A critical role for interleukin-1β in the progression of autoimmune diseases

A critical role for interleukin-1β in the progression of autoimmune diseases

International Immunopharmacology 17 (2013) 658–669 Contents lists available at ScienceDirect International Immunopharmacology journal homepage: www...

878KB Sizes 0 Downloads 9 Views

International Immunopharmacology 17 (2013) 658–669

Contents lists available at ScienceDirect

International Immunopharmacology journal homepage: www.elsevier.com/locate/intimp

Review

A critical role for interleukin-1β in the progression of autoimmune diseases Ruijuan Zhao, Hongyan Zhou, Shao Bo Su ⁎ The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China

a r t i c l e

i n f o

Article history: Received 16 July 2013 Received in revised form 19 August 2013 Accepted 19 August 2013 Available online 5 September 2013 Keywords: Interleukin-1β Autoimmune diseases Biological therapy

a b s t r a c t Interleukin-1β (IL-1β) belongs to IL-1 family and is a potent pro-inflammatory cytokine. It is known to be also involved in a variety of cellular activities, including cell proliferation, differentiation and apoptosis. In addition to its pathophysiologic role in host protection, IL-1β promotes the progression of a number of autoimmune diseases. Most of such diseases can be controlled by anti-IL-1β treatment. This review discusses the contribution of IL-1β to the course of autoimmune diseases, such as rheumatic diseases, uveitis, autoimmune thyroid diseases (AITD), insulin-dependent diabetes mellitus (IDDM), autoimmune inner ear disease (AIED), multiple sclerosis (MS), myocarditis, hepatitis and kidney diseases. The critical involvement of IL-1β in the pathogenesis of autoimmune diseases provides targets for developing therapeutic treatment. © 2013 Elsevier B.V. All rights reserved.

Contents 1. 2. 3.

Introduction . . . . . . . . . . . . . . . . . . . Interleukin-1β . . . . . . . . . . . . . . . . . . Interleukin-1β and autoimmune diseases . . . . . . 3.1. Autoimmune rheumatic diseases (AIRD) . . . 3.1.1. Rheumatoid arthritis (RA) . . . . . 3.1.2. Systemic lupus erythematosus (SLE) 3.1.3. Primary Sjögren's syndrome (pSS) . 3.2. Autoimmune uveitis . . . . . . . . . . . . 3.3. Autoimmune thyroid diseases (AITD) . . . . 3.4. Insulin dependent diabetes mellitus (IDDM) . 3.5. Autoimmune inner ear disease (AIED) . . . . 3.6. Multiple sclerosis (MS) . . . . . . . . . . . 3.7. Myocarditis . . . . . . . . . . . . . . . . 3.8. Autoimmune hepatitis (AIH) . . . . . . . . 3.9. Kidney disease . . . . . . . . . . . . . . . 4. Conclusions and perspectives . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

1. Introduction Interleukin-1 (IL-1) is a family of cytokines that play an important role in initiating the cascade of immunoinflammatory responses by ⁎ Corresponding author at: The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 S Xianlie Road, Guangzhou 510060, China. Tel.: +86 20 87330402; fax: +86 20 87330403. E-mail address: [email protected] (S.B. Su). 1567-5769/$ – see front matter © 2013 Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.intimp.2013.08.012

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . .

658 659 660 660 660 662 662 662 663 663 663 664 664 664 664 665 666 666

linking the innate and acquired immune systems [1]. IL-1 was discovered in the 1970s and was first described as lymphocyte-activating factor (LAF) stimulating both central and peripheral T lymphocytes [2,3] or leukocytic pyrogen (LP) mediating fever [4]. In human, initially, IL-1 gene family was thought to be composed of IL-1α, IL-1β and IL-1Ra which are located within 450 kb on chromosome 2q14–q21 [5]. Now it is widely believed that the members of IL-1 family consist of IL-1 agonist forms (IL-1α and IL-1β), the natural IL-1 receptor antagonist (IL-1Ra), IL-18, IL-33, IL-36, IL-36Ra, IL-37 and IL-38 [6–11] (Table 1).

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

Both IL-1α and IL-1β are synthesized as large precursor proteins and, whereas IL-1β is secreted, IL-1α remains in the cytoplasm or is membrane-associated and released into circulation primarily during severe diseases [12]. IL-1β precursor, the 31 kDa pro-IL-1β, is induced by activation of NF-κB pathway in response to the stimuli of pathogen associated molecular patterns (PAMPs) and localizes in the cytosol in its biologically inactive form. It is active 17.5 kDa IL-1β until cleaved by caspase-1 which is released from inflammasome in response to further stimuli of PAMPs [12–15] (Fig. 1). In resting cells, caspase-1 is in pro-caspase-1 form and binds to inflammasome that prevents its activation. Inflammasome was originally understood as a platform for the activation of caspase-1 and cleavage of the pro-forms of IL-1β and IL-18 into active cytokines [15,16]. It is now believed that inflammasome controls innate and adaptive immune responses [17,18]. It was reported that caspase-11 had also been linked to inflammasome signaling and was essential for the maturation of IL-1β by processing of pro-caspase-1 into caspase-1 [16]. IL-1β releases via a non-conventional secretion mechanism without a signal sequence [15]. In contrast to IL-1β, pro-IL-1α is biologically active and is cleaved by calpain to generate the mature protein. The amino-acid homology between human IL-1α and IL-1β is 22% and that of IL-1α to IL-1Ra is 18%; the homology of IL-1β to IL-1Ra is 26%. Hence, IL-1β is more closely related to IL-1Ra than to IL-1α. The striking difference between IL-1β and IL-1α is that both pro- and mature IL-1α can bind to their receptors and induce cellular responses, whereas pro-IL-1β is biologically inactive and requires ICE to generate the active protein. IL-1α is in acidic form while IL-1β is a neutral molecule. Furthermore, both forms of IL-1α remain intracellular and are normally found in the circulation or in inflammatory fluids unless released by a dying cell, whereas active IL-1β is secreted [19]. Because of these characteristics, more studies focus on IL-1β. In recent years, the critical role of the IL-1 family as regulators of inflammation and immunity has become apparent. IL-1 maintains a central function against infection by pathogenic organisms including bacteria [20], fungi [21] and viruses [22]. The synthesis, processing and secretion of IL-1, particularly IL-1β, are tightly regulated [22]. IL-1 affects nearly every cell either alone or in a synergistic fashion with other mediators [23,24]. IL-1β is primarily a proinflammatory cytokine to stimulate the expression of genes associated with inflammation and autoimmune diseases [25–27]. Two single nucleotide polymorphisms (SNPs) in the IL-1β gene, one at position −511 (rs16944) in the promoter region and the other in the fifth exon +3954 (rs1143634), are associated with various diseases [28]. By binding to specific highaffinity cell surface receptors, IL-1β shows pleiotropic effects that include costimulation of T lymphocytes, B cell proliferation, growth of fibroblasts, induction of adhesion molecules, stimulation of the

659

production of other cytokines and inflammatory mediators in an autocrine and a paracrine way, as well as growth-inhibitory and cytocidal effect on several cell lines [29,30]. The immune system protects the host against diseases and infection. But the immune system also attacks healthy cells by mistake to cause autoimmune diseases. Autoimmune diseases affect many parts of the body. Disappointingly, the etiology of the majority of autoimmune diseases is unknown. Despite the uncertainty of the precise causes in inflammatory and autoimmune diseases, serum levels of IL-1β are elevated which correlate with disease development and severity. Clinically, the role of IL-1β has been validated by treatment of numerous diseases with anti-IL-1β treatment. Since many IL-1β-induced inflammatory diseases have been summarized in excellent reviews by other investigators, this article will focus on the current knowledge about the role of IL-1β in the pathogenic process of autoimmune diseases. 2. Interleukin-1β IL-1β exerts a range of inflammatory and immunomodulatory activities [31]. IL-1β is produced primarily by monocytes and macrophages [32] and also by T cells, NK cells, endothelial cells, fibroblasts, astrocytes, microglial cells, adrenal cortical cells [25,33] and pancreas β-cells [34]. It is a pleiotropic cytokine involved in host responses to microbial invasion, inflammation, immune regulation, metabolic reactions, hematopoietic processes and tumor progression [35]. IL-1β affects important cellular functions, such as reducing DNA content, decreasing protein synthesis and intracellular energy production, inducing β-cell apoptosis and necrosis [36]. IL-1β expression and function are tightly regulated by a complex system of IL-1 family members and their receptors. IL-1β binds to the IL-1 receptor type I (IL-1RI) expressed on all nucleated cells, which triggers the recruitment of the IL-1RAcP to form the active signaling complex. IL-1β also binds to a second receptor, IL-1 receptor type II (IL-1RII), which competes with IL-1RI for IL-1β and for the IL-1RI co-receptor, IL-1RAcP. Additionally, IL-1RII exists in both membrane bound and soluble forms (sIL-1RII) that have biological properties similar to a decoy receptor and a binding protein. The natural inhibitor IL-1Ra binds to both IL-1RI and IL-1RII, but does not allow the recruitment of IL-1RAcP [37]. IL-1β can elicit responses of the cells with a low receptor number because it activates a complex cascade resulting in signal amplification [38]. The signal transduction initiates via the 213-amino acid cytoplasmic domain of IL-1RI. The juxtaposition of the intracellular IL-1 receptor domains of IL-1RI and IL-1RAcP after IL-1β binding is necessary and sufficient for triggering intracellular signaling, which results in activation

Table 1 IL-1 family [7–11,23,37,180–187]. Chromosomal location

Expression cells

Target cells

Signaling

Regulation

IL-1α IL-1β IL-1Ra IL-18 IL-33

2q14 2q14 2q14.2 2q23.1 9p24.1

IL-1Ra, IL-1RI, IL-4, IL-10 IL-1Ra, IL-4, IL-10, DUSP1, IFN-I, 1,25(OH)2D3, matrilin-3 IL-18BP, IL-37, NF- B, CRH IFN-γ, TNF-α, IL-1β

2q12-q14.1 2q14 2q12-q21 2q14 2q12-q14.1

Mo and many cells Mo and many cells Many cells T, NK, basophils, MC, spleen cells Th2, MC, natural helper cells, basophils, eosinophils T cells, KCs, BMDCs

MAPKs, NF-κB MAPKs, NF-κB, ROCKs, AKT N/A NF-κB NF-κB

IL-36α IL-36β IL-36γ IL-36Ra IL-37

Mo, MØ, DCs and many cells Mo, MØ, DCs and many cells Mo, Neu, KCs, splenocytes MØ and DCs Epithelial and endothelial cells, fibroblasts KCs

MAPKs, NF-κB

TNF-α, Th17 cytokines

Mo, B cells, DCs, KCs MØ, Mo, epithelial cells, PBMCs, DCs

BMDCs, DCs, TH17 cells Mo and epithelial cells

N/A MAPKs, FAK, Pyk2, paxillin,

IL-38

2q13

Skin cells, B cells

T-cells, DCs

Unknown

IL-1α, IL-17A, TNF-α TGF-β1, IL-18, IFN-γ, IL-1β, TNF, CpG, IL-4, GM-CSF Unknown

Mo: monocyte; MØ: macrophage; Neu: neutrophils; DCs: dendritic cells; MC: mast cells; SMCs: smooth muscle cells; KCs: keratinocytes; PBMCs: peripheral blood mononuclear cells; BMDCs: bone marrow-derived dendritic cells; MAPK: mitogen-activated protein kinase; NF-κB: nuclear factor-κ-gene binding; FAK: focal adhesion kinase; Pyk2: proline-rich tyrosine kinase 2; C/EBP: CCAAT/enhancer-binding protein; ROCKs: Rho/Rho-associated coiled-coil forming kinases; DEC1: differentiated embryo-chondrocyte expressed gene 1; N/A: no application; GM-CSF: granulocyte-macrophage colony-stimulating factor; BMDCs: bone marrow-derived cells.

