IL‑33 and kidney disease (Review)

  • Authors:
    • Feifei Yang
    • Ping Zhu
    • Lihua Duan
    • Lin Yang
    • Jiajun Wang
  • View Affiliations

  • Published online on: November 6, 2015     https://doi.org/10.3892/mmr.2015.4516
  • Pages: 3-8
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Interleukin (IL)‑33, is a novel member of the IL‑1 superfamily, and act as a dual‑function molecule as a nuclear factor and cytokine. The expression of IL‑33 can be detected in several tissues and cells in humans and in mice. In addition to the conventional secretion approach for cytokines, full‑length IL‑33 can also be released into the extracellular space following cell damage or mechanical injury. IL‑33 mediates its biological effects by interacting with the receptors, suppression of tumorigenicity 2 (ST2) and IL‑1 receptor accessory protein, activating intracellular molecules in the nuclear factor‑κB and mitogen‑activated protein kinase signaling pathways, which drive the production of type 2 cytokines, including IL‑4, IL‑5 and IL‑13, from polarized T helper 2 cells. Increasing evidence indicates that IL‑33 is important in chronic kidney disease, and may be involved in the progression of renal fibrosis associated with systemic lupus erythematosus and renal graft damage. In addition, IL‑33 contributes to acute kidney injury. In the present review, the biology of IL‑33, and the association of IL‑33 with kidney diseases are discussed.

1. Introduction

Interleukin (IL-33) is a cytokine, which belongs to the IL-1 superfamily, and induces T helper (Th) cells to produce type 2 cytokines (1). In 1999, Onda et al identified the Dvs27 gene in canine vasospastic cerebral arteries following subarachnoid hemorrhage (2). Nuclear factor from high endothelial venule (NF-HEV) was cloned in 2003 (3), and 2 years later, Schmitz et al determined the Dvs27 and NF-HEV definitions as the same type of molecule, which was termed IL-33 (4). IL-33 mediates its biological effects by interacting with the receptors suppression of tumorigenicity 2 (ST2) and IL-1 receptor accessory protein (IL-1RAcP), activating intracellular molecules in the NF-κB and mitogen-activated protein kinase signaling pathways, which drive the production of type 2 cytokines, including IL-4, IL-5 and IL-13 from polarized Th2 cells (5). The induction of type 2 cytokines by IL-33 in vivo is considered to induce severe pathological changes in mucosal organs (6).

The focus of the present review is on the properties of this cytokine in kidney diseases, as well as in renal graft damage associated with renal transplantation. Understanding the involvement of IL-33 in the pathogenesis of kidney diseases may assist in identifying novel therapeutic strategies to mitigate or prevent kidney diseases.

2. Structure of IL-33

IL-33, also termed IL-F11, is a novel member of the IL-1 superfamily. The human IL-33 gene is located on chromosome 9p24.1, comprises 270 amino acids and the relative molecular mass of full-length proteins is ~30 kDa. The mouse IL-33 gene can be found on the chromosome 19qC1 region and encodes 266 amino acid polypeptides, corresponding to full-length proteins with a calculated mass of 29.9 kDa (4). IL-33 has, in its amino-terminal portion, a helix-corner-helix structural pattern, which involves a chromatin binding motif and nuclear localization signal. In its carboxy-terminal portion is an IL-1-like β-trefoil domain, which binds with the orphan receptor, ST2 (7). Roussel et al showed that IL-33 combines to chromatin by an acidic pocket area of histone H2A-H2B (8).

3. Distribution of IL-33

In humans, the constitutive and widespread expression of IL-33 can be detected in several normal tissues. For example, IL-33 is constitutively expressed in human secondary lymphoid tissues, including the lymph nodes and appendix (9), and widespread expression is observed along the vascular tree, including large and small blood vessels from normal tissues, including the liver, skeletal muscle, kidney and prostate, despite the microcirculation of the brain and kidney glomeruli (9). In certain tissues exposed to the external environment, high levels of IL-33 have also been found, including the skin, mucosal surfaces and gastric glands in the stomach, as well as in tonsillar crypts and salivary glands (9). Furthermore, the accumulation of IL-33 has been reported in adenocarcinoma of the kidney, stomach, liver, pancreas, lung, breast and colon (9). In addition, IL-33 is substantially elevated in lymphoid tissues, the synovium in chronically inflamed rheumatoid arthritis and the intestines in Crohn's disease (10). Together, these results indicate that IL-33 is broadly expressed in normal, tumor and chronically inflamed human tissues. High mRNA expression levels of IL-33 have been found in stomach, lung, spinal cord, brain and skin in the mouse, whereas low mRNA expression levels of IL-33 have been detected in mouse lymph tissue, the spleen, pancreas, kidney and heart (3).

