Nürnberg, This Bcytokine storm^ or Bcytokine cascade^ is likely re-
Erlangen, Germany sponsible for the diverse, local and remote, signs associated
Semin Immunopathol

with infection (Fig. 1). Eventually, when a threshold is inflammation [10, 11]. Actually, it appears that innate immu-
crossed, a severe clinical syndrome, Bsepsis,^ can be ob- nity can be trained and be based on a memory that will last
served. It has a dramatic impact on morbidity and mortality. through epigenetic regulation [12, 13].
The term Bcytokine storm^ was first used by Ferrara et al. in Innate immunity is of variable forms but is remarkably
1993 to describe the effects of graft-versus-host disease conserved through evolution, and most of the species share a
(GVHD) [1]. In 2003, the cytokine storm was shown to be bundle of common responses to infectious stimuli. The leuko-
associated with reaction to influenza [2] and subsequently to cytes that are considered to be the key actors of the innate
various viral, bacterial, or fungal infections [3]. However, response are the monocytes/macrophages, neutrophils, eosin-
there is no definition of what a cytokine storm is. ophils, basophils, and natural killers (NKs).
The understanding that the global effects of an infection are The activation of innate immunity is made through the
mostly due to the host response was acknowledged in the first pattern recognition receptors (PRRs).
consensual definition of sepsis in the early 1990s as the Leukocyte activation by the PRRs will occur after the bind-
Bsystemic response to an infection^ which is characterized ing of a wide variety of molecules originating from the infecting
by a widespread inflammation [4]. Thus, the diagnosis of sep- microorganism, the pathogen-associated molecular patterns
sis necessitated the presence of a systemic inflammatory re- (PAMPs), or from necrotic cells, the damage-associated molec-
sponse syndrome (SIRS) and a proven or suspected infection. ular patterns (DAMPs). The recognition of PAMPs or DAMPs
In 2016, a new definition of sepsis emerged (sepsis 3.0) and will lead to a cascade of activation/phosphorylation inducing a
states that BSepsis is life-threatening organ dysfunction due to stereotyped inflammatory answer.
a dysregulated host response to infection^ [5]. The clinical To date, four types of PRRs have been identified in verte-
criteria for the diagnosis of sepsis have evolved to an elevation brates: the Toll-like receptors (TLRs), the Nod-like receptors
of at least 2 points of the sepsis occurrence of failing organs (NLRs), the RIG-like receptors (RLRs), and the C-type lectin
score (SOFA) and a suspected or proven infection. Recent receptors (CLRs). The most studied PRRs are the TLRs, par-
data regarding sepsis epidemiology show that the incidence ticularly TLR4. Pioneer work by Drs. Hoffman, Beutler,
of sepsis is dramatically high, close to 450 per 100,000 inhab- Janeway, and Medzhitov (among others) [14, 15] led to the
itants [6], far more than that of myocardial infarction [7] or the discovery of TLRs in both drosophil and man and paved the
combination of prostate, breast, and lung cancer [8]. It is esti- way to the discovery of the innate immune system. The TLR4
mated that there are between 15 and 30 million of sepsis cases was identified as the receptor for lipopolysaccharide (LPS)
worldwide every year [6, 9]. With a nearly 20% mortality rate, [16, 17], which is a wall compound of gram-negative bacteria
it clearly appears that this cytokine storm-induced syndrome is and is studied in depth to investigate host response after
a major public health issue. Unfortunately, despite immense Bendotoxemia,^ a model aimed to mimic the response to a
progress in the Bomics^ (genomic, transcriptomic, proteomic, bacterium. To date, 10 TLRs have been identified in man
metabolomic) of sepsis, all clinical trials aiming to dampen the (12 in mouse) [18]. TLRs have a leucin-rich repeat extracel-
inflammatory response or targeting the cytokines failed. The lular domain and an intracellular Toll-interleukin-1 receptor
translation of basic science findings and clinical data into ef- domain (TIR) [19]. The list of the ligands binding to TLRs
fective therapeutics is highly complex and will necessitate an is still expanding (Table 1).
integrated view of the orchestration of the cytokine storm and The interaction between TLRs and their ligands induces,
its effects. through the TIR domain, a cascade of activation via various
In this review, we will detail the mechanisms leading to the mediators such as myeloid differentiation protein 88
induction of the cytokine storm during sepsis and describe (MyD88), TIR domain-containing adaptor protein (TIRAP),
how and what cytokines are released and how they may con- TIR receptor-inducing interferon-beta (TRIF), TRIF-related
tribute to cell or organ damage. Finally, we will discuss the adaptor molecule (TRAM), and several tyrosine kinases. As
various therapeutic interventions tried or to be tried to stop or a result of these signaling cascades, transcription factors such
quell this deleterious process. as NFκB will induce the generation of inflammatory cyto-
kines such as TNF-α, IL-1β, interferon regulatory factor 3
(IRF3), IRF7, or adaptor-protein 1 (AP-1) [18, 20]. These
Sepsis pathophysiology—activation of innate pathways are tightly modulated by cytoplasmic (IRAK-M,
immunity Tollip, SOCS1) or membrane-bound (SIGIRR, ST2) proteins
[21]. Regulation of TLR signaling is also mediated by a tight
When microorganisms penetrate into the body, they will even- control of TLR expression at the cell membrane. Septic pa-
tually be spotted by the innate immune system. Innate immu- tients have higher levels of TLR4 mRNA and higher levels of
nity is the ensemble of cellular and humoral mechanisms that TLR2 receptors [22, 23].
does not need training (i.e., previous exposure to a germ) and NLRs are soluble cytosolic PRRs; they share a common
that will act quasi-automatically against an insult and generate nucleotide-binding oligomerization domain (NOD) and have
Semin Immunopathol

Fig. 1 Cytokine cascade during sepsis

a LRR domain (like the TLRs). The two most studied NLRs There are three RLRs discovered to date (RIG-I,
are NOD1 and NOD2, which bind bacterial peptidoglycan. MDA5, and LGP2), and they bind double-stranded viral
Activation of the NLR pathway is under the control of the RNA (dsRNA) [32].
RICK kinase and induces NFκB and AP-1. Other NLRs As said earlier, PRRs are activated not only by exogenous
(NLRP, NLRC4) will be involved in the constitution of a PAMPs but also by endogenous DAMPs. Microorganism cell
protein complex named inflammasome. Inflammasome in- toxicity and toxins in addition to the effects of released cyto-
duces the cleavage of pro-caspase 1 into active caspase 1 that kines will induce a large amount of cell death that will release
in turn will cleave pro-IL-1β and pro-IL-18 intro IL-1β and DAMPs. Thus, an autoamplified activation cascade will be
IL-18, two inflammatory cytokines [24, 25]. initiated and will rapidly be independent of any stimuli com-
The CLR family includes dectins, DC-SIGN, or mannose- ing from the invading microorganism.
binding lectin. Dectins signal through Src and Syk and induce In recent years, a significant body of literature has been
ROS production [26–28]. DC-SIGN is involved in the recog- published to show the very crucial role of the endothelium
nition of patterns from viruses (HIV, HCV, dengue virus, etc.), during infection and sepsis [33–35]. Endothelial cells should
leishmaniae, and fungi [29, 30]. Its signaling pathway goes be considered not only as a victim of the inflammatory acti-
through Raf-1 [31]. vation but also as a conveyor and amplifier belt for