660

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

IL-1

Stimuli 2 Stimuli 1

Extracellular

NLR / PYHIN

P I B

ASC

pro-caspase-1

NF- B Inflammasome ProIL-1 Nucleus Cytoplasm NF- B

Nuclear TranscriptIon

Fig. 1. Induction and maturation of IL-1β. IL-1β precursor is induced by activation of NF-κB pathway in response to the stimuli of PAMPs and localizes in the cytosol in its biologically inactive form. It is active 17.5 kDa IL-1β until cleaved by caspase-1 which is released from inflammasome in response to further stimuli of PAMPs.

of transcription factors (Fig. 2). The mitogen-activated protein kinase (MAPK) signal transduction pathways are activated in response to IL-1β, which are ubiquitous and regulate diverse cellular programs by relaying extracellular signals to intracellular responders. Three groups of MAPKs are activated by IL-1β: the p42 and p44 MAP kinases (also termed extracellular signal regulated kinase, ERK1, and ERK2), protein 38 MAP kinase (p38 MAPK) and the stress-activated protein kinase (SAPK, also known as the c-Jun NH2-terminal kinase, JNK) [38,39]. p38 MAPK and JNK pathways promote the stabilization of induced mRNA and regulate nuclear transcription [38,40]. MAPKs are activated by combined tyrosine/threonine phosphorylation catalyzed by MAPKK kinases (MAPKKKs) and MAPK kinases (MAPKKs), forming an evolutionarily conserved signal transduction system in Eukaryotes [41]. IL1β/IL-1RI/IL-1RAcP complex recruits intracellular adaptor proteins and kinases, including MyD88 (myeloid differentiation factor 88) and IRAKs (IL-1R associated kinases). IRAK4 phosphorylates and activates IRAK1, which further recruits TRAF6 (tumor necrosis factor receptorassociated factor 6). IRAK1 and TRAF6 complex released from the receptor complex causes TRAF6 autoubiquitination. TRAF6 is polyubiquitinated by IRAK2 as well. Ubiquitinated TRAF6 subsequently leads to the recruitment of TGF-β-activated kinase 1 (TAK1)–TAK1-binding protein 1 (TAB1)– TAB2 complex, resulting in the activation of TAK1 (a MAPKKK) then initiating MAPK cascades [38,39,42,43]. TRAF6 also catalyzes the synthesis of unanchored Lys 63 polyubiquitin chains, which bind to TAB2, leading to the activation of TAK1 directly [44]. The activated MAPKs regulate several transcription factors such as NF-κB, AP-1, and CCAAT/enhancer-binding protein (C/EBP) [45,46]. Activated TAK1 also activates transcription factors NF-κB directly. In resting cells, NF-κB combines with the regulatory protein inhibitors of κB (IκB) in the cytoplasm in an inactive form. Phosphorylation of IκB, an important step for NF-κB activation, is mediated by IκB kinase (IKK) [47,48]. Activated TAK1 activates downstream kinases IKK-α and IKK-β, which phosphorylate IκB proteins, thereby freeing NFκB which then translocates into the nucleus and binds to the target promoter to increase transcription [38,39,49]. Another kinase TNF/ IL-1-induced protein kinase (TIP/β casein kinase) also associates with

IL-1β activation cascade [50]. In addition to these pathways, IL-1β also activates the Rho/Rho-associated coiled-coil forming kinase (ROCK) pathway associated with the remodeling of actin filaments [43]. The complex cascade of IL-1β signaling contributes to the development and progression of various diseases. 3. Interleukin-1β and autoimmune diseases The IL-1 receptor-bearing cells are extremely sensitive to IL-1 and the overproduction of IL-1 often leads to serious disorders [6]. Absence of IL-1Ra expression during an immune response, or other molecules that oppose the IL-1β inflammatory cascade, promotes the development of autoimmune diseases [51]. The widespread distribution of IL-1β and imbalanced IL-1β and IL-1Ra ratio in serum or tissue have been implicated not only in altered susceptibility to infections but also in other conditions such as autoimmune diseases, as exemplified in the following sections. 3.1. Autoimmune rheumatic diseases (AIRD) Autoimmune rheumatic diseases (AIRD) are a category of refractory diseases based on their ambiguous pathogenesis. Rheumatoid factor, antinuclear antibodies and anti-neutrophil cytoplasmic antibodies are key parameters in the diagnosis of AIRD. Among AIRD, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and primary Sjögren's syndrome (pSS) are the most common diseases and have received particular attention by physician and basic researchers [31,52]. 3.1.1. Rheumatoid arthritis (RA) Rheumatoid arthritis (RA) is an autoimmune systemic disease characterized by persistent inflammation of the diartrodial joints with synovial hyperplasia and progressive joint destruction [31]. The irreversible destruction of the cartilage, tendon, and bone that comprise synovial joints is the hallmark of RA [53].

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

IL-1Ra IL-1RI

IL-1 β

IL-1 β

IL-1RI

IL-1RII

AcP

Extracellular matrix

661

No signal

MyD88 No signal IRAK4 IRAK1

IRAK1

P

TRAF6

TRAF6

TAB1 IκB α

TAB2

Ub Ub

P

NF- κB

IκB α

P IKK- α

NF- κB

IKK- β

TAK1

TAK1

MEK1/2

MKK3/6

MKK4/7

ERK1/2

p38 MAPK

SAPK/ JNK1,2,3

Cytoplasm

Nucleus Nuclear TranscriptIon

mRNA Stabilization

Fig. 2. IL-1β signaling pathway. Matured IL-1β/IL-1RI/IL-1RAcP complex recruits MyD88 and IRAKs. IRAK4 phosphorylates and activates IRAK1, which further recruits TRAF6. IRAK1 and TRAF6 complex released from the receptor complex causes TRAF6 autoubiquitination. Ubiquitinated TRAF6 subsequently leads to recruitment of the TAB1–TAB2 complex, which results in the activation of TAK1 to initiate the cascades of ERK1/2, p38 MAPK and JNK which regulate nuclear transcription. p38 MAPK and JNK enhance the stabilization of mRNA as well. Activated TAK1 also stimulated downstream kinases IKK-α and IKK-β, which phosphorylate IκB, thereby freeing NF-κB which then translocates into the nucleus and binds to the target promoter to increase transcription.

In the pathogenesis of RA, cytokines contribute to the activation of fibroblast-like synoviocytes (FLS) that leads to erosion of cartilage and bone [54]. It is believed that TNF-α, IL-1β and IL-6 are primary cytokines involved in the progression of chronic joint inflammation and the concomitant erosive changes in cartilage and bone [55–57]. As the cytokine derived from synovial membrane, IL-1β is pivotal in initiating cartilage erosion [54,58,59]. Serum and synovial concentrations of IL-1β are higher in RA patients with active disease than in those in remission [31,60]. Furthermore, cartilage from arthritis patients contains upregulated IL-1β mRNA [59]. Numerous observations have shown that tissue destruction and the resulting disability of RA patients are partly the result of extracellular matrix degradation by proteolytic enzymes including matrix metalloproteinases (MMPs) and the release of the mineral phase (Ca2 + release) by prostaglandin E2 (PGE2) [19]. IL-1β triggers the expression of MMP genes such as collagenases and elastase, stimulates the secretion of PGE2 and several proinflammatory mediators involved in joint destruction, and induces the expression of adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-l) by synovial cells [31,61]. MMPs, induced

by IL-1β, degrade all components of the extracellular matrix. The collagenases, MMP-1 and MMP-13, have predominant roles in RA because they are rate limiting in the process of collagen degradation [19]. IL-1β induction of MMP-1 requires pathways different from those required by induction of MMP-13. MMP-1 induction depends on p38 and MEK, while p38, JNK, and NF-κB are required for induction of MMP-13 by IL-1β [62]. IL-1β potently induces MMP-13 by activation of p38 MAPK pathway, transcription factor AP-1 and the tissuespecific Runx-2 [63]. IL-1β-activated AP-1 binds to an evolutionarily conserved DNA sequence ∼ 20 kb upstream relative to the MMP-13 transcription start site (TSS) [64]. Understanding of RA pathophysiological mechanisms has led to the development of a new therapy designed against more precise targets involved in disease course. The side effects and/or poor efficiency of traditional pharmacological therapies (non-steroidal antiinflammatory drugs, NSAIDS and disease-modifying anti-rheumatic drugs, DMARDS) for RA prompted the development of anti-cytokine strategies. Targeting pathways triggered by IL-1β may provide a promising therapeutic opportunity for RA. IL-1Ra has been proven effective in the treatment of

662

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

RA patients [31,65–67]. Anakinra, a recombinant, nonglycosylated human IL-1Ra, is a relatively safe and modestly efficacious therapeutic agent for RA. Anakinra shows down the progression of radiographic joint damage [68]. Anakinra is originally developed for the treatment of moderate–severe RA that has been unresponsive to initial DMARD therapy [69]. It is recommended for the treatment of active RA after an adequate trial of another conventional DMARD, alone or with methotrexate (MTX) [70–72]. Biological therapy with Anakinra in RA has been approved by the US Food and Drug Administration (FDA) in clinical phase IV trial. However, 30% RA patients showed no response to IL-1Ra treatment and the responses were highly variable from one patient to another [55]. Targeting IL-1β with specific antibodies has also shown benefit for the treatment of RA. Gevokizumab (XOMA 052, a recombinant humanized anti-IL-1β antibody) was effective in preventing and treating joint inflammation, at lower and less frequent doses than Anakinra. It was hypothesized that XOMA 052 may have increased efficacy for RA treatment due to its higher affinity for IL-1β and longer halflife in vivo [73]. 3.1.2. Systemic lupus erythematosus (SLE) SLE is a systemic autoimmune disease characterized by increased production of autoantibodies against a relatively limited range of nuclear antigens. The dysregulated immune system produces autoantibodies resulting in the formation of immune complexes that deposit in tissues, thereby attacking the skin, joints, kidneys, heart, and brain [74]. Cytokines are essential for the control of the specific immune responses that characterize SLE [75]. IL-1 is crucial for the synthesis of IgG autoantibodies in patients with SLE [76]. High levels of IL-1 in the course of SLE have been documented [75,77,78]. Although the role of IL-1β in SLE has not been fully defined, there is evidence that IL-1β secretion by PBMC is higher in patients with SLE [79]. Enhanced steady state levels of mRNA for TNF-α and IL-1β, but not IL-1α, have been detected in the renal cortices of animals with lupus nephritis. It was hypothesized that TNF and IL-1, especially IL-1β, contribute to nephritis in murine models of lupus [80]. The importance of secretable IL-1β, rather than cell-associated IL-1α, in the immunostimulatory and inflammatory responses that mediate the pathogenesis of experimental SLE has been suggested [81]. IL-1 receptor-associated kinase 1 (IRAK1) is part of the IL-1/Toll receptor and NF-κB signaling pathway and has been associated with the onset of SLE [82]. In a cross-sectional study, increased serum concentration of IL-1Ra is a pathophysiologic feature of active SLE [83]. Lower levels of IL-1Ra have also been detected in lupus nephritis and coincide with kidney involvement in SLE [31,84]. Thus, high concentrations of IL-1Ra were restricted to patients with extra-renal disease and serum levels in patients with kidney involvement were low or only moderately increased [84]. These data add further evidence for the IL-1β cascade involvement in the pathogenesis of SLE. It has been reported that IL-1β polymorphism influences the susceptibility to acquire SLE and IL-1β +3953 allele was protective from SLE [31]. Mice deficient in IL-1β produce lower levels of antidouble-stranded DNA (anti-dsDNA) antibodies, characteristic of SLE and central to the diagnosis of the disease. Overall, disease severity was milder in mice deficient in IL-1β. Thus, IL-1β is an important factor in the induction and pathogenesis of experimental SLE [81]. Anakinra might be an interesting therapeutic alternative for individual patients with SLE not responding to conventional treatment [85]. Anakinra has been shown to be safe and effective in improving arthritis in a trial of four SLE patients. Therefore, targeting the IL-1β pathway provides a therapeutic opportunity for SLE. 3.1.3. Primary Sjögren's syndrome (pSS) Sjögren's syndrome may exist as a primary disorder (primary Sjögren's syndrome) or be associated with other autoimmune diseases (secondary Sjögren's syndrome) such as RA, SLE, or systemic sclerosis [86,87]. pSS is characterized by chronic lymphocytic or mononuclear cell infiltration near epithelial cells of exocrine glands and the presence

