REVIEW

JOURNAL OF INTERFERON & CYTOKINE RESEARCH Volume 00, Number 00, 2014 ª Mary Ann Liebert, Inc. DOI: 10.1089/jir.2014.0119

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

The Growing Spectrum of Anti-Inflammatory Interleukins and Their Potential Roles in the Development of Sepsis Hong-qiang Zhao,1,2,* Wei-min Li,3,* Zhong-qiu Lu,4 Zhi-yong Sheng,2 and Yong-ming Yao 2

Sepsis, recognized as a deadly immunological disorder, is one of the major causes of death in intensive care units globally. Traditionally, sepsis was characterized by an excessive systemic proinflammatory response to invasive microbial pathogens. However, failures of highly sophisticated trials directed toward the uncontrolled inflammatory reaction have led to an appeal by experts for reevaluation of the present approach toward sepsis. With accumulated evidence, a principal role for immunosuppression in severe sepsis has been evaluated. Different pathways of negative regulation in the pathophysiological process of sepsis have been investigated. Significant among these regulatory elements are the anti-inflammatory cytokines. In the past few years, several interleukins (ILs) have been identified and characterized, among which IL-35 and IL-37 represent newly identified ones in the spectrum of anti-inflammatory cytokines. In this study, we focus on regulatory cytokines of the IL family (including the old members: IL-4, IL-10, and IL-13, and newly discovered ones: IL-35 and IL37) to address current knowledge regarding their structural and functional characteristics as well as their roles in the development of sepsis. Although the exact roles for these cytokines are pending further elucidation, the current advances in our understanding of mechanisms that regulate the immune responses during severe sepsis may lead to the identification of new diagnostic or treatment targets.

sepsis was considered as 1 of the 5 conditions where hospitalization expenses were the most expensive (Chalupka and Talmor 2012).

Introduction

S

epsis, an intricate clinical syndrome arising from the interaction between microbes and host, can end in severe sepsis and septic shock (Levy and others 2003). Sepsis, severe sepsis, and septic shock are major healthcare problems affecting millions of people around the world each year, accounting for one-fourth of deaths, and are increasing in incidence (Dellinger and others 2013). Septic shock had been reported in an epidemiological study as the most common cause of death in noncoronary intensive care units and the 10th leading cause of death in high-income countries (Angus and others 2001). As the growing population at risk for the development of septic complications comprises the increase in the number of elderly and immunocompromised patients, there is a steady rise in the incidence of severe sepsis (Martin and others 2003). Although there is progress in the area of supportive care and immunomodulatory therapies, the mortality rate of severe sepsis remains high (Brun-Buisson 2000). Apart from a clinical challenge, sepsis was also believed to be a large economic burden for global healthcare systems. It was reported that in the United States

Pathophysiology of Septic Response In recent decades, a great number of scientific articles have been published aiming to elucidate the intricate and dynamic pathophysiological mechanisms that cause the heterogeneous sepsis syndrome. It has been shown that the imbalance between proinflammatory and anti-inflammatory responses caused by amplified and subsequently dysregulated host reaction to a microbe infection leads to the development of sepsis (Cohen 2002). Initial host immune response started when invading microorganisms were detected by an innate immune system through pathogen recognition receptors (PRRs), which are expressed on immune cells such as dendritic cells and macrophages as well as epithelial barriers (Akira and others 2006). PRRs recognize the pathogen-associated molecular patterns (PAMPs), which means the macromolecular motives from microorganisms usually comprise lipopolysaccharide (LPS; the main virulence

1

Medical School of Chinese People’s Liberation Army, the Chinese PLA General Hospital, Beijing, People’s Republic of China. Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People’s Republic of China. 3 Department of Hepatobiliary Surgery, the 309th Hospital of Chinese PLA, Beijing, People’s Republic of China. 4 Emergency Department, First Affiliated Hospital of Wenzhou Medical College, Wenzhou, People’s Republic of China. *These authors contributed equally to this study. 2

1

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

2

factor of gram-negative bacteria), lipoteichoic acid (a cell wall component of gram-positive bacteria), bacterial DNA, flagellin, and peptidoglycan (van der Poll and Opal 2008). The interaction between PRRs and PAMPs gives rise to downstream signaling cascades, which ultimately cause the activation of a transcriptional response program. The activation of nuclear factor kB represents one of such responses, which results in the secretion of cytokines, chemokines, and nitric oxide (Akira and Takeda 2004; O’Neill 2011). Generally speaking, sepsis was characterized by an excessive systemic proinflammatory reaction to invasive microbial pathogens. The innate immune response started by interaction between PRRs and PAMPs results in the excessive secretion of cytokines, chemokines, and other inflammatory mediators. A wide variety of inflammatory reactions, including migration of immune cells to the site of infection, which is critical for infection localization, are regulated by cytokines. However, a dysregulated cytokine release may lead to harm the host. One of such typical risks is the endothelial dysfunction with the feature of vasodilation and increased capillary permeability. The frequent symptoms in septic settings, such as hypotension, hemoconcentration, macromolecular extravasation, and edema, are believed to be associated with the leakage syndrome (Rivers and others 2001). Furthermore, accumulated evidence has manifested that various human body physiological courses, including coagulation, metabolism, and neuroendocrine activation, are tightly interlaced with the immune and inflammatory reactions (Atsumi and others 2007; Emonts and others 2007; Levi and van der Poll 2010). Dysregulation of the coagulation system induced by inflammation significantly aggravates the disturbance in coagulation of sepsis, leading to lethal disseminated intravascular coagulation (Hook and Abrams 2012). With the progress of the research, the host immune response in sepsis has been recognized to develop in different, but overlapping, stages. During the course of sepsis, the original excessive systemic proinflammatory response, termed as systemic inflammatory response syndrome (SIRS), overlaps with the counter-regulatory phase, which came to be known as the compensatory anti-inflammatory response syndrome (CARS) (Bone 1996). Functionally, the immune responses during CARS are suggested to work as a brake on systemic inflammation. The entire immune state of 1 patient is dependent on predomination of either SIRS or CARS since they overlap significantly (van der Poll and van Deventer 1999). Immoderate CARS in sepsis is also believed to be harmful to the host. The immunosuppressive state during the periseptic period is mediated by the compensatory secretion of anti-inflammatory mediators and it is believed to be of considerable clinical significance, which is proved by the more virulent infection and higher mortality found in septic patients who get obvious immunoparalysis (Appel and others 1989). Worldwide, researchers have reiterated that studies for sepsis need new directions in research since the failure of a couple of high-profile clinical experiments directed toward SIRS in sepsis (Cohen and others 2012; Wenzel and Edmond 2012). The cause of death for septic patients was highlighted by the postmortem studies, and the immunological defects manifested as impairment of host immunity was highlighted (Boomer and others 2011). Beneficial effects have gained in the treatment of severe sepsis shown by couples of small phase-2 clinical trials of immune-enhancing drugs, thereby substantiating

ZHAO ET AL.

the concept that immunosuppression may play an essential role in the development of septic complications (Meisel and others 2009).