Table 1 Toll-like receptor (TLR)


ligands TLR1 Triacyl lipoproteins
TLR2 Lipoproteins, gram-positive peptidoglycan, lipotechoic acid, amphotericin B, saturated fatty acids,
glycoproteins gB and gH, zymosan, HA
TLR3 Double-stranded RNA, polyinosinic-polycytidylic acid (poly I:C)
TLR4 Lipopolysaccharide, RSV fusion protein
TLR5 Flagellin
TLR6 Diacyl lipoproteins, lipotechoic acid, zymosan
TLR7 Single-stranded RNA
TLR8 Imidazoquinolines
TLR9 Bacterial DNA, CpG deoxyribonucleotide
TLR10 Unknown
Semin Immunopathol

inflammation. Pro-inflammatory cytokines have a deep im- with the severity of pneumonia [49, 50]. Most of the cytokine,
pact on endothelial cells by promoting (i) expression of adhe- receptor, or transcription factor genes have SNPs that were
sion molecules such as ICAM1 or VCAM1 that will allow studied in the context of severe infections, and these genetic
activated immune cells to adhere to the vascular wall and enter variations often have an impact on sepsis outcome.
inflamed tissues, (ii) the synthesis of chemokines to attract
immune cells, (iii) secretion/exhibition of procoagulant fac-
tors, and also (iv) endothelial cell dysfunction and necrosis Cytokines in sepsis pathophysiology
that contribute to increased vascular permeability, a major
feature of severe infection; increased production of NO, a The production and release of potential cytotoxic factors by
vasodilatory agent; and organ dysfunction (induced by both immune cells was reported in the 1960s by the discovery of
cellular infiltration and ischemia). the antitumor effects of the Blymphotoxin^ produced by lym-
phocytes [51, 52]. In 1975, Carswell et al. reported the pro-
Individual response and genetic polymorphisms duction of another cytotoxic factor by macrophages, the
Btumor necrosis factor^ (TNF) [53], which will later be iden-
Infections lead to a wide variety of clinical signs with a wide tified to be the same molecule as that named Bcachectin^ [54].
range of intensities. The individual host response to infection Clark et al. then emitted the hypothesis that TNF could be
is related to numerous factors such as the microorganism, the responsible for the symptoms observed during malaria and
site of infection, the immune status, the balance between acti- endotoxemia [55–57]. This was the angular stone of the con-
vating factors of the gene network involved in sepsis patho- ceptual changes in the pathophysiology of infections.
physiology, the epigenetic control of gene transcription, and Infection causes the release of cytotoxic factors, and this phe-
genetic polymorphisms of sepsis-associated genes. While nomenon is observed with different kinds of infections.
most of these factors may vary a lot during a lifetime, the Tracey et al. then showed that TNF was able to induce shock
nuclear DNA is stable; therefore, genetic polymorphisms that and tissue injury [58] and that anti-TNF antibodies could pre-
are shown to have an impact on sepsis morbidity or outcome vent the lethality induced by bacteremia [59]. In 1989, the
can be identified and could theoretically lead to a better prog- same group proposed that viral disease, especially influenza,
nostication of the disease and/or the choice of more adequate could also have an inflammatory cytokine origin [60].
treatments.
Dozens of genetic polymorphisms were identified as hav- Cytokines
ing an impact on the course of sepsis [36–38], and each new
large-scale, genome wide or not, association study of genetic Cytokines are a broad category of relatively small proteins
polymorphisms in sepsis cohorts brings new players in the (<40 kDa) that are produced and released with the final aim of
game [39, 40]. Bioinformatic advances help to identify geno- cell signaling [61]. They cover autocrine, paracrine, and endo-
typic and phenotypic clusters and could improve our under- crine activities and play an immunomodulating function. After
standing of pathways involved in the host response and reveal binding to specific receptors on various types of cells, cytokines
potential therapeutic targets. Among the identified single nu- induce activation, proliferation, or migration of target cells.
cleotide polymorphisms (SNPs), cytokine genes are well rep- Cytokine taxonomy is highly complex and is based both on
resented. TNF-α levels vary greatly in septic patients. One timing of discovery and on association by structure, target, or
possible explanation lies in the occurrence of a SNP in the function. The term Bcytokine^ encompasses roughly 100 sep-
TNF-α gene promoter in the -308 position (G308A). Thus, arate genes coding for the cytokine of cytokine-like proteins
two alleles of the gene exist, one named TNF1 and the other [61]. This dense cytokine network supports and promotes the
TNF2; TNF2 is associated with high levels of TNF-α and an inflammatory process. Cytokines can be divided into several
increased severity of infections such as meningococcemia or categories: interleukins, chemokines, interferons, tumor ne-
in a cohort of septic patients [41, 42]. The ILRN*2 allele of the crosis factor, and growth factors.
interleukin-1 receptor antagonist (IL-1Ra) is associated with Interleukins are the most important group of cytokines re-
high levels of IL-1α [43] and lower levels of IL-1Ra [44] leased during infectious processes. They encompass a large
while the rs315952 SNP is associated with survival [45]. variety of proteins secreted by leukocytes and endothelial cells
The CD14 gene promoter SNP C(-159)T increases suscepti- (among others) and that contribute to cell signaling and pro-
bility to gram-positive sepsis [46]; the Asp299Gly allele of the mote activation, proliferation, death, and/or motility of im-
TLR4 gene also may have a negative impact on sepsis [47] mune cells. They are artificially divided into pro- and anti-
and the TLR1 (-7202G) SNP induces high NFκB activation inflammatory interleukins. Pro-inflammatory interleukins are
and therefore higher levels of cytokines [48]. This is also supposed to be responsible for cell activation, tissue damage,
observed with anti-inflammatory cytokines, e.g., the G- and necrosis while anti-inflammatory interleukins aim to
1082A SNP of the IL-10 gene was shown to be associated dampen and finally reverse the inflammatory process.
Semin Immunopathol

Among the numerous pro-inflammatory cytokines studied deletion of Tregs and secretion of IL-21 can restore the produc-
during sepsis, IL-1β, IL-6, IL-12, and IL-17 are of crucial tion of IL-17 [76]. TGFβ, through the nuclear receptor
importance. IL-1β is also referred to as catabolin. It is a mem- RORγt, regulates IL-17 in a complex fashion. TGFβ induces
ber of the IL-1 family (11 genes) that is produced after acti- both RORγt expression and reduces its ability to induce IL-17
vation of the inflammasome (especially NLRP3). Activation production. When inflammatory mediators are added,
of the IL-1 receptor leads to the activation of the JNK and p38- RORγt-directed IL-17 production is restored [77].
MAPK pathways and increases the activity of NFκB. IL-1β Interferons are classified into three major types according
promotes the amplification cascade and induces the synthesis to their receptor specificity. IFNAR1/IFNAR2 is the heterodi-
of various inflammatory genes such as IL-6, IL-8, MCP-1, meric receptor for type I interferon (IFN-α and IFN-β).
COX-2, IκBα, IL-1α, IL-1β, and MKP-1. It is interesting to IFN-γR1/IFN-γR2 is the receptor for type II IFN (IFN-γ),
note that the IL-1 pathway works in parallel with the IL-18 and interferon-λ is the third type of IFN, mainly studied during
and IL-33 pathways [62]. During sepsis, levels of IL-1β were viral infections. IFN-γ is mainly produced by CD4 and CD8 T
shown to be higher in patients who died than in survivors, cells. NK cells also produce IFN-γ but to a lesser extent [78,
suggesting an association of high levels of IL-1β and sepsis 79]. IFN is usually used to define TH1-type cells. The IFN-γ
outcome [63]. IL-6 (also known as interferon-ß2, B cell stim- signaling pathway relies mainly on JAK1, JAK2, p38-MAPK,
ulatory factor 2 (BSF-2)) is a pleiotropic interleukin; its role and STAT1 and induces factors such as IRF1 or SMAD7.
during the cytokine storm is complex and controversial. IL-6 INFγ promotes the inflammatory response during sepsis, but
is actually not a single protein but includes a family of mole- INFγ production is dampened during sepsis, probably due to
cules like IL-11, oncostatin M, ciliary neutrophilic factor, or the hyporesponsiveness of lymphocytes during the immuno-
cardiothrophin-like cytokine. IL-6 cytokine receptors are suppressed state [80, 81].
made of two subunits: CD126 and CD130 (gp130) [64]. The Chemokines (CKs) are small molecules (8–12 kDa), usu-
main source of IL-6 is tissue macrophages. High levels of ally soluble, not only specialized in the recruitment but also
IL-6 were observed in many inflammatory diseases such activation of immune cells. CKs need specific receptors
as cardiovascular disease, autoimmune diseases, or can- coupled to G proteins. Chemoattraction of cells requires a
cer. Similarly to IL-1β, high levels of IL-6 were con- gradient of concentration of chemokines. There are four types
sistently shown to be associated with the severity of of CKs that are classified according to their amino acid se-
sepsis and the highest levels associated with the worse quence and the spacing between their two cysteine residues:
outcome [65–67]. The exact contribution of IL-6 to sep- CC, CXC, CX3C, and XC CKs. The most studied CKs in the
sis is still under investigation and is possibly linked to context of infectious disease are MCP-1/CCL2, CXCL8/IL-8,
the activation of the complement pathway [68, 69] and CXCL1/GRO-a, and CXCL2/MIP2a. In contrast to other cy-
capillary leakage. Modulation of IL-6 is also made tokines which have broad effects on many cells, CKs are usu-
through the release of a soluble receptor [70]. ally cell specific. For instance, MCP-1 or CX3CL1 are nearly
IL-12 is also elevated during sepsis. This cytokine, pro- specific for monocytes, CXCL1 and CXCL2 for neutrophils,
duced by dendritic cells, macrophages, and lymphoblastoid and CXCR3 for T lymphocytes. Their role in sepsis patho-
cells, induces the differentiation of naïve T cells into type 1 physiology was deeply investigated, and they were found to
helper T cells (TH1) and activates NK cells; as a result, these play a major orchestrating role during the host response to
cells will produce a high amount of INFγ [71]. IL-17 is pro- infection. CKs induce not only leukocyte recruitment to the
duced by the so-called TH17 T cells after stimulation by IL-23 site of infection but also the release of immune cells from the
and induces the synthesis of a wide variety of cytokines: GM- bone marrow or spleen. Once recruited, leukocytes will clear
CSF, pro-inflammatory interleukins (IL-1β, IL-6), TNF-α, bacteria and dead cells and generate inflammation, but they
and chemokines (IL-8, Gro-α, CCR2/MCP-1), by many cell will also be activated and then directly propagate inflamma-
types such as endothelial cells, epithelial cells, fibroblasts, or tion throughout the entire organism. Lack of CKs or their
macrophages [72, 73]. Generation of IL-17-producing TH17 receptors leads to a quasi-immunosuppressed state and makes
cells is finely tuned. TGFβ, IL-1β, IL-6, IL-21, and IL-23 the organism more susceptible to infection-induced lethality
were shown to be sufficient to induce TH17 cells from naïve [82–85].
CD4+ cells. However, several, and sometimes opposed, pro- Growth factors are deeply involved in sepsis pathogenesis.
cesses occur. TGFβ can induce both regulatory T cells (Tregs) Among the different types of growth factors secreted during
and TH17 [74]; addition of IL-6 restricts the development of sepsis, the most involved in the generation of the cytokine
Tregs. Interestingly, IL-6 can also induce the production of IL- storm are the hematopoietic targeted colony-stimulating fac-
10, which restrains the expression of IL-17 [75]. Therefore, tors (CSFs): granulocyte-macrophage colony-stimulating fac-
there is a balance of pro-inflammatory mediators that will tor (GM-CSF), macrophage colony-stimulating factor (M-
favor TH17 generation. As a proof of the crucial role of IL-6, CSF), and granulocyte colony-stimulating factor (G-CSF).
mice lacking IL-6 cannot develop TH17 cells. In IL-6−/− mice, In addition, with other factors such as IL-3, CSFs will induce
Semin Immunopathol