of anti-Ro/SSA and anti-La/SSB antibodies associated with glandular dysfunction [88,89]. In the disease process, lacrimal and salivary glands are affected and keratoconjunctivitis sicca and xerostomia appear. Although the pathophysiology of pSS is not yet fully understood, there is evidence that IL-1β, TNF-α and IL-6 are important mediators of the autoimmune responses and may cause tissue destruction in salivary and lachrymal glands [90,91]. High levels of IL-1 β in parotid acinar and lacrimal gland cells in mouse model or patients with Sjögren's syndrome have been detected [92,93]. IL-1β is also secreted by peripheral blood mononuclear cells (PBMC) from patients with pSS [88,94]. The abnormal local and systemic production of IL-1β suggests a pathogenic role of IL-1β in pSS. Exogenous addition of IL-1β inhibits neurotransmitter release and lacrimal gland protein secretion [86]. Evidence also suggests that IL-1β may be involved in the destruction of salivary and lacrimal glands in SS [95]. IL-1β may have a proteolytic activity for degradation of the basement membrane and leads to the disruption of acinar or ductal structure in salivary glands [90]. It has been shown that IL-1β inhibits neurotransmitter release in the lacrimal gland resulting in impaired protein secretion and dry eye [96]. The iNOS is known to be induced in the presence of pro-inflammatory cytokines in several secretory epithelial cell types. Lacrimal gland acinar cells are able to produce iNOS and NO in response to IL-1β. This pathway of iNOS induction and overproduction of NO may be a factor in lacrimal gland cell death, a significant pathophysiological pathway of dry eye syndrome [92]. In addition, IL-1β-induced MMP-9 expression and corneal cell migration were mediated through the p42/p44 MAPK- and JNK1/2-dependent AP-1 and NF-kB pathways resulting in dry eye. IL1β causes marked p42/p44 MAPK translocation from the cytosol into the nucleus in corneal cells with yet an unknown mechanism [39]. In addition, IL-1β gene polymorphisms may affect the susceptibility to pSS [95]. The efficacy of an IL-1Ra, Anakinra, is currently studied in a short-term placebo-controlled trial in pSS and may be a potential treatment of pSS [97]. 3.2. Autoimmune uveitis Uveitis is an umbrella term for intraocular inflammation of a variety of etiology. There are numerous causes for uveitis including infection, systemic autoimmune disorders and mechanical injury. The classic autoimmune uveitis includes Behçet's disease (BD), sarcoidosis and Vogt–Koyanagi–Harada (VKH) disease. The initiating factors for uveitis are not well understood. Experimental autoimmune uveitis (EAU) serves as an animal model for the study of human chronic uveitis induced in Lewis rats or mice by immunization with S-antigen or interphotoreceptor retinoid-binding protein (IRBP). Endotoxininduced uveitis (EIU) is an animal model for acute ocular inflammation induced by injection of LPS. Recent studies in mice and with human cells showed that IL-1β was essential for the development of Th17 cells [98], which are increasingly recognized as the key effector cells responsible for autoimmune uveitis [99,100]. IL-1 gene cluster polymorphisms, including IL-1A and IL-1B genes, are associated with an increased risk for Behçet's disease [101]. In addition, IL-1β participates in the pathogenesis of autoimmune uveitis [102,103], and it is effective to use IL-1Ra or anti-IL-1β antibody for treatment [104–106]. NF-κB, activated by IL-1β, plays an important role in the development of EAU and EIU [107]. It has been reported that NF-κB inhibitor, STA-5326, is a promising therapeutic modality for refractory uveitis in human [107]. Kitamei et al. showed several efficacious NF-κB inhibitors on EAU [108]. Acute uveitis was ameliorated by inhibition of IκB kinase β (IKKβ) in rats, particularly when the disease was induced by IL-1β and TNF-α [109]. IL-1β expression was promoted by NF-κB and IL-1β in turn activates IKK. This positive regulatory loop amplifies local inflammatory responses [108]. Ugurlu et al. described a patient with juvenile Behçet's syndrome whose diseased eye was refractory to conventional immunosuppressive agent and corticosteroids. But the patient was treated

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

successfully with a single dose of Canakinumab, a human antiinterleukin-1β antibody [110,111]. Interestingly, the patient did not respond to Anakinra, IL-1Ra that blocks both IL-1α and IL-1β. This may imply a predominance function for IL-1β on uveitis. 3.3. Autoimmune thyroid diseases (AITD) Thyroid auto-immunity is clinically divided into two major subtypes, with over functioning (hyperthyroidism: Graves' disease (GD)) or under functioning (hypothyroidism: Hashimoto thyroiditis (HT)) of the gland. During the development of autoimmune thyroid diseases (AITD), environmental factors, immune system and target tissue are simultaneously involved [112]. Proinflammatory cytokines such as IL-1β and TNF-α are known to affect thyroid function by stimulating IL-6 secretion and modifying epithelium integrity and altering junction proteins such as ZO-1, Claudin and JAM-A. The expression, localization and organization of junction proteins that ensure the epithelial barrier function in the thyroid tissue of patients with AITD (HT and GD) are altered. Therefore, IL-1β modifies thyroid epithelial tightness of human thyrocytes through altering the expression and localization of junction proteins [113]. Paolieri et al. [114] demonstrated that IL-1β was the only cytokine able to induce both Fas expression on Fas-negative thyroid follicular cells (TFC) and B7.1. Fas/Apo1/CD95, an important participant in apoptosis, and B7.1, a member of a family of “co-stimulatory” molecules crucial for efficient antigen presentation, are two additive surface functional molecules of TFC from HT patients. IL-1β induces the tissue damage via suicide Fas/FasL-mediated TFC interaction and the perpetuation of autoimmune reaction via co-stimulatory molecules. Therefore, IL-1β plays a crucial role in the pathogenesis of thyroid autoimmunity. The thyroid dysfunction in GD can be treated effectively. However no reliable, specific, and effective medical therapies have yet been developed for thyroid-associated ophthalmopathy (TAO) [115]. TAO, an autoimmune component of Graves' disease, encompasses specifically to the orbital and periorbital manifestations [115]. It is the most common orbital disease in adults also called Graves' ophthalmopathy (GO) or thyroid eye disease [116]. TAO is associated with profound connective tissue remodeling and fibrosis that appear to involve selective activation of orbital fibroblasts with aberrant accumulation of extracellular matrix molecules glycosaminoglycans (GAGs) in retro-ocular tissue [117]. GAGs then in turn are responsible for the development of clinical signs and symptoms. Oxygen free radicals which are induced by IL-1β are involved in GAG accumulation [118]. Recently, Han et al. demonstrated that orbital fibroblasts treated with IL-1β expressed high levels of tissue inhibitor of metalloproteinase-1 (TIMP-1). The effect of IL-1β was at the pretranslational level and involves elevating the TIMP-1 gene promoter. TIMP-1 is an important modulator of matrix metalloproteinase (MMP) activity. Thus, the important balance between MMPs and TIMPs in human orbit may become disrupted in TAO and that results in fibrosis. IL-1β activates ERK1/2 pathway in fibroblasts and interrupting the pathway with PD98059, a chemical inhibitor of MEK, or by transfecting cells with a dominant negative ERK 1 plasmid results in the attenuation of TIMP-1 induction [117]. Thus, anti-IL-1β treatment may represent an important therapy for modifying the proteolytic environment and restore natural tissue remodeling in TAO. In addition, IL-1β decreases tear production by lacrimal gland [96]. IL-1β, IL-6, and IL-8 concentrations in tears were significantly higher in active TAO than in inactive TAO patients and controls. There was a positive correlation between tear IL-1β, IL-6 and IL-8 levels and clinical activity score (CAS) in TAO patients [119]. 3.4. Insulin dependent diabetes mellitus (IDDM) Insulin-dependent diabetes mellitus (IDDM) is an autoimmune disease characterized by lymphocytic infiltration of the pancreatic islets (insulitis) and the presence of islet cell autoantibodies [120].

663

Proinflammatory cytokines released as part of the insulitis process have been implicated as effector molecules that participate in both islet inflammation and beta-cell lesion during the development of IDDM [121]. A possible diabetogenic effect of cytokines may be mediated by their induction of abnormal expression of islet cell autoantigens. In all stages of IDDM higher levels of IL-1β and TNF-α were detected in patient serum [122]. IL-1β, together with IFN-γ and TNF-α, mediates pancreatic β-cell destruction and death [36,123–126]. It is demonstrated that IL-1β induces β-cell damage by a NO-mediated mechanism [127]. Pancreatic β-cells are more sensitive to IL-1β than to other NO donors [36]. The synthesis of both isoforms of islet enzyme glutamate decarboxylase (GAD), GAD65 and GAD67, is decreased in the presence of IL-1β. GAD is a major target of β-cell autoimmunity in IDDM and IL-1β is the primary cytokine affecting GAD-65 expression [128,129]. In addition, activation of the transcription factor NF-κB is necessary for cytokine-induced pancreatic β-cell death. IL-1β induces NF-κB activation and has a more pronounced pro-apoptotic effect than TNF-α [130–132]. Wilson et al. reported that in the administration of a high dose IL-1β accelerated the onset of IDDM compared to saline-injected controls, while in a low dose IL-1β significantly reduced the frequency of IDDM compared to placebo and high dose IL-1β treatment groups. The results demonstrate that exogenous administration of IL-1β affects rat idiopathic autoimmune diabetes in a dose-dependent manner [133]. Cailleau et al. showed that treatment with neutralizing antibodies for IL1β prevented cyclophosphamide-induced diabetes in nonobese diabetic mice [134]. Whether control of inflammatory pathways mediated by IL-1β using the recombinant IL-1Ra Anakinra will yield measurable decreases in the expression of genes that are otherwise overexpressed as a consequence of IL-1β effect in diabetes requires further investigation. It is believed that control of IL-1β pathways will be benefit the preservation of insulin secretory capacity [135]. A phase I therapy with Rilonacept, a soluble decoy receptor, in adolescents and adults with IDDM is ongoing. These observations support the inhibition of IL-1β may limit the immune responses and reduce the rate of β-cell destruction in IDDM. 3.5. Autoimmune inner ear disease (AIED) Autoimmune inner ear disease (AIED), originally defined as autoimmune sensorineural hearing loss, is an inscrutable disorder. It is characterized by recurring episodes of sudden or progressive sensorineural hearing loss and bilateral sensorineural hearing loss. The disease responds to treatment by immunosuppressives, and occurs in people between the ages of 30 and 60 years [51,136,137]. Patients with AIED usually experience multiple episodes of rapid hearing loss either concurrently or sequentially in both ears. Of those with AIED, up to 30% may have a systemic autoimmune disease such as Cogan syndrome, Wegener granulomatosis, SLE, and various systemic vasculitides [138]. For patients who experience an acute, sensorineural decline in hearing, timely corticosteroid administration may result in preservation of some or all of the hearing. Corticosteroids are widely used in the treatment of AIED and have been the standard treatment for the disease. Unfortunately, not all patients respond to the steroids, and in responders, some develop resistance over time. To date, no effective treatment has been identified for patients with either AIED who do not respond to steroids. Recently, treatment for corticosteroid non-responders with Anakinra is effective and resulted in reduction in IL-1β release, suggesting that IL-1β blockade may be a choice of therapy for these patients. Pathak et al. [51] demonstrated that circulating plasma levels of IL-1β in corticosteroid non-responders are higher as compared with corticosteroid-responders. IL-1β and MMP-9 are overexpressed in PBMC of corticosteroid resistant AIED patients but dexamethasone fails to regulate the expression of these proteins. Treatment of corticosteroid non-responders with Anakinra resulted in the inhibition of IL-1β and MMP-9 release. IL-1β has the ability to induce MMP-9 expression, which cleaves IL-1RII and then enhances IL-1β-induced signal pathway. Therefore, a greater capacity of Anakinra