Anti-Inflammatory Cytokines in Sepsis Although imperfect, our comprehension on cellular and molecular mechanisms underlying sepsis-caused immunosuppression has largely been expanded during the last 2 decades. Different pathways of negative regulation in the pathophysiological process of sepsis have been investigated (Luan and others 2012). Prominent members among these regulatory components are the anti-inflammatory cytokines. Under pathological conditions, anti-inflammatory cytokines may either provide insufficient control over proinflammatory activities or overcompensate and inhibit the immune response, rendering the host risky to systemic infection (Fig. 1). Multiple factors, including the time of release, the local circumstances where they act, and the existence of competing or synergistic factors, might contribute to the final effect of anti-inflammatory cytokines (Opal and DePalo 2000). In recent years, some newly discovered anti-inflammatory cytokines of interleukin (IL) families such as IL-35 and IL-37 have been studied. In this study, we will discuss the old triad (IL-4, IL-10, and IL-13) and the newcomers (IL-35 and IL-37) of anti-inflammatory cytokines of IL families regarding their basic biological characteristics and potential roles in the development of sepsis (see Table 1 as a summary).

Interleukin-4 IL-4, a multifunctional pleiotropic cytokine with a molecular weight of 15 kDa, is not only produced mainly by activated T cells but also by mast cells, basophils, and eosinophils (Nelms and others 1999). IL-4 shares sequence homology, cell surface receptors, intracellular signaling, and a partial functional effect on cells with IL-13 (LaPorte and others 2008). IL-4 binds to and signals through 2 combinations of 3 receptor subunits: IL-4Ra, IL-13Ra1, and the common gchain (gc). The type I IL-4 receptor (IL-4R), a heterodimer of IL-4Ra and gc, is the main receptor on hematopoietic cells. The type II IL-4R, a heterodimer of IL-4Ra and IL13Ra1 chains, can bind both IL-4 and IL-13 and is thought to be the primary receptor utilized by nonhematopoietic cells (Pillai and Bix 2011). Soluble IL-4 binds with high affinity to the extracellular domain of IL-4Ra and nearly undetectable affinity to either soluble gc or soluble IL13Ra1 (Kraich and others 2006). By contrast, the IL-4/IL4Ra complex binds with detectable though low affinity to soluble gc and IL-13Ra1. Consistent with this, IL-4 signaling complexes are thought to be assembled in a stepwise manner started by the binding of soluble IL-4 to the IL-4Ra subunit, followed by recruitment in the plane of the membrane of either gc or IL-13Ra1 to the IL-4/IL-4Ra complex. Complex assembly initiates intracellular signaling through activation of the Janus kinase ( JAK) 1/3-signal transducer and activator of the transcription (STAT) 6 and IRS1/2phosphatidylinositol 3-kinase (PI3K) pathways. Functionally, IL-4 plays a key role in the regulation of T lymphocyte differentiation. It promotes the differentiation of Th2 cells while inhibiting that of Th1 cells (Seder and others 1992). Being a primary cytokine produced by Th2

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

ANTI-INFLAMMATORY INTERLEUKINS AND THIER ROLES IN SEPSIS

3

FIG. 1. Schematic depiction of immune response directed mainly by cytokines in the development of sepsis, including secretion phase, biological effects, clinical significance, and host outcome. The initial excessive systemic proinflammatory response in sepsis, termed as systemic inflammatory response syndrome (SIRS), overlaps with the counter-regulatory phase referred to as the compensatory anti-inflammatory response syndrome (CARS). SIRS and CARS overlap significantly; hence, the overall immune status of the patient is dependent on which response predominates. TNF, tumor necrosis factor; IL, interleukin; IFN, interferon.

lymphocytes, IL-4 leads to a further enhanced secretion of itself and other anti-inflammatory cytokines, and thus causes the suppression of monocyte-derived proinflammatory cytokines (Opal and DePalo 2000). The role of IL-4 for the pathophysiology of sepsis is profoundly studied. It has been revealed that IL-4 increased survival of mice exposed to lethal doses of LPS (Baumhofer and others 1998). The activation of the STAT6 pathway induced by IL-4 contributes to the suppression of cellmediated immunity and death in animals with severe sepsis (Song and others 2000). However, deleterious effects of IL4 were also described in Staphylococcus aureus-challenged murine septic setting, which indicated a relationship between the effect of IL-4 and the host’s genetic background (Hultgren and others 1999). Human-based studies have shown that the mRNA expression of IL-4 was associated with the survival of patients with severe sepsis; however, no obvious difference of plasma IL-4 levels in septic patients of the day, on admission, were found between survivors and nonsurvivors (Wu and others 2008). Enzyme-linked immunosorbent assays (ELISA) in 56 patients with severe trauma who developed sepsis, of whom 36 died, indicated that IL-4 had no significant correlation with the severity and outcome of sepsis (Surbatovic and others 2007). Recently, an association of IL-4 polymorphism and sepsis has been reported. It was reported that IL-4 promoter polymorphisms might influence the balance between Th1 and Th2 responses, and thus predispose trauma patients to the development of sepsis (Gu and others 2011). Studies up to now have demonstrated that IL-4 plays a critical role in the pathogenesis of septic complication; however, more work needs

to be done to elucidate its precise mechanism during the course of the disease.

Interleukin-10 IL-10 was initially described as a cytokine synthesis inhibitory factor produced by Th2 cells that inhibit the function of Th1 cells (Fiorentino and others 1989). It is now recognized that numerous types of immune cells, such as monocytes, macrophages, B and T lymphocytes, and natural killer cells, can express and secrete this 35-kDa homodimeric cytokine-IL-10 (Latifi and others 2002). Two distinct chains (IL-10R1 and IL-10R2), which are part of the class II cytokine receptor family (CRF2), constitute IL-10R (Sabat and others 2010). In 1993, the murine IL-10R1 was first cloned by Moore and others (Ho and others 1993). The sequence of human IL-10R1, which is 60% identical and 73% similar to the murine chain, was published by the same group 1 year later (Liu and others 1994). IL-10R1 whose encoding gene is located on chromosome 11 has been identified as a glycosylated protein with the molecular weight of 90–110 kDa (Taniyama and others 1995). Subsequently, IL-10R2 is suggested to be the protein previously known as CRF2-4 since 1993 (Kotenko and others 1997), and the gene for IL-10R2 is located on chromosome 21. It has been demonstrated that the ligandreceptor binding for IL-10 is initiated by its combination to IL-10R1 and this interaction alters the cytokine conformation enabling the subsequent association of the IL-10/IL10R1 complex with IL-10R2 (Yoon and others 2006). Immune cells, especially monocytes and macrophages, express most IL-10R1 (Wolk and others 2002). In comparison with

4

Activated T cells, Mast cells, basophils, eosinophils

Th2 cells, basophils, mast cells, natural killer cells, fibroblasts, dendritic cells, alveolar macrophages

CD4 + CD25 + regulatory T cells

Peripheral blood mononuclear cells, monocytes, plasma cells, dendritic cells, epithelial cells, carcinoma cells, testis, thymus, and uterus/ no homolog in mouse was found