myeloid cell differentiation and proliferation; thus, a larger In humans, the white blood cell count will increase until it
number of activated cells will be put into movement and con- reaches a peak at 6 h. Leukocytosis is highly dose dependent
tribute to the synthesis of more cytokines and therefore pro- and reflects both cell release from the bone marrow and cell
mote the cytokine storm. It is also suggested that CSFs not adhesion and recruitment to the inflamed tissues. Leukocyte
only play a numerical role but also a functional one by en- response is also mainly a neutrophilia while the lymphocyte
hancing response to early-phase molecules such as IL-1β or number is usually decreasing.
TNF-α [86]. A recent report shows that during sepsis, a major Regarding cytokines, the main inflammatory molecules
source of GM-CSF is the newly discovered IRA B cells [87], (TNF-α, IL-6, IL-1β) will have more or less the same kinetics
which are innate-like B cells derived from B1 cells and that with a rapid peak between 1.5 and 2 h and a rapid decrease to
relocate into infected tissues. the basal levels (nearly undetectable) after 6 h. Anti-
The most studied anti-inflammatory interleukins are IL- inflammatory molecules (IL-10, IL-1RA, TNF-SRI) and
1RA, IL-4, and IL-10. IL-1RA is secreted by immune cells chemokines (IL-8, KC, MIP-2, GRO-α) will have a parallel
or epithelial cells and binds to IL-1R, thus blocking the action evolution [100–103].
of IL-1α or IL-1β inflammatory signals [88, 89]. Treatment A major work by Calvano et al. in 2005 consisted in
with IL-1RA improved sepsis induced by polymicrobial peri- the study of the transcriptome response of white blood
tonitis or gram-negative pneumonia [90, 91]. IL-4 is mainly cells after endotoxin injection in humans [104]. Using
produced by T cells and promotes both proliferation of B and serial blood sampling after the onset of endotoxemia,
T cells and a shift of T cells towards a TH2 profile [92, 93]. the authors showed that pro-inflammatory gene expres-
The role of IL-4 in sepsis is under debate; a possible role of IL- sions such as TNFSF2 (TNF), IL-1α, IL-1β, CXCL1
4 in its pathophysiology was inconstantly reported. IL-10 is (GRO-α), CXCL2 (GRO-b), CCL2 (MCP-1), CXCL8
the central anti-inflammatory cytokine [94]. It has the ability (IL-8), and CXCL10 peaked at 2–4 h with a subsequent
to block the production of inflammatory cytokines by myeloid increase in transcriptions factors NFκB1, NFκB2, RelA,
cells, NK cells, and T cells [95]. The levels of IL-10, even as a and RelB. The 4–6-h time period appeared to be critical
marker of anti-inflammation, are directly related to the degree because of the increase in both initial response genes
of inflammation (as reflected by the levels of IL-6) demon- and regulation genes of the transcription factor family:
strating that pro- and anti-inflammation are closely related. STAT genes, c-AMP-response element binding protein
High IL-10 levels are also associated with more important (CREB), SOCS3, and IKKB, or anti-inflammatory cyto-
features of sepsis-induced immunosuppression [96]. kines: IL1RAP, IL1R1, IL-10, and TNFRSF1A. The
IL-3 is a cytokine discovered in the 1980s and also referred global response to endotoxin comprises a network of
to as multi-CSF [97]. It is a pleiotropic growth factor that acti- 1556 genes. More recently, Shalova et al. performed a
vates proliferation of hematopoietic stem cells and progenitors. transcriptomic characterization of human blood mono-
Until recently, the role of IL-3 during sepsis was unknown. We cytes isolated from septic patients [105]; 1170 genes
identified IL-3 as a key regulator of the cytokine storm during were found to be affected by sepsis (561 upregulated
sepsis [98]. IL-3 is produced by IRA B cells and fuels the and 609 downregulated), and immune response genes
inflammatory cascade by promoting emergency myelopoiesis. were found to be the more significantly altered. Sepsis
Injection promotes high levels of pro-inflammatory cytokines monocytes have a clearly different transcriptomic pro-
through proliferation of myeloid cells but not their activation. A gram compared to basal monocytes. Monocytes upregu-
recent study confirms the role of IL-3 in the immune regulation late a large number of cytokines such as IL-1β, IL-6,
and potential response to corticosteroids during sepsis [99]. CCL3, CCL5, and IL-10 and transcription factors such
as NFκb and the regulatory phospho-IκBa during sepsis.
Cytokine kinetics Therefore, findings from endotoxemia trials are con-
firmed in septic patients. The genomic response of sep-
The endotoxemia model was widely used to study the type tic shock patients was also evaluated in a recent work
and the kinetics of the cytokine response. Sensitivity to endo- by Cazalis et al. [106]. The authors found that both pro-
toxin varies greatly among species. For example, to generate and anti-inflammatory transcriptome responses were ac-
an IL-6 response around 1500 pg/mL, several milligrams per tivated since the admission of the patients and rapidly
kilogram of LPS are needed in mice while only few nano- evolved over the first 48 h. Payen et al. studied the
grams per kilogram are necessary in humans. Clinically, after evolution of the transcriptome over the first week and
LPS injection, the heart rate increases within 30 min to reach a the first month after sepsis [107]. Among the genes
peak at 2–4 h in humans while it will be roughly stable in whose expression was modified over time (in survivors),
mice. Body temperature increases in both species between 1 some were cytokine related like IL-3 (IL3RA) or
and 2 °C, but the peak time is slightly shifted from 1 h in mice ISTF3G (interferon-stimulated transcription factor 3
to 2–4 h in humans. gamma).
Semin Immunopathol