664

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

to reduce MMP-9 expression in nonresponders than responders may be due to a reduction of circulating IL-1β. Thus, IL-1β may play a pivotal role in corticosteroid non-responder AIED. Nevertheless, the exact mechanism of AIED and how IL-1β participates in the progression of this disease remain uncertain. Research is required to delineate the pathophysiologic mechanisms. 3.6. Multiple sclerosis (MS) Multiple sclerosis (MS) is a chronic inflammatory disease mediated by activated T cells, B cells and monocytes (MO), with an autoimmune origin [1]. It is characterized by an altered balance between cytokines followed by selective destruction of myelin and gliosis in the central nervous system (CNS) [139,140]. Experimental autoimmune encephalomyelitis (EAE) is a mouse chronic demyelinating disease model for MS. Filion et al. reveal that MO secreted significant levels of proinflammatory/regulatory cytokines, specifically IL-1β, IL-6 and TNFα, in MS patients as compared to healthy individuals [140]. T effector cells can cause CNS inflammation and demyelinating lesions [141] and IL-17 is important in the immunopathogenesis of autoimmune demyelinating disease [142]. IL-1β is essential for the development of Th1 and Th17 cells [98,143]. Studies showed increased levels of IL-1β in the cerebral spinal fluid (CSF) and sera of MS patients. High levels of IL-1β have also been observed in MS brain lesions while IL-1β has been rarely demonstrated in normal CNS tissue [144]. Huitinga et al. suggested that IL-1β produced by activated glial cells in the hypothalamus of MS patients may contribute to the activation of the hypothalamic CRH neurons, while reduced expression of neuronal IL-1β in MS patients may have consequences for neuroendocrine, behavioural or autonomic functioning [145]. Rats with active EAE, treated with human recombinant IL-1Ra, during the induction phase of disease, developed milder signs of EAE [146]. Circulating serum levels of IL-1Ra are normal during the remission phase of relapsing–remitting MS, but it increases significantly during exacerbation or in response to interferon-β treatment [147]. Therefore, IL-1Ra has a therapeutical potential for MS during exacerbation. Dujmovic et al. revealed that IL-1Ra and/or IL-1RII were increased in the sera after steroid treatment for relapsing MS and suggest the important role of IL-1β. Thus, induction of IL-1RII and IL-1Ra in MS may be beneficial [1]. The production of IL-1 by peripheral blood mononuclear cells (PBMCs) was assessed in patients with relapsing MS in both the active and inactive phases and in chronic progressive MS patients. IL-1α production was significantly higher in MS patients than in healthy subjects while IL-1β in MS patients was not significantly elevated [148]. Therefore, the role of IL-1β in MS needs further research. 3.7. Myocarditis Myocarditis is a major cause of heart failure and sudden death in adolescents and young adults. Autoimmunity plays an important role in myocarditis, in particular, a reaction to cardiac myosin following viral infection may contribute to the development of myocarditis [149]. Experimental autoimmune myocarditis (EAM) is an animal model of human giant cell myocarditis, characterized by extremely severe myocardial damage and the appearance of multinucleated giant cells [150]. One of the leading causes of autoimmune myocardial lesion in patients with myocarditis is imbalance between pro-inflammatory and antiinflammatory cytokines [151]. IFN-γ, IL-1β, and TNF-α have been reported to possess negative inotropic effects against the myocardium and directly cause myocarditis through induction of NO [152]. Rose et al. found that increased production of IL-1β and TNF-α during the early innate response to virus infection is necessary and sufficient to induce a later heart-specific autoimmune disease [153]. Circulating IL-1β and TNF-α, which activate cytotoxic reactions, are markedly increased in patients with myocarditis [151,154,155], and the levels of IL-1β and TNF-α are correlated with the severity of the disease [156]. Inhibition

of these cytokines can prevent the development of EAM [155–157]. IL-1β has multiple effects on cardiac function. In patients with myocarditis myocardial contractility is depressed. Stein et al. demonstrated that IL-1β has direct negative inotropic effects on cardiomyocytes which may lead to the contractile dysfunction [158]. In synergy with TNF-α, IL-1β considerably enhances collagen synthesis and production of MMP 2 and MMP 9, two matrix metalloproteins that are associated with cardiac remodeling [153]. IL-1β activates NF-κB which in turn increases IL-1β transcription to cause myocardial damage in EAM [159]. These results suggest that IL-1β is a significant cytokine in the pathogenesis of myocarditis and blockade of IL-1β may be a treatment. 3.8. Autoimmune hepatitis (AIH) Hepatitis is an inflammation of the liver and can be caused by one of many factors including microbial infection, liver injury by toxins, and attack by host immune system. Autoimmune hepatitis (AIH) is a chronic inflammatory disorder of unknown etiology characterized by periportal inflammation, increased serum aminotransferase, hypergammaglobulinemia, circulating autoantibodies, and a remarkable response to immunosuppressive therapy [160,161]. Cytokines are thought to play a central role in liver metabolism and in the immune responses to viral components. IL-1β production is an important initiating factor in the cascade of events resulting in liver inflammation [30]. IL-1β and IL-6 genes are expressed in the liver infected by HBV, either chronically or acutely, with the exception of fulminant HBV hepatitis [162]. IL-1β is significantly elevated in the serum or within the liver of patients with hepatitis [30,163,164]. Gramantieri et al. found a mild reduction in the IL-1β/IL-1Ra ratio in minimal/mild chronic active hepatitis (CAH) and a strong increase in moderate/severe CAH [30]. ICAM-I is one of cell-surface molecules which mediates the interaction between hepatocytes and cells of the immune system [165]. Circulating levels of ICAM-I are significantly elevated in patients with various autoimmune liver diseases with low or undetectable levels in normal tissues. Inhibition of tissue ICAM-I expression and reduction in sICAMI levels may be important consequences of the actions of various treatments with immunosuppressive agents [165–167]. Incubation with inflammatory cytokines (IL-1β or TNF-α combined with IFN-γ) showed an increase in ICAM-I RNA levels in hepatocytes [165]. Th0 lymphocytes differentiate into Th1 or Th2 cells, while IL-1β and IL-6 favor differentiation into Th17 cells which have been demonstrated to play an important role in the pathogenesis of AIH [160,168]. Geneva-Popova M et al. suggest that IL-1β, IL-6 and TNF-α exist at high levels in the sera of patients with a self-limited form of acute hepatitis B infection (A-HBV). Their data indicate that these cytokines may be related either to multiple immune abnormalities or to favorable outcome of the disease [169]. Furthermore, IL-1β may directly target IFN-αβ signaling in hepatocytes, which are the only established treatment for viral hepatitis. Attenuation of IFN-αβ signaling in the liver by IL-1β could be one of the mechanisms underlying the resistance to IFN therapy [164]. 3.9. Kidney disease Cytokines such as IL-1β, IL-6 and TNF-α contribute to the progression of many kidney diseases and are associated with the development of common chronic kidney disease (CKD) complications [170–172]. The capacity of LPS-stimulated isolated erythrocyte-perfused rat kidney (IEPK) to produce monocyte chemoattractant protein-1 (MCP-1), IL-1β, and TNF-α mRNA has been demonstrated [173]. Both MAPKs and NF-κB are involved in the pathological conditions in the kidney, and are triggered by IL-1β. Thus, IL-1β plays an important role in kidney diseases by increasing MAPK and NF-κB signaling. The influence of cytokine gene polymorphisms on various immunologic complications after solid kidney transplantation is evaluated [174]. Kidney transplant recipients of the IL-1β-511CC genotype are at higher risk for the onset of

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

665

Table 2 The role of IL-1β in autoimmune diseases [31,51,54,62,74,86,88,90,95,98,107,113,114,117,118,123,139,145,150,158,161,164,175]. Disease

Pathological process

The role of IL-1β

RA SLE pSS

Activation of FLS, erosion of cartilage and bone Multisystem inflammation with the generation of autoantibodies Lym or Mon infiltration to epithelial cells of exocrine glands and dryness of the eyes and mouth Autoreactive T-cells to destroy photoreceptor cells in retina Lym infiltration and the presence of autoantibodies of the thyroid gland and developed thyroid tissue destroyed Tissue remodeling and fibrosis; accumulation of GAG in retro-ocular tissue Lym infiltration and the presence of autoantibodies of the pancreatic islets Antibodies or immune cells which are attacking the inner ear Selective destruction of myelin and gliosis in CNS Myocardial damage and the appearance of MGCs Periportal inflammation, increased serum aminotransferase, hypergammaglobulinemia, circulating autoantibodies Abnormal buildup of Ig and the presence of autoantibodies against the kidney

Expression of MMPs, adhesion molecules and proinflammatory mediators Susceptibility to acquire SLE Degradation of the basement membrane, disruption of acinar or ductal structure in salivary glands, induced iNOS Th1/17 cell differentiation, activates NF-κB Enhances IL-6 secretion and modifies epithelium integrity, induces Fas expression and the tissue damage Involves in GAG accumulation, induces TIMP-1 Mediates β-cell destruction and death through NO Induces MMP-9 expression Th1/Th17 cell differentiation, activates the hypothalamic CRH neurons Exacerbates cardiac myocyte remodeling and myocardial damage Increases ICAM-I RNA, favors Th0 Lym differentiation into Th17 cells, targets IFN-αβ signaling Higher risk for the onset of infection after kidney transplantation

Uveitis AITD TAO IDDM AIED MS Myocarditis AIH Kidney diseases

FLS: fibroblast-like synoviocytes; GAG: glycosaminoglycans; TIMP-1: tissue inhibitor of metalloproteinase-1; ICAM-l: intercellular adhesion molecule-1; MGCs: multinucleated giant cells; MMP: matrix metalloproteinase.