IL-4

IL-13

IL-35

IL-37

STAT1/STAT4

Not entirely clear/act as transcriptional modulator or act through SMAD3

Not clear/the binging to the IL-18Ra was observed

JAK-STAT6; PI3K, STAT3, MAPK

IL-4Ra, IL-13Ra1

IL-12b2/gp130

JAK1/3-STAT6; IRS1/2-PI3K

JAK1/ TYK2-STAT3

Signal cascade

IL-4Ra, IL-13Ra1, gc

IL-10R (consists of IL-10R1 and IL-10R2)

Receptor

Anti-inflammatory cytokine/ suppress T cell proliferation and effector functions; induce the conversion of iTR35 Anti-inflammatory cytokine/ limiting tissue injury during infections

Inhibitor of proinflammatory cytokine production/ upregulate MHC-II and CD23

Th1 inhibition/Th2 differentiation/enhance other anti-inflammatory cytokines release/suppress monocyte-derived proinflammatory cytokines secretion

Affect functions of monocytes/ block Th17 proliferation/ promote Tregs survival/ suppress proinflammatory mediator production

Main function

IL-37 transgenic mice are protected against LPS challenge

Not clear

Sepsis models: conflicting Human sepsis: not exactly clear—plasma levels seemed to be not associated with outcome of sepsis/IL-4 promoter may have predictive value Sepsis models: protective role in the development of sepsis/regulate organ-specific inflammation during sepsis Human sepsis: controversial

Sepsis models: conflicting Human sepsis: not exactly clear—might have diagnostic value

Involvement in sepsis and sepsis models

Banchereau and others (2012) Fiorentino and others (1991) Gogos and others (2000) Howard and others (1993) Latifi and others (2002) Moore and others (2001) O’Garra and others (2004) Song and others (1999) Baumhofer and others (1998) Gu and others (2011) Hultgren and others (1999) Nelms and others (1999) Opal and DePalo (2000) Pillai and Bix (2001) Seder and others (1992) Surbatovic and others (2007) Burd and others (1995) Cao and others (2012) Collighan and others (2004) Hancock and others (1998) Hoshino and others (1999) Mannon and Reinisch (2012) Matsukawa and others (2000) Minty and others (1993) Minty and others (1997) Punnonen and others (1993) van der Poll and others (1997) Zurawski and de Vries (1994) Collison and others (2007) Collison and others (2010) Collison and others (2012) Niedbala and others (2007) Olson and others (2013) Kumar and others (2002) Nold and others (2003) Nold and others (2010) Nold and others (2011) Pan and others (2001) Sharma and others (2008) Tete and others (2012)

References

gc, g-chain; JAK, Janus kinase; LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol 3-kinase; STAT, signal transducer and activator of transcription; Tregs, regulatory T cells.

Monocytes, macrophages, T cells, B cells, NK cells

Main sources

IL-10

Cytokine

Table 1. Summary of the Main Features of Anti-Inflammatory Interleukins

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

ANTI-INFLAMMATORY INTERLEUKINS AND THIER ROLES IN SEPSIS

IL-10R1, IL-10R2 can be widely and abundantly found in most cells and tissues (Wolk and others 2004; Kunz and others 2006). Unlike the specificity for IL-10R1 to IL-10R complex, IL-10R2 simultaneously constitutes other receptor complexes. Engagement of IL-10 to its receptor induces the phosphorylation of JAK1 and TYK2 and STAT3 (Moore and others 2001). Functionally, IL-10 acts on a wide variety of cells, including monocytes, macrophages, dendritic cells, B and T cells, and even regulatory T cells (Tregs) (O’Garra and others 2004). IL-10 affects many important functions of monocytes, macrophages, and dendritic cells from phagocytosis to the production of cytokines, the expression of costimulators, and the processing as well as presentation of antigens. In addition, IL-10 directly acts on proinflammatory Th17 cells by blocking the proliferation. However, for CD4 + Foxp3 + Tregs, IL-10 promotes its survival and contributes to the function (Banchereau and others 2012). In vitro, IL-10 suppresses the production of proinflammatory mediators, including tumor necrosis factor (TNF)-a, interferon (IFN)-g, IL-1, and granulocyte-macrophage colony-stimulating factor (GM-CSF), from immune cells (Fiorentino and others 1991). These results were also confirmed in vivo studies. The effects of IL-10 in animal infection models are conflicting. It was clearly shown in early studies that IL-10 possessed a protective effect in the pathology of the LPSinduced experimental murine model. Administration of the recombinant IL-10 protein protected mice from lethal endotoxemia, even when injection started 30 min after the LPS challenge (Howard and others 1993). Nevertheless, later studies using the cecal ligation and puncture (CLP) model of polymicrobial sepsis did not always show the beneficial effect of IL-10. Indeed, improved survivals were observed in the CLP model when IL-10 was inhibited at 12 h after onset (Song and others 1999). However, the effect turned to be lethal when administration of neutralizing IL-10 antibodies at the time of the CLP model commenced (van der Poll and others 1995). As multiple biological properties of IL-10 have been demonstrated, it is becoming increasingly apparent that IL-10 as a target of treatment is fraught with theoretical and practical concerns. While as Gogos and others (2000) report, IL-10 might have a diagnostic value for septic patients. They found in their study that the constant overproduction of IL-10 appeared to be the master predictor of severity and deadly outcome, although both the inflammatory and anti-inflammatory responses were deeply augmented. This is of great significance as the definition of the immune status of the patient with sepsis before therapeutic manipulations can be helpful in the selection of patients who may benefit from it.

Interleukin-13 IL-13, a 33 amino acid peptide cytokine, possesses the gene location at human chromosome 5q31, which is resided within a cluster of cytokine genes that consist of IL-3, IL-4, IL-5, and GM-CSF (McKenzie and others 1993). As a Th2 family cytokine, IL-13 was originally identified as a product of activated CD4 + T cells (Minty and others 1993). However, in humans, it has also been reported that basophils, mast cells, natural killer cells, fibroblasts, dendritic cells, and alveolar macrophages have a capacity to produce IL-13