Compared with its expression in tissue, IL-33 mRNA is more restricted at the cellular level. Activated dendritic cells and macrophages are the only hematopoietic cells to exhibit low mRNA expression levels of human IL-33. By contrast, IL-33 mRNA has been found in resting dendritic cells and activated macrophages in mice (11). Human smooth muscle cells (SMCs) of various tissues, as well as epithelial cells forming the bronchus or small airways exhibit constitutive expression of IL-33 mRNA (11). In addition, high expression levels of IL-33 have been confirmed in activated dermal fibroblasts, activated and resting bronchial SMCs, resting pulmonary artery SMCs, resting coronary artery SMCs and bronchial epithelial cells (4). The distributions of IL-33 in humans in mice are presented in Table I.

Table I

Distribution of interleukin-33 in humans and mice.

Table I

Distribution of interleukin-33 in humans and mice.

SiteHumanMouseReference
Tissue(3,9,10)
 Appendix+NA
 Brain+
 Colon+
 Kidney++
 Liver++
 Lung++
 Lymph nodes++
 Pancreas++
 Prostate+NA
 Salivary glands+NA
 Skeletal muscle+NA
 Skin++
 Spinal cordNA+
 Stomach++
 Tonsillar crypts+NA
Cells(4,811)
 Activated dermal fibroblasts+NA
 Activated macrophages++
 Epithelial cells++
 Resting dendritic cells+
 Smooth muscle cells+NA

[i] +, positive; −, negative; NA, not available.

4. Cleavage of IL-33

It has been suggested that IL-33 is produced as a pro-form IL-33 (pro-IL-33), and is digested into a mature form with a lower molecular weight when it is secreted from the cells (12). Mature IL-33 was initially considered to be the active form, however, subsequent reports have shown that the active pro-form of IL-33 is digested into an inactive mature form (4,13). The initial investigation of IL-33 suggested that it is activated via caspase-1-dependent proteolysis, similar to the proinflammatory cytokines, IL-1β and IL-18 (14). By contrast, Cayrol and Girard reported that full-length IL-33 (1-270) is active, and that processing by caspase-1 results in IL-33 inactivation, rather than activation (13). Another previous report independently arrived at the conclusion that the executioner caspase-3 and caspase-7 inactivate IL-33 by cleaving the carboxy-terminal IL-1-like structure to prevent an inappropriate immune response during apoptosis, but not in necrosis (15).

5. Secretion of IL-33

In addition to the conventional secretion approaches for cytokines, including autocrine, paracrine, intracrine, juxtacrine and retrocrine pathways, full-length IL-33, as with high mobility group protein-1, can also be released into the extracellular space following cell damage or mechanical injury (13). The release of IL-33 by necrotic cells is another recognized mechanism for a cytokine to exert its function, termed a 'necrocrine' pathway (16). The necrocrine pathway can be deleted by endogenous apoptotic caspases in cells undergoing apoptosis (15,17). Therefore, IL-33 functions as an extracellular 'danger signal' in a necrocrine manner, to alert the immune system during infectious and autoimmune diseases.

6. Receptors of IL-33

IL-33R is a heterodimer comprised of IL-1RL1, also ST2, and IL-1RAcP (18). ST2, which exhibits marked homology to the ligand-binding subunits of the IL-1 and IL-18 receptor complexes, was identified in 1989, prior the identification of IL-33, and has been termed an 'orphan receptor' (19). The human ST2 gene is located on chromosome 2, and its germline sequence is conserved (20). IL-33 mediates signal transduction through ST2, which is expressed on mast cells and Th2 cella, but not Th1 cells (21,22). Enhancing the expression of ST2 is associated with an increased risk of developing atopic dermatitis (23).