infection after kidney transplantation [175]. In addition, Wan et al. showed that recipients with IL-1β-511CC genotype or IL-1β-511C allele were more prone to the development of bacteremia within the first year after kidney transplantation [176]. Neutrophil recruitment induced by IL-1 β-stimulated chemokines is likely to play a key role in the disease and may provide novel therapeutic targets for acute tubular necrosis (ATN) [177]. 4. Conclusions and perspectives IL-1β participates in nearly all autoimmune diseases. There are complicated mechanisms for IL-1β to contribute to these diseases (Table 2). Because of the synergistic, and in some cases, redundant effects of many other proinflammatory cytokines such as TNFα and IL-6, it is expected that effective treatment of autoimmune diseases will require simultaneous inhibition of multiple cytokines. Combination therapy based on IL-1β may prove to be beneficial in curing many immunological disorders. In addition, DNA vaccines targeting IL-1β have been considered as good candidates for eradicating or alleviating autoimmune diseases [178]. It has been demonstrated that decreased production of IL-1β in patients with autoimmune disease leads to immune defects. Therefore, development of an efficient IL-1 inhibitor for clinical use is of importance. Clinical trials using recombinant IL-1 blocking agents are ongoing. Currently four IL-1β targeted agents have been approved by FDA: IL-1Ra Anakinra (Kineret, blocks both IL-1α and IL-1β), the neutralizing monoclonal antibody Canakinumab (specifically targets IL-1β) and Gevokizumab (XOMA 052, a recombinant humanized

anti-IL-1β antibody), the soluble decoy receptor rilonacept (binds IL-1β N IL-1α N IL-1Ra) [106,110,179] (Table 3). Other therapeutic approaches, including a monoclonal antibody directed against the IL-1 receptor and orally active small molecules that target the release of active IL-1β, have also been developed [179]. Anakinra has a half life of 4 to 6 h with a FDA approved recommended dose of 100 mg/day subcutaneously for the treatment of RA. Phase 1 clinical trials evaluating the safety and pharmacokinetics (PK) of Gevokizumab administered to patients with active, stable, moderate to severe RA have been completed. FDA grants orphan drug status to Gevokizumab in August 2012 and its safety and efficacy in the treatment of active non-infectious intermediate, posterior, or pan-uveitis clinical trials are in phase 3 trial. Anakinra may be able to treat BD with fewer side effects. Phase 2 clinical trials are projected to complete in August 2013. Clinical trials of the effect of Anakinra on insulin production in patients with new onset type 1 diabetes are in phase 3 trial and therapy in children with newly diagnosed type 1 DM (by ADA criteria) in phase 2 is underway. Gevokizumab may inhibit beta-cell destruction and enhance beta-cell regeneration. The effects of Gevokizumab on insulin production and β cell function in subjects with well-controlled type 1 diabetes in phase 2 trial are ongoing. A phase 1 open label clinical trial of Anakinra for corticosteroid-resistant patients is ongoing to determine if this therapy may improve hearing thresholds in patients with AIED. The clinical trials of IL-1Ra in chronic kidney disease and cardiovascular disease also have begun. Although these inhibitors have shown efficacy in a number of diseases, new therapeutic options with more potent inhibitors need development.

Table 3 Anti-IL-1 β therapy. (Reference: http://www.clinicaltrials.gov/).

IL-1Ra (Anakinra)

Human monoclonal IL-1β antibody (Canakinumab) Humanized IL-1β antibody (Gevokizumab)

Decoy receptor (Rilonacept)

Disease

Effect of therapy

Clinical trial

RA IDDM pSS AIED MS SLE RA BD IDDM RA AIED Uveitis IDDM IDDM

Retards the progression of radiographic joint damage; effective to moderate–severe RA Insulin production Be a potential strategy for future treatment Improves hearing thresholds Therapeutic effect during exacerbations Safety and efficacy in improving arthritis in SLE patients Retards the progression of radiographic joint damage Safety and efficacy Therapeutic intervention of IDDM Prevents and treats joint inflammation Reduces MMP-9 expression Treats successfully Inhibits β-cell destruction and enhances β-cell regeneration Slows the loss of the body's ability to secrete insulin

Phase 4 Phase 3 None Phase1/2 None None Phase 2 Phase 2 Phase 2 Phase 2 None Phase 3 Phase 2 Phase 1

666

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

Acknowledgments We are grateful for the critical comments by Dr. Ji Ming Wang (NCI, NIH). This project was supported in part by the grants from Guangdong Natural Science Foundation (S2011010006048) and the Fundamental Research Funds of State Key Laboratory of Ophthalmology (303060202400439). References [1] Dujmovic I, Mangano K, Pekmezovic T, Quattrocchi C, Mesaros S, Stojsavljevic N, et al. The analysis of IL-1 beta and its naturally occurring inhibitors in multiple sclerosis: the elevation of IL-1 receptor antagonist and IL-1 receptor type II after steroid therapy. J Neuroimmunol 2009;207(1–2):101–6. [2] Gery I, Waksman BH. Potentiation of the T-lymphocyte response to mitogens. II. The cellular source of potentiating mediator(s). J Exp Med 1972;136(1):143–55. [3] Gery I, Gershon RK, Waksman BH. Potentiation of the T-lymphocyte response to mitogens. I. The responding cell. J Exp Med 1972;136(1):128–42. [4] Dinarello CA, Renfer L, Wolff SM. Human leukocytic pyrogen: purification and development of a radioimmunoassay. Proc Natl Acad Sci U S A 1977;74(10):4624–7. [5] Steinkasserer A, Spurr NK, Cox S, Jeggo P, Sim RB. The human IL-1 receptor antagonist gene (IL1RN) maps to chromosome 2q14–q21, in the region of the IL-1 alpha and IL-1 beta loci. Genomics 1992;13(3):654–7. [6] Kluczyk A, Siemiona IZ, Wieczorek Z. The “two-headed” peptide inhibitors of interleukin-1 action. Peptides 2000;21(9):1411–20. [7] Tripodi D, Conti F, Rosati M, Maccauro G, Saggini A, Cianchetti E, et al. IL-36 a new member of the IL-1 family cytokines. J Biol Regul Homeost Agents 2012;26(1):7–14. [8] Pecaric-Petkovic T, Didichenko SA, Kaempfer S, Spiegl N, Dahinden CA. Human basophils and eosinophils are the direct target leukocytes of the novel IL-1 family member IL-33. Blood 2009;113(7):1526–34. [9] Boraschi D, Lucchesi D, Hainzl S, Leitner M, Maier E, Mangelberger D, et al. IL37: a new anti-inflammatory cytokine of the IL-1 family. Eur Cytokine Netw 2011;22(3):127–47. [10] van de Veerdonk FL, Stoeckman AK, Wu G, Boeckermann AN, Azam T, Netea MG, et al. IL-38 binds to the IL-36 receptor and has biological effects on immune cells similar to IL-36 receptor antagonist. Proc Natl Acad Sci U S A 2012;109(8):3001–5. [11] Johnston A, Xing X, Guzman AM, Riblett M, Loyd CM, Ward NL, et al. IL-1F5, -F6, -F8, and -F9: a novel IL-1 family signaling system that is active in psoriasis and promotes keratinocyte antimicrobial peptide expression. J Immunol 2011;186(4):2613–22. [12] Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature 1992;356(6372):768–74. [13] Andrei C, Dazzi C, Lotti L, Torrisi MR, Chimini G, Rubartelli A. The secretory route of the leaderless protein interleukin 1beta involves exocytosis of endolysosomerelated vesicles. Mol Biol Cell 1999;10(5):1463–75. [14] Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 2002;10(2):417–26. [15] Lopez-Castejon G, Brough D. Understanding the mechanism of IL-1beta secretion. Cytokine Growth Factor Rev 2011;22(4):189–95. [16] Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol 2012;13(4):333–42. [17] Barker BR, Taxman DJ, Ting JP. Cross-regulation between the IL-1beta/IL-18 processing inflammasome and other inflammatory cytokines. Curr Opin Immunol 2011;23(5):591–7. [18] Lamkanfi M, Walle LV, Kanneganti TD. Deregulated inflammasome signaling in disease. Immunol Rev 2011;243(1):163–73. [19] Burger D, Dayer JM, Palmer G, Gabay C. Is IL-1 a good therapeutic target in the treatment of arthritis? Best Pract Res Clin Rheumatol 2006;20(5):879–96. [20] Hsu LC, Ali SR, McGillivray S, Tseng PH, Mariathasan S, Humke EW, et al. A NOD2– NALP1 complex mediates caspase-1-dependent IL-1beta secretion in response to Bacillus anthracis infection and muramyl dipeptide. Proc Natl Acad Sci U S A 2008;105(22):7803–8. [21] Gross O, Poeck H, Bscheider M, Dostert C, Hannesschlager N, Endres S, et al. Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature 2009;459(7245):433–6. [22] Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, Franchi L, et al. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem 2006;281(48):36560–8. [23] Dinarello CA. Interleukin-1 and interleukin-1 receptor antagonist. Nutrition 1995;11(5 Suppl.):492–4. [24] Dinarello CA. Interleukin-1 and interleukin-1 receptor antagonist production during haemodialysis: which cytokine is a surrogate marker for dialysis-related complications? Nephrol Dial Transplant 1995;10(Suppl. 3):25–8. [25] Dinarello CA. Biologic basis for interleukin-1 in disease. Blood 1996;87(6):2095–147. [26] Dinarello CA. The IL-1 family and inflammatory diseases. Clin Exp Rheumatol 2002;20(5 Suppl. 27):S1–S13. [27] Contassot E, Beer HD, French LE. Interleukin-1, inflammasomes, autoinflammation and the skin. Swiss Med Wkly 2012;142:w13590. [28] Garcia-Gonzalez MA, Aisa MA, Strunk M, Benito R, Piazuelo E, Jimenez P, et al. Relevance of IL-1 and TNF gene polymorphisms on interleukin-1beta and

[29] [30]

[31]

[32]

[33]

[34]

[35]

[36]

[37] [38] [39]

[40] [41] [42]

[43]

[44] [45]

[46]

[47] [48] [49]

[50]

[51]

[52]

[53] [54] [55]

[56] [57] [58] [59]

[60]