5

(Burd and others 1995; Hancock and others 1998; Hoshino and others 1999). Two cell surface receptors have been suggested to be bound by IL-13, but just one of them takes the predominant signal events. IL-13Ra1, which binds IL-13 alone with low affinity, forms high affinity type1 IL-13Rs by dimerizing with IL-4Ra. This type 1 IL-13R transduces intracellular signals through phosphorylation of STAT6 through JAK kinases. The other second messenger molecule has also been suggested to be activated in various cell types, including PI3K, STAT3, and mitogen-activated protein kinase (MAPK). The type 2 IL-13R (IL-13Ra2), which has less definite signaling properties, acts chiefly as a monomer. Affinity for IL-13 Ra2 to IL-13 is higher than that of the dimeric type 1 IL-13Ra1/IL-4Ra receptor. The precise role for IL13Ra2 seemed to be as a sink for IL-13, binding the cytokine and preventing it from working with type I receptor, thereby downregulating the transduction of the intracellular signal (decoy receptor) (Mannon and Reinisch 2012). IL-13 is defined as an anti-inflammatory cytokine that has been shown to downregulate the expression of a number of proinflammatory cytokines (Minty and others 1997). It was reported that LPS-induced IL-6 as well as the mRNA of other proinflammatory cytokines, particularly TNF-a, IL1b, and IL-8, were strongly inhibited by IL-13 (Minty and others 1993; Muchamuel and others 1997; Wong and others 1997). Expression of certain surface antigens such as MHC class II antigens and CD23 was upregulated by this antiinflammatory cytokine (Punnonen and others 1993; Zurawski and de Vries 1994). Through inhibiting the effects of IL-10 and IFN-g, IL-13 suppresses indirectly antibodydependent cellular cytotoxicity in monocytes. It also restrains the expression of tissue factor induced by LPS and protects endothelial and monocyte activation (Zurawski and de Vries 1994). It has been demonstrated that in the murine CLP model, CLP-induced systemic inflammatory response could be balanced by a series of regulatory cytokines (Walley and others 1996; Hack and others 1997). Such fine balance prevents the inflammatory response from converting to pathological, and is self-destructive and fatal to the host (Kellum and Decker 1996). Rapidly, after CLP onset, IL-13 was found to be increased in tissues, including the liver, lungs and kidneys, without elevation of IL-13 levels found in either peritoneal fluid or serum (DiPiro 1997). Neutralization of IL-13 was shown to be detrimental to survival in a murine sepsis model, suggesting a protective role of IL-13 during the development of septic peritonitis (Matsukawa and others 2000). In another study, IL-13 was suggested to be required for mouse survival as a result of CLP and could be a powerful stimulus for chemokine C10 expression in peritoneal macrophages (Steinhauser and others 2000). These studies suggest that IL-13 is critically involved in regulating organ-specific inflammation during the pathological process of sepsis. Indeed, a recent study indicated that the intestinal concentration of IL-13 was dramatically increased in rats with sepsis as compared with healthy normal rats (Cao and others 2012). The potential role of IL-13 in human sepsis seems to be controversial. van der Poll and others (1997) carried out a study based on 42 septic patients and 7 healthy volunteers who received endotoxin intravenously. It was revealed that IL-13 was not found in most of the patients and was not

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

6

induced by endotoxin. These results were not in accordance with animal experiments. However, Collihgan and others (2004) investigated 31 patients with severe sepsis or sepsis shock. They observed that serum IL-13 level in the shock group from the day of admission to day 3 was remarkably higher, while it decreased subsequently to the level similar to that in the nonshock group. IL-13 level was found to be significantly correlated with TNF-a concentration in the study. Another study reported that IL-13 levels were lower in patients with sepsis, and low IL-13 levels in the early hours of sepsis were associated with poor prognosis. However, no correlation between IL-13 levels and other cytokines (IL-1b, IL-10, IL-12, TNF-a) was found (Blanco-Quiro´s and others 2005).

Interleukin-35 IL-35 was found to be a heterodimeric cytokine belonging to the IL-12 family, which is a group of cytokines composing 1 of 5 subunits [p19, p28, p35, p40, and Epstein-Barr virus-induced gene 3 (Ebi3)] that aggregate in various combinations to form IL-12, IL-23, IL-27, and IL-35 (Vignali and Kuchroo 2012). Two subunits, the a subunit (p35, which is encoded by Il12a) and the b subunit (Ebi3, which is encoded by Ebi3), constitute IL-35 (Niedbala and others 2007). Recently, the composition and signaling pathways of the IL-35Rs were characterized in mice. The receptor of IL-35 comprises IL-12b2 and gp130, which are also associated with IL-12 and IL-27Rs, respectively (Collison and others 2012). Following binding of IL-35 to its receptors, the signal is transduced through STAT1 and STAT4 to form a unique heterodimer and results in the expression of the target gene, including p35 and Ebi3, thus leading to a feedback loop enhancing IL-35 expression. Different from other members of the IL-12 family, IL-35 can also regulate the signal through a homodimer of its receptor subunit. However, when such binding happens, only one branch of the signal transduction pathway is activated (either STAT1 or STAT4 for gp130:gp130 or IL12b2:IL-12b2 homodimers, respectively). Although there is a diversity of the ligand-binding mode of IL-35 and its receptors, it seems that IL-35 can exert full functions only by binding to the IL-12b2-gp130 heterodimer receptors. Compared with other IL-12 family members, another feature of IL-35 is that rather than being expressed primarily by antigen-presenting cells IL-35 is expressed primarily by Tregs. Although dozens of reports have been published describing the expression of IL-35 in both thymus-derived and peripheral Tregs, the results taken together suggest that IL-35 may be closely associated with the suppressive activity of Tregs in peripheral tissues rather than a constitutive marker of Tregs (Olson and others 2013). The biological effect for IL-35 is incompletely understood and most of the available data are results from in vitro studies and animal models. With accumulated evidence, IL-35 is recognized as a typical anti-inflammatory cytokine, and the predominant mechanism of suppression associated with the activity of IL-35 is its ability to suppress T cell proliferation and effector functions (Collison and others 2007). Such an observation was not only proved by numerous models by several groups of investigators but also by studies using recombinant agent rIL-35 (Collison and others 2010; Wirtz and others 2011; Tirotta and others 2013).

ZHAO ET AL.

It has been demonstrated that Tregs, lacking p35 or EBI3, show decreased regulatory capabilities in vitro. These cells are unable to control T cell proliferation and inhibit the development of inflammatory bowel disease. IL-35 alone is sufficient to inhibit T cell proliferation in vitro. These findings suggest that optimal Tregs suppressive function may require the presence of IL-35 (Collison and others 2007, 2009). In addition to the direct inhibitory effect on naive T cells, IL-35 treatment of human or murine CD4 + T cells could induce their conversion to IL-35-producing T cells, known as iTr35 cells (Collison and others 2010; Chaturvedi and others 2011). It has been reported that maximal Tregs suppression requires not only IL-35 expression but also its contact with naive T cells that can subsequently be converted into iTr35 cells, and thus highlights the importance of IL-35 for the function of Tregs. The suppressive activity of IL-35 is not limited to CD4 + Tregs, as a population of CD8 + CTLA-4 + Tregs was noted to suppress the proliferation of autologous T cells in a contact independent and IL-35-dependent manner (Olson and others 2012). IL-35 has even been shown to exert an effect on suppressing Th17 responses. Tregs expressing IL-35 or rIL35 have been shown to reduce Th17 differentiation as well as the function of Th17 cells (Niedbala and others 2007; Kochetkova and others 2010; Liu and others 2012). Given the direct immunosuppressive potential of IL-35, there has been interest in evaluating the role of IL-35 in the development of a variety of diseases. Several diseases have been shown to be associated with increased IL-35 expression, including various inflammatory diseases, coronary artery disease, and cancer. Niedbala and colleagues found in their studies that IL-35 decreased the severity of collageninduced arthritis in animals (Niedbala and others 2007). Such an effect was mediated by the induction of a CD4 + CD39 + regulatory T population and by the release of IL-10 (Kochetkova and others 2010). The disease severity of a chronic colitis model was also attenuated following the injection of IL-35 (Wirtz and others 2011). In an asthma model, intratracheal instillation of IL-35 decreased disease severity by diminishing the Th2 cell counts and the release of IgE and IgG (Huang and others 2011). Moreover, Icos + Tregs-produced IL-35 downregulated airway hypersensitivity in mice by reducing the production of IL-17 (Whitehead and others 2012). However, the role of IL-35 in the pathological process of sepsis has not been highlighted. Further study is warranted to reveal the exact mechanism of IL-35 in the pathogenesis of sepsis.