Targeting cytokines during sepsis: a therapeutic treatments are given without the exact same timing. This leads
challenge to the more important potential cause. Septic patients do not
represent a homogeneous category of patients. Sepsis presen-
Despite the increasing amount of evidence on the key patho- tations are diverse, and patients included in the study have
physiological role of cytokines during infection, there is no different sources of sepsis, with different evolutions, different
specific treatment targeting inflammation that was shown to levels of inflammation, and therefore different responses to
be effective in sepsis. treatment. Applying the same treatment for sepsis is unlikely
As recently reviewed by Opal et al. [108], nearly 30 years to show any beneficial effects. Patients included in studies
of clinical trials were unsuccessful despite promising preclin- need to be selected based on specific parameters or bio-
ical and single-center results. markers. For instance, patients with levels of pro-
The first immunomodulatory attempt in sepsis was with the inflammatory cytokines are more likely to respond to the
use of corticosteroids [109]. Corticosteroids are well known to anti-cytokine treatments.
dampen inflammation and inhibit the activation of the NFκb Another approach to treat the effects of the cytokine storm
and AP-1 pathways [110]. Annane et al. showed that low-dose during infectious diseases is to target the direct tissue conse-
corticosteroids could reduce mortality in severe sepsis and quences of the inflammation cascade. Blood vessels’ in-
septic shock [111]. Unfortunately, a large-scale clinical trial, creased permeability is a hallmark feature of serious infec-
the CORTICUS study [112], showed no clinical benefits of tions; thus, London et al. [122] decided to target an
the systematic use of corticosteroids during sepsis. endothelium-specific pathway: Slit-Robo4. Activation of the
More targeted therapies were also tried. Antibodies and cap- Slit-Robo4 pathway (Robo4 is the membrane receptor for Slit)
ture of molecules such as TNF-α, IL-1, and LPS had great is associated with a dramatic reduction in vascular permeabil-
preclinical results, in mice models, but these treatments failed ity and organ injury in various models of infections such as
to reduce overall sepsis mortality [113–117]. However, a recent polymicrobial sepsis or H5N1 influenza. In these two models,
meta-analysis showed that anti-TNF treatment was associated Slit treatment does not seem to reduce the amount of circulat-
with a modest but significant reduction in sepsis mortality ing inflammatory cytokines while it appears to be effective in
[118]. This could justify a new trial with more selected patients. this task in the endotoxemia model [123]. This innovative
The blocking of the TLR4 pathway also faced clinical failure approach is a potential effective treatment of serious infections
[114]. The interaction between coagulation and inflammation that do not aim to block the already self-sustaining inflamma-
was shown in numerous experimental and clinical studies tory cascade but to limit what is eventually killing patients,
[119]. The coagulation cascade induces blood clot and endo- vascular-induced tissue damage.
thelial and immune cell activation and amplifies inflammation. Sphingosine-1-phosphate (S1P) was identified as a good
Therefore, targeting this coagulation disorder could be effective candidate to target the infection-associated cytokine storm.
in reducing inflammation. These statements led to the use of S1P is a signaling lysophospholipid that promotes cytokine
activated protein C (APC) (drotrecogin alfa) in sepsis. The first synthesis and secretion [124]. Ceramides are a component
large-scale study, the PROWESS trial [120], showed great po- of the cell membrane; once deacetylated into sphingosine
tential for APC during sepsis. Twenty-eight-day mortality was and a fatty acid, sphingosine is phosphorylated by sphingo-
assessed in a randomized study including nearly 1700 patients. sine kinase 1 or 2 into its active form, S1P. S1P actions are
The use of APC led to a significant reduction in mortality mediated by five G protein-coupled receptors expressed on
(30.8% in the placebo group vs 24.7% in the treatment group, various cell types like immune cells (macrophages, dendrit-
a nearly 20% relative risk reduction). Nevertheless, when a ic cells, T cells, B cells, NK cells), endothelial cells, or
larger study attempted to replicate these results, the use of epithelial cells. In 2011, Teijaro et al. showed that the
APC was not associated with mortality reduction [121]. After S1P1 receptor expressed on endothelial cells and lympho-
publication of these results, the molecule was withdrawn from cytes was a key regulator in immune and vascular response
the market by the pharmaceutical company producing it. after influenza infection [125]. Activation of the S1P1 path-
Failure of sepsis therapy could have several explanations. way with chemical agonists named AAL-R or CYM-5442
The appropriateness of the preclinical models, especially mice dramatically reduced cytokine production and cell recruit-
models or in vitro experiments, is a major source of concern. ment and improved survival after influenza infection. These
Immune response between species varies greatly; the transpo- effects were mediated by a reduction in chemokine produc-
sition of results obtained in animal experiments into effective tion by endothelial cells.
treatment for humans is not straightforward. Another issue The study of these two pathways, Slit-Robo4 and S1P-
linked to animal models is that they are Bcalibrated,^ meaning S1P1, confirm that using endothelial activation as a therapeu-
that treatments are administered at a precise time point before tic target to quell the cytokine storm after infection is highly
or after the onset of the disease. In clinical practice, it is nearly effective and is a valid alternative to direct cytokine
impossible to date precisely the beginning of sepsis; therefore, antagonism.
Semin Immunopathol