tumor necrosis factor-alpha gastric mucosal production. Hum Immunol 2009;70(11):935–45. Onozaki K, Matsushima K, Aggarwal BB, Oppenheim JJ. Human interleukin 1 is a cytocidal factor for several tumor cell lines. J Immunol 1985;135(6):3962–8. Gramantieri L, Casali A, Trere D, Gaiani S, Piscaglia F, Chieco P, et al. Imbalance of IL1 beta and IL-1 receptor antagonist mRNA in liver tissue from hepatitis C virus (HCV)-related chronic hepatitis. Clin Exp Immunol 1999;115(3):515–20. Camargo JF, Correa PA, Castiblanco J, Anaya JM. Interleukin-1beta polymorphisms in Colombian patients with autoimmune rheumatic diseases. Genes Immun 2004;5(8):609–14. Wewers MD, Dare HA, Winnard AV, Parker JM, Miller DK. IL-1 beta-converting enzyme (ICE) is present and functional in human alveolar macrophages: macrophage IL-1 beta release limitation is ICE independent. J Immunol 1997;159(12):5964–72. Aggelakis K, Zacharaki F, Dardiotis E, Xiromerisiou G, Tsimourtou V, Ralli S, et al. Interleukin-1B and interleukin-1 receptor antagonist gene polymorphisms in Greek multiple sclerosis (MS) patients with bout-onset MS. Neurol Sci 2010;31(3):253–7. Heitmeier MR, Arnush M, Scarim AL, Corbett JA. Pancreatic beta-cell damage mediated by beta-cell production of interleukin-1. A novel mechanism for virus-induced diabetes. J Biol Chem 2001;276(14):11151–8. Oberholzer A, Oberholzer C, Moldawer LL. Cytokine signaling—regulation of the immune response in normal and critically ill states. Crit Care Med 2000;28(4 Suppl.): N3–N12. Sparre T, Christensen UB, Gotfredsen CF, Larsen PM, Fey SJ, Hjerno K, et al. Changes in expression of IL-1 beta influenced proteins in transplanted islets during development of diabetes in diabetes-prone BB rats. Diabetologia 2004;47(5):892–908. Dinarello CA. Interleukin-1, interleukin-1 receptors and interleukin-1 receptor antagonist. Int Rev Immunol 1998;16(5–6):457–99. Rothwell NJ, Luheshi GN. Interleukin 1 in the brain: biology, pathology and therapeutic target. Trends Neurosci 2000;23(12):618–25. Tseng HC, Lee IT, Lin CC, Chi PL, Cheng SE, Shih RH, et al. IL-1beta promotes corneal epithelial cell migration by increasing MMP-9 expression through NF-kappaB- and AP-1-dependent pathways. PLoS One 2013;8(3):e57955. Dunne A, O'Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduction during inflammation and host defense. Sci STKE 2003;2003(171):re3. Nishida E, Gotoh Y. The MAP kinase cascade is essential for diverse signal transduction pathways. Trends Biochem Sci 1993;18(4):128–31. Walsh MC, Kim GK, Maurizio PL, Molnar EE, Choi Y. TRAF6 autoubiquitinationindependent activation of the NFkappaB and MAPK pathways in response to IL-1 and RANKL. PLoS One 2008;3(12):e4064. Lie PP, Cheng CY, Mruk DD. The biology of interleukin-1: emerging concepts in the regulation of the actin cytoskeleton and cell junction dynamics. Cell Mol Life Sci 2012;69(4):487–500. Xia ZP, Sun L, Chen X, Pineda G, Jiang X, Adhikari A, et al. Direct activation of protein kinases by unanchored polyubiquitin chains. Nature 2009;461(7260):114–9. Chun J, Choi RJ, Khan S, Lee DS, Kim YC, Nam YJ, et al. Alantolactone suppresses inducible nitric oxide synthase and cyclooxygenase-2 expression by down-regulating NF-kappaB, MAPK and AP-1 via the MyD88 signaling pathway in LPS-activated RAW 264.7 cells. Int Immunopharmacol 2012;14(4):375–83. Persichini T, Maio N, di Patti MC, Rizzo G, Toscano S, Colasanti M, et al. Interleukin1beta induces ceruloplasmin and ferroportin-1 gene expression via MAP kinases and C/EBPbeta, AP-1, and NF-kappaB activation. Neurosci Lett 2010;484(2):133–8. Thanos D, Maniatis T. NF-kappa B: a lesson in family values. Cell 1995;80(4):529–32. Verma IM, Stevenson J. IkappaB kinase: beginning, not the end. Proc Natl Acad Sci U S A 1997;94(22):11758–60. Chen R, Li M, Zhang Y, Zhou Q, Shu HB. The E3 ubiquitin ligase MARCH8 negatively regulates IL-1beta-induced NF-kappaB activation by targeting the IL1RAP coreceptor for ubiquitination and degradation. Proc Natl Acad Sci U S A 2012;109(35):14128–33. Guesdon F, Knight CG, Rawlinson LM, Saklatvala J. Dual specificity of the interleukin 1- and tumor necrosis factor-activated beta casein kinase. J Biol Chem 1997;272(48):30017–24. Pathak S, Goldofsky E, Vivas EX, Bonagura VR, Vambutas A. IL-1beta is overexpressed and aberrantly regulated in corticosteroid nonresponders with autoimmune inner ear disease. J Immunol 2011;186(3):1870–9. Galarza C, Valencia D, Tobon GJ, Zurita L, Mantilla RD, Pineda-Tamayo R, et al. Should rituximab be considered as the first-choice treatment for severe autoimmune rheumatic diseases? Clin Rev Allergy Immunol 2008;34(1):124–8. Burrage PS, Mix KS, Brinckerhoff CE. Matrix metalloproteinases: role in arthritis. Front Biosci 2006;11:529–43. Breedveld F. New tumor necrosis factor-alpha biologic therapies for rheumatoid arthritis. Eur Cytokine Netw 1998;9(3):233–8. Bansard C, Lequerre T, Derambure C, Vittecoq O, Hiron M, Daragon A, et al. Gene profiling predicts rheumatoid arthritis responsiveness to IL-1Ra (anakinra). Rheumatology (Oxford) 2011;50(2):283–92. Olsen NJ, Stein CM. New drugs for rheumatoid arthritis. N Engl J Med 2004;350(21):2167–79. Smolen JS, Aletaha D, Koeller M, Weisman MH, Emery P. New therapies for treatment of rheumatoid arthritis. Lancet 2007;370(9602):1861–74. van den Berg WB. Anti-cytokine therapy in chronic destructive arthritis. Arthritis Res 2001;3(1):18–26. Alten R, Gram H, Joosten LA, van den Berg WB, Sieper J, Wassenberg S, et al. The human anti-IL-1 beta monoclonal antibody ACZ885 is effective in joint inflammation models in mice and in a proof-of-concept study in patients with rheumatoid arthritis. Arthritis Res Ther 2008;10(3):R67. Miyata S, Ohkubo Y, Mutoh S. A review of the action of tacrolimus (FK506) on experimental models of rheumatoid arthritis. Inflamm Res 2005;54(1):1–9.

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669 [61] Lafyatis R, Remmers EF, Roberts AB, Yocum DE, Sporn MB, Wilder RL. Anchorageindependent growth of synoviocytes from arthritic and normal joints. Stimulation by exogenous platelet-derived growth factor and inhibition by transforming growth factor-beta and retinoids. J Clin Invest 1989;83(4):1267–76. [62] Mengshol JA, Vincenti MP, Coon CI, Barchowsky A, Brinckerhoff CE. Interleukin-1 induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p38, c-Jun N-terminal kinase, and nuclear factor kappaB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum 2000;43(4):801–11. [63] Mengshol JA, Vincenti MP, Brinckerhoff CE. IL-1 induces collagenase-3 (MMP13) promoter activity in stably transfected chondrocytic cells: requirement for Runx-2 and activation by p38 MAPK and JNK pathways. Nucleic Acids Res 2001;29(21):4361–72. [64] Schmucker AC, Wright JB, Cole MD, Brinckerhoff CE. Distal interleukin-1beta (IL-1beta) response element of human matrix metalloproteinase-13 (MMP-13) binds activator protein 1 (AP-1) transcription factors and regulates gene expression. J Biol Chem 2012;287(2):1189–97. [65] Le Loet X, Nordstrom D, Rodriguez M, Rubbert A, Sarzi-Puttini P, Wouters JM, et al. Effect of anakinra on functional status in patients with active rheumatoid arthritis receiving concomitant therapy with traditional disease modifying antirheumatic drugs: evidence from the OMEGA Trial. J Rheumatol 2008;35(8):1538–44. [66] den Broeder AA, de Jong E, Franssen MJ, Jeurissen ME, Flendrie M, van den Hoogen FH. Observational study on efficacy, safety, and drug survival of anakinra in rheumatoid arthritis patients in clinical practice. Ann Rheum Dis 2006;65(6):760–2. [67] Cohen S, Hurd E, Cush J, Schiff M, Weinblatt ME, Moreland LW, et al. Treatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor antagonist, in combination with methotrexate: results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum 2002;46(3):614–24. [68] Bresnihan B, Cobby M. Clinical and radiological effects of anakinra in patients with rheumatoid arthritis. Rheumatology (Oxford) 2003;42(Suppl. 2):ii22–8. [69] Mertens M, Singh JA. Anakinra for rheumatoid arthritis. Cochrane Database Syst Rev 2009;1:CD005121. [70] Bresnihan B. The safety and efficacy of interleukin-1 receptor antagonist in the treatment of rheumatoid arthritis. Semin Arthritis Rheum 2001;30(5 Suppl. 2):17–20. [71] Fleischmann RM, Schechtman J, Bennett R, Handel ML, Burmester GR, Tesser J, et al. Anakinra, a recombinant human interleukin-1 receptor antagonist (r-metHuIL1ra), in patients with rheumatoid arthritis: a large, international, multicenter, placebo-controlled trial. Arthritis Rheum 2003;48(4):927–34. [72] Fleischmann RM, Tesser J, Schiff MH, Schechtman J, Burmester GR, Bennett R, et al. Safety of extended treatment with anakinra in patients with rheumatoid arthritis. Ann Rheum Dis 2006;65(8):1006–12. [73] Owyang AM, Issafras H, Corbin J, Ahluwalia K, Larsen P, Pongo E, et al. XOMA 052, a potent, high-affinity monoclonal antibody for the treatment of IL-1beta-mediated diseases. MAbs 2011;3(1):49–60. [74] Askanase A, Shum K, Mitnick H. Systemic lupus erythematosus: an overview. Soc Work Health Care 2012;51(7):576–86. [75] Dean GS, Tyrrell-Price J, Crawley E, Isenberg DA. Cytokines and systemic lupus erythematosus. Ann Rheum Dis 2000;59(4):243–51. [76] Jandl RC, George JL, Dinarello CA, Schur PH. The effect of interleukin 1 on IgG synthesis in systemic lupus erythematosus. Clin Immunol Immunopathol 1987;45(3):384–94. [77] Segal R, Bermas BL, Dayan M, Kalush F, Shearer GM, Mozes E. Kinetics of cytokine production in experimental systemic lupus erythematosus: involvement of T helper cell 1/T helper cell 2-type cytokines in disease. J Immunol 1997;158(6):3009–16. [78] Handwerger BS, Rus V, da Silva L, Via CS. The role of cytokines in the immunopathogenesis of lupus. Springer Semin Immunopathol 1994;16(2–3):153–80. [79] Aringer M, Smolen JS. Cytokine expression in lupus kidneys. Lupus 2005;14(1):13–8. [80] Brennan DC, Yui MA, Wuthrich RP, Kelley VE. Tumor necrosis factor and IL-1 in New Zealand Black/White mice. Enhanced gene expression and acceleration of renal injury. J Immunol 1989;143(11):3470–5. [81] Voronov E, Dayan M, Zinger H, Gayvoronsky L, Lin JP, Iwakura Y, et al. IL-1 betadeficient mice are resistant to induction of experimental SLE. Eur Cytokine Netw 2006;17(2):109–16. [82] Connolly JJ, Hakonarson H. Role of cytokines in systemic lupus erythematosus: recent progress from GWAS and sequencing. J Biomed Biotechnol 2012;2012:798924. [83] Suzuki H, Takemura H, Kashiwagi H. Interleukin-1 receptor antagonist in patients with active systemic lupus erythematosus. Enhanced production by monocytes and correlation with disease activity. Arthritis Rheum 1995;38(8):1055–9. [84] Sturfelt G, Roux-Lombard P, Wollheim FA, Dayer JM. Low levels of interleukin-1 receptor antagonist coincide with kidney involvement in systemic lupus erythematosus. Br J Rheumatol 1997;36(12):1283–9. [85] Lee WW, Lee N, Fujii H, Kang I. Active hexose correlated compound promotes T helper (Th) 17 and 1 cell responses via inducing IL-1beta production from monocytes in humans. Cell Immunol 2012;275(1–2):19–23. [86] Zoukhri D, Macari E, Choi SH, Kublin CL. c-Jun NH2-terminal kinase mediates interleukin-1beta-induced inhibition of lacrimal gland secretion. J Neurochem 2006;96(1):126–35. [87] Fox RI, Tornwall J, Michelson P. Current issues in the diagnosis and treatment of Sjogren's syndrome. Curr Opin Rheumatol 1999;11(5):364–71. [88] Willeke P, Schotte H, Schluter B, Erren M, Becker H, Dyong A, et al. Interleukin 1beta and tumour necrosis factor alpha secreting cells are increased in the peripheral blood of patients with primary Sjogren's syndrome. Ann Rheum Dis 2003;62(4):359–62. [89] Tapinos NI, Polihronis M, Tzioufas AG, Moutsopoulos HM. Sjogren's syndrome. Autoimmune epithelitis. Adv Exp Med Biol 1999;455:127–34.