Interleukin-37 The IL-1 family of ligands is related to acute and chronic inflammation and plays an essential role in the nonspecific innate response to infection. The biological properties of the IL-1 family of ligands are typically proinflammatory; however, IL-37 sharing the structural pattern of the IL-1 family, but particularly that of IL-18, functions as an antiinflammation cytokine. IL-37, formerly termed IL-1F7, stems from hematopoietic cells and is naturally expressed in organs that comprise epithelial cells (Tete and others 2012). The IL-1F7 cytokine is transcribed as 5 different splice variants, namely IL-1F7 a, b, c, d, and e. IL-1F7b is found to be the largest isoform. It has been reported that IL-37 is bound to the IL-18Ra and it

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

ANTI-INFLAMMATORY INTERLEUKINS AND THIER ROLES IN SEPSIS

was also observed in IL-37 transgenic mice (Pan and others 2001; Kumar and others 2002; Nold and others 2011). However, IL-37 appears not to exert its biological effect as a classical receptor antagonist for IL-18. It has been reported that high concentrations of IL-37 do not inhibit the expression of IL-18-induced IFN-g, while IL-18-induced IFNg in the presence of low concentrations of the IL-18-binding protein can be modestly reduced by recombinant IL-37 (Bufler and others 2002). Like IL-1 and IL-18, IL-37 is produced as a precursor that must be cleaved by caspase-1 to be activated. Being activated by caspase-1, IL-37 translocates to the nucleus and serves as a transcriptional regulator and, in turn, enhances the production of LPS-stimulated proinflammatory cytokines (Sharma and others 2008). Other studies also pointed out that the mechanism of IL-37 as an anti-inflammatory cytokine appeared to require SMAD3, which was a downstream kinase of transforming growth factor-b (Grimsby and others 2004; Nold and others 2010). In the form of mRNA and protein, IL-37 has been found in multiple organs, tissues and cell types, including peripheral blood mononuclear cells, monocytes, plasma cells, dendritic cells, epithelial cells, carcinoma cells, testis, thymus, and the uterus. IL-37 is the only IL-1 family member that lacks the homolog in the mouse. IL-37, as a potent anti-inflammatory cytokine, plays a significant role in restricting tissue injury during infections by downregulating the duration and intensity of immune as well as inflammatory responses in animal models (Tete and others 2012). A strain of transgenic mice has been generated to reveal the in vivo biological effect of IL-37, as a mouse homolog for human IL-37 has not been identified (Nold and others 2010). Both heterozygous and homozygous IL-37 transgenic mice breed normally and give rise to no obvious phenotype. IL-37 is not constitutively expressed in these mice even in mRNA levels. However, the expression of IL37 increases upon stimulation with IL-1b or LPS after 4– 24 h, and IL-37 precursor can be detected in peripheral blood cells separated from the transgenic mice. IL-37 transgenic mice were better protected against LPS challenge compared with similarly challenged wild-type mice. They displayed markedly less hypothermia, acidosis, hyperkalemia, hepatitis, and dehydration secondary to LPS challenge. In addition, circulating cytokines were significantly reduced as well as cytokines induced in whole blood cultures and in the lung and spleen cell homogenates. The anti-inflammatory activity of IL-37 was also manifested in a marked reduction in the expression of CD86 and MHC II on dendritic cells isolated from the spleen of IL-37 transgenic mice after LPS challenge (Nold and others 2010).

Conclusion A highly complex network of control elements modulates the human immune response, among which the antiinflammatory cytokines are of profound significance. Under physiological conditions, these cytokine inhibitors serve as immunomodulatory roles that attenuate the injury of sustained or excess inflammatory reactions. However, under pathological conditions, including severe sepsis and septic shock, these anti-inflammatory agents may either inhibit the proinflammatory responses that are insufficient or overcompensate, causing the host a state of immunoparalysis (Kasai and others 1997; Opal and DePalo 2000).

7

Although great efforts have been made worldwide in the past decades to elucidate the exact relationship between anti-inflammatory cytokines and septic response, it still seemed mysterious to us as illustrated by this review. The significances of most reviewed ILs in the pathological process of sepsis are not exactly clear both in septic models or human sepsis. Moreover, limited information is available to show marked diagnostic value for septic patients, such as IL-10 and IL-13. The findings of the growing number of new antiinflammatory cytokines and the new features of old cytokines will further elucidate the complicated and dynamic pathophysiological mechanisms underlying severe sepsis. Better understanding of the significance of these anti-inflammatory ILs might provide not only some validation on immune status of septic patients but also potential valuable diagnostic or therapeutic strategies.

Acknowledgments This work was supported, in part, by grants from the National Natural Science Foundation (Nos. 81130035, 81372054, 81272090, 81121004) and the National Basic Research Program of China (No. 2012CB518102).

Author Disclosure Statement The authors declare that they have no competing interests.

References Akira S, Takeda K. 2004. Toll-like receptor signalling. Nat Rev Immunol 4(7):499–511. Akira S, Uematsu S, Takeuchi O. 2006. Pathogen recognition and innate immunity. Cell 124(4):783–801. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. 2001. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 29(7):1303–1310. Appel SH, Wellhausen SR, Montgomery R, DeWeese RC, Polk HC Jr. 1989. Experimental and clinical significance of endotoxin-dependent HLA-DR expression on monocytes. J Surg Res 47(1):39–44. Atsumi T, Cho YR, Leng L, McDonald C, Yu T, Danton C, Hong EG, Mitchell RA, Metz C, Niwa H, Takeuchi J, Onodera S, Umino T, Yoshioka N, Koike T, Kim JK, Bucala R. 2007. The proinflammatory cytokine macrophage migration inhibitory factor regulates glucose metabolism during systemic inflammation. J Immunol 179(8):5399–5406. Banchereau J, Pascual V, O’Garra A. 2012. From IL-2 to IL-37: the expanding spectrum of anti-inflammatory cytokines. Nat Immunol 13(10):925–931. Baumhofer JM, Beinhauer BG, Wang JE, Brandmeier H, Geissler K, Losert U, Philip R, Aversa G, Rogy MA. 1998. Gene transfer with IL-4 and IL-13 improves survival in lethal endotoxemia in the mouse and ameliorates peritoneal macrophages immune competence. Eur J Immunol 28(2):610–615. Blanco-Quiro´s A, Casado-Flores J, Garrote Adrados JA, Moro MN, Anto´n JA, Sanz EA. 2005. Interleukin-13 is involved in the survival of children with sepsis. Acta Paediatr 94(12): 1828–1831. Bone RC. 1996. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med 24(7):1125–1128. Boomer JS, To K, Chang KC, Takasu O, Osborne DF, Walton AH, Bricker TL, Jarman SD 2nd, Kreisel D, Krupnick AS,