ST2 has two major forms: soluble (s)ST2 and membrane-bound ST2 (ST2 L), which are produced from the IL-1RL1 gene as a result of alternative splicing under the control of two distinct promoters (2426). sST2 is a soluble ST2, which has no transmembrane sequence, therefore, it can be excreted outside cells. Increased levels of soluble ST2 have been associated with several human diseases, including acute myocardial infarction, asthma with acute exacerbation, eosinophilic pneumonia, sepsis and trauma, and exacerbated idiopathic pulmonary fibrosis (2732). ST2L is the trans-membrane ST2, possessing a transmembrane sequence, and is considered to be a functional component of IL-33R, whereas sST2 is regarded as a decoy receptor for IL-33 (33). T cell-associated ST2L augments Th2 immune responses, however, macrophage-associated ST2L has been reported to exhibit anti-inflammatory activity (34). There are two splice variants of ST2: ST2V and ST2LV, produced via loss of the third immunoglobulin motif and alternative splicing in the C-terminal portion of ST2 (35).

7. IL-33/ST2 signaling pathway

IL-33 can transfer extracellular information through binding of a receptor complex comprised of ST2 and IL-1RAcP as a cytokine-alarmin (9). ST2L and IL-1RAcP are necessary for IL-33 action (36,37). Extracellular IL-33 signals results in recruitment of myeloid differentiation primary response gene 88, IL-1 receptor-associated kinase 1 and tumor necrosis factor (TNF) receptor-associated factor 6, leading to activation of the transcription factor NF-kB, c-Jun N-terminal kinase1/2 and extracellular regulated protein kinase 1/2, and finally causing inflammatory responses (4,3638). IL-33-mediated signalling can be inhibited by single Ig IL-1-related molecule, also known as Toll IL-1R8, through interactions with the IL-33 receptor complex (39) (Fig. 1).

8. Function of IL-33

Similar to the chromatin-associated cytokine, high-mobility group box 1 (HMGB1), IL-33 acts as a dual-function molecule, as a nuclear factor and cytokine. As a nuclear factor, the transcriptional repressor function of IL-33 may be involved in the nucleus. Küchler et al showed that nuclear IL-33 is rapidly downregulated during wound healing and is lost in tumor endothelium. In addition, activation of endothelial cell cultures with either TNF-α or vascular endothelial growth factor and subcutaneous injection of these cytokines also leads to a downregulation in vascular IL-33 (40). The above evidence supports the hypothesis that the transcriptional repressor function of IL-33 may be involved in the control of endothelial cell activation.

As a cytokine, IL-33 promotes the polarization of T cells towards a Th2 cell phenotype and is involved in Th2-type responses through stimulating the production of IL-5, IL-6, IL-13 and granulocyte-macrophage colony-stimulating factor in vivo (41). In addition, IL-33 has been referred to as an 'endogenous danger signal' or 'alarmin', similar to HMGB1, in order to alert the immune system of tissue damage and infection, and to promote the initiation of a healing responses (42).

9. IL-33 and kidney disease

IL-33 and chronic kidney disease (CKD)

A study by Bao et al (43) on CKD aimed to examine the association between serum levels of IL-33 and sST2, and disease severity. This involved comparing the serum concentrations of IL-33 and sST2 between patients with CKD and healthy individuals. The results showed no difference in the serum concentration of IL-33 between the patients with CKD and healthy individuals, whereas a higher serum level of sST2 was found in the patients with CKD. Therefore, the results revealed a significant correlation between the serum level of sST2 and disease severity. In addition, higher levels of sST2 correlated with elevated parathyroid hormone, serum phosphorus and serum calcium (43). However, the expression of IL-33 was observed to be increased in aortic endothelial cells from a mouse model of CKD (44). Additionally, higher concentrations of sST2 appeared to be associated with impaired kidney function in a study involving participants with cardiovascular disease (45). Together, these findings indicate that the levels of IL-33 and sST2 are relevant to the progressive deterioration of kidney function.