667

[90] Azuma M, Motegi K, Aota K, Hayashi Y, Sato M. Role of cytokines in the destruction of acinar structure in Sjogren's syndrome salivary glands. Lab Investig; J Tech Methods Pathol 1997;77(3):269–80. [91] Garcic-Carrasco M, Font J, Filella X, Cervera R, Ramos-Casals M, Siso A, et al. Circulating levels of Th1/Th2 cytokines in patients with primary Sjogren's syndrome: correlation with clinical and immunological features. Clin Exp Rheumatol 2001;19(4):411–5. [92] Beauregard C, Brandt PC, Chiou GC. Induction of nitric oxide synthase and overproduction of nitric oxide by interleukin-1beta in cultured lacrimal gland acinar cells. Exp Eye Res 2003;77(1):109–14. [93] Yamakawa M, Weinstein R, Tsuji T, McBride J, Wong DT, Login GR. Age-related alterations in IL-1beta, TNF-alpha, and IL-6 concentrations in parotid acinar cells from BALB/c and non-obese diabetic mice. J Histochem Cytochem 2000;48(8):1033–42. [94] Szodoray P, Alex P, Brun JG, Centola M, Jonsson R. Circulating cytokines in primary Sjogren's syndrome determined by a multiplex cytokine array system. Scand J Immunol 2004;59(6):592–9. [95] Muraki Y, Tsutsumi A, Takahashi R, Suzuki E, Hayashi T, Chino Y, et al. Polymorphisms of IL-1 beta gene in Japanese patients with Sjogren's syndrome and systemic lupus erythematosus. J Rheumatol 2004;31(4):720–5. [96] Zoukhri D, Hodges RR, Byon D, Kublin CL. Role of proinflammatory cytokines in the impaired lacrimation associated with autoimmune xerophthalmia. Invest Ophthalmol Vis Sci 2002;43(5):1429–36. [97] Becker H, Pavenstaedt H, Willeke P. Emerging treatment strategies and potential therapeutic targets in primary Sjogren's syndrome. Inflamm Allergy Drug Targets 2010;9(1):10–9. [98] Lasiglie D, Traggiai E, Federici S, Alessio M, Buoncompagni A, Accogli A, et al. Role of IL-1 beta in the development of human T(H)17 cells: lesson from NLPR3 mutated patients. PLoS One 2011;6(5):e20014. [99] Chi W, Yang P, Li B, Wu C, Jin H, Zhu X, et al. IL-23 promotes CD4 + T cells to produce IL-17 in Vogt–Koyanagi–Harada disease. J Allergy Clin Immunol 2007;119(5):1218–24. [100] Amadi-Obi A, Yu CR, Liu X, Mahdi RM, Clarke GL, Nussenblatt RB, et al. TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med 2007;13(6):711–8. [101] Karasneh J, Hajeer AH, Barrett J, Ollier WE, Thornhill M, Gul A. Association of specific interleukin 1 gene cluster polymorphisms with increased susceptibility for Behcet's disease. Rheumatology (Oxford) 2003;42(7):860–4. [102] Su SB, Silver PB, Grajewski RS, Agarwal RK, Tang J, Chan CC, et al. Essential role of the MyD88 pathway, but nonessential roles of TLRs 2, 4, and 9, in the adjuvant effect promoting Th1-mediated autoimmunity. J Immunol 2005;175(10):6303–10. [103] Pay S, Erdem H, Pekel A, Simsek I, Musabak U, Sengul A, et al. Synovial proinflammatory cytokines and their correlation with matrix metalloproteinase-3 expression in Behcet's disease. Does interleukin-1beta play a major role in Behcet's synovitis? Rheumatol Int 2006;26(7):608–13. [104] Xuan B, Chiou GC, Chen Z, Yamasaki T, Okawara T. Effective treatment of experimental uveitis with interleukin-1 blockers, CK 123 and CK 124. J Ocul Pharmacol Ther 1998;14(1):31–44. [105] Duzgun N, Ayaslioglu E, Tutkak H, Aydintug OT. Cytokine inhibitors: soluble tumor necrosis factor receptor 1 and interleukin-1 receptor antagonist in Behcet's disease. Rheumatol Int 2005;25(1):1–5. [106] Gul A, Tugal-Tutkun I, Dinarello CA, Reznikov L, Esen BA, Mirza A, et al. Interleukin1beta-regulating antibody XOMA 052 (gevokizumab) in the treatment of acute exacerbations of resistant uveitis of Behcet's disease: an open-label pilot study. Ann Rheum Dis 2012;71(4):563–6. [107] Keino H. Therapeutic effect of the low molecular weight inhibitor of the NF-kappaB signaling pathway on experimental autoimmune uveoretinitis. Nihon Ganka Gakkai Zasshi 2010;114(11):944–54. [108] Kitamei H, Iwabuchi K, Namba K, Yoshida K, Yanagawa Y, Kitaichi N, et al. Amelioration of experimental autoimmune uveoretinitis (EAU) with an inhibitor of nuclear factor-kappaB (NF-kappaB), pyrrolidine dithiocarbamate. J Leukoc Biol 2006;79(6):1193–201. [109] Lennikov A, Kitaichi N, Noda K, Ando R, Dong Z, Fukuhara J, et al. Amelioration of endotoxin-induced uveitis treated with an IkappaB kinase beta inhibitor in rats. Mol Vis 2012;18:2586–97. [110] Ugurlu S, Ucar D, Seyahi E, Hatemi G, Yurdakul S. Canakinumab in a patient with juvenile Behcet's syndrome with refractory eye disease. Ann Rheum Dis 2012;71(9):1589–91. [111] Kone-Paut I, Lachmann HJ, Kuemmerle-Deschner JB, Hachulla E, Leslie KS, Mouy R, et al. Sustained remission of symptoms and improved health-related quality of life in patients with cryopyrin-associated periodic syndrome treated with canakinumab: results of a double-blind placebo-controlled randomized withdrawal study. Arthritis Res Ther 2011;13(6):R202. [112] Weetman AP. The potential immunological role of the thyroid cell in autoimmune thyroid disease. Thyroid 1994;4(4):493–9. [113] Rebuffat SA, Kammoun-Krichen M, Charfeddine I, Ayadi H, Bougacha-Elleuch N, Peraldi-Roux S. IL-1beta and TSH disturb thyroid epithelium integrity in autoimmune thyroid diseases. Immunobiology 2013;218(3):285–91. [114] Paolieri F, Salmaso C, Battifora M, Montagna P, Pesce G, Bagnasco M, et al. Possible pathogenetic relevance of interleukin-1 beta in “destructive” organ-specific autoimmune disease (Hashimoto's thyroiditis). Ann N Y Acad Sci 1999;876:221–8. [115] Naik V, Khadavi N, Naik MN, Hwang C, Goldberg RA, Tsirbas A, et al. Biologic therapeutics in thyroid-associated ophthalmopathy: translating disease mechanism into therapy. Thyroid 2008;18(9):967–71. [116] Garrity JA, Bahn RS. Pathogenesis of graves ophthalmopathy: implications for prediction, prevention, and treatment. Am J Ophthalmol 2006;142(1):147–53.

668

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669

[117] Han R, Smith TJ. Induction by IL-1 beta of tissue inhibitor of metalloproteinase-1 in human orbital fibroblasts: modulation of gene promoter activity by IL-4 and IFNgamma. J Immunol 2005;174(5):3072–9. [118] Lu R, Wang P, Wartofsky L, Sutton BD, Zweier JL, Bahn RS, et al. Oxygen free radicals in interleukin-1beta-induced glycosaminoglycan production by retro-ocular fibroblasts from normal subjects and Graves' ophthalmopathy patients. Thyroid 1999;9(3):297–303. [119] Huang D, Xu N, Song Y, Wang P, Yang H. Inflammatory cytokine profiles in the tears of thyroid-associated ophthalmopathy. Graefes Arch Clin Exp Ophthalmol 2012;250(4):619–25. [120] Jun HS, Yoon JW. Initiation of autoimmune type 1 diabetes and molecular cloning of a gene encoding for islet cell-specific 37kd autoantigen. Adv Exp Med Biol 1994;347:207–20. [121] Corbett JA, Kwon G, Marino MH, Rodi CP, Sullivan PM, Turk J, et al. Tyrosine kinase inhibitors prevent cytokine-induced expression of iNOS and COX-2 by human islets. Am J Physiol 1996;270(6 Pt 1):C1581–7. [122] Dogan Y, Akarsu S, Ustundag B, Yilmaz E, Gurgoze MK. Serum IL-1beta, IL-2, and IL-6 in insulin-dependent diabetic children. Mediators Inflamm 2006;2006(1):59206. [123] Corbett JA, Kwon G, Turk J, McDaniel ML. IL-1 beta induces the coexpression of both nitric oxide synthase and cyclooxygenase by islets of Langerhans: activation of cyclooxygenase by nitric oxide. Biochemistry 1993;32(50):13767–70. [124] Rabinovitch A. An update on cytokines in the pathogenesis of insulin-dependent diabetes mellitus. Diabetes Metab Rev 1998;14(2):129–51. [125] Reddy S, Young M. IL-1beta expression in islet cells of the NOD mouse and its spatial relationship to beta cells and inducible nitric oxide synthase. Ann N Y Acad Sci 2002;958:190–3. [126] Sprinkel AM, Andersen NA, Mandrup-Poulsen T. Glucose potentiates interleukin-1 beta (IL-1 beta)-induced p38 mitogen-activated protein kinase activity in rat pancreatic islets of Langerhans. Eur Cytokine Netw 2001;12(2):331–9. [127] Xenos ES, Stevens RB, Gores PF, Casanova D, Farney AC, Sutherland DE, et al. IL-1 beta-induced inhibition of beta-cell function is mediated through nitric oxide. Transplant Proc 1993;25(1 Pt 2):994. [128] Baekkeskov S, Aanstoot HJ, Christgau S, Reetz A, Solimena M, Cascalho M, et al. Identification of the 64 K autoantigen in insulin-dependent diabetes as the GABAsynthesizing enzyme glutamic acid decarboxylase. Nature 1990;347(6289):151–6. [129] Schmidli RS, Faulkner-Jones BE, Harrison LC, James RF, DeAizpurua HJ. Cytokine regulation of glutamate decarboxylase biosynthesis in isolated rat islets of Langerhans. Biochem J 1996;317(Pt 3):713–9. [130] Giannoukakis N, Rudert WA, Trucco M, Robbins PD. Protection of human islets from the effects of interleukin-1beta by adenoviral gene transfer of an Ikappa B repressor. J Biol Chem 2000;275(47):36509–13. [131] Ortis F, Pirot P, Naamane N, Kreins AY, Rasschaert J, Moore F, et al. Induction of nuclear factor-kappaB and its downstream genes by TNF-alpha and IL-1beta has a pro-apoptotic role in pancreatic beta cells. Diabetologia 2008;51(7):1213–25. [132] Ortis F, Miani M, Colli ML, Cunha DA, Gurzov EN, Allagnat F, et al. Differential usage of NF-kappaB activating signals by IL-1beta and TNF-alpha in pancreatic beta cells. FEBS Lett 2012;586(7):984–9. [133] Wilson CA, Jacobs C, Baker P, Baskin DG, Dower S, Lernmark A, et al. IL-1 beta modulation of spontaneous autoimmune diabetes and thyroiditis in the BB rat. J Immunol 1990;144(10):3784–8. [134] Cailleau C, Diu-Hercend A, Ruuth E, Westwood R, Carnaud C. Treatment with neutralizing antibodies specific for IL-1beta prevents cyclophosphamide-induced diabetes in nonobese diabetic mice. Diabetes 1997;46(6):937–40. [135] Sumpter KM, Adhikari S, Grishman EK, White PC. Preliminary studies related to anti-interleukin-1beta therapy in children with newly diagnosed type 1 diabetes. Pediatr Diabetes 2011;12(7):656–67. [136] McCabe BF. Autoimmune sensorineural hearing loss. Ann Otol Rhinol Laryngol 1979;88(5 Pt 1):585–9. [137] Solares CA, Edling AE, Johnson JM, Baek MJ, Hirose K, Hughes GB, et al. Murine autoimmune hearing loss mediated by CD4+ T cells specific for inner ear peptides. J Clin Invest 2004;113(8):1210–7. [138] Ruckenstein MJ. Autoimmune inner ear disease. Curr Opin Otolaryngol Head Neck Surg 2004;12(5):426–30. [139] Trapp BD, Bo L, Mork S, Chang A. Pathogenesis of tissue injury in MS lesions. J Neuroimmunol 1999;98(1):49–56. [140] Filion LG, Graziani-Bowering G, Matusevicius D, Freedman MS. Monocyte-derived cytokines in multiple sclerosis. Clin Exp Immunol 2003;131(2):324–34. [141] Lovett-Racke AE, Yang Y, Racke MK. Th1 versus Th17: are T cell cytokines relevant in multiple sclerosis? Biochim Biophys Acta 2011;1812(2):246–51. [142] Segal BM. Th17 cells in autoimmune demyelinating disease. Semin Immunopathol 2010;32(1):71–7. [143] Madera RF, Wang JP, Libraty DH. The combination of early and rapid type I IFN, IL-1alpha, and IL-1beta production are essential mediators of RNA-like adjuvant driven CD4+ Th1 responses. PLoS One 2011;6(12):e29412. [144] Cannella B, Raine CS. The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol 1995;37(4):424–35. [145] Huitinga I, van der Cammen M, Salm L, Erkut Z, van Dam A, Tilders F, et al. IL-1beta immunoreactive neurons in the human hypothalamus: reduced numbers in multiple sclerosis. J Neuroimmunol 2000;107(1):8–20. [146] Badovinac V, Mostarica-Stojkovic M, Dinarello CA, Stosic-Grujicic S. Interleukin-1 receptor antagonist suppresses experimental autoimmune encephalomyelitis (EAE) in rats by influencing the activation and proliferation of encephalitogenic cells. J Neuroimmunol 1998;85(1):87–95. [147] Nicoletti F, Patti F, DiMarco R, Zaccone P, Nicoletti A, Meroni P, et al. Circulating serum levels of IL-1ra in patients with relapsing remitting multiple sclerosis are