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

8

Srivastava A, Swanson PE, Green JM, Hotchkiss RS. 2011. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306(23):2594–2605. Brun-Buisson C. 2000. The epidemiology of the systemic inflammatory response. Intensive Care Med 26 Suppl 1:S64–S74. Bufler P, Azam T, Gamboni-Robertson F, Reznikov LL, Kumar S, Dinarello CA, Kim SH. 2002. A complex of the IL-1 homologue IL-1F7b and IL-18-binding protein reduces IL-18 activity. Proc Natl Acad Sci U S A 99(21):13723–13728. Burd PR, ThompsonWC, Max EE, Mills FC. 1995. Activated mast cells produce interleukin 13. J Exp Med 181(4):1373–1380. Cao YZ, Tu YY, Chen X, Wang BL, Zhong YX, Liu MH. 2012. Protective effect of Ulinastatin against murine models of sepsis: inhibition of TNF-a and IL-6 and augmentation of IL10 and IL-13. Exp Toxicol Pathol 64(6):543–547. Chalupka AN, Talmor D. 2012. The economics of sepsis. Crit Care Clin 28(1):57–76. Chaturvedi V, Collison LW, Guy CS, Workman CJ, Vignali DA. 2011. Cutting edge: human regulatory T cells require IL35 to mediate suppression and infectious tolerance. J Immunol 186(12):6661–6666. Cohen J. 2002. The immunopathogenesis of sepsis. Nature 420(6917):885–891. Cohen J, Opal S, Calandra T. 2012. Sepsis studies need new direction. Lancet Infect Dis 12(7):503–505. Collighan N, Giannoudis PV, Kourgeraki O, Perry SL, Guillou PJ, Bellamy MC. 2004. Interleukin 13 and inflammatory markers in human sepsis. Br J Surg 91(6):762–768. Collison LW, Chaturvedi V, Henderson AL, Giacomin PR, Guy C, Bankoti J, Finkelstein D, Forbes K, Workman CJ, Brown SA, Rehg JE, Jones ML, Ni HT, Artis D, Turk MJ, Vignali DA. 2010. IL-35-mediated induction of a potent regulatory T cell population. Nat Immunol 11(12):1093–1101. Collison LW, Delgoffe GM, Guy CS, Vignali KM, Chaturvedi V, Fairweather D, Satoskar AR, Garcia KC, Hunter CA, Drake CG, Murray PJ, Vignali DA. 2012. The composition and signaling of the IL-35 receptor are unconventional. Nat Immunol 13(3):290–299. Collison LW, Pillai MR, Chaturvedi V, Vignali DA. 2009. Regulatory T cell suppression is potentiated by target T cells in a cell contact, IL-35- and IL-10-dependent manner. J Immunol 182(10):6121–6128. Collison LW, Workman CJ, Kuo TT, Boyd K, Wang Y, Vignali KM, Cross R, Sehy D, Blumberg RS, Vignali DA. 2007. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 450(7169):566–569. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE, Sprung CL, Douglas IS, Jaeschke R, Osborn TM, Nunnally ME, Townsend SR, Reinhart K, Kleinpell RM, Angus DC, Deutschman CS, Machado FR, Rubenfeld GD, Webb SA, Beale RJ, Vincent JL, Moreno R. 2013. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 41(2):580–637. DiPiro JT. 1997. Cytokine networks with infection: mycobacterial infections, leishmaniasis, human immunodeficiency virus infection, and sepsis. Pharmacotherapy 17(2):205–223. Emonts M, Sweep FC, Grebenchtchikov N, Geurts-Moespot A, Knaup M, Chanson AL, Erard V, Renner P, Hermans PW, Hazelzet JA, Calandra T. 2007. Association between high levels of blood macrophage migration inhibitory factor, inappropriate adrenal response, and early death in patients with severe sepsis. Clin Infect Dis 44(10):1321–1328. Fiorentino DF, Bond MW, Mosmann TR. 1989. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that

ZHAO ET AL.

inhibits cytokine production by Th1 clones. J Exp Med 170(6):2081–2095. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. 1991. IL-10 inhibits cytokine production by activated macrophages. J Immunol 147(11):3815–3822. Gogos CA, Drosou E, Bassaris HP, Skoutelis A. 2000. Proversus anti-inflammatory cytokine profile in patients with severe sepsis: a marker for prognosis and future therapeutic options. J Infect Dis 181(1):176–180. Grimsby S, Jaensson H, Dubrovska A, Lomnytska M, Hellman U, Souchelnytskyi S. 2004. Proteomics-based identification of proteins interacting with Smad3: SREBP-2 forms a complex with Smad3 and inhibits its transcriptional activity. FEBS Lett 577 (1–2):93–100. Gu W, Zeng L, Zhang LY, Jiang DP, Du DY, Hu P, Wang HY, Liu Q, Huang SN, Jiang JX. 2011. Association of interleukin 4-589T/C polymorphism with T(H)1 and T(H)2 bias and sepsis in Chinese major trauma patients. J Trauma 71(6):1583–1587. Hack CE, Aarden LA, Thijs LG. 1997. Role of cytokines in sepsis. Adv Immunol 66:101–195. Hancock A, Armstrong L, Gama R, Millar A. 1998. Production of interleukin 13 by alveolar macrophages from normal and fibrotic lung. Am J Respir Cell Mol Biol 18(1):60–65. Ho AS, Liu Y, Khan TA, Hsu DH, Bazan JF, Moore KW. 1993. A receptor for interleukin 10 is related to interferon receptors. Proc Natl Acad Sci U S A 90(23):11267–11271. Hook KM, Abrams CS. 2012. The loss of homeostasis in hemostasis: new approaches in treating and understanding acute disseminated intravascular coagulation in critically ill patients. Clin Transl Sci 5(1):85–92. Hoshino T, Winkler-Pickett RT, Mason AT, Ortaldo JR, Young HA. 1999. IL-13 production by NK cells: IL-13 producing NK and T cells are present in vivo in the absence of IFN-g. J Immunol 162(1):51–59. Howard M, Muchamuel T, Andrade S, Menon S. 1993. Interleukin 10 protects mice from lethal endotoxemia. J Exp Med 177(4):1205–1208. Huang CH, Loo EX, Kuo IC, Soh GH, Goh DL, Lee BW, Chua KY. 2011. Airway inflammation and IgE production induced by dust mite allergen-specific memory/effector Th2 cell line can be effectively attenuated by IL-35. J Immunol 187(1):462–471. Hultgren O, Kopf M, Tarkowski A. 1999. Outcome of Staphylococcus aureus-triggered sepsis and arthritis in IL-4deficient mice depends on the genetic background of the host. Eur J Immunol 29(8):2400–2405 Kasai T, Inada K, Takakuwa T, Yamada Y, Inoue Y, Shimamura T, Taniguchi S, Sato S, Wakabayashi G, Endo S. 1997. Anti-inflammatory cytokine levels in patients with septic shock. Res Commun Mol Pathol Pharmacol 98(1):34–42. Kellum JA, Decker JM. 1996. The immune system: relation to sepsis and multiple organ failure. AACN Clin Issues 7(3): 339–350. Kochetkova I, Golden S, Holderness K, Callis G, Pascual DW. 2010. IL-35 stimulation of CD39 + regulatory T cells confers protection against collagen II-induced arthritis via the production of IL-10. J Immunol 184(12):7144–7153. Kotenko SV, Krause CD, Izotova LS, Pollack BP, Wu W, Pestka S. 1997. Identification and functional characterization of a second chain of the interleukin-10 receptor complex. EMBO J 16(19):5894–5903. Kraich M, Klein M, Patin˜o E, Harrer H, Nickel J, Sebald W, Mueller TD. 2006. A modular interface of IL-4 allows for scalable affinity without affecting specificity for the IL-4 receptor. BMC Biol 4:13.