IL-33 and systemic lupus erythematosus (SLE) nephropathy (LN)

Renal fibrosis is the common pathway of chronic kidney disease eventually lead to kidney failure, which is one of the most serious complications of SLE (46). Fibrotic disease is characterized by the excess accumulation of extracellular matrix components, including colla gen, and re quires eosinophils and RAG-dependent lymphocytes (47). Of note, IL-33 mediates the regulation of several extracellular matrix-associated genes, including collagen VI, collagen III and tissue inhibitor of metalloproteases-1. In addition, the administration of IL-33 resultes in IL-33R-dependent accumulation of eosinophils, RAG-dependent lymphocytes and CD3+ lymphocytes (48). The levels of IL-33 in patients with SLE have been found to be greater than those in a healthy control group, and were correlated with elevated erythrocyte sedimentation rate and C-reactive protein, suggesting that the abnormal increase in serum IL-33 is closely associated with the development of SLE and may be involved in the acute phase reaction of SLE (49). Shui-Lian et al indicated that the IL-33/ST2 axis has a detrimental effect in the pathogenesis of renal fibrosis associated with LN (46). Therefore, IL-33 may involved in renal fibrosis associated with SLE. Further mechanistic investigations examining the precise physiological and pathophysiological roles of IL-33 in SLE are required.

IL-33 and diabetic nephropathy

The principle of the pathogeny of type 2 diabetes and its complications, including diabetic nephropathy, remain to be fully elucidated, however predicting the potential complications of diabetic patients can assist in early treatment. Whether IL-33 can be used for predicting the early stage of kidney injury in diabetic patients remains to be elucidated. In a study by Caner et al identified three groups: Healthy group; diabetes mellitus (DM) group without any known kidney disease; and DM+microalbuminuria (MA) group, assumed to have nephropathy. Following assessment of the concentrations of IL-33 in the three groups, it was found that the level of IL-33 in the DM group was greater than that in the healthy group; and the level of IL-33 in the DM+MA group was greater than that in the healthy group; although no difference was observed between the DM and DM+MA group. Therefore, IL-33 cannot be used in the early recognition of diabetic nephropathy (50). A study by Miller et al showed that the levels of sST2 in individuals largely without vascular disease, are associated principally with markers associated with diabetes, and support a role for sST2 in diabetes (51). These findings support the hypothesis that the increase in IL-33 levels in diabetic nephropathy is not associated with kidney injury, but that the increase may be a result of diabetes. Further investigations are required to clarify the value of IL-33 in DM and the early stage of kidney injury.

IL-33 and renal transplantation

Ischemia-reperfusion injury (IRI) contributes to the develop ment of renal graft damage associated with renal transplantation (52). Inflammatory and immune responses are involved in kidney IRI (52). IL-33 has been identified as an alarmin, capable of mediating danger signals during tissue damage (53). Thierry et al addressed the role of IL-33 in IRI following human kidney transplantation (54). This involved analysis of the levels of IL-33 in a cohort of 26 deceased renal transplant recipients, and revealed that the level of IL-33 was significantly increased as soon as 30 min post-reperfusion, which supported the potential role of IL-33 as an immune mediator following transplantation during kidney IRI in humans (54). Consistent with this, invariant natural killer T cells, which have been suggested to be crucial in IRI and targeted by IL-33, exhibited a state of early activation in kidney transplant recipients (54). In addition, a significant correlation was found between serum and urinary levels of IL-33 levels and cold ischemia duration, between 30 min and 3 days post-tranplantation (55). In conclusion, these results emphasize the possible role of IL-33 as an innate-immune mediator during IRI in humans.

IL-33 and acute kidney injury (AKI)

AKI contributes to significant morbidity and mortality rates in intensive care units (56). Alterations in renal hemodynamics, inflammation, endothelial dysfunction, tubular obstruction and glomerular thrombosis are involved in the pathogenesis of AKI (57). To determine whether IL-33 promotes AKI, a study by Akcay et al examined the protein expression of IL-33 in the kidney using an AKI mouse model. Following neutralizing IL-33 activity with sST2, these mice had fewer CD4 T cells infiltrating the kidney, lower levels of serum creatinine, and reduced acute tubular necrosis and apoptosis, compared with cisplatin-induced AKI in the untreated mice. By contrast, the administration of recombinant IL-33 exacerbated cisplatin-induced AKI (58). Of note, IL-33 mediates cisplatin-induced AKI by acting as an proinflammatory cytokine, whereas IL-10 protects against cisplatin-induced AKI by acting as an anti-inflammatory cytokine (59). In addition, high expression levels of IL-33 have been observed in lipopolysaccharide-induced acute glomerular injury (57). The inhibition of IL-33 may provide a novel strategy in the treatment of AKI (Table II).