[148]

[149]

[150]

[151]

[152] [153] [154]

[155]

[156] [157]

[158]

[159]

[160]

[161] [162]

[163]

[164]

[165]

[166]

[167]

[168] [169]

[170]

[171] [172] [173]

[174]

[175]

[176]

normal during remission phases but significantly increased either during exacerbations or in response to IFN-beta treatment. Cytokine 1996;8(5):395–400. Matsuda M, Tsukada N, Miyagi K, Yanagisawa N. Increased interleukin-1 production by peripheral blood mononuclear cells in patients with multiple sclerosis. J Neurol Sci 1991;102(1):100–4. Futamatsu H, Suzuki J, Kosuge H, Yokoseki O, Kamada M, Ito H, et al. Attenuation of experimental autoimmune myocarditis by blocking activated T cells through inducible costimulatory molecule pathway. Cardiovasc Res 2003;59(1):95–104. Kodama M, Matsumoto Y, Fujiwara M, Masani F, Izumi T, Shibata A. A novel experimental model of giant cell myocarditis induced in rats by immunization with cardiac myosin fraction. Clin Immunol Immunopathol 1990;57(2):250–62. Paleev NR, Paleev FN, Suchkov SV, Kotova AN, Pronina OA. Cytokines as a diagnostic and therapeutic tool in patients with myocarditis. Vestn Ross Akad Med Nauk 2005;5:8–13. Ing DJ, Zang J, Dzau VJ, Webster KA, Bishopric NH. Modulation of cytokine-induced cardiac myocyte apoptosis by nitric oxide, Bak, and Bcl-x. Circ Res 1999;84(1):21–33. Rose NR. Critical cytokine pathways to cardiac inflammation. J Interferon Cytokine Res 2011;31(10):705–10. Matsumori A, Yamada T, Suzuki H, Matoba Y, Sasayama S. Increased circulating cytokines in patients with myocarditis and cardiomyopathy. Br Heart J 1994;72(6):561–6. Matsui Y, Inobe M, Okamoto H, Chiba S, Shimizu T, Kitabatake A, et al. Blockade of T cell costimulatory signals using adenovirus vectors prevents both the induction and the progression of experimental autoimmune myocarditis. J Mol Cell Cardiol 2002;34(3):279–95. Zhang H, Song Y, Zhang Z. Glycyrrhizin administration ameliorates coxsackievirus B3-induced myocarditis in mice. Am J Med Sci 2012;344(3):206–10. Mito S, Watanabe K, Harima M, Thandavarayan RA, Veeraveedu PT, Sukumaran V, et al. Curcumin ameliorates cardiac inflammation in rats with autoimmune myocarditis. Biol Pharm Bull 2011;34(7):974–9. Stein B, Frank P, Schmitz W, Scholz H, Thoenes M. Endotoxin and cytokines induce direct cardiodepressive effects in mammalian cardiomyocytes via induction of nitric oxide synthase. J Mol Cell Cardiol 1996;28(8):1631–9. Yokoseki O, Suzuki J, Kitabayashi H, Watanabe N, Wada Y, Aoki M, et al. cis Element decoy against nuclear factor-kappaB attenuates development of experimental autoimmune myocarditis in rats. Circ Res 2001;89(10):899–906. Zhao L, Tang Y, You Z, Wang Q, Liang S, Han X, et al. Interleukin-17 contributes to the pathogenesis of autoimmune hepatitis through inducing hepatic interleukin-6 expression. PLoS One 2011;6(4):e18909. Makol A, Watt KD, Chowdhary VR. Autoimmune hepatitis: a review of current diagnosis and treatment. Hepat Res Treat 2011;2011:390916. Devergne O, Peuchmaur M, Humbert M, Navratil E, Leger-Ravet MB, Crevon MC, et al. In vivo expression of IL-1 beta and IL-6 genes during viral infections in human. Eur Cytokine Netw 1991;2(3):183–94. Shindo M, Mullin GE, Braun-Elwert L, Bergasa NV, Jones EA, James SP. Cytokine mRNA expression in the liver of patients with primary biliary cirrhosis (PBC) and chronic hepatitis B (CHB). Clin Exp Immunol 1996;105(2):254–9. Tian Z, Shen X, Feng H, Gao B. IL-1 beta attenuates IFN-alpha beta-induced antiviral activity and STAT1 activation in the liver: involvement of proteasome-dependent pathway. J Immunol 2000;165(7):3959–65. Satoh S, Nussler AK, Liu ZZ, Thomson AW. Proinflammatory cytokines and endotoxin stimulate ICAM-1 gene expression and secretion by normal human hepatocytes. Immunology 1994;82(4):571–6. Adams DH, Mainolfi E, Burra P, Neuberger JM, Ayres R, Elias E, et al. Detection of circulating intercellular adhesion molecule-1 in chronic liver diseases. Hepatology 1992;16(3):810–4. Thomson AW, Satoh S, Nussler AK, Tamura K, Woo J, Gavaler J, et al. Circulating intercellular adhesion molecule-1 (ICAM-1) in autoimmune liver disease and evidence for the production of ICAM-1 by cytokine-stimulated human hepatocytes. Clin Exp Immunol 1994;95(1):83–90. Liberal R, Grant CR, Mieli-Vergani G, Vergani D. Autoimmune hepatitis: a comprehensive review. J Autoimmun 2013;41:126–39. Geneva-Popova M, Murdjeva M. Study on proinflammatory cytokines (IL-1 beta, IL-6, TNF-alpha) and IL-2 in patients with acute hepatitis B. Folia Med (Plovdiv) 1999;41(1):78–81. Carrero JJ, Park SH, Axelsson J, Lindholm B, Stenvinkel P. Cytokines, atherogenesis, and hypercatabolism in chronic kidney disease: a dreadful triad. Semin Dial 2009;22(4):381–6. Kilis-Pstrusinska K. Genetic factors in the development and progression of chronic kidney disease. Postepy Hig Med Dosw (Online) 2010;64:50–7. Zyga S, Christopoulou G, Malliarou M. Malnutrition–inflammation–atherosclerosis syndrome in patients with end-stage renal disease. J Ren Care 2011;37(1):12–5. Xia Y, Feng L, Yoshimura T, Wilson CB. LPS-induced MCP-1, IL-1 beta, and TNFalpha mRNA expression in isolated erythrocyte-perfused rat kidney. Am J Physiol 1993;264(5 Pt 2):F774–80. Cartwright NH, Keen LJ, Demaine AG, Hurlock NJ, McGonigle RJ, Rowe PA, et al. A study of cytokine gene polymorphisms and protein secretion in renal transplantation. Transpl Immunol 2001;8(4):237–44. Rodrigo E, Sanchez-Velasco P, Ruiz JC, Fernandez-Fresnedo G, Lopez-Hoyos M, Pinera C, et al. Cytokine polymorphisms and risk of infection after kidney transplantation. Transplant Proc 2007;39(7):2219–21. Wan QQ, Ye QF, Ma Y, Zhou JD. Genetic association of interleukin-1beta (-511C/T) and its receptor antagonist (86-bpVNTR) gene polymorphism with susceptibility to bacteremia in kidney transplant recipients. Transplant Proc 2012;44(10):3026–8.

R. Zhao et al. / International Immunopharmacology 17 (2013) 658–669 [177] Berry M, Clatworthy MR. Immunotherapy for acute kidney injury. Immunotherapy 2012;4(3):323–34. [178] Shao HJ, Chen L, Shen MS, Yu GF. Enhancement of immune responses to the hepatitis B virus core protein through DNA vaccines with a DNA fragment encoding human IL-1beta 163-171 peptide. Acta Virol 2003;47(4):217–21. [179] Dinarello CA, Simon A, van der Meer JW. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov 2012;11(8):633–52. [180] Kong J, Grando SA, Li YC. Regulation of IL-1 family cytokines IL-1alpha, IL-1 receptor antagonist, and IL-18 by 1,25-dihydroxyvitamin D3 in primary keratinocytes. J Immunol 2006;176(6):3780–7. [181] Jayasuriya CT, Goldring MB, Terek R, Chen Q. Matrilin-3 Induction of IL-1 receptor antagonist is required for up-regulating collagen II and aggrecan and downregulating ADAMTS-5 gene expression. Arthritis Res Ther 2012;14(5):R197.

669

[182] Prefontaine D, Lajoie-Kadoch S, Foley S, Audusseau S, Olivenstein R, Halayko AJ, et al. Increased expression of IL-33 in severe asthma: evidence of expression by airway smooth muscle cells. J Immunol 2009;183(8):5094–103. [183] Nold MF, Nold-Petry CA, Zepp JA, Palmer BE, Bufler P, Dinarello CA. IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol 2010;11(11):1014–22. [184] Essner R, Rhoades K, McBride WH, Morton DL, Economou JS. IL-4 down-regulates IL-1 and TNF gene expression in human monocytes. J Immunol 1989;142(11):3857–61. [185] Murray PJ, Smale ST. Restraint of inflammatory signaling by interdependent strata of negative regulatory pathways. Nat Immunol 2012;13(10):916–24. [186] Yang Y, Hahm E, Kim Y, Kang J, Lee W, Han I, et al. Regulation of IL-18 expression by CRH in mouse microglial cells. Immunol Lett 2005;98(2):291–6. [187] Carrier Y, Ma HL, Ramon HE, Napierata L, Small C, O'Toole M, et al. Inter-regulation of Th17 cytokines and the IL-36 cytokines in vitro and in vivo: implications in psoriasis pathogenesis. J Invest Dermatol 2011;131(12):2428–37.