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

ANTI-INFLAMMATORY INTERLEUKINS AND THIER ROLES IN SEPSIS

Kumar S, Hanning CR, Brigham-Burke MR, Rieman DJ, Lehr R, Khandekar S, Kirkpatrick RB, Scott GF, Lee JC, Lynch FJ, Gao W, Gambotto A, Lotze MT. 2002. Interleukin-1F7B (IL-1H4/IL-1F7) is processed by caspase-1 and mature IL1F7B binds to the IL-18 receptor but does not induce IFNgamma production. Cytokine 18(2):61–71. Kunz S, Wolk K, Witte E, Witte K, Doecke WD, Volk HD, Sterry W, Asadullah K, Sabat R. 2006. Interleukin (IL)-19, IL-20 and IL-24 are produced by and act on keratinocytes and are distinct from classical ILs. Exp Dermatol 15(12):991– 1004. LaPorte SL, Juo ZS, Vaclavikova J, Colf LA, Qi X, Heller NM, Keegan AD, Garcia KC. 2008. Molecular and structural basis of cytokine receptor pleiotropy in the interleukin-4/13 system. Cell 132(2):259–272. Latifi SQ, O’Riordan MA, Levine AD. 2002. Interleukin-10 controls the onset of irreversible septic shock. Infect Immun 70(8):4441–4446. Levi M, van der Poll T. 2010. Inflammation and coagulation. Crit Care Med 38(2 Suppl):S26–S34. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, Cohen J, Opal SM, Vincent JL, Ramsay G. 2003. SCCM/ ESICM/ACCP/ATS/SIS. 2001 SCCM/ESICM/ACCP/ATS/ SIS International Sepsis Definitions Conference. Crit Care Med 31(4):1250–1256. Liu JQ, Liu Z, Zhang X, Shi Y, Talebian F, Carl JW Jr., Yu C, Shi FD, Whitacre CC, Trgovcich J, Bai XF. 2012. Increased Th17 and regulatory T cell responses in EBV-induced gene 3-deficient mice lead to marginally enhanced development of autoimmune encephalomyelitis. J Immunol 188(7):3099– 3106. Liu Y, Wei SH, Ho AS, de Waal Malefyt R, Moore KW. 1994. Expression cloning and characterization of a human IL-10 receptor. J Immunol 152(4):1821–1829. Luan YY, Yao YM, Sheng ZY. 2012. Update on the immunological pathway of negative regulation in acute insults and sepsis. J Interferon Cytokine Res 32(7):288–298. Mannon P, Reinisch W. 2012. Interleukin 13 and its role in gut defence and inflammation. Gut 61(12):1765–1773. Martin GS, Mannino DM, Eaton S, Moss M. 2003. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med 348(16):1546–1554. Matsukawa A, Hogaboam CM, Lukacs NW, Lincoln PM, Evanoff HL, Strieter RM, Kunkel SL. 2000. Expression and contribution of endogenous IL-13 in an experimental model of sepsis. J Immunol 164(5):2738–2744. McKenzie AN, Li X, Largaespada DA, Sato A, Kaneda A, Zurawski SM, Doyle EL, Milatovich A, Francke U, Copeland NG, et al. 1993. Structural comparison and chromosomal localization of the human and mouse IL-13 genes. J Immunol 150(12):5436–5444. Meisel C, Schefold JC, Pschowski R, Baumann T, Hetzger K, Gregor J, Weber-Carstens S, Hasper D, Keh D, Zuckermann H, Reinke P, Volk HD. 2009. Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: a double-blind, randomized, placebo-controlled multicenter trial. Am J Respir Crit Care Med 180(7):640–648. Minty A, Chalon P, Derocq JM, Dumont X, Guillemot JC, Kaghad M, Labit C, Leplatois P, Liauzun P, Miloux B, et al. 1993. Interleukin-13 is a new human lymphokine regulating inflammatory and immune responses. Nature 362(6417):248–250. Minty A, Ferrara P, Caput D. 1997. Interleukin-13 effects on activated monocytes lead to novel cytokine secretion profiles intermediate between those induced by interleukin-10 and by interferon-gamma. Eur Cytokine Netw 8(2):189–201.

9

Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A. 2001. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol 19:683–765. Muchamuel T, Menon S, Pisacane P, Howard MC, Cockayne DA. 1997. IL-13 protects mice from lipopolysaccharideinduced lethal endotoxemia: correlation with down-modulation of TNF-alpha, IFN-gamma, and IL-12 production. J Immunol 158(6):2898–2903. Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE. 1999. The IL-4 receptor: signaling mechanisms and biologic functions. Annu Rev Immunol 17:701–738. Niedbala W, Wei XQ, Cai B, Hueber AJ, Leung BP, McInnes IB, Liew FY. 2007. IL-35 is a novel cytokine with therapeutic effects against collagen-induced arthritis through the expansion of regulatory T cells and suppression of Th17 cells. Eur J Immunol 37(11):3021–3029. Nold M, Hauser IA, Ho¨fler S, Goede A, Eberhardt W, Ditting T, Geiger H, Pfeilschifter J, Mu¨hl H. 2003. IL-18BPa:Fc cooperates with immunosuppressive drugs in human whole blood. Biochem Pharmacol 66(3):505–510. Nold MF, Nold-Petry CA, Zepp JA, Lo C, Garlanda C, Mantovani A, Bufler P, Dinarello C. 2011. Interleukin 37 exerts its anti-inflammatory functions by associating with IL-18Ra and SIGIRR. Cytokine 56(1):12. Nold MF, Nold-Petry CA, Zepp JA, Palmer BE, Bufler P, Dinarello CA. 2010. IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol 11 (11):1014–1022. O’Garra A, Vieira PL, Vieira P, Goldfeld AE. 2004. IL-10producing and naturally occurring CD4 + Tregs: limiting collateral damage. J Clin Invest 114(10):1372–1378. Olson BM, Jankowska-Gan E, Becker JT, Vignali DA, Burlingham WJ, McNeel DG. 2012. Human prostate tumor antigen-specific CD8 + regulatory T cells are inhibited by CTLA-4 or IL-35 blockade. J Immunol 189(12):5590–5601. Olson BM, Sullivan JA, Burlingham WJ. 2013. Interleukin 35: a key mediator of suppression and the propagation of infectious tolerance. Front Immunol 4:315. O’Neill LA. 2011. A critical role for citrate metabolism in LPS signalling. Biochem J 438(3):e5–e6. Opal SM, DePalo VA. 2000. Anti-inflammatory cytokines. Chest 117(4):1162–1172. Pan G, Risser P, Mao W, Baldwin DT, Zhong AW, Filvaroff E, Yansura D, Lewis L, Eigenbrot C, Henzel WJ, Vandlen R. 2001. IL-1H, an interleukin 1-related protein that binds IL-18 receptor/IL-1Rrp. Cytokine 13(1):1–7. Pillai MR, Bix M. 2011. Evolution of IL-4 and pathogen antagonism. Growth Factors 29(4):153–160. Punnonen J, Aversa G, Cocks BG, McKenzie AN, Menon S, Zurawski G, et al. 1993. Interleukin 13 induces interleukin 4independent IgG4 and IgE synthesis and CD23 expression by human B cells. Proc Natl Acad Sci USA 90(8):3730–3734. Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, Peterson E, Tomlanovich M, Early Goal-Directed Therapy Collaborative Group. 2001. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med 345(19):1368–1377. Sabat R, Gru¨tz G, Warszawska K, Kirsch S, Witte E, Wolk K, Geginat J. 2010. Biology of interleukin-10. Cytokine Growth Factor Rev 21(5):331–344. Seder RA, Paul WE, Davis MM, Fazekas de St Groth B. 1992. The presence of interleukin 4 during in vitro priming determines the lymphokine-producing potential of CD4 + T cells from T cell receptor transgenic mice. J Exp Med 176(4):1091–1098. Sharma S, Kulk N, Nold MF, Gra¨f R, Kim SH, Reinhardt D, Dinarello CA, Bufler P. 2008. The IL-1 family member 7b