Table II

Role of IL-33 in kidney diseases.

Table II

Role of IL-33 in kidney diseases.

Kidney diseaseRole of IL-33Reference
CKDSerum levels of IL-33 not correlated with disease severity in CKD.(44,45)
Serum levels of sST2 are significantly correlated with disease severity in CKD.
Systemic lupus erythematosus nephropathyMediates regulation of several extracellular matrix-associated genes, resulting in IL-33R-dependent accumulation of eosinophils, RAG-dependent lymphocytes and CD3+ lymphocytes.(4749)
Diabetic nephropathyExpression of IL-33 is not associated with kidney injury, but the increase may be a result of diabetes.(50,51)
Renal transplantationImmune mediator following transplantation during kidney IRI in humans; correlated with cold ischemia duration; activates invariant natural killer T cells in kidney transplant recipients.(5355)
Acute kidney injuryStimulates CD4+ T cell infiltration in the kidney, induces higher levels of serum creatinine, acute tubular necrosis and apoptosis.(5759)

[i] IL-33, interleukin-33; CKD, chronic kidney disease; IRI, ischemia reperfusion injury.

10. Conclusion

In conclusion, increasing evidence indicates that the IL-33/ST2 axis has a significant effect in the pathogenesis of kidney disease. Although the mechanism underlying the effects of IL-33 in kidney disease remains to be fully elucidated, accumulating evidence links IL-33 to the nephropathies, indicating that the antagonism of IL-33 may be a novel strategy for the treatment of kidney disease. Further detailed investigations of the association between IL-33 and kidney disease are required in the future.

Acknowledgments

This study was supported by the Natural Science Foundation of Hubei province (grant. no. 2012FFB03708) and the Natural Science Foundation of Yichang City (grant. no. A13301-020).

References

1 

Louten J, Rankin AL, Li Y, Murphy EE, Beaumont M, Moon C, Bourne P, McClanahan TK, Pflanz S and de Waal Malefyt R: Endogenous IL-33 enhances Th2 cytokine production and T-cell responses during allergic airway inflammation. Int Immunol. 23:307–315. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Onda H, Kasuya H, Takakura K, et al: Identification of genes differentially expressed in canine vasospastic cerebral arteries after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 19:1279–1288. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Baekkevold ES, Roussigné M, Yamanaka T, Johansen FE, Jahnsen FL, Amalric F, Brandtzaeg P, Erard M, Haraldsen G and Girard JP: Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol. 163:69–79. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, Zurawski G, Moshrefi M, Qin J, Li X, Gorman DM, et al: IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 23:479–490. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Marian AJ: Pathogenesis of diverse clinical and pathological phenotypes in hypertrophic cardiomyopathy. Lancet. 355:58–60. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Bartemes KR, Iijima K, Kobayashi T, Kephart GM, McKenzie AN and Kita H: IL-33-responsive lineage- CD25+ CD44(hi) lymphoid cells mediate innate type 2 immunity and allergic inflammation in the lungs. J Immunol. 188:1503–1513. 2012. View Article : Google Scholar :

7 

Miller AM and Liew FY: The IL-33/ST2 pathway–A new therapeutic target in cardiovascular disease. Pharmacol Ther. 131:179–186. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Roussel L, Erard M, Cayrol C and Girard JP: Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep. 9:1006–1012. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Moussion C, Ortega N and Girard JP: The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: A novel 'alarmin'? PloS one. 3:e33312008. View Article : Google Scholar : PubMed/NCBI

10 

Carriere V, Roussel L, Ortega N, Lacorre DA, Americh L, Aguilar L, Bouche G and Girard JP: IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci USA. 104:282–287. 2007. View Article : Google Scholar :