Conclusion Current estimates and limitations. Am J Respir Crit Care


Med 193(3):259–272. doi:10.1164/rccm.201504-0781OC
7. Yeh RW, Sidney S, Chandra M, Sorel M, Selby JV, Go AS (2010)
The cytokine storm induced by infectious diseases is the result Population trends in the incidence and outcomes of acute myocar-
of an autoamplifying phenomenon aiming to destroy the in- dial infarction. N Engl J Med 362(23):2155–2165. doi:10.1056/
vader and restore homeostasis that will eventually lead to sep- NEJMoa0908610
8. Siegel R, Ma J, Zou Z, Jemal A (2014) Cancer statistics, 2014. CA
sis. While the understanding of the orchestration of the
Cancer J Clin 64(1):9–29. doi:10.3322/caac.21208
cytokine-induced cell response during sepsis is in constant 9. Adhikari NK, Fowler RA, Bhagwanjee S, Rubenfeld GD
progress, we still lack a global integrative view, at the cell (2010) Critical care and the global burden of critical illness
level, in time and space. The organization of the host response in adults. Lancet 376(9749):1339–1346. doi:10.1016/S0140-
6736(10)60446-1
to infection, locally and remotely, is still incompletely under-
10. Janeway CA Jr, Medzhitov R (2002) Innate immune recognition.
stood. Despite the identification of key molecules, such as the Annu Rev Immunol 20:197–216. doi:10.1146/annurev.immunol.
emblematic tumor necrosis factor alpha, every attempt aiming 20.083001.084359
to dampen this cytokine storm activation or consequences 11. Akira S, Uematsu S, Takeuchi O (2006) Pathogen recogni-
tion and innate immunity. Cell 124(4):783–801. doi:10.
failed in clinical trials. While the exact triggers or connectors
1016/j.cell.2006.02.015
that should be targeted are still to be identified, the endothelium 12. Netea MG (2013) Training innate immunity: the changing concept
as a node of immune response appears to be a valid and prom- of immunological memory in innate host defence. Eur J Clin
ising target. The next decade will probably see a more precise Investig 43(8):881–884. doi:10.1111/eci.12132
depiction of in-cell, in-organ, and global response to infection; 13. Netea MG, Quintin J, van der Meer JW (2011) Trained immunity:
a memory for innate host defense. Cell Host Microbe 9(5):355–
there is no doubt that future discoveries will challenge our ac- 361. doi:10.1016/j.chom.2011.04.006
tual concepts and will make us revisit our therapeutic targets; 14. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA
this makes the fight against this dramatic response to infection (1996) The dorsoventral regulatory gene cassette spatzle/Toll/cac-
an exciting challenge. tus controls the potent antifungal response in Drosophila adults.
Cell 86(6):973–983
15. Medzhitov R, Preston-Hurlburt P, Janeway CA Jr (1997) A
Acknowledgements This work is supported by a public grant overseen human homologue of the Drosophila Toll protein signals
by the French National Research Agency (ANR) (Project BCMOS,^ activation of adaptive immunity. Nature 388(6640):394–
ANR-15-CE15-0019). 397. doi:10.1038/41131
16. Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X,
Compliance with ethical standards Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M,
Ricciardi-Castagnoli P, Layton B, Beutler B (1998) Defective
Conflict of interest The authors declare that they have no conflicts of LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations
interest. in Tlr4 gene. Science 282(5396):2085–2088
17. Poltorak A, Smirnova I, He X, Liu MY, Van Huffel C, McNally O,
Birdwell D, Alejos E, Silva M, Du X, Thompson P, Chan EK,
Ledesma J, Roe B, Clifton S, Vogel SN, Beutler B (1998) Genetic
References and physical mapping of the Lps locus: identification of the toll-4
receptor as a candidate gene in the critical region. Blood Cells Mol
Dis 24(3):340–355
1. Ferrara JL, Abhyankar S, Gilliland DG (1993) Cytokine storm of
18. Kawai T, Akira S (2011) Toll-like receptors and their crosstalk
graft-versus-host disease: a critical effector role for interleukin-1.
with other innate receptors in infection and immunity. Immunity
Transplant Proc 25(1 Pt 2):1216–1217
34(5):637–650. doi:10.1016/j.immuni.2011.05.006
2. Yokota S (2003) Influenza-associated encephalopathy—patho- 19. Gao H, Leaver SK, Burke-Gaffney A, Finney SJ (2008) Severe
physiology and disease mechanisms. Nihon Rinsho 61(11): sepsis and Toll-like receptors. Semin Immunopathol 30(1):29–40.
1953–1958 doi:10.1007/s00281-007-0101-4
3. Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze 20. Annane D, Bellissant E, Cavaillon JM (2005) Septic shock.
MG (2012) Into the eye of the cytokine storm. Microbiol Mol Biol Lancet 365(9453):63–78. doi:10.1016/S0140-6736(04)17667-8
Rev 76(1):16–32. doi:10.1128/MMBR.05015-11 21. Akira S, Takeda K (2004) Toll-like receptor signalling. Nat Rev
4. Bone RC, Sibbald WJ, Sprung CL (1992) The ACCP- Immunol 4(7):499–511. doi:10.1038/nri1391
SCCM consensus conference on sepsis and organ failure. 22. Armstrong L, Medford AR, Hunter KJ, Uppington KM, Millar
Chest 101(6):1481–1483 AB (2004) Differential expression of Toll-like receptor (TLR)-2
5. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, and TLR-4 on monocytes in human sepsis. Clin Exp Immunol
Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, 136(2):312–319. doi:10.1111/j.1365-2249.2004.02433.x
Coopersmith CM, Hotchkiss RS, Levy MM, Marshall JC, 23. Harter L, Mica L, Stocker R, Trentz O, Keel M (2004) Increased
Martin GS, Opal SM, Rubenfeld GD, van der Poll T, Vincent expression of toll-like receptor-2 and -4 on leukocytes from pa-
JL, Angus DC (2016) The third international consensus defini- tients with sepsis. Shock 22(5):403–409
tions for sepsis and septic shock (Sepsis-3). JAMA 315(8):801– 24. Mariathasan S, Monack DM (2007) Inflammasome adaptors and
810. doi:10.1001/jama.2016.0287 sensors: intracellular regulators of infection and inflammation. Nat
6. Fleischmann C, Scherag A, Adhikari NK, Hartog CS, Rev Immunol 7(1):31–40. doi:10.1038/nri1997
Tsaganos T, Schlattmann P, Angus DC, Reinhart K, 25. Zamboni DS, Kobayashi KS, Kohlsdorf T, Ogura Y, Long EM,
International Forum of Acute Care T (2016) Assessment Vance RE, Kuida K, Mariathasan S, Dixit VM, Flavell RA,
of global incidence and mortality of hospital-treated sepsis. Dietrich WF, Roy CR (2006) The Birc1e cytosolic pattern-
Semin Immunopathol

recognition receptor contributes to the detection and control of 43. Witkin SS, Gerber S, Ledger WJ (2002) Influence of interleukin-1
Legionella pneumophila infection. Nat Immunol 7(3):318–325. receptor antagonist gene polymorphism on disease. Clin Infect Dis
doi:10.1038/ni1305 34(2):204–209. doi:10.1086/338261
26. Brown GD, Herre J, Williams DL, Willment JA, Marshall AS, 44. Arnalich F, Lopez-Maderuelo D, Codoceo R, Lopez J, Solis-
Gordon S (2003) Dectin-1 mediates the biological effects of be- Garrido LM, Capiscol C, Fernandez-Capitan C, Madero R,
ta-glucans. J Exp Med 197(9):1119–1124. doi:10.1084/jem. Montiel C (2002) Interleukin-1 receptor antagonist gene polymor-
20021890 phism and mortality in patients with severe sepsis. Clin Exp
27. Brown GD, Taylor PR, Reid DM, Willment JA, Williams DL, Immunol 127(2):331–336
Martinez-Pomares L, Wong SY, Gordon S (2002) Dectin-1 is a 45. Meyer NJ, Ferguson JF, Feng R, Wang F, Patel PN, Li M, Xue C,
major beta-glucan receptor on macrophages. J Exp Med 196(3): Qu L, Liu Y, Boyd JH, Russell JA, Christie JD, Walley KR, Reilly
407–412 MP (2014) A functional synonymous coding variant in the IL1RN
28. Gross O, Gewies A, Finger K, Schafer M, Sparwasser T, Peschel gene is associated with survival in septic shock. Am J Respir Crit
C, Forster I, Ruland J (2006) Card9 controls a non-TLR signalling Care Med 190(6):656–664. doi:10.1164/rccm.201403-0586OC
pathway for innate anti-fungal immunity. Nature 442(7103):651– 46. Surbatovic M, Grujic K, Cikota B, Jevtic M, Filipovic N, Romic P,
656. doi:10.1038/nature04926 Strelic N, Magic Z (2010) Polymorphisms of genes encoding tu-
29. Brown GD, Gordon S (2001) Immune recognition. A new recep- mor necrosis factor-alpha, interleukin-10, cluster of
tor for beta-glucans. Nature 413(6851):36–37. doi:10.1038/ differentiation-14 and interleukin-1ra in critically ill patients. J
35092620 Crit Care 25(3):542 e541-548. doi:10.1016/j.jcrc.2009.12.003
30. Dennehy KM, Brown GD (2007) The role of the beta-glucan 47. Lorenz E, Mira JP, Frees KL, Schwartz DA (2002) Relevance of
receptor Dectin-1 in control of fungal infection. J Leukoc Biol mutations in the TLR4 receptor in patients with gram-negative
82(2):253–258. doi:10.1189/jlb.1206753 septic shock. Arch Intern Med 162(9):1028–1032
31. den Dunnen J, Gringhuis SI, Geijtenbeek TB (2009) Innate sig- 48. Wurfel MM, Gordon AC, Holden TD, Radella F, Strout J,
naling by the C-type lectin DC-SIGN dictates immune responses. Kajikawa O, Ruzinski JT, Rona G, Black RA, Stratton S, Jarvik
Cancer Immunol, Immunother 58(7):1149–1157. doi:10.1007/ GP, Hajjar AM, Nickerson DA, Rieder M, Sevransky J, Maloney
s00262-008-0615-1 JP, Moss M, Martin G, Shanholtz C, Garcia JG, Gao L, Brower R,
32. Fitzgerald ME, Rawling DC, Vela A, Pyle AM (2014) An evolv- Barnes KC, Walley KR, Russell JA, Martin TR (2008) Toll-like
ing arsenal: viral RNA detection by RIG-I-like receptors. Curr receptor 1 polymorphisms affect innate immune responses and
Opin Microbiol 20:76–81. doi:10.1016/j.mib.2014.05.004 outcomes in sepsis. Am J Respir Crit Care Med 178(7):710–
33. Aird WC (2003) The role of the endothelium in severe sepsis and 720. doi:10.1164/rccm.200803-462OC
multiple organ dysfunction syndrome. Blood 101(10):3765–3777. 49. Gallagher PM, Lowe G, Fitzgerald T, Bella A, Greene CM,
doi:10.1182/blood-2002-06-1887 McElvaney NG, O’Neill SJ (2003) Association of IL-10 polymor-
34. Ince C, Mayeux PR, Nguyen T, Gomez H, Kellum JA, Ospina- phism with severity of illness in community acquired pneumonia.
Tascon GA, Hernandez G, Murray P, De Backer D, Workgroup Thorax 58(2):154–156
AX (2016) The endothelium in sepsis. Shock 45(3):259–270. doi: 50. Azab SF, Abdalhady MA, Elsaadany HF, Elkomi MA, Elhindawy
10.1097/SHK.0000000000000473 EM, Sarhan DT, Salam MM, Allah MA, Emam AA, Noah MA,
35. Vallet B (2003) Bench-to-bedside review: endothelial cell dys- Abdelsalam NI, Abdellatif SH, Rass AA, Ismail SM, Gheith T,
function in severe sepsis: a role in organ dysfunction? Crit Care Aziz KA, Hamed ME, Abdelrahman HM, Ahmed AR, Nabil RM,
7(2):130–138 Abdulmaksoud RS, Yousef HY (2016) Interleukin-10 -1082 G/A
36. Sutherland AM, Walley KR (2009) Bench-to-bedside review: as- gene polymorphisms in Egyptian children with CAP: a case-
sociation of genetic variation with sepsis. Crit Care 13(2):210. doi: control study. Medicine (Baltimore) 95(26):e4013. doi:10.1097/
10.1186/cc7702 MD.0000000000004013
37. Arcaroli J, Fessler MB, Abraham E (2005) Genetic polymor- 51. Kolb WP, Granger GA (1968) Lymphocyte in vitro cytotoxicity:
phisms and sepsis. Shock 24(4):300–312 characterization of human lymphotoxin. Proc Natl Acad Sci U S A
38. Texereau J, Pene F, Chiche JD, Rousseau C, Mira JP (2004) 61(4):1250–1255
Importance of hemostatic gene polymorphisms for susceptibility 52. Ruddle NH, Waksman BH (1967) Cytotoxic effect of
to and outcome of severe sepsis. Crit Care Med 32(5 Suppl): lymphocyte-antigen interaction in delayed hypersensitivity.
S313–S319 Science 157(3792):1060–1062
39. Chung LP, Waterer GW (2011) Genetic predisposition to respira- 53. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B
tory infection and sepsis. Crit Rev Clin Lab Sci 48(5–6):250–268. (1975) An endotoxin-induced serum factor that causes necrosis of
doi:10.3109/10408363.2011.641517 tumors. Proc Natl Acad Sci U S A 72(9):3666–3670
40. Cornell TT, Wynn J, Shanley TP, Wheeler DS, Wong HR (2010) 54. Beutler B, Greenwald D, Hulmes JD, Chang M, Pan YC, Mathison J,
Mechanisms and regulation of the gene-expression response to Ulevitch R, Cerami A (1985) Identity of tumour necrosis factor and
sepsis. Pediatrics 125(6):1248–1258. doi:10.1542/peds.2009- the macrophage-secreted factor cachectin. Nature 316(6028):552–554
3274 55. Clark IA, Cowden WB (1989) Is TNF a key to acute infectious
41. Mira JP, Cariou A, Grall F, Delclaux C, Losser MR, Heshmati F, illness? Today’s Life Sci 1:26–29
Cheval C, Monchi M, Teboul JL, Riche F, Leleu G, Arbibe L, 56. Clark IA, Virelizier JL, Carswell EA, Wood PR (1981) Possible
Mignon A, Delpech M, Dhainaut JF (1999) Association of importance of macrophage-derived mediators in acute malaria.
TNF2, a TNF-alpha promoter polymorphism, with septic shock Infect Immun 32(3):1058–1066
susceptibility and mortality: a multicenter study. JAMA 282(6): 57. Clark IA (1982) Suggested importance of monokines in patho-
561–568 physiology of endotoxin shock and malaria. Klin Wochenschr
42. Calvano JE, Um JY, Agnese DM, Hahm SJ, Kumar A, Coyle SM, 60(14):756–758
Calvano SE, Lowry SF (2003) Influence of the TNF-alpha and 58. Tracey KJ, Beutler B, Lowry SF, Merryweather J, Wolpe S,
TNF-beta polymorphisms upon infectious risk and outcome in Milsark IW, Hariri RJ, Fahey TJ 3rd, Zentella A, Albert JD et al
surgical intensive care patients. Surg Infect 4(2):163–169. doi: (1986) Shock and tissue injury induced by recombinant human
10.1089/109629603766956951 cachectin. Science 234(4775):470–474
Semin Immunopathol

59. Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC, 76. Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, Oukka
Lowry SF, Cerami A (1987) Anti-cachectin/TNF monoclonal an- M, Kuchroo VK (2007) IL-21 initiates an alternative pathway to
tibodies prevent septic shock during lethal bacteraemia. Nature induce proinflammatory T(H)17 cells. Nature 448(7152):484–
330(6149):662–664. doi:10.1038/330662a0 487. doi:10.1038/nature05970
60. Tracey KJ, Cerami A (1989) Cachectin/tumor necrosis fac- 77. Manel N, Unutmaz D, Littman DR (2008) The differentiation of
tor and other cytokines in infectious disease. Curr Opin human T(H)-17 cells requires transforming growth factor-beta and
Immunol 1(3):454–461 induction of the nuclear receptor RORgammat. Nat Immunol 9(6):
61. Dinarello CA (2007) Historical insights into cytokines. Eur J 641–649. doi:10.1038/ni.1610
Immunol 37(Suppl 1):S34–S45. doi:10.1002/eji.200737772 78. Schroder K, Hertzog PJ, Ravasi T, Hume DA (2004) Interferon-
62. Weber A, Wasiliew P, Kracht M (2010) Interleukin-1 (IL-1) path- gamma: an overview of signals, mechanisms and functions. J
way. Sci Signal 3(105):cm1. doi:10.1126/scisignal.3105cm1 Leukoc Biol 75(2):163–189. doi:10.1189/jlb.0603252
63. Mera S, Tatulescu D, Cismaru C, Bondor C, Slavcovici A, Zanc V, 79. Schoenborn JR, Wilson CB (2007) Regulation of interferon-
Carstina D, Oltean M (2011) Multiplex cytokine profiling in pa- gamma during innate and adaptive immune responses. Adv
tients with sepsis. APMIS 119(2):155–163. doi:10.1111/j.1600- Immunol 96:41–101. doi:10.1016/S0065-2776(07)96002-2
0463.2010.02705.x 80. Romero CR, Herzig DS, Etogo A, Nunez J, Mahmoudizad R,
64. Jones SA, Scheller J, Rose-John S (2011) Therapeutic strategies Fang G, Murphey ED, Toliver-Kinsky T, Sherwood ER (2010)
for the clinical blockade of IL-6/gp130 signaling. J Clin Invest The role of interferon-gamma in the pathogenesis of acute intra-
121(9):3375–3383. doi:10.1172/JCI57158 abdominal sepsis. J Leukoc Biol 88(4):725–735. doi:10.1189/jlb.
65. Gouel-Cheron A, Allaouchiche B, Guignant C, Davin F, Floccard 0509307
B, Monneret G, AzuRea G (2012) Early interleukin-6 and slope of 81. Ono S, Ueno C, Aosasa S, Tsujimoto H, Seki S, Mochizuki H
monocyte human leukocyte antigen-DR: a powerful association to (2001) Severe sepsis induces deficient interferon-gamma and
predict the development of sepsis after major trauma. PLoS One interleukin-12 production, but interleukin-12 therapy improves
7(3):e33095. doi:10.1371/journal.pone.0033095 survival in peritonitis. Am J Surg 182(5):491–497
66. Wu HP, Chen CK, Chung K, Tseng JC, Hua CC, Liu YC, Chuang 82. Cai S, Batra S, Lira SA, Kolls JK, Jeyaseelan S (2010) CXCL1
DY, Yang CH (2009) Serial cytokine levels in patients with severe regulates pulmonary host defense to Klebsiella infection via
sepsis. Inflamm Res 58(7):385–393. doi:10.1007/s00011-009- CXCL2, CXCL5, NF-kappaB, and MAPKs. J Immunol
0003-0 185(10):6214–6225. doi:10.4049/jimmunol.0903843
67. Kellum JA, Kong L, Fink MP, Weissfeld LA, Yealy DM, Pinsky 83. Robben PM, LaRegina M, Kuziel WA, Sibley LD (2005)
MR, Fine J, Krichevsky A, Delude RL, Angus DC, Gen IMSI Recruitment of Gr-1+ monocytes is essential for control of acute
(2007) Understanding the inflammatory cytokine response in toxoplasmosis. J Exp Med 201(11):1761–1769. doi:10.1084/jem.
pneumonia and sepsis: results of the Genetic and Inflammatory 20050054
Markers of Sepsis (GenIMS) Study. Arch Intern Med 167(15): 84. Lionakis MS, Swamydas M, Fischer BG, Plantinga TS, Johnson
1655–1663. doi:10.1001/archinte.167.15.1655 MD, Jaeger M, Green NM, Masedunskas A, Weigert R, Mikelis
68. Riedemann NC, Guo RF, Hollmann TJ, Gao H, Neff TA, Reuben C, Wan W, Lee CC, Lim JK, Rivollier A, Yang JC, Laird GM,
JS, Speyer CL, Sarma JV, Wetsel RA, Zetoune FS, Ward PA Wheeler RT, Alexander BD, Perfect JR, Gao JL, Kullberg BJ,
(2004) Regulatory role of C5a in LPS-induced IL-6 production Netea MG, Murphy PM (2013) CX3CR1-dependent renal macro-
by neutrophils during sepsis. FASEB J 18(2):370–372. doi:10. phage survival promotes Candida control and host survival. J Clin
1096/fj.03-0708fje Invest 123(12):5035–5051. doi:10.1172/JCI71307
69. Riedemann NC, Neff TA, Guo RF, Bernacki KD, Laudes IJ, 85. Chousterman BG, Boissonnas A, Poupel L, Baudesson de
Sarma JV, Lambris JD, Ward PA (2003) Protective effects of IL- Chanville C, Adam J, Tabibzadeh N, Licata F, Lukaszewicz AC,
6 blockade in sepsis are linked to reduced C5a receptor expres- Lombes A, Deterre P, Payen D, Combadiere C (2016) Ly6Chigh
sion. J Immunol 170(1):503–507 monocytes protect against kidney damage during sepsis via a
70. Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM (2001) CX3CR1-dependent adhesion mechanism. J Am Soc Nephrol
The soluble interleukin 6 receptor: mechanisms of production and 27(3):792–803. doi:10.1681/ASN.2015010009
implications in disease. FASEB J 15(1):43–58. doi:10.1096/fj.99- 86. Hamilton JA (2008) Colony-stimulating factors in inflammation
1003rev and autoimmunity. Nat Rev Immunol 8(7):533–544. doi:10.1038/
71. Hsieh CS, Macatonia SE, Tripp CS, Wolf SF, O’Garra A, Murphy nri2356
KM (1993) Development of TH1 CD4+ T cells through IL-12 87. Rauch PJ, Chudnovskiy A, Robbins CS, Weber GF, Etzrodt M,
produced by Listeria-induced macrophages. Science 260(5107): Hilgendorf I, Tiglao E, Figueiredo JL, Iwamoto Y, Theurl I,
547–549 Gorbatov R, Waring MT, Chicoine AT, Mouded M, Pittet MJ,
72. Costa VS, Mattana TC, da Silva ME (2010) Unregulated IL-23/ Nahrendorf M, Weissleder R, Swirski FK (2012) Innate response
IL-17 immune response in autoimmune diseases. Diabetes Res activator B cells protect against microbial sepsis. Science
Clin Pract 88(3):222–226. doi:10.1016/j.diabres.2010.03.014 335(6068):597–601. doi:10.1126/science.1215173
73. Reynolds JM, Angkasekwinai P, Dong C (2010) IL-17 family 88. Schreuder H, Tardif C, Trump-Kallmeyer S, Soffientini A,
member cytokines: regulation and function in innate immunity. Sarubbi E, Akeson A, Bowlin T, Yanofsky S, Barrett RW
Cytokine Growth Factor Rev 21(6):413–423. doi:10.1016/j. (1997) A new cytokine-receptor binding mode revealed by the
cytogfr.2010.10.002 crystal structure of the IL-1 receptor with an antagonist. Nature
74. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B 386(6621):194–200. doi:10.1038/386194a0
(2006) TGFbeta in the context of an inflammatory cytokine milieu 89. Dinarello CA (1997) Induction of interleukin-1 and interleukin-1
supports de novo differentiation of IL-17-producing T cells. receptor antagonist. Semin Oncol 24(3 Suppl 9):S9-81–S89-93
Immunity 24(2):179–189. doi:10.1016/j.immuni.2006.01.001 90. Alexander HR, Doherty GM, Venzon DJ, Merino MJ, Fraker DL,
75. McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein Norton JA (1992) Recombinant interleukin-1 receptor antagonist
W, McClanahan T, Cua DJ (2007) TGF-beta and IL-6 drive the (IL-1ra): effective therapy against gram-negative sepsis in rats.
production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell- Surgery 112(2):188–193 discussion 193-184
mediated pathology. Nat Immunol 8(12):1390–1397. doi:10. 91. Herold S, Tabar TS, Janssen H, Hoegner K, Cabanski M, Lewe-
1038/ni1539 Schlosser P, Albrecht J, Driever F, Vadasz I, Seeger W,
Semin Immunopathol