Journal of Interferon & Cytokine Research Downloaded from online.liebertpub.com by Uc Davis Libraries University of California Davis on 01/29/15. For personal use only.

10

translocates to the nucleus and down-regulates proinflammatory cytokines. J Immunol 180(8):5477–5482. Song GY, Chung CS, Chaudry IH, Ayala A. 1999. What is the role of interleukin 10 in polymicrobial sepsis: anti-inflammatory agent or immunosuppressant? Surgery 126(2):378–383. Song GY, Chung CS, Chaudry IH, Ayala A. 2000. IL-4-induced activation of the Stat6 pathway contributes to the suppression of cell-mediated immunity and death in sepsis. Surgery 128(2):133–138. Steinhauser ML, Hogaboam CM, Matsukawa A, Lukacs NW, Strieter RM, Kunkel SL. 2000. Chemokine C10 promotes disease resolution and survival in an experimental model of bacterial sepsis. Infect Immun 68(11):6108–6114. Surbatovic M, Filipovic N, Radakovic S, Stankovic N, Slavkovic Z. 2007. Immune cytokine response in combat casualties: blast or explosive trauma with or without secondary sepsis. Mil Med 172(2):190–195. Taniyama T, Takai S, Miyazaki E, Fukumura R, Sato J, Kobayashi Y, Hirakawa T, Moore KW, Yamada K. 1995. The human interleukin-10 receptor gene maps to chromosome 11q23.3. Hum Genet 95(1):99–101. Tete S, Tripodi D, Rosati M, Conti F, Maccauro G, Saggini A, Cianchetti E, Caraffa A, Antinolfi P, Toniato E, Castellani ML, Conti P, Theoharides TC. 2012. IL-37 (IL-1F7) the newest anti-inflammatory cytokine which suppresses immune responses and inflammation. Int J Immunopathol Pharmacol 25(1):31–38. Tirotta E, Duncker P, Oak J, Klaus S, Tsukamoto MR, Gov L, Lane TE. 2013. Epstein-Barr virus-induced gene 3 negatively regulates neuroinflammation and T cell activation following coronavirus-induced encephalomyelitis. J Neuroimmunol 254(1–2):110–116. van der Poll T, de Waal Malefyt R, Coyle SM, Lowry SF. 1997. Antiinflammatory cytokine responses during clinical sepsis and experimental endotoxemia: sequential measurements of plasma soluble interleukin (IL)-1 receptor type II, IL-10, and IL-13. J Infect Dis 175(1):118–122. van der Poll T, Marchant A, Buurman WA, Berman L, Keogh CV, Lazarus DD, Nguyen L, Goldman M, Moldawer LL, Lowry SF. 1995. Endogenous IL-10 protects mice from death during septic peritonitis. J Immunol 155(11):5397–5401. van der Poll T, Opal SM. 2008. Host-pathogen interactions in sepsis. Lancet Infect Dis 8(1):32–43. van der Poll T, van Deventer SJ. 1999. Cytokines and anticytokines in the pathogenesis of sepsis. Infect Dis Clin North Am 13(2):413–426. Vignali DA, Kuchroo VK. 2012. IL-12 family cytokines: immunological playmakers. Nat Immunol 13(8):722–728. Walley KR, Lukacs NW, Standiford TJ, Strieter RM, Kunkel SL. 1996. Balance of inflammatory cytokines related to se-

ZHAO ET AL.

verity and mortality of murine sepsis. Infect Immun 64(11): 4733–4738. Wenzel RP, Edmond MB. 2012. Septic shock: evaluating another failed treatment. N Engl J Med 366(22):2122–2124. Whitehead GS, Wilson RH, Nakano K, Burch LH, Nakano H, Cook DN. 2012. IL-35 production by inducible costimulator (ICOS)-positive regulatory T cells reverses established IL17-dependent allergic airways disease. J Allergy Clin Immunol 129(1):207–215.e1–e5. Wirtz S, Billmeier U, Mchedlidze T, Blumberg RS, Neurath MF. 2011. Interleukin-35 mediates mucosal immune responses that protect against T-cell-dependent colitis. Gastroenterology 141(5):1875–1886. Wolk K, Kunz S, Asadullah K, Sabat R. 2002. Cutting edge: immune cells as sources and targets of the IL-10 family members? J Immunol 168(11):5397–5402. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. 2004. IL-22 increases the innate immunity of tissues. Immunity 21(2):241–254. Wong M, Bongiorno PB, Rettori V, McCann SM, Licinio J. 1997. Interleukin (IL) 1b, IL-1 receptor antagonist, IL-10, and IL-13 gene expression in the central nervous system and anterior pituitary during systemic inflammation: pathophysiological implications. Proc Natl Acad Sci USA 94(1): 227–232. Wu HP, Wu CL, Chen CK, Chung K, Tseng JC, Liu YC, Chuang DY. 2008. The interleukin-4 expression in patients with severe sepsis. J Crit Care 23(4):519–524. Yoon SI, Logsdon NJ, Sheikh F, Donnelly RP, Walter MR. 2006. Conformational changes mediate interleukin-10 receptor 2 (IL-10R2) binding to IL-10 and assembly of the signaling complex. J Biol Chem 281(46):35088–35096. Zurawski G, de Vries JE. 1994. Interleukin 13, an interleukin 4like cytokine that acts on monocytes and B cells, but not on T cells. Immunol Today 15(1):19–26.

Address correspondence to: Dr. Yong-ming Yao Trauma Research Center First Hospital Affiliated to the Chinese PLA General Hospital Fucheng Road 51 Haidian District Beijing 100048 People’s Republic of China E-mail: [email protected] Received 21 June 2014/Accepted 22 September 2014

The growing spectrum of anti-inflammatory interleukins and their potential roles in the development of sepsis.

Sepsis, recognized as a deadly immunological disorder, is one of the major causes of death in intensive care units globally. Traditionally, sepsis was...
253KB Sizes 7 Downloads 8 Views