11 

Nakajima A: Application of cellular gene therapy for rheumatoid arthritis. Mod Rheumatol. 16:269–275. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Tsuda H, Komine M, Karakawa M, Etoh T, Tominaga S and Ohtsuki M: Novel splice variants of IL-33: Differential expression in normal and transformed cells. J Invest Dermatol. 132:2661–2664. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Cayrol C and Girard JP: The IL-1-like cytokine IL-33 is inactivated after maturation by caspase-1. Proc Natl Acad Sci USA. 106:9021–9026. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Creagh EM, Conroy H and Martin SJ: Caspase-activation pathways in apoptosis and immunity. Immunol Rev. 193:10–21. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Luthi AU, Cullen SP, McNeela EA, Duriez PJ, Afonina IS, Sheridan C, Brumatti G, Taylor RC, Kersse K, Vandenabeele P, et al: Suppression of interleukin-33 bioactivity through proteolysis by apoptotic caspases. Immunity. 31:84–98. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Zhao W and Hu Z: The enigmatic processing and secretion of interleukin-33. Cell Mol Immunol. 7:260–262. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Lamkanfi M and Dixit VM: IL-33 raises alarm. Immunity. 31:5–7. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Nakae S, Morita H, Ohno T, Arae K, Matsumoto K and Saito H: Role of interleukin-33 in innate-type immune cells in allergy. Allergol Int. 62:13–20. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Tominaga S: A putative protein of a growth specific cDNA from BALB/c-3T3 cells is highly similar to the extracellular portion of mouse interleukin 1 receptor. FEBS Lett. 258:301–304. 1989. View Article : Google Scholar : PubMed/NCBI

20 

Luzina IG, Pickering EM, Kopach P, Kang PH, Lockatell V, Todd NW, Papadimitriou JC, McKenzie AN and Atamas SP: Full-length IL-33 promotes inflammation but not Th2 response in vivo in an ST2-independent fashion. J Immunol. 189:403–410. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Löhning M, Stroehmann A, Coyle AJ, Grogan JL, Lin S, Gutierrez-Ramos JC, Levinson D, Radbruch A and Kamradt T: T1/ST2 is preferentially expressed on murine Th2 cells, independent of interleukin 4, interleukin 5 and interleukin 10 and important for Th2 effector function. Proc Natl Acad Sci USA. 95:6930–6935. 1998. View Article : Google Scholar

22 

Yanagisawa K, Naito Y, Kuroiwa K, Arai T, Furukawa Y, Tomizuka H, Miura Y, Kasahara T, Tetsuka T and Tominaga S: The expression of ST2 gene in helper T cells and the binding of ST2 protein to myeloma-derived RPMI8226 cells. J Biochm. 121:95–103. 1997. View Article : Google Scholar

23 

Shimizu M, Matsuda A, Yanagisawa K, Hirota T, Akahoshi M, Inomata N, Ebe K, Tanaka K, Sugiura H, Nakashima K, et al: Functional SNPs in the distal promoter of the ST2 gene are associated with atopic dermatitis. Hum Mol Genet. 14:2919–2927. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Lin J, Zhang L, Zhao G, Su Z, Deng R, Pflugfelder SC and Li DQ: A novel interleukin 33/ST2 signaling regulates inflammatory response in human corneal epithelium. PloS One. 8:e609632013. View Article : Google Scholar : PubMed/NCBI

25 

Oboki K, Ohno T, Kajiwara N, Saito H and Nakae S: IL-33 and IL-33 receptors in host defense and diseases. Allergol Int. 59:143–160. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Smith DE: IL-33: A tissue derived cytokine pathway involved in allergic inflammation and asthma. Clin Exp Allergy. 40:200–208. 2010. View Article : Google Scholar