Steinmueller M, Lohmeyer J (2011) Exudate macrophages atten- changes in gene expression in septic shock patients: a genome-
uate lung injury by the release of IL-1 receptor antagonist in gram- wide approach. Intensive Care Med Exp 2(1):20. doi:10.1186/
negative pneumonia. Am J Respir Crit Care Med 183(10):1380– s40635-014-0020-3
1390. doi:10.1164/rccm.201009-1431OC 107. Payen D, Lukaszewicz AC, Belikova I, Faivre V, Gelin C,
92. Wynn TA (2015) Type 2 cytokines: mechanisms and therapeutic Russwurm S, Launay JM, Sevenet N (2008) Gene profiling in
strategies. Nat Rev Immunol 15(5):271–282. doi:10.1038/nri3831 human blood leucocytes during recovery from septic shock.
93. Luzina IG, Keegan AD, Heller NM, Rook GA, Shea-Donohue T, Intensive Care Med 34(8):1371–1376. doi:10.1007/s00134-008-
Atamas SP (2012) Regulation of inflammation by interleukin-4: a 1048-1
review of Balternatives^. J Leukoc Biol 92(4):753–764. doi:10. 108. Opal SM, Dellinger RP, Vincent JL, Masur H, Angus DC (2014)
1189/jlb.0412214 The next generation of sepsis clinical trial designs: what is next
94. Couper KN, Blount DG, Riley EM (2008) IL-10: the master reg- after the demise of recombinant human activated protein C?*. Crit
ulator of immunity to infection. J Immunol 180(9):5771–5777 Care Med 42(7):1714–1721. doi:10.1097/CCM.
95. Opal SM, DePalo VA (2000) Anti-inflammatory cytokines. Chest 0000000000000325
117(4):1162–1172 109. Hahn EO, Houser HB, Rammelkamp CH Jr, Denny FW,
96. Hotchkiss RS, Monneret G, Payen D (2013) Sepsis-induced im- Wannamaker LW (1951) Effect of cortisone on acute streptococ-
munosuppression: from cellular dysfunctions to immunotherapy. cal infections and poststreptococcal complications. J Clin Invest
Nat Rev Immunol 13(12):862–874. doi:10.1038/nri3552 30(3):274–281. doi:10.1172/JCI102441
97. Yang YC, Ciarletta AB, Temple PA, Chung MP, Kovacic S, 110. Rhen T, Cidlowski JA (2005) Antiinflammatory action of gluco-
Witek-Giannotti JS, Leary AC, Kriz R, Donahue RE, Wong GG corticoids—new mechanisms for old drugs. N Engl J Med
et al (1986) Human IL-3 (multi-CSF): identification by expression 353(16):1711–1723. doi:10.1056/NEJMra050541
cloning of a novel hematopoietic growth factor related to murine
111. Annane D, Sebille V, Charpentier C, Bollaert PE, Francois B,
IL-3. Cell 47(1):3–10
Korach JM, Capellier G, Cohen Y, Azoulay E, Troche G,
98. Weber GF, Chousterman BG, He S, Fenn AM, Nairz M, Anzai A,
Chaumet-Riffaud P, Bellissant E (2002) Effect of treatment with
Brenner T, Uhle F, Iwamoto Y, Robbins CS, Noiret L, Maier SL,
low doses of hydrocortisone and fludrocortisone on mortality in
Zonnchen T, Rahbari NN, Scholch S, Klotzsche-von Ameln A,
patients with septic shock. JAMA 288(7):862–871
Chavakis T, Weitz J, Hofer S, Weigand MA, Nahrendorf M,
112. Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K,
Weissleder R, Swirski FK (2015) Interleukin-3 amplifies acute
Weiss YG, Benbenishty J, Kalenka A, Forst H, Laterre PF,
inflammation and is a potential therapeutic target in sepsis.
Reinhart K, Cuthbertson BH, Payen D, Briegel J, Group CS
Science 347(6227):1260–1265. doi:10.1126/science.aaa4268
(2008) Hydrocortisone therapy for patients with septic shock. N
99. Bentzer P, Fjell C, Walley KR, Boyd J, Russell JA (2016) Plasma
Engl J Med 358(2):111–124. doi:10.1056/NEJMoa071366
cytokine levels predict response to corticosteroids in septic shock.
Intensive Care Med 42(12):1970–1979. doi:10.1007/s00134-016- 113. Opal SM, Fisher CJ Jr, Dhainaut JF, Vincent JL, Brase R, Lowry
4338-z SF, Sadoff JC, Slotman GJ, Levy H, Balk RA, Shelly MP, Pribble
100. Copeland S, Warren HS, Lowry SF, Calvano SE, Remick D, JP, LaBrecque JF, Lookabaugh J, Donovan H, Dubin H,
Inflammation, the Host Response to Injury I (2005) Acute inflam- Baughman R, Norman J, DeMaria E, Matzel K, Abraham E,
matory response to endotoxin in mice and humans. Clin Diagn Seneff M (1997) Confirmatory interleukin-1 receptor antagonist
Lab Immunol 12(1):60–67. doi:10.1128/CDLI.12.1.60-67.2005 trial in severe sepsis: a phase III, randomized, double-blind, pla-
101. Bahador M, Cross AS (2007) From therapy to experimental mod- cebo-controlled, multicenter trial. The Interleukin-1 Receptor
el: a hundred years of endotoxin administration to human subjects. Antagonist Sepsis Investigator Group. Crit Care Med 25(7):
J Endotoxin Res 13(5):251–279. doi:10.1177/0968051907085986 1115–1124
102. Dorresteijn MJ, Visser T, Cox LA, Bouw MP, Pillay J, 114. Opal SM, Laterre PF, Francois B, LaRosa SP, Angus DC, Mira JP,
Koenderman AH, Strengers PF, Leenen LP, van der Hoeven JG, Wittebole X, Dugernier T, Perrotin D, Tidswell M, Jauregui L,
Koenderman L, Pickkers P (2010) C1-esterase inhibitor attenu- Krell K, Pachl J, Takahashi T, Peckelsen C, Cordasco E, Chang
ates the inflammatory response during human endotoxemia. Crit CS, Oeyen S, Aikawa N, Maruyama T, Schein R, Kalil AC, Van
C a r e M e d 3 8 ( 11 ) : 2 1 3 9 – 2 1 4 5 . d o i : 1 0 . 1 0 9 7 / C C M . Nuffelen M, Lynn M, Rossignol DP, Gogate J, Roberts MB,
0b013e3181f17be4 Wheeler JL, Vincent JL, Group AS (2013) Effect of eritoran, an
103. van Deventer SJ, Buller HR, ten Cate JW, Aarden LA, Hack CE, antagonist of MD2-TLR4, on mortality in patients with severe
Sturk A (1990) Experimental endotoxemia in humans: analysis of sepsis: the ACCESS randomized trial. JAMA 309(11):1154–
cytokine release and coagulation, fibrinolytic, and complement 1162. doi:10.1001/jama.2013.2194
pathways. Blood 76(12):2520–2526 115. Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S,
104. Calvano SE, Xiao W, Richards DR, Felciano RM, Baker HV, Cho Levy H, Bone R, Wenzel RP, Balk R, Allred R et al (1995)
RJ, Chen RO, Brownstein BH, Cobb JP, Tschoeke SK, Miller- Efficacy and safety of monoclonal antibody to human tumor ne-
Graziano C, Moldawer LL, Mindrinos MN, Davis RW, crosis factor alpha in patients with sepsis syndrome. A random-
Tompkins RG, Lowry SF, Inflamm, Host Response to Injury ized, controlled, double-blind, multicenter clinical trial. TNF-
Large Scale Collab. Res P (2005) A network-based analysis of alpha MAb Sepsis Study Group. JAMA 273(12):934–941
systemic inflammation in humans. Nature 437(7061):1032– 116. McCloskey RV, Straube RC, Sanders C, Smith SM, Smith CR
1037. doi:10.1038/nature03985 (1994) Treatment of septic shock with human monoclonal anti-
105. Shalova IN, Lim JY, Chittezhath M, Zinkernagel AS, Beasley F, body HA-1A. A randomized, double-blind, placebo-controlled tri-
Hernandez-Jimenez E, Toledano V, Cubillos-Zapata C, Rapisarda al. CHESS Trial Study Group. Ann Intern Med 121(1):1–5
A, Chen J, Duan K, Yang H, Poidinger M, Melillo G, Nizet V, 117. Payen DM, Guilhot J, Launey Y, Lukaszewicz AC, Kaaki M,
Arnalich F, Lopez-Collazo E, Biswas SK (2015) Human mono- Veber B, Pottecher J, Joannes-Boyau O, Martin-Lefevre L,
cytes undergo functional re-programming during sepsis mediated Jabaudon M, Mimoz O, Coudroy R, Ferrandiere M, Kipnis E,
by hypoxia-inducible factor-1alpha. Immunity 42(3):484–498. Vela C, Chevallier S, Mallat J, Robert R, Group A (2015) Early
doi:10.1016/j.immuni.2015.02.001 use of polymyxin B hemoperfusion in patients with septic shock
106. Cazalis MA, Lepape A, Venet F, Frager F, Mougin B, Vallin H, due to peritonitis: a multicenter randomized control trial. Intensive
Paye M, Pachot A, Monneret G (2014) Early and dynamic Care Med 41(6):975–984. doi:10.1007/s00134-015-3751-z
Semin Immunopathol