27 

Oshikawa K, Kuroiwa K, Tago K, Iwahana H, Yanagisawa K, Ohno S, Tominaga SI and Sugiyama Y: Elevated soluble ST2 protein levels in sera of patients with asthma with an acute exacerbation. Am J Respir Crit Care Med. 164:277–281. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Oshikawa K, Kuroiwa K, Tokunaga T, Kato T, Hagihara SI, Tominaga SI and Sugiyama Y: Acute eosinophilic pneumonia with increased soluble ST2 in serum and bronchoalveolar lavage fluid. Respir Med. 95:532–533. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Weinberg EO, Shimpo M, De Keulenaer GW, MacGillivray C, Tominaga S, Solomon SD, Rouleau JL and Lee RT: Expression and regulation of ST2, an interleukin-1 receptor family member, in cardiomyocytes and myocardial infarction. Circulation. 106:2961–2966. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Tajima S, Oshikawa K, Tominaga S and Sugiyama Y: The increase in serum soluble ST2 protein upon acute exacerbation of idiopathic pulmonary fibrosis. Chest. 124:1206–1214. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Brunner M, Krenn C, Roth G, Moser B, Dworschak M, Jensen-Jarolim E, Spittler A, Sautner T, Bonaros N, Wolner E, et al: Increased levels of soluble ST2 protein and IgG1 production in patients with sepsis and trauma. Intensive Care Med. 30:1468–1473. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Shimpo M, Morrow DA, Weinberg EO, Sabatine MS, Murphy SA, Antman EM and Lee RT: Serum levels of the interleukin-1 receptor family member ST2 predict mortality and clinical outcome in acute myocardial infarction. Circulation. 109:2186–2190. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Ohno T, Morita H, Arae K, Matsumoto K and Nakae S: Interleukin-33 in allergy. Allergy. 67:1203–1214. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Brint EK, Xu D, Liu H, Dunne A, McKenzie AN, O'Neill LA and Liew FY: ST2 is an inhibitor of interleukin 1 receptor and toll-like receptor 4 signaling and maintains endotoxin tolerance. Nat Immunol. 5:373–379. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Kakkar R and Lee RT: The IL-33/ST2 pathway: Therapeutic target and novel biomarker. Nat Rev Drug Discov. 7:827–840. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Kurowska-Stolarska M, Kewin P, Murphy G, Russo RC, Stolarski B, Garcia CC, Komai-Koma M, Pitman N, Li Y, Niedbala W, et al: IL-33 induces antigen-specific IL-5+ T cells and promotes allergic-induced airway inflammation independent of IL-4. J Immunol. 181:4780–4790. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Chackerian AA, Oldham ER, Murphy EE, Schmitz J, Pflanz S and Kastelein RA: IL-1 receptor accessory protein and ST2 comprise the IL-33 receptor complex. J Immunol. 179:2551–2555. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Kurowska-Stolarska M, Hueber A, Stolarski B and McInnes IB: Interleukin-33: A novel mediator with a role in distinct disease pathologies. J Internal Med. 269:29–35. 2011. View Article : Google Scholar

39 

Bulek K, Swaidani S, Qin J, Lu Y, Gulen MF, Herjan T, Min B, Kastelein RA, Aronica M, Kosz-Vnenchak M and Li X: The essential role of single Ig IL-1 receptor-related molecule/Toll IL-1R8 in regulation of Th2 immune response. J Immunol. 182:2601–2609. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Küchler AM, Pollheimer J, Balogh J, Sponheim J, Manley L, Sorensen DR, De Angelis PM, Scott H and Haraldsen G: Nuclear interleukin-33 is generally expressed in resting endothelium but rapidly lost upon angiogenic or proinflammatory activation. Am J Pathol. 173:1229–1242. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Smithgall MD, Comeau MR, Yoon BR, Kaufman D, Armitage R and Smith DE: IL-33 amplifies both Th1- and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells. Int Immunol. 20:1019–1030. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Bianchi ME: DAMPs, PAMPs and alarmins: All we need to know about danger. J Leukoc Biol. 81:1–5. 2007. View Article : Google Scholar

43 

Bao YS, Na SP, Zhang P, Jia XB, Liu RC, Yu CY, Mu SH and Xie RJ: Characterization of interleukin-33 and soluble ST2 in serum and their association with disease severity in patients with chronic kidney disease. J Clin Immunol. 32:587–594. 2012. View Article : Google Scholar

44 

Wiese CB, Toth CL, Tabet F, Taylor RC, Landstreet SR, Rye KA, Hofmeister LH, Harrison DG, Kon V and Vickers KC: HDL-microRNA-92a and interleukin-33 axis underlies endothelial dysfunction associated with atherosclerosis and chronic kidney disease. Arterioscler Thromb Vasc Biol. 34:A3. 2014.