118. Qiu P, Cui X, Sun J, Welsh J, Natanson C, Eichacker PQ (2013) 122. London NR, Zhu W, Bozza FA, Smith MC, Greif DM, Sorensen
Antitumor necrosis factor therapy is associated with improved LK, Chen L, Kaminoh Y, Chan AC, Passi SF, Day CW, Barnard
survival in clinical sepsis trials: a meta-analysis. Crit Care Med DL, Zimmerman GA, Krasnow MA, Li DY (2010) Targeting
41(10):2419–2429. doi:10.1097/CCM.0b013e3182982add Robo4-dependent Slit signaling to survive the cytokine storm in
119. Schouten M, Wiersinga WJ, Levi M, van der Poll T (2008) sepsis and influenza. Sci Transl Med 2(23):23ra19. doi:10.1126/
Inflammation, endothelium, and coagulation in sepsis. J Leukoc scitranslmed.3000678
Biol 83(3):536–545. doi:10.1189/jlb.0607373 123. Zhao H, Anand AR, Ganju RK (2014) Slit2-Robo4 pathway mod-
120. Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, ulates lipopolysaccharide-induced endothelial inflammation and
Lopez-Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, its expression is dysregulated during endotoxemia. J Immunol
Ely EW, Fisher CJ Jr, Recombinant human protein CWEiSSsg 192(1):385–393. doi:10.4049/jimmunol.1302021
(2001) Efficacy and safety of recombinant human activated pro- 124. Maceyka M, Harikumar KB, Milstien S, Spiegel S (2012)
tein C for severe sepsis. N Engl J Med 344(10):699–709. doi:10. Sphingosine-1-phosphate signaling and its role in disease.
1056/NEJM200103083441001 Trends Cell Biol 22(1):50–60. doi:10.1016/j.tcb.2011.09.003
121. Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, 125. Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E, Scott
Finfer S, Gardlund B, Marshall JC, Rhodes A, Artigas A, Payen F, Martinborough E, Peach R, Oldstone MB, Rosen H (2011)
D, Tenhunen J, Al-Khalidi HR, Thompson V, Janes J, Macias WL, Endothelial cells are central orchestrators of cytokine amplifica-
Vangerow B, Williams MD, Group P-SS (2012) Drotrecogin alfa tion during influenza virus infection. Cell 146(6):980–991. doi:10.
(activated) in adults with septic shock. N Engl J Med 366(22): 1016/j.cell.2011.08.015
2055–2064. doi:10.1056/NEJMoa1202290

You might also like