45 

Januzzi JL Jr, Peacock WF, Maisel AS, Chae CU, Jesse RL, Baggish AL, O'Donoghue M, Sakhuja R, Chen AA, van Kimmenade RR, et al: Measurement of the interleukin family member ST2 in patients with acute dyspnea: Results from the PRIDE (Pro-Brain natriuretic peptide investigation of dyspnea in the emergency department) study. J Am Coll Cardiol. 50:607–613. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Yu SL, Wong CK and Tam LS: The alarmin functions of high-mobility group box-1 and IL-33 in the pathogenesis of systemic lupus erythematosus. Expert Rev Clin Immunol. 9:739–749. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Wynn TA: Cellular and molecular mechanisms of fibrosis. J Pathol. 214:199–210. 2008. View Article : Google Scholar

48 

Rankin AL, Mumm JB, Murphy E, Turner S, Yu N, McClanahan TK, Bourne PA, Pierce RH, Kastelein R and Pflanz S: IL-33 induces IL-13-dependent cutaneous fibrosis. J Immunol. 184:1526–1535. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Yang Z, Liang Y, Xi W, Li C and Zhong R: Association of increased serum IL-33 levels with clinical and laboratory characteristics of systemic lupus erythematosus in Chinese population. Clin Exp Med. 11:75–80. 2011. View Article : Google Scholar

50 

Caner S, Usluoğullari CA, Balkan F, Büyükcam F, Kaya C, Saçıkara M, Koca C, Ersoy R and Çakır B: Is IL-33 useful to detect early stage of renal failure? Ren Fail. 36:78–80. 2014. View Article : Google Scholar

51 

Miller AM, Purves D, McConnachie A, Asquith DL, Batty GD, Burns H, Cavanagh J, Ford I, McLean JS, Packard CJ, et al: Soluble ST2 associates with diabetes but not established cardiovascular risk factors: A new inflammatory pathway of relevance to diabetes? PloS One. 7:e478302012. View Article : Google Scholar : PubMed/NCBI

52 

Basile DP, Anderson MD and Sutton TA: Pathophysiology of acute kidney injury. Compr Physiol. 2:1303–1353. 2012.PubMed/NCBI

53 

Galli SJ, Nakae S and Tsai M: Mast cells in the development of adaptive immune responses. Nat Immunol. 6:135–142. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Thierry A, Giraud S, Robin A, Barra A, Bridoux F, Ameteau V, Hauet T, Girard JP, Touchard G, Gombert JM and Herbelin A: The alarmin concept applied to human renal transplantation: Evidence for a differential implication of HMGB1 and IL-33. PloS One. 9:e887422014. View Article : Google Scholar : PubMed/NCBI

55 

Gombert JM, Thierry A, Robin A, Barra A, Thierry H, Touchard G and Herbelin A: Study of alarmin release during ischemia reperfusion injury after human renal transplantation (P2214). J Immunol. 190:69.452013.

56 

Singbartl K and Kellum JA: AKI in the ICU: Definition, epidemiology, risk stratification, and outcomes. Kidney Int. 81:819–825. 2012. View Article : Google Scholar

57 

Lee SJ, Borsting E, Declèves AE, Singh P and Cunard R: Podocytes express IL-6 and lipocalin 2/neutrophil gelatinase-associated lipocalin in lipopolysaccharide-induced acute glomerular injury. Nephron Exp Nephrol. 121:e86–96. 2012. View Article : Google Scholar

58 

Akcay A, Nguyen Q, He Z, Turkmen K, Won D, Hernando AA, Altmann C, Toker A, Pacic A, Ljubanovic DG, et al: IL-33 exacerbates acute kidney injury. J Am Soc Nephrol. 22:2057–2067. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Ozkok A and Edelstein CL: Pathophysiology of cisplatin-induced acute kidney injury. Biomed Res Int. 2014:9678262014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 13 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang F, Zhu P, Duan L, Yang L and Wang J: IL‑33 and kidney disease (Review). Mol Med Rep 13: 3-8, 2016
APA
Yang, F., Zhu, P., Duan, L., Yang, L., & Wang, J. (2016). IL‑33 and kidney disease (Review). Molecular Medicine Reports, 13, 3-8. https://doi.org/10.3892/mmr.2015.4516
MLA
Yang, F., Zhu, P., Duan, L., Yang, L., Wang, J."IL‑33 and kidney disease (Review)". Molecular Medicine Reports 13.1 (2016): 3-8.
Chicago
Yang, F., Zhu, P., Duan, L., Yang, L., Wang, J."IL‑33 and kidney disease (Review)". Molecular Medicine Reports 13, no. 1 (2016): 3-8. https://doi.org/10.3892/mmr.2015.4516